Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Nanobiotechnology ; 22(1): 122, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38504208

RESUMEN

Endocrine therapy is standard for hormone receptor-positive (HR+) breast cancer treatment. However, current strategies targeting estrogen signaling pay little attention to estradiol metabolism in the liver and is usually challenged by treatment failure. In a previous study, we demonstrated that the natural compound naringenin (NAR) inhibited HR+ breast cancer growth by activating estrogen sulfotransferase (EST) expression in the liver. Nevertheless, the poor water solubility, low bio-barrier permeability, and non-specific distribution limited its clinical application, particularly for oral administration. Here, a novel nano endocrine drug NAR-cell penetrating peptide-galactose nanoparticles (NCG) is reported. We demonstrated that NCG presented specific liver targeting and increased intestinal barrier permeability in both cell and zebrafish xenotransplantation models. Furthermore, NCG showed liver targeting and enterohepatic circulation in mouse breast cancer xenografts following oral administration. Notably, the cancer inhibition efficacy of NCG was superior to that of both NAR and the positive control tamoxifen, and was accompanied by increased hepatic EST expression and reduced estradiol levels in the liver, blood, and tumor tissue. Moreover, few side effects were observed after NCG treatment. Our findings reveal NCG as a promising candidate for endocrine therapy and highlight hepatic EST targeting as a novel therapeutic strategy for HR+ breast cancer.


Asunto(s)
Neoplasias de la Mama , Flavanonas , Nanopartículas , Humanos , Ratones , Animales , Femenino , Neoplasias de la Mama/patología , Pez Cebra/metabolismo , Receptores de Estrógenos/metabolismo , Estrógenos/metabolismo , Estrógenos/uso terapéutico , Tamoxifeno/farmacología , Estradiol/farmacología , Hígado/metabolismo
2.
Biochem J ; 477(18): 3583-3598, 2020 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-32686824

RESUMEN

Estrogen sulfotransferase (SULT1E1) metabolically inactivates estrogen and SULT1E1 expression is tightly regulated by multiple nuclear receptors. Human fetal, but not adult, livers express appreciable amounts of SULT1E1 protein, which is mimicked in human hepatoma-derived HepG2 cells cultured in high glucose (450 mg/dl) medium. Here, we have investigated this glucose signal that leads to phosphorylation of nuclear receptor RORα (NR1F1) at Ser100 and the transcription mechanism by which phosphorylated RORα transduces this signal to nuclear receptor HNF4α, activating the SULT1E1 promoter. The promoter is repressed by non-phosphorylated RORα which binds a distal enhancer (-943/-922 bp) and interacts with and represses HNF4α-mediated transcription. In response to high glucose, RORα becomes phosphorylated at Ser100 and reverses its repression of HNF4α promoter activation. Moreover, the casein kinase CK1α, which is identified in an enhancer-bound nuclear protein complex, phosphorylates Ser100 in in vitro kinase assays. During these dynamic processes, both RORα and HNF4α remain on the enhancer. Thus, RORα utilizes phosphorylation to integrate HNF4α and transduces the glucose signal to regulate the SULT1E1 gene in HepG2 cells and this phosphorylation-mediated mechanism may also regulate SULT1E1 expressions in the human liver.


Asunto(s)
Caseína Quinasa Ialfa/metabolismo , Estrógenos/metabolismo , Glucosa/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Transducción de Señal , Sulfotransferasas/metabolismo , Animales , Células COS , Caseína Quinasa Ialfa/genética , Chlorocebus aethiops , Estrógenos/genética , Glucosa/genética , Células Hep G2 , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Humanos , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Fosforilación , Sulfotransferasas/genética
3.
J Am Soc Nephrol ; 31(7): 1496-1508, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32424001

RESUMEN

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.


Asunto(s)
Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/prevención & control , Hígado/metabolismo , Sulfotransferasas/genética , Sulfotransferasas/metabolismo , Lesión Renal Aguda/etiología , Animales , Calcitriol/farmacología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Riñón/metabolismo , Masculino , Ratones , Ratones Noqueados , Orquiectomía , Ovariectomía , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Daño por Reperfusión/complicaciones , Factores Sexuales , Transducción de Señal , Sulfotransferasas/antagonistas & inhibidores , Triclosán/farmacología
4.
J Pept Sci ; 26(10): e3274, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32633098

RESUMEN

Estrogen sulfotransferase (SULT1E) mainly catalyzes the sulfation of estrogens, which are known to prevent the pathogenesis of atherosclerosis. Recently, we found that peptides with a YKDG sequence specifically bind to oxidized low-density lipoprotein (Ox-LDL), which plays a major role in the pathogenesis of atherosclerosis. Here, we investigated the interaction between human SULT1E1 (hSULT1E1), which has a YKEG sequence (residues 61-64) unlike other human SULTs, and Ox-LDL. Results from polyacrylamide gel electrophoresis and western blotting demonstrated that hSULT1E1 specifically binds to Ox-LDL and its major lipid component (lysophosphatidylcholine; LPC), and platelet-activating factor (PAF), which bears a marked resemblance to LPC in terms of structure and activity. Moreover, an N-terminally fluorescein isothiocyanate (FITC)-labeled decapeptide (MIYKEGDVEK; FITC-hSULT1E1-P10) corresponding to residues 59-68 of hSULT1E1 specifically binds to Ox-LDL, LPC, and PAF. Unveiling the specific interaction between hSULT1E1 and Ox-LDL, LPC, and PAF provides important information regarding the mechanisms underlying various diseases caused by Ox-LDL, LPC, and PAF, such as atherosclerosis. In addition, FITC-hSULT1E1-P10 could be used as an efficient fluorescent probe for the detection of Ox-LDL, LPC, and PAF, which could facilitate the mechanistic study, identification, diagnosis, prevention, and treatment of atherosclerosis.


Asunto(s)
Colorantes Fluorescentes/química , Isotiocianatos/química , Lipoproteínas LDL/química , Oligopéptidos/química , Sulfotransferasas/química , Secuencia de Aminoácidos , Electroforesis en Gel de Poliacrilamida , Colorantes Fluorescentes/metabolismo , Humanos , Isotiocianatos/metabolismo , Lipoproteínas LDL/metabolismo , Estructura Molecular , Oligopéptidos/síntesis química , Oligopéptidos/metabolismo , Sulfotransferasas/metabolismo
5.
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
6.
J Biol Chem ; 290(23): 14754-64, 2015 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-25922074

RESUMEN

Estrogen sulfotransferase (EST) regulates estrogen homeostasis by sulfonating and deactivating estrogens. Liver ischemia and reperfusion (I/R) involves both hypoxia during the ischemic phase and oxidative damage during the reperfusion phase. In this report, we showed that the expression of EST was markedly induced by I/R. Mechanistically, oxidative stress-induced activation of Nrf2 was responsible for the EST induction, which was abolished in Nrf2(-/-) mice. EST is a direct transcriptional target of Nrf2. In female mice, the I/R-responsive induction of EST compromised estrogen activity. EST ablation attenuated I/R injury as a result of decreased estrogen deprivation, whereas this benefit was abolished upon ovariectomy. The effect of EST ablation was sex-specific because the EST(-/-) males showed heightened I/R injury. Reciprocally, both estrogens and EST regulate the expression and activity of Nrf2. Estrogen deprivation by ovariectomy abolished the I/R-responsive Nrf2 accumulation, whereas the compromised estrogen deprivation in EST(-/-) mice was associated with increased Nrf2 accumulation. Our results suggested a novel I/R-responsive feedback mechanism to limit the activity of Nrf2 in which Nrf2 induces the expression of EST, which subsequently increases estrogen deactivation and limits the estrogen-responsive activation of Nrf2. Inhibition of EST, at least in females, may represent an effective approach to manage hepatic I/R injury.


Asunto(s)
Hígado/patología , Estrés Oxidativo , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Sulfotransferasas/genética , Animales , Células Cultivadas , Estrógenos/metabolismo , Femenino , Eliminación de Gen , Células Hep G2 , Humanos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Daño por Reperfusión/metabolismo , Factores Sexuales , Sulfotransferasas/metabolismo , Regulación hacia Arriba
7.
FASEB J ; 27(9): 3860-70, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23756648

RESUMEN

Environmental contaminants are suspected to be involved in the epidemic incidence of metabolic disorders, food ingestion being a primarily route of exposure. We hypothesized that life-long consumption of a high-fat diet that contains low doses of pollutants will aggravate metabolic disorders induced by obesity itself. Mice were challenged from preconception throughout life with a high-fat diet containing pollutants commonly present in food (2,3,7,8-tetrachlorodibenzo-p-dioxin, polychlorinated biphenyl 153, diethylhexyl phthalate, and bisphenol A), added at low doses in the tolerable daily intake range. We measured several blood parameters, glucose and insulin tolerance, hepatic lipid accumulation, and gene expression in adult mice. Pollutant-exposed mice exhibited significant sex-dependent metabolic disorders in the absence of toxicity and weight gain. In males, pollutants increased the expression of hepatic genes (from 36 to 88%) encoding proteins related to cholesterol biosynthesis and decreased (40%) hepatic total cholesterol levels. In females, there was a marked deterioration of glucose tolerance, which may be related to the 2-fold induction of estrogen sulfotransferase and reduced expression of estrogen receptor α (25%) and estrogen target genes (>34%). Because of the very low doses of pollutants used in the mixture, these findings may have strong implications in terms of understanding the potential role of environmental contaminants in food in the development of metabolic diseases.


Asunto(s)
Hígado/efectos de los fármacos , Hígado/metabolismo , Animales , Compuestos de Bencidrilo/toxicidad , Western Blotting , Peso Corporal/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Fenoles/toxicidad , Bifenilos Policlorados/toxicidad , Dibenzodioxinas Policloradas/toxicidad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Protein Pept Lett ; 30(10): 821-829, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37724676

RESUMEN

Estrogen plays a key role in the development and progression of many malignant tumours, and the regulation of estrogen levels involves several metabolic pathways. Among these pathways, estrogen sulfotransferase (SULT1E1) is the enzyme with the most affinity for estrogen and is primarily responsible for catalysing the metabolic reaction of estrogen sulphation. Relevant studies have shown significant differences in the expression of SULT1E1 in different malignant tumours, suggesting that SULT1E1 plays a dual role in malignant tumours, both inhibiting the growth of malignant tumours and promoting their development. In addition, the expression level of SULT1E1 may be regulated by a variety of factors, which in turn affect the growth and therapeutic effects of malignant tumours. The aim of this paper is to review the mechanism of action of SULT1E1 in malignant tumours and the mechanisms that are regulated, in order to provide potential targets for the treatment of malignant tumour patients in the future and theoretical support for the realisation of more personalised and effective therapeutic regimens.


Asunto(s)
Estrógenos , Neoplasias , Humanos , Estrógenos/metabolismo , Sulfotransferasas/genética , Sulfotransferasas/metabolismo
9.
Protein J ; 41(1): 179-188, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35059953

RESUMEN

Cytosolic estrogen sulfotransferase (SULT1E1) mainly catalyzes the sulfoconjugation and deactivation of estrogens that are known to exert potent anti-atherogenic effects. However, it remains unknown about the connection between SULT1E1 and atherosclerosis. Recently, we reported that SULT1E1 is highly expressed in the aorta with plaques of high fat-fed ApoE knockout (KO) mice (mouse model of atherosclerosis), and interacts with oxidized low-density lipoprotein (Ox-LDL) known as a major component of atherosclerotic lesions. In this study, immunohistochemical staining for SULT1E1 in the aorta of high fat-fed ApoE KO mice showed that SULT1E1 is detected in vascular endothelial cells overlying atherosclerotic plaques. Results from Western blotting showed that Ox-LDL induces the protein expression of both SULT1E1 and peroxisome proliferator-activated receptor (PPAR) γ in human umbilical vein endothelial cells (HUVECs), and then that a PPARγ antagonist GW9662, but not a PPARα antagonist GW6471, inhibited the protein expression of SULT1E1 induced by Ox-LDL. Moreover, GW9662 significantly increased the proliferation of HUVECs induced by Ox-LDL. Our results suggest that SULT1E1 and PPARγ, both of which are increased by Ox-LDL, may interact with each other, and then may reduce cooperatively Ox-LDL-induced proliferation of vascular endothelial cells overlying atherosclerotic plaques, leading to against atherosclerosis.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Sulfotransferasas , Animales , Aterosclerosis/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , PPAR gamma/metabolismo , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Sulfotransferasas/genética , Sulfotransferasas/metabolismo
10.
Methods Mol Biol ; 2418: 41-51, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35119658

RESUMEN

Estrogen receptor α (ERα) conserves a phosphorylation motif at Serine 216. This site constitutes a protein kinase C phosphorylation motif located within the DNA binding domain (DBD) of ERα. The liver plays a critical role in the regulation of metabolism of various xenobiotics, fatty acids, and cholesterol or endogenous compounds. Moreover, numerous metabolizing enzymes are mainly expressed in the liver. In this chapter, we describe several practical experimental procedures confirming that mouse ERα is phosphorylated at serine 216 in livers upon phenobarbital (PB) treatment. Also, this phosphorylation regulates the expression of estrogen sulfotransferase gene (SULT1E1) which has an important role to sulfate and inactivate estrogen. In response to PB, the conserved motif within the DBD activates the Sult1e1 gene. When this motif was mutated, the activation of Sult1e1 was suppressed significantly. This chapter also describes the use of a phospho-peptide antibody (αP-S216) in the chromatin immunoprecipitation (ChIP) assay, and the co-immunoprecipitation (Co-IP) assay visualized by Western blot analysis.


Asunto(s)
Receptor alfa de Estrógeno , Serina , Animales , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Inmunoprecipitación , Hígado/metabolismo , Ratones , Fosforilación/fisiología , Serina/metabolismo
11.
Biochem Pharmacol ; 190: 114662, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34157297

RESUMEN

Circulating estrogens levels significantly decrease in menopause and levels off in postmenopausal women. Accordingly, the liver represses levels of enzymes and membrane transporters, thereby decreasing capability of inactivating and excreting estrogens. Women increasingly develop type 2 diabetes during or after menopause. Estrogens are known to promote liver diseases in these women. Here, we have found that the estrogen inactivating sulfotransferase (SULT1E1) and an ATP-binding cassette subfamily G member 2 (ABCG2), a gene encoding breast cancer resistance protein that exports sulfated estrogens, increased their expression levels in diabetic women but not men. For the sulfotransferase gene, phosphorylated nuclear receptors ERα and RORα, at Ser212 and Ser100, respectively, bind their response elements to activate the SULT1E1 promoter in women. This coordinated increase in estrogen inactivation and excretion, and the phosphorylated nuclear receptor-mediated gene activation could be a defense mechanism against toxicities of estrogens through inactivation and excretion in the livers of women.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Receptor alfa de Estrógeno/metabolismo , Proteínas de Neoplasias/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Sulfotransferasas/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Adulto , Anciano , Animales , Células COS , Chlorocebus aethiops , Receptor alfa de Estrógeno/genética , Femenino , Regulación de la Expresión Génica , Humanos , Hígado , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Regiones Promotoras Genéticas , Unión Proteica , Factores Sexuales , Sulfotransferasas/genética
12.
Andrology ; 9(2): 657-664, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33290605

RESUMEN

BACKGROUND: Decreased testosterone (T) to LH ratio and increased 17ß-estradiol (E2) serum concentrations represent a common finding among patients with severe spermatogenic failure, suggesting a concurrent Leydig cell steroidogenic dysfunction. Aromatase overexpression has been associated with increased serum and intratesticular E2 in these patients. However, it is unknown whether the sulfatase pathway contributes to the increased availability of active estrogens in patients with primary spermatogenic failure. OBJECTIVES: To assess estrogen sulfotransferase (SULT1E1) and steroid sulfatase (STS) mRNA abundance in testicular tissue of patients with Sertoli cell-only syndrome (SCOS) and normal tissues, its association with serum and intratesticular hormone levels, and to explore the mRNA and protein testicular localization of both enzymes. MATERIALS AND METHODS: Testicular tissues of 23 subjects with SCOS (cases) and 22 patients with obstructive azoospermia and normal spermatogenesis (controls) were obtained after biopsy. SULT1E1 and STS transcripts accumulation was quantified by RT-qPCR. For mRNA and protein localization, we performed RT-qPCR in Leydig cell clusters and seminiferous tubules isolated by laser-capture microdissection and immunofluorescence in testicular tissues. Serum and intratesticular hormones were measured by immunoradiometric assays. RESULTS: SULT1E1 mRNA accumulation was similar in both groups. The amount of STS mRNA was higher in cases (p = 0.007) and inversely correlated with T/LH ratio (r = -0.402; p = 0.02). Also, a near significant correlation was observed with intratesticular E2 (r = 0.329, p = 0.057), in agreement with higher intratesticular E2 in cases (p < 0.001). Strong STS immunoreaction was localized in the wall of small blood vessels but not in Leydig cells. Both SULT1E1 and STS mRNA abundance was similar in Leydig cell clusters and the tubular compartment, except for lower SUTL1E1 mRNA in the seminiferous tubules of SCOS patients (p = 0.001). CONCLUSIONS: Our results suggest that an unbalance of the STS/SULT1E1 pathway contributes to the testicular hyperestrogenic microenvironment in patients with primary spermatogenic failure and Leydig cell dysfunction.


Asunto(s)
Células Intersticiales del Testículo , Síndrome de Sólo Células de Sertoli/enzimología , Esteril-Sulfatasa/metabolismo , Testículo/enzimología , Adulto , Azoospermia/enzimología , Azoospermia/genética , Azoospermia/fisiopatología , Microambiente Celular , Hormonas Esteroides Gonadales/sangre , Humanos , Masculino , ARN Mensajero , Síndrome de Sólo Células de Sertoli/genética , Síndrome de Sólo Células de Sertoli/metabolismo , Síndrome de Sólo Células de Sertoli/fisiopatología , Espermatogénesis , Esteril-Sulfatasa/genética , Sulfotransferasas/genética , Sulfotransferasas/metabolismo
13.
Pathol Oncol Res ; 27: 589649, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34257538

RESUMEN

Sex-specific differences in the incidence of urinary bladder carcinomas are well known, and the possible involvement of sex steroids has been proposed. We previously reported the association of the loss of androgen receptors and androgen-producing enzymes with tumor progression of urinary bladder cancer patients. Clinically, the selective estrogen receptor modulators (SERMs) were reported to suppress the progression of these tumors but the status of estrogen receptors (ERs) has not been well studied in patients with bladder urinary cancer. Moreover, not only ERs but also estrogen-related enzymes, such as aromatase, steroid sulfatase (STS), and estrogen sulfotransferase (EST), have been reported in the biological/clinical behavior of various hormone-dependent carcinomas but not studied in urinary bladder carcinoma. Therefore, in this study, we immunolocalized ERs as well as estrogen metabolizing enzymes in urinary bladder carcinoma and performed immunoblotting and cell proliferation assays using the bladder urothelial carcinoma cell line, T24. The results revealed that the loss of STS and aromatase was significantly correlated with advanced stages of the carcinoma. In vitro studies also revealed that T24 cell proliferation rates were significantly ameliorated after treatment with estradiol or diarylpropionitrile (DPN). EST and aromatase were also significantly correlated with the nuclear grade of the carcinoma. The results of our present study, for the first time, demonstrated that biologically active estrogens that bind to ERs could suppress tumor progression and the inactive ones could promote its progression and the potential clinical utility of SERM treatment in selective patients with urinary bladder carcinoma.


Asunto(s)
Carcinoma de Células Transicionales/metabolismo , Receptor beta de Estrógeno/metabolismo , Estrógenos/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo , Aromatasa/metabolismo , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Transicionales/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Estradiol/farmacología , Receptor beta de Estrógeno/agonistas , Femenino , Humanos , Nitrilos/farmacología , Propionatos/farmacología , Esteril-Sulfatasa/metabolismo , Sulfotransferasas/metabolismo , Neoplasias de la Vejiga Urinaria/patología
14.
Protein J ; 40(2): 192-204, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33665770

RESUMEN

Cytosolic estrogen sulfotransferase (SULT1E) mainly catalyzes the sulfate conjugation of estrogens, which decrease atherosclerosis progression. Recently we reported that a YKEG sequence in human SULT1E1 (hSULT1E1) corresponding to residues 61-64 can bind specifically to oxidized low-density lipoprotein (Ox-LDL), which plays a major role in the pathogenesis of atherosclerosis; its major oxidative lipid component lysophosphatidylcholine (LPC), and its structurally similar lipid, platelet-activating factor (PAF). In this study, we investigated the effect of Ox-LDL on the sulfating activity of hSULT1E1. In vivo experiments using a mouse model of atherosclerosis showed that the protein expression of SULT1E1 was higher in the aorta of mice with atherosclerosis compared with that in control animals. Results from a sulfating activity assay of hSULT1E1 using 1-hydroxypyrene as the substrate demonstrated that Ox-LDL, LPC, and PAF markedly decreased the sulfating activity of hSULT1E1, whereas native LDL and 1-palmitoyl-2-(5'-oxo-valeroyl)-sn-glycero-3-phosphocholine (POVPC) as one of the oxidized phosphatidylcholines showed the opposite effect. The sulfating activity greatly changed in the presence of LPC, PAF, and POVPC in their concentration-dependen manner (especially above their critical micelle concentrations). Moreover, Ox-LDL specifically recognized dimeric hSULT1E1. These results suggest that the effects of Ox-LDL and native LDL on the sulfating activity of hSULT1E1 might be helpful in elucidating the novel mechanism underlying the pathogenesis of atherosclerosis, involving the relationship between estrogen metabolism, LDL, and Ox-LDL.


Asunto(s)
Lipoproteínas LDL , Sulfotransferasas , Animales , Aterosclerosis , Humanos , Lipoproteínas LDL/química , Lipoproteínas LDL/metabolismo , Lisofosfatidilcolinas/metabolismo , Masculino , Ratones , Ratones Noqueados , Factor de Activación Plaquetaria/metabolismo , Unión Proteica , Sulfotransferasas/química , Sulfotransferasas/metabolismo
15.
J Pers Med ; 11(3)2021 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-33799763

RESUMEN

Estrogen sulfotransferase (SULT1E1) is a phase II enzyme that sulfates estrogens to inactivate them and regulate their homeostasis. This enzyme is also involved in the sulfation of thyroid hormones and several marketed medicines. Though the profound action of SULT1E1 in molecular/pathological biology has been extensively studied, its genetic variants and functional studies have been comparatively rarely studied. Genetic variants of this gene are associated with some diseases, especially sex-hormone-related cancers. Comprehending the role and polymorphisms of SULT1E1 is crucial to developing and integrating its clinical relevance; therefore, this study gathered and reviewed various literature studies to outline several aspects of the function, molecular regulation, and polymorphisms of SULT1E1.

16.
Endocrinology ; 161(1)2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31837219

RESUMEN

Hemorrhagic shock (HS) is a potential life-threatening condition that may lead to injury to multiple organs, including the lung. The estrogen sulfotransferase (EST, or SULT1E1) is a conjugating enzyme that sulfonates and deactivates estrogens. In this report, we showed that the expression of Est was markedly induced in the liver but not in the lung of female mice subject to HS and resuscitation. Genetic ablation or pharmacological inhibition of Est effectively protected female mice from HS-induced acute lung injury (ALI), including interstitial edema, neutrophil mobilization and infiltration, and inflammation. The pulmonoprotective effect of Est ablation or inhibition was sex-specific, because the HS-induced ALI was not affected in male Est-/- mice. Mechanistically, the pulmonoprotective phenotype in female Est-/- mice was accompanied by increased lung and circulating levels of estrogens, attenuated pulmonary inflammation, and inhibition of neutrophil mobilization from the bone marrow and neutrophil infiltration to the lung, whereas the pulmonoprotective effect was abolished upon ovariectomy, suggesting that the protection was estrogen dependent. The pulmonoprotective effect of Est ablation was also tissue specific, as loss of Est had little effect on HS-induced liver injury. Moreover, transgenic reconstitution of human EST in the liver of global Est-/- mice abolished the pulmonoprotective effect, suggesting that it is the EST in the liver that sensitizes mice to HS-induced ALI. Taken together, our results revealed a sex- and tissue-specific role of EST in HS-induced ALI. Pharmacological inhibition of EST may represent an effective approach to manage HS-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda/patología , Choque Hemorrágico/complicaciones , Sulfotransferasas/metabolismo , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/prevención & control , Animales , Estrógenos/metabolismo , Femenino , Hígado/enzimología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Resucitación , Factores Sexuales , Choque Hemorrágico/terapia
17.
Biochem Pharmacol ; 180: 114197, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32798464

RESUMEN

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.


Asunto(s)
Ayuno/metabolismo , Glucosa/administración & dosificación , Hígado/metabolismo , Receptor X de Pregnano/metabolismo , Serina/metabolismo , Sulfotransferasas/biosíntesis , Animales , Células COS , Chlorocebus aethiops , Femenino , Células Hep G2 , Humanos , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación/fisiología , Receptor X de Pregnano/química , Receptor X de Pregnano/genética , Estructura Secundaria de Proteína , Serina/genética , Sulfotransferasas/antagonistas & inhibidores , Sulfotransferasas/genética
18.
Expert Opin Drug Metab Toxicol ; 15(4): 329-339, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30822161

RESUMEN

INTRODUCTION: Biotransformation is important in the metabolism of endobiotics and xenobiotics. This process comprises the activity of phase I and phase II enzymes. Estrogen sulfotransferase (SULT1E1 or EST) is a phase II conjugating enzyme that belongs to the family of cytosolic sulfotransferases. The expression of SULT1E1 can be detected in many tissues, including the liver. SULT1E1 catalyzes the transfer of a sulfate group from 3'-phosphoadenosine-5'-phosphosulfate (PAPS) to any available hydroxyl group in estrogenic molecules. The substrates of SULT1E1 include the endogenous and synthetic estrogens. Upon SULT1E1-mediated sulfation, the hydrosolubility of estrogens increases, preventing the binding between the sulfated estrogens and the estrogen receptor (ER). This sulfated state of the estrogens is not irreversible, as the steroid sulfatase (STS) can convert sulfoconjugated estrogens to free estrogens. The expression of SULT1E1 is inducible by several diseases that involve tissue inflammation, such as type 2 diabetes, sepsis, and ischemia-reperfusion injury. Areas covered: This systematic literature review aims to summarize the role of SULT1E1 in the metabolism of estrogenic drugs and xenobiotics, and the role of SULT1E1 in the pathogenesis of several diseases, including cancer, metabolic disease, sepsis, liver injury, and cystic fibrosis. Meanwhile, ablation or pharmacological inhibition of SULT1E1 can affect the outcomes of the aforementioned diseases. Expert opinion: In addition to its role in metabolizing estrogenic drugs, SULT1E1 is unexpectedly being unveiled as a mediator for the disease effect on estrogen metabolism and homeostasis. Meanwhile, because the expression and activity of SULT1E1 can affect the outcome of diseases, the same sulfotransferase and the reversing enzymes STS can be potential therapeutic targets to prevent or manage diseases. Accumulating evidence suggest that the physiological and pathophysiological effects of SULT1E1 can be estrogen-independent and it is necessary to elucidate what other possible substrates may be recognized by the enzyme. Moreover, human studies are paramount to confirm the human relevance of the animal studies.


Asunto(s)
Estrógenos/metabolismo , Sulfotransferasas/metabolismo , Xenobióticos/metabolismo , Animales , Citosol/enzimología , Citosol/metabolismo , Regulación Enzimológica de la Expresión Génica , Humanos , Hígado/enzimología , Hígado/metabolismo , Sulfotransferasas/genética
19.
Biomed J Sci Tech Res ; 22(5): 16960-16967, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-36312461

RESUMEN

Estrogens are known for their proliferative effects, resulting in tumorigenesis and causing cancers. The majority of breast cancers are estrogen-dependent. Estrogenb-locking drugs developed for treating estrogen-dependent breast cancers include antagonists of estrogen receptors (ERs) and inhibitors of estrogen synthesis enzymes such as aromatase and steroid sulfatase (STS). However, drugs targeting estrogen inactivation enzyme, estrogen sulfotransferase (SULT1E1), have not been developed for estrogen-dependent cancer treatment or prevention. Estrogens must be inactivated after their actions in vivo by SULT1E1, uncontrolled estrogen activity in certain tissues will cause cancers. The majority of human breast cancer cell lines are known to express very low levels of SULT1E1 compared to normal mammary cells. Therefore, gene up-regulation of SULT1E1 could provide novel possibilities for the treatment of estrogen-dependent breast and endometrial cancers. Our data suggest that certain nutritional flavonoids induce SULT1E1 and inhibit cell proliferation in estrogen-dependent breast cancer MCF-7 cells. Our results also suggest that SULT1E1 inducer has an additive or synergistic effect on the inhibition of MCF-7 cell proliferation when combined with other known breast cancer drugs. Naturally occurring flavonoids are safe and inexpensive; they could be used for the prevention of certain breast cancers and/or used as co-drug for the treatment of estrogen-dependent cancers. These results may lead to creating innovative approaches for the treatment or prevention of estrogen-dependent cancers and may lead to the discovery of novel drugs or co-drugs.

20.
Mol Cell Endocrinol ; 489: 45-65, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30326245

RESUMEN

Endometrial cancer (EC) is the most common malignancy of the female gynaecological tract and increased exposure to estrogens is a risk factor. EC cells are able to produce estrogens locally using precursors like, among others, adrenal steroids present in the serum. This is referred to as local estrogen metabolism (or intracrinology) and consists of a complex network of multiple enzymes. Particular relevant to the final generation of active estrogens in endometrial cells are: steroid sulfatase (STS), estrogen sulfotransferase (SULT1E1), aromatase (CYP19A1), 17ß-hydroxysteroid dehydrogenase (HSD17B) type 1 and type 2. During the last decades, a plethora of studies explored the level of these enzymes in EC but contrasting data were reported, which generated vigorous debate and controversies. Several reviews attempted at clarifying some of the debated issues, but published reviews are based on investigator-defined bibliography selection and not on systematic analysis. Therefore, we performed a systematic review of the literature reporting about the level of STS, SULT1E1, CYP19A1, HSD17B1 and HSD17B2 in EC. Additional intracrine enzymes and networks (e.g., HSD17Bs other than types 1 and 2, aldo-keto reductases, progesterone and androgen metabolism) were non-systematically reviewed as well.


Asunto(s)
Neoplasias Endometriales/metabolismo , Estrógenos/metabolismo , 17-Hidroxiesteroide Deshidrogenasas/metabolismo , Aromatasa/metabolismo , Neoplasias Endometriales/diagnóstico , Neoplasias Endometriales/terapia , Femenino , Humanos , Transducción de Señal , Esteril-Sulfatasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA