Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
1.
Nat Chem Biol ; 17(6): 684-692, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33846619

RESUMO

Heparan sulfate (HS) proteoglycans bind extracellular proteins that participate in cell signaling, attachment and endocytosis. These interactions depend on the arrangement of sulfated sugars in the HS chains generated by well-characterized biosynthetic enzymes; however, the regulation of these enzymes is largely unknown. We conducted genome-wide CRISPR-Cas9 screens with a small-molecule ligand that binds to HS. Screening of A375 melanoma cells uncovered additional genes and pathways impacting HS formation. The top hit was the epigenetic factor KDM2B, a histone demethylase. KDM2B inactivation suppressed multiple HS sulfotransferases and upregulated the sulfatase SULF1. These changes differentially affected the interaction of HS-binding proteins. KDM2B-deficient cells displayed decreased growth rates, which was rescued by SULF1 inactivation. In addition, KDM2B deficiency altered the expression of many extracellular matrix genes. Thus, KDM2B controls proliferation of A375 cells through the regulation of HS structure and serves as a master regulator of the extracellular matrix.


Assuntos
Proteínas F-Box/antagonistas & inibidores , Estudo de Associação Genômica Ampla , Heparitina Sulfato/metabolismo , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Algoritmos , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Descoberta de Drogas , Matriz Extracelular/genética , Ensaios de Triagem em Larga Escala , Humanos , Ligação Proteica/genética , RNA-Seq , Sulfotransferases/antagonistas & inibidores
2.
J Biol Chem ; 296: 100353, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33524392

RESUMO

Polychlorinated bisphenols (PCBs) continue to contaminate food chains globally where they concentrate in tissues and disrupt the endocrine systems of species throughout the ecosphere. Hydroxylated PCBs (OH-PCBs) are major PCB metabolites and high-affinity inhibitors of human estrogen sulfotransferase (SULT1E1), which sulfonates estrogens and thus prevents them from binding to and activating their receptors. OH-PCB inhibition of SULT1E1 is believed to contribute significantly to PCB-based endocrine disruption. Here, for the first time, the molecular basis of OH-PCB inhibition of SULT1E1 is revealed in a structure of SULT1E1 in complex with OH-PCB1 (4'-OH-2,6-dichlorobiphenol) and its substrates, estradiol (E2), and PAP (3'-phosphoadenosine-5-phosphosulfate). OH-PCB1 prevents catalysis by intercalating between E2 and catalytic residues and establishes a new E2-binding site whose E2 affinity and positioning are greater than and competitive with those of the reactive-binding pocket. Such complexes have not been observed previously and offer a novel template for the design of high-affinity inhibitors. Mutating residues in direct contact with OH-PCB weaken its affinity without compromising the enzyme's catalytic parameters. These OH-PCB resistant mutants were used in stable transfectant studies to demonstrate that OH-PCBs regulate estrogen receptors in cultured human cell lines by binding the OH-PCB binding pocket of SULT1E1.


Assuntos
Inibidores Enzimáticos/farmacologia , Estrogênios/farmacologia , Bifenilos Policlorados/farmacologia , Sulfotransferases/antagonistas & inibidores , Inibidores Enzimáticos/química , Estrogênios/química , Humanos , Hidroxilação , Modelos Moleculares , Bifenilos Policlorados/química , Receptores de Estrogênio/metabolismo , Sulfotransferases/química , Sulfotransferases/metabolismo
3.
Med Sci Monit ; 26: e926780, 2020 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-33079922

RESUMO

BACKGROUND Cervical cancer threatens women's health worldwide. Verteporfin (VP), a small-molecule YAP1 inhibitor, inhibits cancer cell growth. This study investigated whether VP could inhibit the proliferation and promote the apoptosis of cervical cancer cells by decreasing SULT2B1 expression. MATERIAL AND METHODS Normal and cancerous cervical cell proliferation after VP treatment was detected by CCK-8 assay. HeLa cell migration, invasion, and apoptosis after VP treatment and transfection were analyzed by wound healing assay, transwell assay, and TUNEL assay, respectively. The expression of related proteins was determined by western blot analysis. Western blot and RT-qPCR analysis detected mRNA and protein expression of SULT2B1. RESULTS Different VP concentrations (0.5, 1, 2, and 5 µM) inhibited the viability of HeLa cells and had no obvious effect on H8 cells. Therefore, 5 µM VP was selected for subsequent experiments. VP inhibited the proliferation, migration, and invasion of HeLa cells and promoted their apoptosis. Bcl-2 expression decreased, and expression of Bax, caspase-3, and caspase-9 in VP-treated HeLa cells increased. SULT2B1 expression increased in cervical cancer cells compared with normal cervical cells. Furthermore, SULT2B1 expression increased in HeLa cells and VP suppressed SULT2B1 expression. SULT2B1 overexpression reduced the inhibiting effect of VP on the proliferation, migration, and apoptosis of HeLa cells, and reduced VP effect on apoptosis of HeLa cells. SULT2B1 overexpression upregulated the Bcl-2 expression and downregulated the expression of Bax, caspase-3, and caspase-9 in VP-treated HeLa cells. CONCLUSIONS VP inhibited the proliferation, migration, and invasion and promoted apoptosis of cervical cancer cells by decreasing SULT2B1 expression.


Assuntos
Antineoplásicos , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sulfotransferases , Neoplasias do Colo do Útero/tratamento farmacológico , Verteporfina , Antineoplásicos/farmacologia , Proteínas Reguladoras de Apoptose/metabolismo , Movimento Celular/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Invasividade Neoplásica , Sulfotransferases/antagonistas & inibidores , Sulfotransferases/metabolismo , Neoplasias do Colo do Útero/patologia , Verteporfina/farmacologia
4.
Biochem Pharmacol ; 180: 114197, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32798464

RESUMO

Hepatic estrogen sulfotransferase (SULT1E1), the enzyme that inactivates estrogen, regulates metabolic estrogen homeostasis. Here, we have demonstrated how nuclear receptor PXR regulated the SULT1E1 gene in response to glucose in human hepatoma-derived cells and in response to fasting in mouse livers. The SULT1E1 gene was activated by a nuclear receptor HNF4α-RORα complex binding on an upstream enhancer of the SULT1E1 promoter in cells cultured in high glucose medium (Hu and Negishi, 2020). The SULT1E1 gene was repressed in cells cultured in low glucose medium, in which PXR was phosphorylated at Ser350 by vaccinia virus-related kinase 1. Phosphorylated PXR interacted with this complex, retaining HNF4α on and dissociating RORα from the enhancer as a phosphorylated PXR complex. Therefore, in response to low glucose, phosphorylated PXR transduced a low glucose signal to repress the SULT1E1 gene in cells. Hepatic Sult1e1 mRNA was induced in PXR wild type (WT) male mice in response to fasting, whereas this induction was synergistically increased in phosphorylation-blocking PXR Ser347Ala (Ser350 in human) KI males over that observed in PXR WT males. As phosphorylated PXR repressed the Sult1e1 gene, it increased its binding to the Sult1e1 promoter in WT males. The absence of phosphorylated PXR resulted in the synergistic activation of the Sult1e1 gene in PXR KI males. Apparently, phosphorylated PXR functioned as a transcriptional repressor to the SULT1E1/Sult1e1 gene in human liver cells and mouse livers.


Assuntos
Jejum/metabolismo , Glucose/administração & dosagem , Fígado/metabolismo , Receptor de Pregnano X/metabolismo , Serina/metabolismo , Sulfotransferases/biossíntese , Animais , Células COS , Chlorocebus aethiops , Feminino , Células Hep G2 , Humanos , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação/fisiologia , Receptor de Pregnano X/química , Receptor de Pregnano X/genética , Estrutura Secundária de Proteína , Serina/genética , Sulfotransferases/antagonistas & inibidores , Sulfotransferases/genética
5.
J Am Soc Nephrol ; 31(7): 1496-1508, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32424001

RESUMO

BACKGROUND: Studies have suggested that estrogens may protect mice from AKI. Estrogen sulfotransferase (SULT1E1, or EST) plays an important role in estrogen homeostasis by sulfonating and deactivating estrogens, but studies on the role of SULT1E1 in AKI are lacking. METHODS: We used the renal ischemia-reperfusion model to investigate the role of SULT1E1 in AKI. We subjected wild-type mice, Sult1e1 knockout mice, and Sult1e1 knockout mice with liver-specific reconstitution of SULT1E1 expression to bilateral renal ischemia-reperfusion or sham surgery, either in the absence or presence of gonadectomy. We assessed relevant biochemical, histologic, and gene expression markers of kidney injury. We also used wild-type mice treated with the SULT1E1 inhibitor triclosan to determine the effect of pharmacologic inhibition of SULT1E1 on AKI. RESULTS: AKI induced the expression of Sult1e1 in a tissue-specific and sex-specific manner. It induced expression of Sult1e1 in the liver in both male and female mice, but Sult1e1 induction in the kidney occurred only in male mice. Genetic knockout or pharmacologic inhibition of Sult1e1 protected mice of both sexes from AKI, independent of the presence of sex hormones. Instead, a gene profiling analysis indicated that the renoprotective effect was associated with increased vitamin D receptor signaling. Liver-specific transgenic reconstitution of SULT1E1 in Sult1e1 knockout mice abolished the protection in male mice but not in female mice, indicating that Sult1e1's effect on AKI was also tissue-specific and sex-specific. CONCLUSIONS: SULT1E1 appears to have a novel function in the pathogenesis of AKI. Our findings suggest that inhibitors of SULT1E1 might have therapeutic utility in the clinical management of AKI.


Assuntos
Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/prevenção & controle , Fígado/metabolismo , Sulfotransferases/genética , Sulfotransferases/metabolismo , Injúria Renal Aguda/etiologia , Animais , Calcitriol/farmacologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Rim/metabolismo , Masculino , Camundongos , Camundongos Knockout , Orquiectomia , Ovariectomia , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Traumatismo por Reperfusão/complicações , Fatores Sexuais , Transdução de Sinais , Sulfotransferases/antagonistas & inibidores , Triclosan/farmacologia
6.
Lipids Health Dis ; 18(1): 203, 2019 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-31757214

RESUMO

BACKGROUND: A healthy gastric mucosal epithelium exhibits tumor-suppressive properties. Gastric epithelial cell dysfunction contributes to gastric cancer development. Oxysterols provided from food or cholesterol oxidation in the gastric epithelium may be further sulfated by hydroxysteroid sulfotransferase 2B1 (SULT2B1), which is highly abundant in the gastric epithelium. However, the effects of SULT2B1 on gastric epithelial function and gastric carcinogenesis are unclear. METHODS: A mouse gastric tumor model was established using carcinogenic agent 3-methylcholanthrene (3-MCA). A SULT2B1 deletion (SULT2B1-/-) human gastric epithelial line GES-1 was constructed by CRISPR/CAS9 genome editing system. RESULTS: The gastric tumor incidence was higher in the SULT2B1-/- mice than in the wild-type (WT) mice. In gastric epithelial cells, adenovirus-mediated SULT2B1b overexpression reduced the levels of oxysterols, such as 24(R/S),25-epoxycholesterol (24(R/S),25-EC) and 27-hydroxycholesterol (27HC). This condition also increased PI3K/AKT signaling to promote gastric epithelial cell proliferation, epithelization, and epithelial development. However, SULT2B1 deletion or SULT2B1 knockdown suppressed PI3K/AKT signaling, epithelial cell epithelization, and wound healing and induced gastric epithelial cell malignant transition upon 3-MCA induction. CONCLUSIONS: The abundant SULT2B1 expression in normal gastric epithelium might maintain epithelial function via the PI3K/AKT signaling pathway and suppress gastric carcinogenesis induced by a carcinogenic agent.


Assuntos
Carcinogênese/genética , Regulação Neoplásica da Expressão Gênica , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Neoplasias Gástricas/genética , Sulfotransferases/genética , Animais , Sequência de Bases , Sistemas CRISPR-Cas , Carcinogênese/efeitos dos fármacos , Carcinogênese/metabolismo , Carcinogênese/patologia , Linhagem Celular Tumoral , Proliferação de Células , Colesterol/análogos & derivados , Colesterol/metabolismo , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/enzimologia , Mucosa Gástrica/patologia , Edição de Genes , Humanos , Hidroxicolesteróis/metabolismo , Metilcolantreno/administração & dosagem , Camundongos , Camundongos Knockout , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Neoplasias Gástricas/induzido quimicamente , Neoplasias Gástricas/enzimologia , Neoplasias Gástricas/mortalidade , Sulfotransferases/antagonistas & inibidores , Sulfotransferases/deficiência , Análise de Sobrevida
7.
Chem Res Toxicol ; 32(6): 1281-1288, 2019 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-31046239

RESUMO

Aloe-emodin (AE) is a major anthraquinone ingredient of numerous traditional Chinese medicines with a variety of beneficial biological activities in vitro. Previous studies suggested that AE possessed cytotoxicity and genotoxicity. Nevertheless, the mechanisms of the toxic action of AE have not yet been fully clarified. The present study aimed at characterization of metabolic pathways of AE to better understand the mechanisms of AE-induced cytotoxicity. An AE-derived glutathione conjugate (AE-GSH) was observed in rat liver cytosol incubations containing AE and GSH, along with 3'-phosphoadenosine-5'-phosphosulfate (PAPS). Similar incubation fortified with N-acetylcysteine (NAC) in place of GSH offered an AE-NAC conjugate corresponding to the GSH conjugate. The formation of the two conjugates was found to require PAPS. The two conjugates were respectively detected in bile and urine of rats given AE. Sulfotransferase (SULT) inhibitor pentachlorophenol (PCP) suppressed the production of the observed AE-GSH/NAC conjugates in vivo, which suggested that SULTs participated in the process of the metabolic activation of AE. The presence of PCP attenuated cell susceptibility to AE-induced cytotoxicity. The present study illustrated potential association of sulfation-mediated bioactivation of AE with its cytotoxicity.


Assuntos
Antraquinonas/farmacologia , Sulfotransferases/metabolismo , Ativação Metabólica/efeitos dos fármacos , Animais , Antraquinonas/química , Antraquinonas/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Citosol/química , Citosol/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Células Hep G2 , Humanos , Fígado/química , Fígado/metabolismo , Masculino , Estrutura Molecular , Pentaclorofenol/farmacologia , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Sulfotransferases/antagonistas & inibidores
8.
J Appl Toxicol ; 39(4): 650-657, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30874336

RESUMO

Sulfotransferase 1A (SULT1A) expression is lower in the liver of humans than that of rodents. Therefore, species differences should be taken into consideration when assessing the risk of rodent hepatocarcinogens metabolically activated by SULT1A in humans. Although some renal carcinogens require SULT1A-mediated activation, it is unclear how SULT1A activity in the liver affects renal carcinogens. To explore the effects of SULT1A activity in the liver on genotoxicity induced by SULT1A-activated renal carcinogens, B6C3F1 mice or gpt delta mice of the same strain background were given lucidin-3-O-primeveroside (LuP), a hepatic and renal carcinogen of rodents, for 4 or 13 weeks, respectively, and pentachlorophenol (PCP) as a liver-specific SULT inhibitor, was given from 1 week before LuP treatment to the end of the experiment. A 4 week exposure of LuP induced lucidin-specific DNA adduct formation. The suppression of Sult1a expression was observed only in the liver but not in the kidneys of PCP-treated mice, but co-administration of PCP suppressed LuP-induced DNA adduct formation in both organs. Thirteen-week exposure of LuP increased mutation frequencies and cotreatment with PCP suppressed these increases in both organs. Given that intact levels of SULT activity in the liver were much higher than in the kidneys of rodents, SULT1A may predominantly activate LuP in the liver, consequently leading to genotoxicity not only in the liver but also in the kidney. Thus, species differences should be considered in human risk assessment of renal carcinogens activated by SULT1A as in the case of the corresponding liver carcinogens.


Assuntos
Antraquinonas/toxicidade , Dissacarídeos/toxicidade , Corantes de Alimentos/toxicidade , Rim/efeitos dos fármacos , Fígado/enzimologia , Sulfotransferases/antagonistas & inibidores , Animais , Inibidores Enzimáticos/farmacologia , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos , Pentaclorofenol/farmacologia , Sulfotransferases/genética
9.
J Pharmacol Exp Ther ; 369(3): 389-405, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30918069

RESUMO

Lithocholic acid (LCA) is a bile acid associated with adverse effects, including cholestasis, and it exists in vivo mainly as conjugates known as glyco-LCA (GLCA) and tauro-LCA (TLCA). Tamoxifen has been linked to the development of cholestasis, and it inhibits sulfotransferase 2A1 (SULT2A1)-catalyzed dehydroepiandrosterone (DHEA) sulfonation. The present study was done to characterize the sulfonation of LCA, GLCA, and TLCA and to investigate whether triphenylethylene (clomifene, tamoxifen, toremifene, ospemifene, droloxifene), benzothiophene (raloxifene, arzoxifene), tetrahydronaphthalene (lasofoxifene, nafoxidine), indole (bazedoxifene), and benzopyran (acolbifene) classes of selective estrogen receptor modulator (SERM) inhibit LCA, GLCA, and TLCA sulfonation. Human recombinant SULT2A1, but not SULT2B1b or SULT1E1, catalyzed LCA, GLCA, and TLCA sulfonation, whereas each of these enzymes catalyzed DHEA sulfonation. LCA, GLCA, and TLCA sulfonation is catalyzed by human liver cytosol, and SULT2A1 followed the substrate inhibition model with comparable apparent K m values (≤1 µM). Each of the SERMs inhibited LCA, GLCA, and TLCA sulfonation with varying potency and mode of enzyme inhibition. The potency and extent of inhibition of LCA sulfonation were attenuated or increased by structural modifications to toremifene, bazedoxifene, and lasofoxifene. The inhibitory effect of raloxifene, bazedoxifene, and acolbifene on LCA sulfonation was also observed in HepG2 human hepatocellular carcinoma cells. Overall, among the SERMs investigated, bazedoxifene and raloxifene were the most effective inhibitors of LCA, GLCA, and TLCA sulfonation. These findings provide insight into the structural features of specific SERMs that contribute to their inhibition of SULT2A1-catalyzed LCA sulfonation. Inhibition of LCA, GLCA, and TLCA detoxification by a SERM may provide a biochemical basis for adverse effects associated with a SERM.


Assuntos
Biocatálise/efeitos dos fármacos , Ácido Litocólico/análogos & derivados , Moduladores Seletivos de Receptor Estrogênico/química , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Ácidos Sulfônicos/metabolismo , Sulfotransferases/metabolismo , Ácido Taurolitocólico/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Células Hep G2 , Humanos , Cinética , Ácido Litocólico/metabolismo , Fígado/citologia , Oxirredução , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Sulfotransferases/antagonistas & inibidores
10.
Dermatol Ther ; 31(6): e12741, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30226287

RESUMO

Topical minoxidil is the only US FDA approved OTC drug for the treatment of androgenetic alopecia (AGA). Minoxidil is a pro-drug converted into its active form, minoxidil sulfate, by the sulfotransferase enzymes in the outer root sheath of hair follicles. Previously, we demonstrated that sulfotransferase activity in hair follicles predicts response to topical minoxidil in the treatment of AGA. In the human liver, sulfotransferase activity is significantly inhibited by salicylic acid. Low-dose OTC aspirin (75-81 mg), a derivative of salicylic acid, is used by millions of people daily for the prevention of coronary heart disease and cancer. It is not known whether oral aspirin inhibits sulfotransferase activity in hair follicles, potentially affecting minoxidil response in AGA patients. In the present study, we determined the follicular sulfotransferase enzymatic activity following 14 days of oral aspirin administration. In our cohort of 24 subjects, 50% were initially predicted to be responders to minoxidil. However, following 14 days of aspirin administration, only 27% of the subjects were predicted to respond to topical minoxidil. To the best of our knowledge, this is the first study to report the effect of low-dose daily aspirin use on the efficacy of topical minoxidil.


Assuntos
Alopecia/tratamento farmacológico , Aspirina/administração & dosagem , Inibidores Enzimáticos/administração & dosagem , Folículo Piloso/efeitos dos fármacos , Minoxidil/administração & dosagem , Pró-Fármacos/administração & dosagem , Sulfotransferases/antagonistas & inibidores , Administração Cutânea , Adulto , Alopecia/diagnóstico , Alopecia/fisiopatologia , Aspirina/efeitos adversos , Interações Medicamentosas , Inibidores Enzimáticos/efeitos adversos , Folículo Piloso/enzimologia , Folículo Piloso/crescimento & desenvolvimento , Humanos , Masculino , Minoxidil/análogos & derivados , Minoxidil/metabolismo , Pró-Fármacos/metabolismo , Medição de Risco , Sulfotransferases/metabolismo , Resultado do Tratamento , Adulto Jovem
11.
Biochem J ; 475(15): 2435-2455, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-29934490

RESUMO

Protein tyrosine sulfation is a post-translational modification best known for regulating extracellular protein-protein interactions. Tyrosine sulfation is catalysed by two Golgi-resident enzymes termed tyrosylprotein sulfotransferases (TPSTs) 1 and 2, which transfer sulfate from the cofactor PAPS (3'-phosphoadenosine 5'-phosphosulfate) to a context-dependent tyrosine in a protein substrate. A lack of quantitative tyrosine sulfation assays has hampered the development of chemical biology approaches for the identification of small-molecule inhibitors of tyrosine sulfation. In the present paper, we describe the development of a non-radioactive mobility-based enzymatic assay for TPST1 and TPST2, through which the tyrosine sulfation of synthetic fluorescent peptides can be rapidly quantified. We exploit ligand binding and inhibitor screens to uncover a susceptibility of TPST1 and TPST2 to different classes of small molecules, including the anti-angiogenic compound suramin and the kinase inhibitor rottlerin. By screening the Published Kinase Inhibitor Set, we identified oxindole-based inhibitors of the Ser/Thr kinase RAF (rapidly accelerated fibrosarcoma) as low-micromolar inhibitors of TPST1 and TPST2. Interestingly, unrelated RAF inhibitors, exemplified by the dual BRAF/VEGFR2 inhibitor RAF265, were also TPST inhibitors in vitro We propose that target-validated protein kinase inhibitors could be repurposed, or redesigned, as more-specific TPST inhibitors to help evaluate the sulfotyrosyl proteome. Finally, we speculate that mechanistic inhibition of cellular tyrosine sulfation might be relevant to some of the phenotypes observed in cells exposed to anionic TPST ligands and RAF protein kinase inhibitors.


Assuntos
Imidazóis/química , Proteínas de Membrana , Peptídeos/química , Proteínas Proto-Oncogênicas B-raf , Piridinas/química , Sulfotransferases , Tirosina/química , Humanos , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/química , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas B-raf/química , Sulfotransferases/antagonistas & inibidores , Sulfotransferases/química
12.
Methods Enzymol ; 605: 101-138, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29909823

RESUMO

Sterol sulfates are widely occurring molecules in marine organisms. Their importance has been so far underestimated although many of these compounds are crucial mediators of physiological and ecological functions in other organisms. Biosynthesis of sterol sulfates is controlled by cytosolic sulfotransferases (SULTs), a varied family of enzymes that catalyze the transfer of a sulfo residue (-SO3H) from the universal donor 3'-phosphoadenosine-5'-phosphosulfate to the hydroxyl function at C-3 of the steroid skeleton. The absence of molecular tools has been the main impediment to the development of a biosynthetic study of this class of compounds in marine organisms. In fact, there is very limited information about these enzymes in marine environments. SULT activity has, however, been reported in several marine species, and, recently, the production of sterol sulfates has been linked to the control of growth in marine diatoms. In this chapter, we describe methods for the study of sterol sulfates in this lineage of marine microalgae. The main aim is to provide the tools useful to deal with the biosynthesis and regulation of these compounds and to circumvent the bottleneck of the lack of molecular information. The protocols have been designed for marine diatoms, but most of the procedures can be used for other marine organisms.


Assuntos
Fracionamento Químico/métodos , Esteróis/análise , Sulfatos/análise , Sulfotransferases/isolamento & purificação , Vias Biossintéticas/efeitos dos fármacos , Isótopos de Carbono/química , Fracionamento Químico/instrumentação , Cromatografia Líquida de Alta Pressão/instrumentação , Cromatografia Líquida de Alta Pressão/métodos , Diatomáceas/efeitos dos fármacos , Diatomáceas/fisiologia , Microalgas/efeitos dos fármacos , Microalgas/fisiologia , Fosfoadenosina Fosfossulfato/metabolismo , Quercetina/farmacologia , Coloração e Rotulagem/instrumentação , Coloração e Rotulagem/métodos , Esteróis/química , Esteróis/metabolismo , Sulfatos/química , Sulfatos/metabolismo , Sulfotransferases/antagonistas & inibidores , Sulfotransferases/metabolismo , Espectrometria de Massas em Tandem/instrumentação , Espectrometria de Massas em Tandem/métodos
13.
Expert Rev Mol Diagn ; 18(6): 499-512, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29708446

RESUMO

INTRODUCTION: BRAF mutant colorectal cancer (BRAF MT CRC) is a unique category of colorectal tumour with peculiar molecular, pathological and clinical features and poor prognosis; despite recent research, BRAF mutation predictive value and standard treatment of BRAF MT CRC still have to be defined. In this review, we focused on this challenging topic. Areas covered: The potential use of BRAF mutational status among recent additional prognostic and predictive indicators and current treatment strategy in use in these patients is discussed. Moreover, implications and characteristics of new BRAF mutations other than BRAFV600E are analyzed. An in-deep outlook on the immediate future for clinical and translational research in this subgroup of patients is also presented, such as combination therapy with agents targeting the RAS/RAF/MEK/ERK pathway and standard chemotherapy in order to overcome resistance. We performed a research on Pubmed typing 'BRAF mutation', 'colorectal cancer', 'predictive and prognostic value', 'targeted therapy', 'BRAF inhibition'. Expert commentary: BRAFV600E mutation represents a strong, independent negative prognostic factor in II-III stage MSS CRC and mCRC. The best treatment still has to be identified; currently, in good performance status patients, an intensive-chemotherapy-combination remains the standard of care. Further investigations are warranted to explore new horizons to change BRAF MT mCRC outcomes.


Assuntos
Neoplasias Colorretais , Sistema de Sinalização das MAP Quinases , Glicoproteínas de Membrana , Mutação de Sentido Incorreto , Sulfotransferases , Substituição de Aminoácidos , Animais , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Prognóstico , Sulfotransferases/antagonistas & inibidores , Sulfotransferases/genética , Sulfotransferases/metabolismo
14.
Toxicol Lett ; 294: 116-121, 2018 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-29772265

RESUMO

Sulfonation is a major pathway of estrogen biotransformation with a role in regulating estrogen homeostasis in humans and sheep. Previous in vitro studies found that triclosan is an especially potent competitive inhibitor of ovine placental estrogen sulfotransferase, with Kic of <0.1 nM. As the placenta is the main organ responsible for estrogen synthesis in pregnancy in both women and sheep, and the liver is another site of estrogen biotransformation, this study examined the effects of triclosan exposure of pregnant ewes on placental and hepatic sulfotransferase activity. Triclosan, 0.1 mg/kg/day, or saline vehicle was administered to late gestation fetal sheep for two days either by direct infusion into the fetal circulation or infusion into the maternal blood. On the third day, fetal liver and placenta were harvested and analyzed for triclosan and for cytosolic estradiol sulfotransferase activity. Placenta contained higher concentrations of triclosan than liver in each individual sheep in both treatment groups. There was a negative correlation between triclosan tissue concentration (pmol/g tissue) and cytosolic sulfotransferase activity (pmol/min/mg protein) towards estradiol. These findings demonstrated that in the sheep exposed to very low concentrations of triclosan, this substance is taken up into placenta and reduces estrogen sulfonation.


Assuntos
Anti-Infecciosos Locais/toxicidade , Inibidores Enzimáticos/toxicidade , Fígado/efeitos dos fármacos , Exposição Materna/efeitos adversos , Placenta/efeitos dos fármacos , Sulfotransferases/antagonistas & inibidores , Triclosan/toxicidade , Animais , Anti-Infecciosos Locais/administração & dosagem , Anti-Infecciosos Locais/metabolismo , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/metabolismo , Estradiol/metabolismo , Feminino , Feto/irrigação sanguínea , Feto/efeitos dos fármacos , Feto/metabolismo , Infusões Intravenosas , Fígado/embriologia , Fígado/metabolismo , Placenta/enzimologia , Placenta/metabolismo , Gravidez , Carneiro Doméstico , Sulfotransferases/metabolismo , Distribuição Tecidual , Toxicocinética , Triclosan/administração & dosagem , Triclosan/metabolismo
15.
Sci Rep ; 8(1): 6382, 2018 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-29686391

RESUMO

The misfolding and accumulation of tau protein into intracellular aggregates known as neurofibrillary tangles is a pathological hallmark of neurodegenerative diseases such as Alzheimer's disease. However, while tau propagation is a known marker for disease progression, exactly how tau propagates from one cell to another and what mechanisms govern this spread are still unclear. Here, we report that cellular internalization of tau is regulated by quaternary structure and have developed a cellular assay to screen for genetic modulators of tau uptake. Using CRISPRi technology we have tested 3200 genes for their ability to regulate tau entry and identified enzymes in the heparan sulfate proteoglycan biosynthetic pathway as key regulators. We show that 6-O-sulfation is critical for tau-heparan sulfate interactions and that this modification regulates uptake in human central nervous system cell lines, iPS-derived neurons, and mouse brain slice culture. Together, these results suggest novel strategies to halt tau transmission.


Assuntos
Encéfalo/metabolismo , Glioma/metabolismo , Proteoglicanas de Heparan Sulfato/química , Estrutura Quaternária de Proteína , Sulfotransferases/metabolismo , Enxofre/metabolismo , Proteínas tau/metabolismo , Animais , Encéfalo/fisiologia , Sistemas CRISPR-Cas , Dinamina II/antagonistas & inibidores , Dinamina II/genética , Dinamina II/metabolismo , Genômica , Glioma/genética , Glioma/patologia , Proteoglicanas de Heparan Sulfato/metabolismo , Humanos , Camundongos , N-Acetilglucosaminiltransferases/antagonistas & inibidores , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo , Sulfotransferases/antagonistas & inibidores , Sulfotransferases/genética , Células Tumorais Cultivadas , Proteínas tau/química , Proteínas tau/genética
16.
Biomed Pharmacother ; 103: 362-372, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29669302

RESUMO

Hepatocellular carcinoma (HCC) is characterized by elevation in the activity of sulfatase-2, an extracellular enzyme that catalyzes removal of 6-O-sulfate groups from heparan sulfate. Therefore, we conducted this study to investigate the cytotoxic activity of the strong water-soluble antioxidant, sodium ascorbate, against HCC both in vivo and in vitro. Sodium ascorbate enhanced animal survival in vivo and reduced HepG2 cells survival. The protein levels of heparan sulfate proteoglycans (HSPGs), insulin like growth factor (IGF)-2, sulfatase-2 and glypican-3 were assessed. Inflammation was evaluated by measuring the gene and protein expression of NFκB, TNF-α, IL-1ß, IL-4, IL-6 and IL-10. We found that sodium ascorbate blocked HCC-induced activation of sulfatase-2 leading to restoration of HSPGs receptors associated with reduction in IGF-2 and glypican-3. Sodium ascorbate exerts anti-inflammatory activity by reducing the expression of NFκB, CRP, TNF-α, IL-1ß and IL-6 associated with enhanced expression of the anti-inflammatory cytokines, IL-4 and IL-10. In conclusion, cytotoxic effects of sodium ascorbate against HCC can be partially explained by inhibition of sulfatase-2, restoration of HSPGs receptors and deactivation of the inflammatory pathway.


Assuntos
Ácido Ascórbico/farmacologia , Carcinoma Hepatocelular/enzimologia , Citotoxinas/farmacologia , Neoplasias Hepáticas Experimentais/enzimologia , Sulfotransferases/antagonistas & inibidores , Sulfotransferases/biossíntese , Animais , Ácido Ascórbico/uso terapêutico , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/prevenção & controle , Citotoxinas/uso terapêutico , Relação Dose-Resposta a Droga , Células Hep G2 , Humanos , Neoplasias Hepáticas Experimentais/patologia , Neoplasias Hepáticas Experimentais/prevenção & controle , Masculino , Ratos , Ratos Sprague-Dawley , Sulfatases
17.
Environ Toxicol Pharmacol ; 58: 196-201, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29408762

RESUMO

Polychlorinated biphenyls (PCBs) are ubiquitous environmental contaminants that are associated with varied adverse health effects. Lower chlorinated PCBs are prevalent in indoor and outdoor air and can be metabolized to their hydroxylated derivatives (OH-PCBs) followed by sulfation to form PCB sulfates. Sulfation is also a means of signal termination for steroid hormones. The human estrogen sulfotransferase (SULT1E1) and alcohol/hydroxysteroid sulfotransferase (SULT2A1) catalyze the formation of steroid sulfates that are inactive at steroid hormone receptors. We investigated the inhibition of SULT1E1 (IC50s ranging from 7.2 nM to greater than 10 µM) and SULT2A1 (IC50s from 1.3 µM to over 100 µM) by five lower-chlorinated OH-PCBs and their corresponding PCB sulfates relevant to airborne PCB-exposure. Several congeners of lower chlorinated OH-PCBs relevant to airborne PCB exposures were potent inhibitors of SULT1E1 and SULT2A1 and thus have the potential to disrupt regulation of intracellular concentrations of the receptor-active steroid substrates for these enzymes.


Assuntos
Poluentes Atmosféricos/farmacologia , Bifenilos Policlorados/farmacologia , Sulfatos/farmacologia , Sulfotransferases/antagonistas & inibidores , Estradiol/metabolismo , Humanos , Hidroxilação , Proteínas Recombinantes/metabolismo , Sulfotransferases/metabolismo
18.
Gastrointest Endosc ; 87(4): 1126-1131, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29122598

RESUMO

BACKGROUND AND AIMS: Carbohydrate sulfotransferase 15 (CHST15) promotes tumor growth and invasion and is considered to be an emergent therapeutic target for pancreatic cancer. The aim of this study was to evaluate the safety and efficacy of EUS-guided fine-needle injection (EUS-FNI) of STNM01, the double-stranded RNA oligonucleotide that specifically represses CHST15, for use in patients with pancreatic cancer. METHODS: Six patients with unresectable pancreatic cancer, treated at Tokyo Metropolitan Geriatric Hospital, were used in this open-labeled, investigator-initiated trial. A total of 16 mL STNM01 (250 nM) was injected into the tumor through EUS-FNI. The study's primary endpoint was safety, with a secondary endpoint of tumor response 4 weeks after the initial injection. Some patients received a series of infusions as extensions. The local expression of CHST15 and overall survival (OS) were also evaluated. RESULTS: There were no adverse events. The mean tumor diameter changed from 30.7 to 29.3 mm 4 weeks after injection. Four patients exhibited necrosis of tumor in biopsy specimens. CHST15 was highly expressed at baseline, with 2 patients showing large reductions of CHST15 at week 4. The mean OS of these 2 patients was 15 months, whereas it was 5.7 months for the other 4 patients. CONCLUSIONS: EUS-FNI of STNM01 in pancreatic cancer is safe and feasible. The CHST15 reduction could predict tumor progression and OS. Injections of STNM01 during the beginning of treatment may reduce CHST15 and warrants further investigation.


Assuntos
Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Oligonucleotídeos/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Idoso , Idoso de 80 Anos ou mais , Endossonografia , Feminino , Humanos , Injeções Intralesionais , Masculino , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/metabolismo , Necrose , Oligonucleotídeos/efeitos adversos , Sulfotransferases/antagonistas & inibidores , Sulfotransferases/metabolismo , Taxa de Sobrevida , Carga Tumoral , Ultrassonografia de Intervenção
19.
Sci Rep ; 7(1): 4160, 2017 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-28646214

RESUMO

Sulphotransferases are a diverse group of enzymes catalysing the transfer of a sulfuryl group from 3'-phosphoadenosine 5'-phosphosulphate (PAPS) to a broad range of secondary metabolites. They exist in all kingdoms of life. In Arabidopsis thaliana (L.) Heynh. twenty-two sulphotransferase (SOT) isoforms were identified. Three of those are involved in glucosinolate (Gl) biosynthesis, glycosylated sulphur-containing aldoximes containing chemically different side chains, whose break-down products are involved in stress response against herbivores, pathogens, and abiotic stress. To explain the differences in substrate specificity of desulpho (ds)-Gl SOTs and to understand the reaction mechanism of plant SOTs, we determined the first high-resolution crystal structure of the plant ds-Gl SOT AtSOT18 in complex with 3'-phosphoadenosine 5'-phosphate (PAP) alone and together with the Gl sinigrin. These new structural insights into the determination of substrate specificity were complemented by mutagenesis studies. The structure of AtSOT18 invigorates the similarity between plant and mammalian sulphotransferases, which illustrates the evolutionary conservation of this multifunctional enzyme family. We identified the essential residues for substrate binding and catalysis and demonstrated that the catalytic mechanism is conserved between human and plant enzymes. Our study indicates that the loop-gating mechanism is likely to be a source of the substrate specificity in plants.


Assuntos
Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimologia , Sulfotransferases/química , Sulfotransferases/metabolismo , Difosfato de Adenosina/química , Difosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Proteínas de Arabidopsis/antagonistas & inibidores , Sítios de Ligação , Biocatálise , Cristalografia por Raios X , Análise Mutacional de DNA , Glucosinolatos/química , Cinética , Ligantes , Modelos Biológicos , Mutagênese , Especificidade por Substrato , Sulfotransferases/antagonistas & inibidores
20.
Sci Rep ; 7: 41977, 2017 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-28169314

RESUMO

Defective autophagy and deranged metabolic pathways are common in cancer; pharmacologic targeting of these two pathways could provide a viable therapeutic option. However, how these pathways are regulated by limited availability of growth factors is still unknown. Our study shows that HSulf-1 (endosulfatase), a known tumor suppressor which attenuates heparin sulfate binding growth factor signaling, also regulates interplay between autophagy and lipogenesis. Silencing of HSulf-1 in OV202 and TOV2223 cells (ovarian cancer cell lines) resulted in increased lipid droplets (LDs), reduced autophagic vacuoles (AVs) and less LC3B puncta. In contrast, HSulf-1 proficient cells exhibit more AVs and reduced LDs. Increased LDs in HSulf-1 depleted cells was associated with increased ERK mediated cPLA2S505 phosphorylation. Conversely, HSulf-1 expression in SKOV3 cells reduced the number of LDs and increased the number of AVs compared to vector controls. Furthermore, pharmacological (AACOCF3) and ShRNA mediated downregulation of cPLA2 resulted in reduced LDs, and increased autophagy. Finally, in vivo experiment using OV202 Sh1 derived xenograft show that AACOCF3 treatment effectively attenuated tumor growth and LD biogenesis. Collectively, these results show a reciprocal regulation of autophagy and lipid biogenesis by HSulf-1 in ovarian cancer.


Assuntos
Autofagia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Gotículas Lipídicas/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Sulfotransferases/metabolismo , Animais , Antineoplásicos/farmacologia , Ácidos Araquidônicos/farmacologia , Carboplatina/farmacologia , Combinação de Medicamentos , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Gotículas Lipídicas/efeitos dos fármacos , Camundongos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Fosfolipases A2 Citosólicas/antagonistas & inibidores , RNA Interferente Pequeno/genética , Transdução de Sinais , Sulfotransferases/antagonistas & inibidores , Sulfotransferases/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA