Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 533(3): 458-466, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-32977942

RESUMO

Bile acids (BA) play a vital physiological role in vivo. They are not only detergent of dietary lipids and nutrients, but also important hormones or nutrient signaling molecules in metabolic regulation process. Recent studies have also shown BA involvement in various cancers and diseases such as Parkinson's and Alzheimer's and liver diseases. However, majority of the reported literature about BA is restricted to enterohepatic circulation. Hitherto, there has been no comprehensive study of the BA profile in all the major tissue and biofluids in rat has been reported. In this first bileomics study, BA profile of 14 different rat biological specimens (liver, serum, kidney, heart, stomach, ovary, mammary, uterus, small intestine, big intestine, spleen, brain, feces and urine) were studied by ultra-performance liquid chromatography (UPLC)-tandem mass spectrometry (MS/MS). Here I report the comprehensive identification and measurements of bile acids, the bileome, in rat. PCA analysis show distinct separate clusters of tissues as well as biofluids based on BA composition profile. Furthermore, we found that BA profiles of the organs that are involved in enterohepatic circulation were different than the other organs. Most of BA in brain, spleen, heart, ovary, urine, feces and uterus were in the unamidated form, and LCA and MOCA are the most abundant BAs in these organs. Whereas, most of BAs in liver, serum, mammary, large intestine, small intestine, stomach and kidney existed in amidated form, and TCA and T-ß-MCA are primary BAs. Finally, first time, BAs are found and measured in kidney, heart, stomach, ovary, mammary, uterus, and spleen of rats.


Assuntos
Ácidos e Sais Biliares/metabolismo , Animais , Ácidos e Sais Biliares/química , Feminino , Metaboloma , Ratos , Ratos Wistar , Distribuição Tecidual
2.
Am J Physiol Endocrinol Metab ; 313(5): E528-E539, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28698282

RESUMO

Male hypogonadism results in changes in body composition characterized by increases in fat mass. Resident immune cells influence energy metabolism in adipose tissue and could promote increased adiposity through paracrine effects. We hypothesized that manipulation of circulating sex steroid levels in healthy men would alter adipose tissue immune cell populations. Subjects (n = 44 men, 19-55 yr of age) received 4 wk of treatment with the gonadotropin-releasing hormone receptor antagonist acyline with daily administration of 1) placebo gel, 2) 1.25 g testosterone gel (1.62%), 3) 5 g testosterone gel, or 4) 5 g testosterone gel with an aromatase inhibitor. Subcutaneous adipose tissue biopsies were performed at baseline and end-of-treatment, and adipose tissue immune cells, gene expression, and intra-adipose estrogen levels were quantified. Change in serum total testosterone level correlated inversely with change in the number of CD3+ (ß = -0.36, P = 0.04), CD4+ (ß = -0.34, P = 0.04), and CD8+ (ß = -0.33, P = 0.05) T cells within adipose tissue. Change in serum 17ß-estradiol level correlated inversely with change in the number of adipose tissue macrophages (ATMs) (ß = -0.34, P = 0.05). A negative association also was found between change in serum testosterone and change in CD11c+ ATMs (ß = -0.41, P = 0.01). Overall, sex steroid deprivation was associated with increases in adipose tissue T cells and ATMs. No associations were found between changes in serum sex steroid levels and changes in adipose tissue gene expression. Circulating sex steroid levels may regulate adipose tissue immune cell populations. These exploratory findings highlight a possible novel mechanism that could contribute to increased metabolic risk in hypogonadal men.


Assuntos
Tecido Adiposo/citologia , Tecido Adiposo/imunologia , Hormônios Esteroides Gonadais/fisiologia , Imunidade Celular/fisiologia , Adulto , Inibidores da Aromatase/farmacologia , Antígeno CD11c/metabolismo , Complexo CD3/metabolismo , Antígenos CD4/metabolismo , Estradiol/farmacologia , Regulação da Expressão Gênica , Hormônios Esteroides Gonadais/sangue , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Receptores LHRH/antagonistas & inibidores , Linfócitos T/imunologia , Testosterona/sangue , Testosterona/farmacologia , Adulto Jovem
3.
J Biol Chem ; 290(23): 14754-64, 2015 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-25922074

RESUMO

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.


Assuntos
Fígado/patologia , Estresse Oxidativo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Sulfotransferases/genética , Animais , Células Cultivadas , Estrogênios/metabolismo , Feminino , Deleção de Genes , Células Hep G2 , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Traumatismo por Reperfusão/metabolismo , Fatores Sexuais , Sulfotransferases/metabolismo , Regulação para Cima
4.
J Hepatol ; 64(1): 44-52, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26220752

RESUMO

BACKGROUND & AIMS: Chronic inflammatory liver diseases are associated with estrogen excess and feminization in men, which is thought to be due to compromised liver function to break down estrogens. The goal of this study is to determine whether the inflammatory induction of steroid sulfatase (STS), which converts inactive estrogen sulfates to active estrogens, may have contributed to the estrogen excess in chronic liver disease. METHODS: We performed bioinformatic analysis, real-time PCR, immunohistochemistry, and UPLC/MS-MS to analyze hepatic STS expression and serum estrogen levels in patients with chronic liver diseases. The crosstalk between NF-κB pathway and STS-regulated estrogen signaling was investigated by electrophoretic mobility shift assay, chromatin immunoprecipitation, luciferase assay and gene knockdown experiments in human hepatocytes. RESULTS: Hepatic STS was induced in patients with chronic inflammatory liver diseases, which was accompanied by increased circulating estrogen levels. The human STS gene, but not the mouse Sts gene, was induced by inflammatory stimuli in hepatic cells. Mechanistically, STS was established as a novel NF-κB target gene, whose induction facilitated the conversion of inactive estrogen sulfates to active estrogens, and consequently attenuated the inflammatory response. In contrast, genetic or pharmacological inhibition of STS or a direct blockade of estrogen signaling sensitized liver cells to the transcriptional activation of NF-κB and inflammatory response, possibly through the inhibition of IκB kinase activation. CONCLUSIONS: Our results suggest a negative feedback loop in chronic inflammatory liver diseases, in which the inflammatory activation of NF-κB induces STS gene expression. The induced STS facilitates the conversion of inactive estrogen sulfates to active estrogens, which in return attenuates the NF-κB-mediated inflammation.


Assuntos
Estrogênios/metabolismo , Homeostase , Inflamação/etiologia , Hepatopatias/metabolismo , Esteril-Sulfatase/fisiologia , Células Cultivadas , Doença Crônica , Biologia Computacional , Humanos , Cirrose Hepática Alcoólica/metabolismo , NF-kappa B/fisiologia , Transdução de Sinais
5.
J Biol Chem ; 289(12): 8086-97, 2014 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-24497646

RESUMO

The steroid sulfatase (STS)-mediated desulfation is a critical metabolic mechanism that regulates the chemical and functional homeostasis of endogenous and exogenous molecules. In this report, we first showed that the liver expression of Sts was induced in both the high fat diet (HFD) and ob/ob models of obesity and type 2 diabetes and during the fed to fasting transition. In defining the functional relevance of STS induction in metabolic disease, we showed that overexpression of STS in the liver of transgenic mice alleviated HFD and ob/ob models of obesity and type 2 diabetes, including reduced body weight, improved insulin sensitivity, and decreased hepatic steatosis and inflammation. Interestingly, STS exerted its metabolic benefit through sex-specific mechanisms. In female mice, STS may have increased hepatic estrogen activity by converting biologically inactive estrogen sulfates to active estrogens and consequently improved the metabolic functions, whereas ovariectomy abolished this protective effect. In contrast, the metabolic benefit of STS in males may have been accounted for by the male-specific decrease of inflammation in white adipose tissue and skeletal muscle as well as a pattern of skeletal muscle gene expression that favors energy expenditure. The metabolic benefit in male STS transgenic mice was retained after castration. Treatment with the STS substrate estrone sulfate also improved metabolic functions in both the HFD and ob/ob models. Our results have uncovered a novel function of STS in energy metabolism and type 2 diabetes. Liver-specific STS induction or estrogen/estrogen sulfate delivery may represent a novel approach to manage metabolic syndrome.


Assuntos
Diabetes Mellitus Tipo 2/genética , Fígado/enzimologia , Obesidade/genética , Esteril-Sulfatase/genética , Regulação para Cima , Animais , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/patologia , Metabolismo Energético , Estrogênios/metabolismo , Fígado Gorduroso/enzimologia , Fígado Gorduroso/genética , Fígado Gorduroso/patologia , Feminino , Resistência à Insulina , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Transgênicos , Obesidade/enzimologia , Obesidade/patologia , Esteril-Sulfatase/metabolismo
6.
Proc Natl Acad Sci U S A ; 108(21): 8595-600, 2011 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-21555564

RESUMO

Mesoamerican peoples had a long history of cacao use--spanning more than 34 centuries--as confirmed by previous identification of cacao residues on archaeological pottery from Paso de la Amada on the Pacific Coast and the Olmec site of El Manatí on the Gulf Coast. Until now, comparable evidence from San Lorenzo, the premier Olmec capital, was lacking. The present study of theobromine residues confirms the continuous presence and use of cacao products at San Lorenzo between 1800 and 1000 BCE, and documents assorted vessels forms used in its preparation and consumption. One elite context reveals cacao use as part of a mortuary ritual for sacrificial victims, an event that occurred during the height of San Lorenzo's power.


Assuntos
Arqueologia/métodos , Cacau/história , Teobromina/análise , América , Cerâmica , Comportamento Ritualístico , História Antiga , Humanos , Teobromina/história
7.
Anal Chem ; 85(10): 4951-60, 2013 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-23597399

RESUMO

In humans, steroids play a broad and vital role in regulation of gene expression, secondary sexual characteristics, maturation, reproduction, cardiovascular health, neurological functions, etc., but imbalance in steroid metabolism is also linked to development and progression of many diseases, such as cancer, neurodegenerative diseases, and cardiovascular diseases. Hence, measurement of steroids in biological samples is essential to monitor human health. Currently, there is radioimmunoassay, gas chromatography-mass spectrometry (GC/MS), and liquid chromatography-mass spectrometry (LC-MS) methods developed for steroid measurements in biological samples. However, these methods require elaborate sample preparation procedures and have concerns(s) related to reproducibility, dynamic range, time, costs, and most importantly the total coverage of steroids. Also currently, there is no method available for comprehensive steroid profiling in a single LC-MS run that includes androgens, corticosteroids, progestogens, estrogens, estrogen metabolites, estrogen conjugates, and estrogen-DNA adducts as well as exogenous steroid derivatives. Here, I present a global steroid metabolic profiling method based on liquid-liquid extraction (LLE) followed by ultra performance liquid chromatography (UPLC)-tandem mass spectrometry (MS/MS) for simultaneous measurement of over 100 indigenous as well as exogenous steroids in about 12 min, without derivatization. The method was successfully applied to determine steroid hormone levels in the breast tissue of healthy women. Overall presence of all major classes of steroids as well as estrogen derivatives was detected in breast tissue.


Assuntos
Mama/citologia , Cromatografia Líquida de Alta Pressão/métodos , Metabolômica/métodos , Esteroides/metabolismo , Espectrometria de Massas em Tandem/métodos , Adulto , Mama/metabolismo , Feminino , Humanos , Extração Líquido-Líquido , Pessoa de Meia-Idade , Reprodutibilidade dos Testes , Esteroides/isolamento & purificação
8.
Psychopharmacology (Berl) ; 240(6): 1359-1372, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37129616

RESUMO

RATIONALE: The prepulse inhibition (PPI) of the startle reflex is the best-established index of sensorimotor gating. We documented that the neurosteroid allopregnanolone (AP) is necessary to reduce PPI in response to D1 dopamine receptor agonists. Since Sprague-Dawley (SD) rats are poorly sensitive to the PPI-disrupting effects of these drugs, we hypothesized that AP might increase this susceptibility. OBJECTIVES: We tested whether AP is sufficient to increase the vulnerability of SD rats to PPI deficits in response to the D1 receptor full agonist SKF82958. METHODS: SD rats were tested for PPI after treatment with SKF82958 (0.05-0.3 mg/kg, SC) in combination with either intraperitoneal (1-10 mg/kg) or intracerebral (0.5 µg/µl/side) AP administration into the medial prefrontal cortex (mPFC) or nucleus accumbens shell. To rule out potential confounds, we measured whether SKF82958 affected the endogenous mPFC levels of AP. RESULTS: SD rats exhibited marked PPI deficits in response to the combination of systemic and intra-mPFC AP with SKF82958 but not with the D2 receptor agonist quinpirole (0.3-0.6 mg/kg, SC). SKF82958 did not elevate mPFC levels of AP but enhanced the content of its precursor progesterone. The PPI deficits caused by SKF82958 in combination with AP were opposed by the AP antagonist isoallopregnanolone (10 mg/kg, IP) and the glutamate NMDA receptor positive modulator CIQ (5 mg/kg, IP). CONCLUSION: These results suggest that AP enables the detrimental effects of D1 receptor activation on sensorimotor gating. AP antagonism or glutamatergic modulation counters these effects and may have therapeutic potential for neuropsychiatric disorders characterized by gating deficits.


Assuntos
Pregnanolona , Receptores de Dopamina D1 , Ratos , Animais , Masculino , Ratos Sprague-Dawley , Pregnanolona/farmacologia , Benzazepinas/farmacologia , Reflexo de Sobressalto , Filtro Sensorial , Estimulação Acústica/métodos
9.
Bull Environ Contam Toxicol ; 88(6): 1038-42, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22456730

RESUMO

This study was conducted to evaluate the effect of molinate on retinoids homeostasis in rat testis. Molinate was administrated to male Sprague-Dawley rats (200 mg kg(-1) in corn oil, ip). Retinoid measurements were made at 6, 12, 48 and 168 h time points after administration. Testis levels of retinoic acid decreased (32 %) in a statistically significant manner at the 12 and 48 h time points. However, retinol and retinaldehyde were not significantly affected by molinate. These results suggest that molinate affects retinoic acid synthesis in testis and could contribute to understanding the molecular mechanism of molinate involved testicular toxicity.


Assuntos
Azepinas/toxicidade , Herbicidas/toxicidade , Testículo/efeitos dos fármacos , Tiocarbamatos/toxicidade , Vitamina A/metabolismo , Animais , Masculino , Ratos , Ratos Sprague-Dawley , Retinaldeído/metabolismo , Testículo/metabolismo
10.
Biomarkers ; 16(5): 434-44, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21692648

RESUMO

Damage to DNA by dopamine quinone and/or catechol estrogen quinones may play a significant role in the initiation of Parkinson's disease (PD). Depurinating estrogen-DNA adducts are shed from cells and excreted in urine. The aim of this study was to discover whether higher levels of estrogen-DNA adducts are associated with PD. Forty estrogen metabolites, conjugates, and DNA adducts were analyzed in urine samples from 20 PD cases and 40 matched controls by using ultra performance liquid chromatography/tandem mass spectrometry. The levels of adducts in cases versus controls (P < 0.005) suggest that unbalanced estrogen metabolism could play a causal role in the initiation of PD.


Assuntos
Encéfalo/metabolismo , Estrogênios/metabolismo , Doença de Parkinson/etiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Cromatografia Líquida/métodos , Adutos de DNA/metabolismo , Estrogênios/urina , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Doença de Parkinson/metabolismo , Extração em Fase Sólida , Espectrometria de Massas em Tandem
11.
PLoS One ; 16(1): e0243585, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33406107

RESUMO

BACKGROUND: Preterm delivery is a common pregnancy complication that can result in significant neonatal morbidity and mortality. Limited tools exist to predict preterm birth, and none to predict neonatal morbidity, from early in pregnancy. The objective of this study was to determine if the progesterone metabolites 11-deoxycorticosterone (DOC) and 16-alpha hydroxyprogesterone (16α-OHP), when combined with patient demographic and obstetric history known during the pregnancy, are predictive of preterm delivery-associated neonatal morbidity, neonatal length of stay, and risk for spontaneous preterm delivery prior to 32 weeks' gestation. METHODS AND FINDINGS: We conducted a cohort study of pregnant women with plasma samples collected as part of Building Blocks of Pregnancy Biobank at the Indiana University School of Medicine. The progesterone metabolites, DOC and 16α-OHP, were quantified by mass spectroscopy from the plasma of 58 pregnant women collected in the late first trimester/early second trimester. Steroid levels were combined with patient demographic and obstetric history data in multivariable logistic regression models. The primary outcome was composite neonatal morbidity as measured by the Hassan scale. Secondary outcomes included neonatal length of stay and spontaneous preterm delivery prior to 32 weeks' gestation. The final neonatal morbidity model, which incorporated antenatal corticosteroid exposure and fetal sex, was able to predict high morbidity (Hassan score ≥ 2) with an area under the ROC curve (AUROC) of 0.975 (95% CI 0.932, 1.00), while the model without corticosteroid and fetal sex predictors demonstrated an AUROC of 0.927 (95% CI 0.824, 1.00). The Hassan score was highly correlated with neonatal length of stay (p<0.001), allowing the neonatal morbidity model to also predict increased neonatal length of stay (53 [IQR 22, 76] days vs. 4.5 [2, 31] days, above and below the model cut point, respectively; p = 0.0017). Spontaneous preterm delivery prior to 32 weeks' gestation was also predicted with an AUROC of 0.94 (95% CI 0.869, 1.00). CONCLUSIONS: Plasma levels of DOC and 16α-OHP in early gestation can be combined with patient demographic and clinical data to predict significant neonatal morbidity, neonatal length of stay, and risk for very preterm delivery, though validation studies are needed to verify these findings. Early identification of pregnancies at risk for preterm delivery and neonatal morbidity allows for timely implementation of multidisciplinary care to improve perinatal outcomes.


Assuntos
Biomarcadores/sangue , Nascimento Prematuro/diagnóstico , Nascimento Prematuro/epidemiologia , Esteroides/sangue , Adulto , Feminino , Humanos , Recém-Nascido , Morbidade , Fenótipo , Gravidez , Curva ROC , Análise de Regressão , Adulto Jovem
12.
J Clin Invest ; 131(10)2021 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-33848264

RESUMO

Opioid use disorder (OUD) has become a leading cause of death in the United States, yet current therapeutic strategies remain highly inadequate. To identify potential treatments for OUD, we screened a targeted selection of over 100 drugs using a recently developed opioid self-administration assay in zebrafish. This paradigm showed that finasteride, a steroidogenesis inhibitor approved for the treatment of benign prostatic hyperplasia and androgenetic alopecia, reduced self-administration of multiple opioids without affecting locomotion or feeding behavior. These findings were confirmed in rats; furthermore, finasteride reduced the physical signs associated with opioid withdrawal. In rat models of neuropathic pain, finasteride did not alter the antinociceptive effect of opioids and reduced withdrawal-induced hyperalgesia. Steroidomic analyses of the brains of fish treated with finasteride revealed a significant increase in dehydroepiandrosterone sulfate (DHEAS). Treatment with precursors of DHEAS reduced opioid self-administration in zebrafish in a fashion akin to the effects of finasteride. These results highlight the importance of steroidogenic pathways as a rich source of therapeutic targets for OUD and point to the potential of finasteride as a new treatment option for this disorder.


Assuntos
Inibidores de 5-alfa Redutase/farmacologia , Finasterida/farmacologia , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Animais , Modelos Animais de Doenças , Humanos , Masculino , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Ratos , Ratos Sprague-Dawley , Peixe-Zebra
13.
Hum Reprod Open ; 2020(2): hoaa007, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32274422

RESUMO

STUDY QUESTION: Do maternal serum levels of progesterone metabolites early in pregnancy correspond to an increased risk for very preterm delivery prior to 32 weeks? SUMMARY ANSWER: Maternal serum levels of 11-deoxycorticosterone (DOC) measured during the late first trimester or early second trimester correlate with an increased risk for preterm delivery prior to 32 weeks, and the correlation becomes stronger when the ratio of DOC to 16-alpha-hydroxyprogesterone was measured. WHAT IS KNOWN ALREADY: Progesterone is a pro-gestational steroid hormone that has been shown to decrease the risk of preterm birth in some pregnant women. Progesterone is metabolized by the body into various metabolites including members of the mineralocorticoid and glucocorticoid families. Our group has previously demonstrated that some progesterone metabolites enhance myometrial contractility in an ex vivo system, while others result in myometrial relaxation. The current exploratory study was designed to determine if pre-specified metabolites of progesterone measured early in pregnancy were associated with a woman's risk for delivery prior to 32 weeks, which is referred to as a very preterm delivery. STUDY DESIGN SIZE DURATION: The Building Blocks of Pregnancy Biobank (BBPB) is a biorepository at Indiana University (IU) that follows women prospectively through their pregnancy. A variety of biospecimens are collected at various time points during a woman's pregnancy. Women participating in the IU BBPB who were enrolled after 8 weeks' gestation with pregnancy outcome data were eligible for participation. PARTICIPANTS/MATERIALS SETTING METHODS: Women delivering prior to 37 weeks (preterm) and at or after 37 weeks (term) who had blood samples collected during the late first trimester/early second trimester and/or during the early third trimester were identified. These samples were then processed for mass spectroscopy, and the amount of progesterone and progesterone metabolites in the samples were measured. Mean values of each measured steroid metabolite were calculated and compared among women delivering at less than 32 weeks, less than 37 weeks and greater than or equal to 37 weeks. Receiver operating characteristic (ROC) curves were constructed and threshold levels determined for each compound to identify a level above or below which best predicted a woman's risk for delivery prior to 32 and prior to 37 weeks. Mann-Whitney U nonparametric testing with Holm-Bonferroni correction for multiple comparisons was utilized to identify steroid ratios that could differentiate women delivering spontaneously at less than 32 weeks from all other pregnancies. MAIN RESULTS AND THE ROLE OF CHANCE: Steroid hormone levels and pregnancy outcome data were available for 93 women; 28 delivering prior to 32 weeks, 40 delivering between 32 0/7 and 36 6/7 weeks and 25 delivering at or greater than 37 weeks: the mean gestational age at delivery within the three groups was 27.0, 34.4 and 38.8 weeks, respectively. Among women delivering spontaneously at less than 37 weeks, maternal 11-deoxycorticosterone (DOC) levels drawn in the late first trimester/early second trimester were significantly associated with spontaneous preterm delivery prior to 32 weeks; a threshold level of 47.5 pg/ml had 78% sensitivity, 73% specificity and an AUC of 0.77 (P = 0.044). When DOC levels were analyzed as a ratio with other measured steroid hormones, the ratio of DOC to 16-alpha-hydroxyprogesterone among women delivering spontaneously prior to 37 weeks was able to significantly discriminate women delivering prior to 32 weeks from those delivering at or greater than 32 weeks, with a threshold value of 0.2 with 89% sensitivity, 91% specificity and an AUC of 0.92 (P = 0.002). When the entire study cohort population was considered, including women delivering at term and women having an iatrogenic preterm delivery, the ratio of DOC to 16-alpha-hydroxyprogesterone was able to discriminate women delivering spontaneously prior to 32 weeks from the rest of the population at a threshold of 0.18 and 89% sensitivity, 59% specificity and an AUC of 0.81 (P = 0.003). LIMITATIONS REASONS FOR CAUTION: This is a discovery study, and the findings have not been validated on an independent cohort. To mitigate issues with multiple comparisons, we limited our study to pre-specified metabolites that are most representative of the major metabolic pathways for progesterone, and adjustments for multiple comparisons were made. WIDER IMPLICATIONS OF THE FINDINGS: Spontaneous preterm birth is increasingly being recognized to represent a common end pathway for a number of different disease phenotypes that include infection, inflammation, premature rupture of the membranes, uterine over distension, cervical insufficiency, placental dysfunction and genetic predisposition. In addition to these phenotypes, longitudinal changes in the maternal-fetal hypothalamic-pituitary-adrenal (HPA) axis also likely contribute to a significant proportion of the disease burden of spontaneous preterm birth. Here, we demonstrate that differential production of steroid metabolites is associated with very early preterm birth. The identified biomarkers may hint at a pathophysiologic mechanism and changes in the maternal-fetal dyad that result in preterm delivery. The early identification of abnormal changes in HPA axis metabolites may allow for targeted interventions that reverse the aberrant steroid metabolic profile to a more favorable one, thereby decreasing the risk for early delivery. Further research is therefore required to validate and extend the results presented here. STUDY FUNDING/COMPETING INTERESTS: Funding for this study was provided from the Office of the Vice Chancellor for Research at IUPUI, 'Funding Opportunities for Research Commercialization and Economic Success (FORCES) grant'.Both A.S.P. and C.A.G. are affiliated with Nixxi, a biotech startup. The remaining authors report no conflict of interest. TRIAL REGISTRATION NUMBER: Not applicable.

14.
Biomedicines ; 8(8)2020 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-32796613

RESUMO

Our group and others have previously shown that genistein combined polysaccharide (GCP), an aglycone isoflavone-rich extract with high bioavailability and low toxicity, can inhibit prostate cancer (CaP) cell growth and survival as well as androgen receptor (AR) activity. We now elucidate the mechanism by which this may occur using LNCaP and PC-346C CaP cell lines; GCP can inhibit intracrine androgen synthesis in CaP cells. UPLC-MS/MS and qPCR analyses demonstrated that GCP can mediate a ~3-fold decrease in testosterone levels (p < 0.001) and cause decreased expression of intracrine androgen synthesis pathway enzymes (~2.5-fold decrease of 3ßHSD (p < 0.001), 17ßHSD (p < 0.001), CYP17A (p < 0.01), SRB1 (p < 0.0001), and StAR (p < 0.01)), respectively. Reverse-phase HPLC fractionation and bioassay identified three active GCP fractions. Subsequent NMR and LC-MS analysis of the fraction with the highest level of activity, fraction 40, identified genistein as the primary active component of GCP responsible for its anti-proliferative, pro-apoptotic, and anti-AR activity. GCP, fraction 40, and genistein all mediated at least a ~2-fold change in these biological activities relative to vehicle control (p < 0.001). Genistein caused similar decreases in the expression of 17ßHSD and CYP17A (2.5-fold (p < 0.001) and 1.5-fold decrease (p < 0.01), respectively) compared to GCP, however it did not cause altered expression of the other intracrine androgen synthesis pathway enzymes; 3ßHSD, SRB1, and StAR. Our combined data indicate that GCP and/or genistein may have clinical utility and that further pre-clinical studies are warranted.

15.
Prostate ; 69(1): 41-8, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18816637

RESUMO

BACKGROUND: Although the estrogens estrone and estradiol are recognized to play very important roles in the risk of developing prostate cancer (Pca), the molecular mechanism by which estrogens initiate and/or promote Pca is still unknown. Substantial evidence supports that specific metabolites of estrogens, catechol estrogen quinones, can react with DNA to form depurinating estrogen-DNA adducts. Apurinic sites derived from depurination of these adducts can induce mutations leading to cancer. Once released from DNA, depurinating estrogen-DNA adducts are shed from cells into the bloodstream and excreted in urine. By analyzing profiles of estrogen metabolites, conjugates, and depurinating DNA adducts in urine from men with and without prostate cancer, potential biomarkers of Pca can be detected. The goal of this case-control study was to detect and identify potential biomarkers of Pca. METHODS: Urine samples from fourteen cases, men diagnosed with Pca, and 125 controls, men who had not been diagnosed with Pca, were partially purified by solid phase extraction and analyzed by ultraperformance liquid chromatography/tandem mass spectrometry. The urinary levels of androgens, estrogens, estrogen metabolites, conjugates and depurinating DNA adducts were measured. RESULTS: The ratio of depurinating estrogen-DNA adducts to the sum of the corresponding estrogen metabolites and conjugates was significantly higher in cases (median: 57.34) compared to controls (median: 23.39) (P < 0.001). CONCLUSIONS: This study suggests that depurinating estrogen-DNA adducts could serve as potential biomarkers to predict risk of Pca. They also could be useful tools for early clinical diagnosis and development of suitable strategies to prevent Pca.


Assuntos
Androgênios/urina , Biomarcadores Tumorais/urina , Adutos de DNA/urina , Estrogênios/urina , Neoplasias da Próstata/epidemiologia , Neoplasias da Próstata/urina , Idoso , Idoso de 80 Anos ou mais , Androgênios/biossíntese , Estudos de Casos e Controles , Adutos de DNA/biossíntese , Diagnóstico Precoce , Estrogênios/biossíntese , Humanos , Masculino , Pessoa de Meia-Idade , Razão de Chances , Neoplasias da Próstata/diagnóstico , Fatores de Risco
16.
Biomarkers ; 14(7): 502-12, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19863189

RESUMO

A variety of evidence suggests that estrogens may induce non-Hodgkin lymphoma (NHL). The reaction of catechol estrogen quinones with DNA to form depurinating estrogen-DNA adducts is hypothesized to initiate this process. These adducts are released from DNA, shed from cells into the bloodstream and excreted in urine. The aim of this study was to determine whether or not the depurinating estrogen-DNA adducts might be involved in the aetiology of human NHL. Estrogen metabolites, conjugates and depurinating DNA adducts were identified and quantified in spot urine samples from 15 men with NHL and 30 healthy control men by using ultraperformance liquid chromatography/tandem mass spectrometry. The levels of estrogen-DNA adducts were significantly higher in the men with NHL than in the healthy control men. Thus, formation of estrogen-DNA adducts may play a critical role in the aetiology of NHL, and these adducts could be potential biomarkers of NHL risk.


Assuntos
Biomarcadores Tumorais/urina , Adutos de DNA/urina , Estrogênios/urina , Linfoma não Hodgkin/diagnóstico , Idoso , Cromatografia Líquida de Alta Pressão , DNA/química , DNA/urina , Adutos de DNA/química , Estrogênios/química , Humanos , Linfoma não Hodgkin/tratamento farmacológico , Linfoma não Hodgkin/urina , Masculino , Pessoa de Meia-Idade , Estrutura Molecular , Espectrometria de Massas em Tandem
17.
Nutrition ; 65: 208-215, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31031064

RESUMO

OBJECTIVE: The beneficial effects of tea on health, including obesity, are well known. However, the comparative effects of black, green, white, and oolong teas, which are prepared from the same fresh leaves, on weight gain and the potential mechanisms involved are not yet fully understood. Bile acids (BAs) are shown to be powerful regulators of metabolism; however, to our knowledge, no studies have investigated the effect of tea on BA metabolism. The aim of this study was to investigate the modulatory effects that green, black, white, and oolong teas that were prepared from the same raw tea leaves have on the plasma BA profile. METHODS: Female rats were dosed with the aforementioned tea types as their sole source of drinking fluid for 28 d. We then investigated their weight and effect on BA metabolic profile using advanced ultra-performance liquid chromatography-tandem mass spectrometer (UPLC-MS/MS)-based metabolomics. RESULTS: The UPLC-MS/MS analysis of the plasma show that the levels of murocholic acid, glycochenodeoxycholic acid, glycocholic acid, glycodeoxycholic acid, taurochenodeoxycholic acid, tauroursodeoxycholic acid, taurodeoxycholic acid, tauromuricholic acid, and taurocholic acid were increased; whereas levels of taurolithocholic acid and isolithocholic acid were decreased after drinking green, oolong, and white tea types compared with control. Surprisingly, oolong tea significantly influenced reduction in relative weight compared with control, black, and green tea; whereas black, green, and white teas had no effects on weight compared with control. CONCLUSIONS: Green, black, oolong, and white teas altered the BA metabolism. This change in BA metabolism could be associated with the health benefit effects of tea. Oolong tea was most effective in reducing weight.


Assuntos
Ácidos e Sais Biliares/sangue , Extratos Vegetais/farmacologia , Folhas de Planta , Chá , Aumento de Peso/efeitos dos fármacos , Animais , Cromatografia Líquida , Feminino , Ratos , Espectrometria de Massas em Tandem
18.
Int J Cancer ; 122(9): 1949-57, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18098283

RESUMO

Estrogens can become endogenous carcinogens via formation of catechol estrogen quinones, which react with DNA to form specific depurinating estrogen-DNA adducts. The mutations resulting from these adducts can lead to cell transformation and the initiation of breast cancer. Estrogen metabolites, conjugates and depurinating DNA adducts in urine samples from 46 healthy control women, 12 high-risk women and 17 women with breast cancer were analyzed. The estrogen metabolites, conjugates and depurinating DNA adducts were identified and quantified by using ultraperformance liquid chromatography/tandem mass spectrometry. The levels of the ratios of depurinating DNA adducts to their respective estrogen metabolites and conjugates were significantly higher in high-risk women (p < 0.001) and women with breast cancer (p < 0.001) than in control subjects. The high-risk and breast cancer groups were not significantly different (p = 0.62). After adjusting for patient characteristics, these ratios were still significantly associated with health status. Thus, the depurinating estrogen-DNA adducts are possible biomarkers for early detection of breast cancer risk and response to preventive treatment.


Assuntos
Biomarcadores Tumorais/urina , Neoplasias da Mama/urina , Adutos de DNA/urina , Estrogênios Conjugados (USP)/urina , Adulto , Idoso , Análise de Variância , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/etiologia , Neoplasias da Mama/metabolismo , Cromatografia Líquida , Estudos Transversais , Adutos de DNA/metabolismo , Estrogênios Conjugados (USP)/metabolismo , Feminino , Humanos , Itália , Modelos Lineares , Pessoa de Meia-Idade , Modelos Estatísticos , Valores de Referência , Medição de Risco , Fatores de Risco , Espectrometria de Massas em Tandem , Estados Unidos
19.
Free Radic Biol Med ; 45(2): 136-45, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18423413

RESUMO

Resveratrol (Resv), a natural occurring phytolexin present in grapes and other foods, possesses chemopreventive effects revealed by its striking modulation of diverse cellular events associated with tumor initiation, promotion, and progression. Catechol estrogens generated in the metabolism of estrogens are oxidized to catechol quinones that react with DNA to form predominantly depurinating estrogen-DNA adducts. This event can generate the mutations responsible for cancer initiation. In this regard, Resv acts as both an antioxidant and an inducer of the phase II enzyme NAD(P)H:quinone oxidoreductase 1 (NQO1). In this report, we present the effects of Resv on the metabolism of estrogens in normal breast epithelial cells (MCF-10F) treated with 4-hydroxyestradiol (4-OHE(2)) or estradiol-3,4-quinone (E(2)-3,4-Q). Resv induced NQO1 in a dose- and time-dependent manner, but did not affect the expression of catechol-O-methyltransferase. Ultraperformance liquid chromatography/tandem mass spectrometry was used to determine the effects of Resv on estrogen metabolism. Preincubation of the cells with Resv for 48 h decreased the formation of depurinating estrogen-DNA adducts from 4-OHE(2) or E(2)-3,4-Q and increased formation of methoxycatechol estrogens. When Resv was also present with the 4-OHE(2) or E(2)-3,4-Q, even greater increases in methoxycatechol estrogens were observed, and the DNA adducts were undetectable. We conclude that Resv can protect breast cells from carcinogenic estrogen metabolites, suggesting that it could be used in breast cancer prevention.


Assuntos
Antioxidantes/farmacologia , Neoplasias da Mama/prevenção & controle , Adutos de DNA/efeitos dos fármacos , Estrogênios/metabolismo , Estilbenos/farmacologia , Linhagem Celular Tumoral , Cromatografia Líquida , Feminino , Humanos , NAD(P)H Desidrogenase (Quinona)/efeitos dos fármacos , Resveratrol , Espectrometria de Massas em Tandem
20.
Endocrinology ; 159(9): 3365-3377, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-30060148

RESUMO

Steroid sulfatase (STS), a desulfating enzyme that converts steroid sulfates to hormonally active steroids, plays an important role in the homeostasis of sex hormones. STS is expressed in the adipose tissue of both male and female mice, but the role of STS in the development and function of adipose tissue remains largely unknown. In this report, we show that the adipose expression of Sts was induced in the high-fat diet (HFD) and ob/ob models of obesity and type 2 diabetes. Transgenic overexpression of the human STS in the adipose tissue of male mice exacerbated the HFD-induced metabolic phenotypes, including increased body weight gain and fat mass, and worsened insulin sensitivity, glucose tolerance, and energy expenditure, which were accounted for by adipocyte hypertrophy, increased adipose inflammation, and dysregulation of adipogenesis. The metabolic harm of the STS transgene appeared to have resulted from increased androgen activity in the adipose tissue, and castration abolished most of the phenotypes. Interestingly, the transgenic effects were sex specific, because the HFD-fed female STS transgenic mice exhibited improved metabolic functions, which were associated with attenuated adipose inflammation. The metabolic benefit of the STS transgene in female mice was accounted for by increased estrogenic activity in the adipose tissue, whereas such benefit was abolished upon ovariectomy. Our results revealed an essential role of the adipose STS in energy homeostasis in sex- and sex hormone-dependent manner. The adipose STS may represent a therapeutic target for the management of obesity and type 2 diabetes.


Assuntos
Tecido Adiposo/metabolismo , Androgênios/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético/genética , Estrogênios/metabolismo , Obesidade/metabolismo , Caracteres Sexuais , Esteril-Sulfatase/genética , Adipogenia , Tecido Adiposo/imunologia , Tecido Adiposo/patologia , Animais , Peso Corporal , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Teste de Tolerância a Glucose , Hormônios Esteroides Gonadais/metabolismo , Humanos , Inflamação , Resistência à Insulina , Lipólise , Masculino , Camundongos , Camundongos Obesos , Camundongos Transgênicos , Obesidade/imunologia , Orquiectomia , Ovariectomia , Esteril-Sulfatase/metabolismo , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA