Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 978
Filtrar
1.
Int J Mol Sci ; 25(7)2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38612635

RESUMO

We previously found that feeding rats with broccoli or cauliflower leads to the formation of characteristic DNA adducts in the liver, intestine and various other tissues. We identified the critical substances in the plants as 1-methoxy-3-indolylmethyl (1-MIM) glucosinolate and its degradation product 1-MIM-OH. DNA adduct formation and the mutagenicity of 1-MIM-OH in cell models were drastically enhanced when human sulfotransferase (SULT) 1A1 was expressed. The aim of this study was to clarify the role of SULT1A1 in DNA adduct formation by 1-MIM-OH in mouse tissues in vivo. Furthermore, we compared the endogenous mouse Sult1a1 and transgenic human SULT1A1 in the activation of 1-MIM-OH using genetically modified mouse strains. We orally treated male wild-type (wt) and Sult1a1-knockout (ko) mice, as well as corresponding lines carrying the human SULT1A1-SULT1A2 gene cluster (tg and ko-tg), with 1-MIM-OH. N2-(1-MIM)-dG and N6-(1-MIM)-dA adducts in DNA were analysed using isotope-dilution UPLC-MS/MS. In the liver, caecum and colon adducts were abundant in mice expressing mouse and/or human SULT1A1, but were drastically reduced in ko mice (1.2-10.6% of wt). In the kidney and small intestine, adduct levels were high in mice carrying human SULT1A1-SULT1A2 genes, but low in wt and ko mice (1.8-6.3% of tg-ko). In bone marrow, adduct levels were very low, independently of the SULT1A1 status. In the stomach, they were high in all four lines. Thus, adduct formation was primarily controlled by SULT1A1 in five out of seven tissues studied, with a strong impact of differences in the tissue distribution of mouse and human SULT1A1. The behaviour of 1-MIM-OH in these models (levels and tissue distribution of DNA adducts; impact of SULTs) was similar to that of methyleugenol, classified as "probably carcinogenic to humans". Thus, there is a need to test 1-MIM-OH for carcinogenicity in animal models and to study its adduct formation in humans consuming brassicaceous foodstuff.


Assuntos
Adutos de DNA , Glucosinolatos , Camundongos , Humanos , Animais , Ratos , Camundongos Knockout , Cromatografia Líquida , Espectrometria de Massas em Tandem , Arilsulfotransferase/genética
2.
Int J Mol Sci ; 24(23)2023 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-38069221

RESUMO

Sulfotransferases (SULTs) are phase II metabolizing enzymes catalyzing the sulfoconjugation from the co-factor 3'-Phosphoadenosine 5'-Phosphosulfate (PAPS) to a wide variety of endogenous compounds, drugs and natural products. Although SULT1A1 and SULT1A3 share 93% identity, SULT1A1, the most abundant SULT isoform in humans, exhibits a broad substrate range with specificity for small phenolic compounds, while SULT1A3 displays a high affinity toward monoamine neurotransmitters like dopamine. To elucidate the factors determining the substrate specificity of the SULT1 isoenzymes, we studied the dynamic behavior and structural specificities of SULT1A1 and SULT1A3 by using molecular dynamics (MD) simulations and ensemble docking of common and specific substrates of the two isoforms. Our results demonstrated that while SULT1A1 exhibits a relatively rigid structure by showing lower conformational flexibility except for the lip (loop L1), the loop L2 and the cap (L3) of SULT1A3 are extremely flexible. We identified protein residues strongly involved in the recognition of different substrates for the two isoforms. Our analyses indicated that being more specific and highly flexible, the structure of SULT1A3 has particularities in the binding site, which are crucial for its substrate selectivity.


Assuntos
Isoenzimas , Sulfotransferases , Humanos , Sulfotransferases/metabolismo , Especificidade por Substrato , Sítios de Ligação , Isoenzimas/metabolismo , Arilsulfotransferase/metabolismo
3.
Biochem Biophys Res Commun ; 665: 133-140, 2023 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-37163933

RESUMO

Coelenterazine (CTZ) is known as a light-emitting source for the bioluminescence reaction in marine organisms. CTZ has two phenolic hydroxy groups at the C2-benzyl and C6-phenyl positions, and a keto-enol type hydroxy group at the C3-position in the core structure of imidazopyrazinone (= 3,7-dihydroimidazopyrazin-3-one). These hydroxy groups in CTZ could be sulfated by sulfotransferase(s), and the sulfates of Watasenia luciferin (CTZ disulfate at the C2- and C6-positions) and Renilla pre-luciferin (CTZ 3-enol sulfate) have been identified in marine organisms. To characterize the sulfation process of CTZ, human cytosolic aryl sulfotransferase SULT1A1 (SUTase) was used as a model enzyme. The sulfated products catalyzed by SUTase with 3'-phosphoadenosine 5'-phosphosulfate (PAPS) were analyzed by LC/ESI-TOF-MS. The product was the monosulfate of CTZ and identified as the C2-benzyl sulfate of CTZ (CTZ C2-benzyl monosulfate), but CTZ disulfate, CTZ 3-enol sulfate, and CTZ C6-phenyl monosulfate were not detected. The non-enzymatic oxidation products of dehydrocoelenterazine (dCTZ, dehydrogenated derivative of CTZ), coelenteramide (CTMD), and coelenteramine (CTM) from CTZ were also identified as their monosulfates.


Assuntos
Arilsulfotransferase , Imidazóis , Humanos , Imidazóis/química , Sulfotransferases , Luciferinas , Sulfatos
4.
Sci Rep ; 13(1): 7256, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-37142702

RESUMO

In the sulfotransferase (SULT) superfamily, members of the SULT1 family mainly catalyse the sulfonation reaction of phenolic compounds, which is involved in the phase II metabolic detoxification process and plays a key role in endocrine homeostasis. A coding variant rs1059491 in the SULT1A2 gene has been reported to be associated with childhood obesity. This study aimed to investigate the association of rs1059491 with the risk of obesity and cardiometabolic abnormalities in adults. This case‒control study included 226 normal weight, 168 overweight and 72 obese adults who underwent a health examination in Taizhou, China. Genotyping of rs1059491 was performed by Sanger sequencing in exon 7 of the SULT1A2 coding region. Chi-squared tests, one-way ANOVA, and logistic regression models were applied. The minor allele frequencies of rs1059491 in the overweight combined with obesity and control groups were 0.0292 and 0.0686, respectively. No differences in weight and body mass index were detected between the TT genotype and GT + GG genotype under the dominant model, but the levels of serum triglycerides were significantly lower in G-allele carriers than in non-G-allele carriers (1.02 (0.74-1.32) vs. 1.35 (0.83-2.13) mmol/L, P = 0.011). The GT + GG genotype of rs1059491 versus the TT genotype reduced the risk of overweight and obesity by 54% (OR 0.46, 95% CI 0.22-0.96, P = 0.037) after adjusting for sex and age. Similar results were observed for hypertriglyceridaemia (OR 0.25, 95% CI 0.08-0.74, P = 0.013) and dyslipidaemia (OR 0.37, 95% CI 0.17-0.83, P = 0.015). However, these associations disappeared after correction for multiple tests. This study revealed that the coding variant rs1059491 is nominally associated with a decreased risk of obesity and dyslipidaemia in southern Chinese adults. The findings will be validated in larger studies including more detailed information on genetic background, lifestyle and weight change with age.


Assuntos
Arilsulfotransferase , Dislipidemias , Obesidade , Sobrepeso , Adulto , Humanos , Alelos , Arilsulfotransferase/genética , Índice de Massa Corporal , Estudos de Casos e Controles , Dislipidemias/genética , População do Leste Asiático , Genótipo , Sobrepeso/genética , Polimorfismo de Nucleotídeo Único , Obesidade/genética
5.
Nat Cancer ; 4(3): 365-381, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36914816

RESUMO

Adult liver malignancies, including intrahepatic cholangiocarcinoma and hepatocellular carcinoma, are the second leading cause of cancer-related deaths worldwide. Most individuals are treated with either combination chemotherapy or immunotherapy, respectively, without specific biomarkers for selection. Here using high-throughput screens, proteomics and in vitro resistance models, we identify the small molecule YC-1 as selectively active against a defined subset of cell lines derived from both liver cancer types. We demonstrate that selectivity is determined by expression of the liver-resident cytosolic sulfotransferase enzyme SULT1A1, which sulfonates YC-1. Sulfonation stimulates covalent binding of YC-1 to lysine residues in protein targets, enriching for RNA-binding factors. Computational analysis defined a wider group of structurally related SULT1A1-activated small molecules with distinct target profiles, which together constitute an untapped small-molecule class. These studies provide a foundation for preclinical development of these agents and point to the broader potential of exploiting SULT1A1 activity for selective targeting strategies.


Assuntos
Alquilantes , Neoplasias Hepáticas , Humanos , Sulfotransferases , Neoplasias Hepáticas/tratamento farmacológico , Arilsulfotransferase
6.
PLoS One ; 17(10): e0276315, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36251663

RESUMO

The luciferin sulfokinase (coelenterazine sulfotransferase) of Renilla was previously reported to activate the storage form, luciferyl sulfate (coelenterazine sulfate) to luciferin (coelenterazine), the substrate for the luciferase bioluminescence reaction. The gene coding for the coelenterazine sulfotransferase has not been identified. Here we used a combined proteomic/transcriptomic approach to identify and clone the sulfotransferase cDNA. Multiple isoforms of coelenterazine sulfotransferase were identified from the anthozoan Renilla muelleri by intersecting its transcriptome with the LC-MS/MS derived peptide sequences of coelenterazine sulfotransferase purified from Renilla. Two of the isoforms were expressed in E. coli, purified, and partially characterized. The encoded enzymes display sulfotransferase activity that is comparable to that of the native sulfotransferase isolated from Renilla reniformis that was reported in 1970. The bioluminescent assay for sensitive detection of 3'-phosphoadenosine 5'-phosphate (PAP) using the recombinant sulfotransferase is demonstrated.


Assuntos
Escherichia coli , Proteômica , Animais , Arilsulfotransferase , Cromatografia Líquida , DNA Complementar , Escherichia coli/genética , Imidazóis , Luciferases/genética , Medições Luminescentes , Pirazinas , Renilla/genética , Sulfatos , Sulfotransferases/genética , Espectrometria de Massas em Tandem
7.
Food Funct ; 13(20): 10558-10573, 2022 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-36156668

RESUMO

Extensive phase II metabolic reactions (i.e., glucuronidation and sulfation) have resulted in low bioavailability and decreased biological effects of curcumin and quercetin. Compared to glucuronidation, information on the sulfation disposition of curcumin and quercetin is limited. In this study, we identified that BCRP and MRP4 played a critical role in the cellular excretion of curcumin-O-sulfate (C-O-S) and quercetin-O-sulfate (Q-O-S) by integrating chemical inhibition with transporter knock-down experiments. Inhibited excretion of sulfate (C-O-S and Q-O-S) caused significant reductions in cellular O-sulfation of curcumin (a maximal 74.4% reduction) and quercetin (a maximal 76.9% reduction), revealing a strong interplay of sulfation with efflux transport. It was further identified that arylsulfatase B (ARSB) played a crucial role in the regulation of cellular O-sulfation by transporters. ARSB overexpression significantly enhanced the reduction effect of MK-571 on the cellular O-sulfation (fmet) of the model compound (38.8% reduction for curcumin and 44.2% reduction for quercetin). On the contrary, ARSB knockdown could reverse the effect of MK-571 on the O-sulfation disposition of the model compound (29.7% increase for curcumin and 47.3% increase for quercetin). Taken together, ARSB has been proven to be involved in cellular O-sulfation, accounting for transporter-dependent O-sulfation of curcumin and quercetin. A better understanding of the interplay beneath metabolism and transport will contribute to the exact prediction of in vivo drug disposition and drug-drug interactions.


Assuntos
Curcumina , N-Acetilgalactosamina-4-Sulfatase , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Arilsulfotransferase , Curcumina/farmacologia , Células HEK293 , Humanos , Proteínas de Membrana Transportadoras , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , N-Acetilgalactosamina-4-Sulfatase/metabolismo , Proteínas de Neoplasias/metabolismo , Propionatos , Quercetina , Quinolinas , Sulfatos/metabolismo
8.
Nutrients ; 14(18)2022 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-36145145

RESUMO

Citrus fruits and juices are a major source of dietary flavanones, and the regular consumption of these foods is inversely associated with the development of cardiometabolic diseases. However, the biological benefits depend on the bioavailability of these compounds, and previous studies have reported a large interindividual variability in the absorption and excretion of these compounds. Different factors, such as age, gender or genetic polymorphism of genes coding enzymes involved in the metabolism and transport of the flavanones, may explain this heterogeneity. This study aimed to assess the impact of single nucleotide polymorphism of sulfotransferases SULT1A1 and SULT1C4, and ABCC2 transporter genes on excretion of phase II flavanone metabolites in volunteers after 24 h of orange juice intake. Forty-six volunteers ingested a single dose of 500 mL of orange juice and 24-h urine was collected. The hesperetin and naringenin phase II metabolites were quantified in urine, and SNPs in SULT1A1, SULT1C4 and ABCC2 genes were genotyped. A significant (p < 0.05) relationship between the SNPs in these genes and the high excretion of phase II flavanone metabolites were observed. These results identified novel polymorphisms associated with higher absorption of flavanones, which may provide bases for future personalized nutritional guidelines for consuming flavanone-rich foods rich in these nutrients for better benefit from their health properties.


Assuntos
Citrus sinensis , Flavanonas , Hesperidina , Arilsulfotransferase/genética , Bebidas/análise , Citrus sinensis/genética , Humanos , Polimorfismo de Nucleotídeo Único , Sulfotransferases/genética
9.
Biochem Pharmacol ; 204: 115243, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36084709

RESUMO

Nevirapine (NVP) is an effective drug for the treatment of HIV infections, but its use is limited by a high incidence of severe skin rash and liver injury. 12-Hydroxynevirapine (12-OH-NVP) is the major metabolite of nevirapine. There is strong evidence that the sulfate of 12-OH-NVP is responsible for the skin rash. While several cytosolic sulfotransferases (SULTs) have been shown to be capable of sulfating 12-OH-NVP, the exact mechanism of sulfation in vivo is unclear. The current study aimed to clarify human SULT(s) and human organs that are capable of sulfating 12-OH-NVP and investigate the metabolic sulfation of 12-OH-NVP using cultured HepG2 human hepatoma cells. Enzymatic assays revealed that of the thirteen human SULTs, SULT1A1 and SULT2A1 displayed strong 12-OH-NVP-sulfating activity. 1-Phenyl-1-hexanol (PHHX), which applied topically prevents the skin rash in rats, inhibited 12-OH-NVP sulfation by SULT1A1 and SULT2A1, implying the involvement of these two enzymes in the sulfation of 12-OH-NVP in vivo. Among five human organ cytosols analyzed, liver cytosol displayed the strongest 12-OH-NVP-sulfating activity, while a low but significant activity was detected with skin cytosol. Cultured HepG2 cells were shown to be capable of sulfating 12-OH-NVP. The effects of genetic polymorphisms of SULT1A1 and SULT2A1 genes on the sulfation of 12-OH-NVP by SULT1A1 and SULT2A1 allozymes were investigated. Two SULT1A1 allozymes, Arg37Asp and Met223Val, showed no detectable 12-OH-NVP-sulfating activity, while a SULT2A1 allozyme, Met57Thr, displayed significantly higher 12-OH-NVP-sulfating activity compared with the wild-type enzyme. Collectively, these results contribute to a better understanding of the involvement of sulfation in NVP-induced skin rash and provide clues to the possible role of SULT genetic polymorphisms in the risk of this adverse reaction.


Assuntos
Exantema , Infecções por HIV , Sulfotransferases/metabolismo , Animais , Arilsulfotransferase/genética , Arilsulfotransferase/metabolismo , Citosol/metabolismo , Exantema/metabolismo , Infecções por HIV/metabolismo , Humanos , Isoenzimas/metabolismo , Nevirapina/metabolismo , Polimorfismo Genético , Ratos , Sulfatos/metabolismo , Sulfotransferases/genética
10.
Chem Res Toxicol ; 35(8): 1418-1424, 2022 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-35926086

RESUMO

The cochaperone Aha1 activates HSP90 ATPase to promote the folding of its client proteins; however, very few client proteins of Aha1 are known. With the use of an ascorbate peroxidase (APEX)-based proximity labeling method, we identified SULT1A1 as a proximity protein of HSP90 that is modulated by genetic depletion of Aha1. Immunoprecipitation followed by Western blot analysis showed the interaction of SULT1A1 with Aha1, but not HSP90. We also observed a reduced level of SULT1A1 protein upon genetic depletion of Aha1 but not upon pharmacological inhibition of HSP90, suggesting that the SULT1A1 protein level is regulated by Aha1 alone. Maturation-dependent interaction assay results showed that Aha1, but not HSP90, binds preferentially to newly synthesized SULT1A1. Reconstitution of Aha1-depleted cells with wild-type Aha1 and its E67K mutant, which is deficient in interacting with HSP90, restored SULT1A1 protein to the same level. Nonetheless, complementation of Aha1-depleted cells with an Aha1 mutant lacking the first 20 amino acids, which disrupts its autonomous chaperone function, was unable to rescue the SULT1A1 protein level. Together, our study revealed, for the first time, Aha1 as an autonomous chaperone in regulating SULT1A1. SULT1A1 is a phase-II metabolic enzyme, where it adds sulfate groups to hydroxyl functionalities in endogenous hormones and xenobiotic chemicals to improve their solubilities and promote their excretion. Thus, our work suggests the role of Aha1 cochaperone in modulating the detoxification of endogenous and environmental chemicals.


Assuntos
Proteínas de Choque Térmico HSP90 , Chaperonas Moleculares , Adenosina Trifosfatases/metabolismo , Arilsulfotransferase/genética , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Chaperonas Moleculares/genética
11.
ChemSusChem ; 15(18): e202201253, 2022 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-35832026

RESUMO

Regioselective sulfation of bioactive compounds is a vital and scarcely studied topic in enzyme-catalyzed transformations and metabolomics. The major bottleneck of enzymatic sulfation consists in finding suitable sulfate donors. In this regard, 3'-phosphoadenosine 5'-phosphosulfate (PAPS)-independent aryl sulfotransferases using aromatic sulfate donors are a favored choice due to their cost-effectiveness. This work presents a unique study of five sulfate donors differing in their leaving group pKa values with a new His-tagged construct of aryl sulfotransferase from Desulfitobacterium hafniense (DhAST-tag). DhAST-tag was purified to homogeneity and biochemically characterized. Two new donors (3-nitrophenyl sulfate and 2-nitrophenyl sulfate) were synthesized. The kinetic parameters of these and other commercial sulfates (4-nitrophenyl, 4-methylumbelliferyl, and phenyl) revealed large differences with respect to the structure of the leaving group. These donors were screened for the sulfation of selected flavonoids (myricetin, chrysin) and phenolic acids (gallate, 3,4-dihydroxyphenylacetate). The donor impact on the sulfation regioselectivity and yield was assessed. The obtained regioselectively sulfated compounds are authentic human metabolites required as standards in clinical trials.


Assuntos
Arilsulfotransferase , Sulfotransferases , Flavonoides , Humanos , Fosfoadenosina Fosfossulfato/metabolismo , Sulfatos/química , Sulfotransferases/metabolismo
12.
ACS Chem Biol ; 17(3): 661-669, 2022 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-35196009

RESUMO

Sulfotransferases (STs) are ubiquitous enzymes that participate in a vast number of biological processes involving sulfuryl group (SO3) transfer. 3'-phosphoadenosine 5'-phosphosulfate (PAPS) is the universal ST cofactor, serving as the "active sulfate" source in cells. Herein, we report the synthesis of three fluorinated PAPS analogues that bear fluorine or trifluoromethyl substituents at the C2 or C8 positions of adenine and their evaluation as substitute cofactors that enable ST activity to be quantified and real-time-monitored by fluorine-19 nuclear magnetic resonance (19F NMR) spectroscopy. Using plant AtSOT18 and human SULT1A3 as two model enzymes, we reveal that the fluorinated PAPS analogues show complementary properties with regard to recognition by enzymes and the working 19F NMR pH range and are attractive versatile tools for studying STs. Finally, we developed an 19F NMR assay for screening potential inhibitors against SULT1A3, thereby highlighting the possible use of fluorinated PAPS analogues for the discovery of drugs for ST-related diseases.


Assuntos
Fosfoadenosina Fosfossulfato , Sulfotransferases , Arabidopsis , Proteínas de Arabidopsis , Arilsulfotransferase , Humanos , Cinética , Espectroscopia de Ressonância Magnética , Sulfotransferases/metabolismo
13.
Pak J Biol Sci ; 25(1): 15-22, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35001571

RESUMO

<b>Background and Objective:</b> Pineapple (<i>Ananas comosus</i>) is a popular fruit worldwide with natural antioxidant properties. This study examined how pineapple modified the expression of drug-metabolizing enzymes (CYP1A2, CYP2C9, CYP3A4, UGT1A6, NAT2 and SULT1A1) and a drug transporter (OATP1B1) in human hepatocarcinoma (HepG2) cells. <b>Materials and Methods:</b> HepG2 cells (2.5×10<sup>5</sup> cells/well in a 24-well plate) were incubated with pineapple juice extract (125-1,000 µg mL<sup>1</sup>) for 48 hrs in phenol red-free medium. Resazurin reduction, ROS, AST and ALT assays were performed. The mRNA expression of target genes was determined by RT/qPCR. <b>Results:</b> Pineapple juice slightly reduced HepG2 cell viability to 80% of the control, while ROS, AST and ALT levels were not changed. Pineapple juice did not alter the expression of CYP1A2, CYP2C9 and UGT1A6 mRNA. All tested concentrations of pineapple juice suppressed CYP3A4, NAT2 and OATP1B1 expression, while SULT1A1 expression was induced. <b>Conclusion:</b> Though pineapple juice slightly decreased the viability of HepG2 cells, cell morphology and cell function remained normal. Pineapple juice disturbed the expression of phase I (CYP3A4) and phase II (NAT2 and SULT1A1) metabolizing genes and the drug transporter OATP1B1. Therefore, the consumption of excessive amounts of pineapple juice poses a risk for drug interactions.


Assuntos
Ananas/metabolismo , Sucos de Frutas e Vegetais/normas , Expressão Gênica/efeitos dos fármacos , Células Hep G2/efeitos dos fármacos , Ananas/microbiologia , Arilamina N-Acetiltransferase/efeitos dos fármacos , Arilamina N-Acetiltransferase/genética , Arilsulfotransferase/efeitos dos fármacos , Arilsulfotransferase/genética , Citocromo P-450 CYP3A/efeitos dos fármacos , Citocromo P-450 CYP3A/genética , Células Hep G2/fisiologia , Humanos
14.
Pak J Biol Sci ; 25(1): 56-66, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35001576

RESUMO

<b>Background and Objective:</b> Dill<i> </i>(<i>Anethum graveolens</i> L.) has the potential to develop as a new alternative medicine due to its pharmacological activities. However, studies into its safety regarding herb-drug interactions have been neglected. This study investigated the risk of dill-induced herb-drug interactions (HDI) by examining its effect on the expression of phase I and II drug-metabolizing enzyme and transporter genes in Caco-2 cells. <b>Materials and Methods:</b> Caco-2 cells (5×10<sup>5</sup> cells/well) were treated with 10 µM ketoconazole, 20 µM rifampicin or dill extract (60-240 µg mL<sup>1</sup>) for 72 hrs. Cell viability was assessed using the resazurin assay and reactive oxygen species (ROS) content was determined with 2 ,7 -dichlorofluorescein diacetate. Aspartate (AST) and alanine aminotransferase (ALT) levels were measured using L-aspartate and L-alanine with α-ketoglutarate as substrate. Expression of phase I (<i>CYP1A2</i>, <i>CYP2C19</i>, <i>CYP2D6</i>, <i>CYP2E1 </i>and <i>CYP3A4</i>) and II (<i>UGT1A6</i>,<i> SULT1A1</i>,<i> NAT1</i>,<i> NAT2 </i>and<i> GSTA1/2</i>) metabolizing genes and transporters (<i>ABCB1</i>,<i> ABCC2</i>,<i> ABCG2 </i>and <i>SLCO1B1</i>) were determined by RT/qPCR. <b>Results:</b> All tested concentrations of dill did not affect cell viability or AST and ALT levels. The highest concentration of dill extract (240 µg mL<sup>1</sup>) significantly lowered the ROS level. Expression of <i>CYP1A2</i>, <i>CYP2C19</i>, <i>SULT1A1</i>, <i>NAT2 </i>and <i>ABCB1 </i>mRNA was significantly up-regulated by dill extract. <b>Conclusion:</b> Dill extract did not directly damage Caco-2 cells but prolonged use of dill may increase the risk of HDI via the up-regulation of the drug-metabolizing genes <i>CYP1A2</i>, <i>CYP2C19</i>, <i>SULT1A1</i>, <i>NAT2 </i>and the transporter <i>ABCB1</i>.


Assuntos
Anethum graveolens/metabolismo , Células CACO-2/efeitos dos fármacos , Regulação para Cima/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/efeitos dos fármacos , Arilamina N-Acetiltransferase/efeitos dos fármacos , Arilsulfotransferase/efeitos dos fármacos , Citocromo P-450 CYP1A2/efeitos dos fármacos , Citocromo P-450 CYP2C19/efeitos dos fármacos , Interações Ervas-Drogas/fisiologia , Humanos , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico
15.
Pharmacol Res Perspect ; 10(1): e00891, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34939338

RESUMO

Opicapone (2,5-dichloro-3-(5-(3,4-dihydroxy-5-nitrophenyl)-1,2,4-oxadiazol-3-yl)-4,6-dimethylpyridine 1-oxide) is a selective catechol-O-methyltransferase inhibitor that has been granted marketing authorization in Europe, Japan, and United States. The present work describes the metabolism and disposition of opicapone in the rat obtained in support to its development and regulatory filling. Plasma levels and elimination of total radioactivity were determined after oral and intravenous administration of [14 C]-opicapone. The maximum plasma concentrations of opicapone-related radioactivity were reached at early time points followed by a gradual return to baseline with a biphasic elimination. Fecal excretion was the primary route of elimination of total radioactivity. Quantitative distribution of drug-related radioactivity demonstrated that opicapone and related metabolites did not distribute to the central nervous system. Opicapone was extensively metabolized in rats resulting in more than 20 phase I and phase II metabolites. Although O-glucuronidation, -sulfation, and -methylation of the nitrocatechol moiety were the principal metabolic pathways, small amount of the N-acetyl derivative was detected, as a result of reduction of the nitro group and subsequent conjugation. Other metabolic transformations included N-oxide reduction to the pyridine derivative and reductive cleavage of 1,2,4-oxadiazole ring followed by further conjugative reactions. Reaction phenotyping studies suggested that SULT 1A1*1 and *2 and UGT1A7, UGT1A8, UGT1A9, and UGT1A10 may be involved in opicapone sulfation and glucuronidation, respectively. However, the reductive metabolic pathways mediated by gut microflora cannot be excluded. Opicapone, in the rat, was found to be rapidly absorbed, widely distributed to peripheric tissues, metabolized mainly via conjugative pathways at the nitro catechol ring, and primarily excreted via feces.


Assuntos
Inibidores de Catecol O-Metiltransferase/farmacocinética , Oxidiazóis/farmacocinética , Administração Intravenosa , Administração Oral , Animais , Arilsulfotransferase/metabolismo , Inibidores de Catecol O-Metiltransferase/administração & dosagem , Glucuronosiltransferase/metabolismo , Masculino , Oxidiazóis/administração & dosagem , Fenótipo , Ratos , Ratos Wistar , Distribuição Tecidual
16.
Arch Toxicol ; 96(2): 673-687, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34921608

RESUMO

Breast cancer etiology is associated with both proliferation and DNA damage induced by estrogens. Breast cancer risk factors (BCRF) such as body mass index (BMI), smoking, and intake of estrogen-active drugs were recently shown to influence intratissue estrogen levels. Thus, the aim of the present study was to investigate the influence of BCRF on estrogen-induced proliferation and DNA damage in 41 well-characterized breast glandular tissues derived from women without breast cancer. Influence of intramammary estrogen levels and BCRF on estrogen receptor (ESR) activation, ESR-related proliferation (indicated by levels of marker transcripts), oxidative stress (indicated by levels of GCLC transcript and oxidative derivatives of cholesterol), and levels of transcripts encoding enzymes involved in estrogen biotransformation was identified by multiple linear regression models. Metabolic fluxes to adducts of estrogens with DNA (E-DNA) were assessed by a metabolic network model (MNM) which was validated by comparison of calculated fluxes with data on methoxylated and glucuronidated estrogens determined by GC- and UHPLC-MS/MS. Intratissue estrogen levels significantly influenced ESR activation and fluxes to E-DNA within the MNM. Likewise, all BCRF directly and/or indirectly influenced ESR activation, proliferation, and key flux constraints influencing E-DNA (i.e., levels of estrogens, CYP1B1, SULT1A1, SULT1A2, and GSTP1). However, no unambiguous total effect of BCRF on proliferation became apparent. Furthermore, BMI was the only BCRF to indeed influence fluxes to E-DNA (via congruent adverse influence on levels of estrogens, CYP1B1 and SULT1A2).


Assuntos
Neoplasias da Mama/metabolismo , Dano ao DNA , Estrogênios/metabolismo , Glândulas Mamárias Humanas/metabolismo , Adulto , Arilsulfotransferase/metabolismo , Índice de Massa Corporal , Neoplasias da Mama/etiologia , Proliferação de Células/fisiologia , Cromatografia Líquida de Alta Pressão , Citocromo P-450 CYP1B1/metabolismo , Feminino , Humanos , Glândulas Mamárias Humanas/patologia , Estresse Oxidativo/fisiologia , Fatores de Risco , Espectrometria de Massas em Tandem
17.
J Cosmet Dermatol ; 21(1): 343-346, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34133836

RESUMO

BACKGROUND: Minoxidil is a widely used over-the-counter topical treatment for hair loss. The response rate for topical minoxidil is relatively low. Minoxidil is a pro-drug, converted to its active form, minoxidil sulfate, by SULT1A1 enzymes located in the scalp. Recently, a novel topical formula that increases the activity of SULT1A1 in hair follicles was reported. AIMS: To evaluate any benefit of applying the SULT1A1 enzyme booster prior to daily 5% minoxidil treatment. METHODS: Male androgenic alopecia patients were recruited to a randomized blinded placebo-controlled study. Patients were randomized to receive 5% topical minoxidil plus the novel formula or minoxidil plus a sham adjuvant. Patient's hair growth was monitored using global photography over 60 days. RESULTS: Twenty-four males with androgenic alopecia (Norwood scale average 4.4, range 2-6) were randomized and completed the trial: 12 in the active arm and 12 in placebo. 75% of the subjects who used the SULT1A1 adjuvant with their daily minoxidil treatments for 60 days regrew hair versus 33% of those using the placebo adjuvant (p = 0.023). CONCLUSIONS: In a small cohort of androgenetic alopecia men, adding the SULT1A1 adjuvant to their daily minoxidil treatment regimen improved hair regrowth.


Assuntos
Minoxidil , Sulfotransferases , Administração Tópica , Alopecia/tratamento farmacológico , Arilsulfotransferase/uso terapêutico , Cabelo , Humanos , Masculino , Sulfotransferases/uso terapêutico , Resultado do Tratamento
18.
Anticancer Drugs ; 33(1): e525-e533, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34387600

RESUMO

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.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Micronutrientes/farmacologia , Quinazolinas/farmacologia , Sulfotransferases/efeitos dos fármacos , Animais , Antimetabólitos Antineoplásicos/administração & dosagem , Arilsulfotransferase/efeitos dos fármacos , Western Blotting , Ciclo Celular , Relação Dose-Resposta a Droga , Feminino , Ácido Fólico/administração & dosagem , Ácido Fólico/farmacologia , Células Hep G2 , Humanos , Leucovorina/administração & dosagem , Leucovorina/farmacologia , Masculino , Metotrexato/administração & dosagem , Metotrexato/farmacologia , Micronutrientes/administração & dosagem , Quinazolinas/administração & dosagem , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Fatores Sexuais
19.
Ann Clin Lab Sci ; 51(6): 868-874, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34921041

RESUMO

Mucosal prolapse syndrome most commonly involves the rectum and presents as solitary rectal ulcer syndrome and proctitis cystica profunda. Symptoms and endoscopic appearances are nonspecific. Histologically, mucosal prolapse is characterized by fibromuscular obliteration of the lamina propria, and displacement of crypts into submucosa and muscularis mucosae. Mucosal prolapse presenting as polyposis is rare and has only been reported involving the rectosigmoid colon. In this report, we describe a case of mucosal prolapse syndrome presenting as diffuse polyposis and colitis cystica profunda involving the hepatic, splenic flexures and descending colon in a teenage boy suffering from refractory fibrostenosing Crohn's disease. This patient was found to have possibly deleterious homozygous single nucleotide polymorphisms in both SULT1A1 and SULT1A2 genes within a unique polygenic variation of altered cell adhesion.


Assuntos
Polipose Adenomatosa do Colo , Arilsulfotransferase/genética , Colectomia/métodos , Doença de Crohn , Mucosa Intestinal , Prolapso Retal , Inibidores do Fator de Necrose Tumoral/uso terapêutico , Polipose Adenomatosa do Colo/diagnóstico , Polipose Adenomatosa do Colo/etiologia , Adolescente , Adesão Celular/genética , Colite/diagnóstico por imagem , Colite/etiologia , Colite/patologia , Colonoscopia/métodos , Doença de Crohn/diagnóstico , Doença de Crohn/genética , Doença de Crohn/fisiopatologia , Testes Genéticos/métodos , Humanos , Mucosa Intestinal/diagnóstico por imagem , Mucosa Intestinal/patologia , Imageamento por Ressonância Magnética/métodos , Masculino , Polimorfismo de Nucleotídeo Único , Prolapso Retal/diagnóstico , Prolapso Retal/etiologia , Índice de Gravidade de Doença
20.
Xenobiotica ; 51(9): 1071-1080, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34328372

RESUMO

Sulfotransferases (SULTs) are phase II detoxification enzymes that is involved in the biotransformation of many compounds including tobacco carcinogens. A polymorphism in the SULT1A1 (Arg213His) gene results in reduced enzyme activity.We investigated the association between the SULT1A1 (Arg213/His) genotype and lung cancer (LC). This case-control study comprised of 550 cases and controls, matched on age, gender and smoking status.The variant genotype exhibited no association with LC risk, even after stratification on basis of histological subtypes. Male LC patients carrying the variant His213 allele (p = 0.02) did not exhibit an increased risk towards LC. Smokers harbouring the Arg/His genotype did demonstrate a reduced risk towards LC (AOR = 0.70; p = 0.019). Furthermore, the LC subjects who were heavy smokers and harbouring the Arg/His genotype (AOR = 0.28; p = 0.019) did not show a genetic predisposition towards LC susceptibility. The subjects who smoked pack years of above 40 and carrying the His/His (AOR = 0.28; p = 0.036) genotype were found to have a reduced risk for LC. Furthermore, 473 subjects were analysed in regards to overall survival, wherein the His/His genotype exhibited better OS than Arg/Arg genotype (11.30 vs. 8.07 months).This study provides evidence of no genetic predisposition towards LC risk associated with SULT1A1 Arg213His polymorphism in relation to tobacco smoking.


Assuntos
Arilsulfotransferase , Neoplasias Pulmonares , Arilsulfotransferase/genética , Estudos de Casos e Controles , Genótipo , Humanos , Neoplasias Pulmonares/genética , Masculino , Polimorfismo Genético , Fatores de Risco , Fumar/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...