Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
MethodsX ; 9: 101775, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35855952

RESUMO

Fractionation of steroids allows for multiple assays to be run on a single low volume liquid biopsy, whereas performing the same number of assays without fractionation would require increasing the sample volume by dilution, rendering the concentration of steroids below the level of detection for most, if not all, downstream assays. Briefly, steroids are extracted from a biofluid sample using solvent phase extraction to separate the aqueous (conjugated) steroids from the non-aqueous (non-conjugated) steroids in the organic phase. The latter is further separated by high-performance liquid chromatography (HPLC) and collected in an automated fraction collector based on the UV detection of internal standards. Commercially available immunoassays are then used to quantify the < ng/ml concentrations of steroids in each fraction. This protocol was designed for small samples of nipple aspirate fluid (minimum 2 µL), but it can be modified to fractionate steroids from homogenized solid tissue samples or other liquid biopsies. Included in this protocol are precautions to help ensure reproducibility and minimize matrix effects and other errors of measurement, given that samples requiring fractionation are fundamentally precious and, like other quantitative procedures of small samples, can be prone to contamination by solvent residues and other factors.•The method permits quantitative analysis of multiple steroids from very small volumes of biofluid.•Fractionation by HPLC provides a highly purified sample for quantification.•The immunoassay end point provides specificity without expensive equipment.

2.
Steroids ; 177: 108937, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34762930

RESUMO

PURPOSE: Although alterations of concentrations in circulating steroids have been linked to single nucleotide polymorphisms (SNPs) of steroidogenic enzymes, we hypothesized that SNPs of such enzymes located within the breast affect local steroid concentrations more than products of such SNPs absorbed from the circulation. METHODS: Steroids (estradiol, estrone, testosterone, androstenedione, DHEA, DHEA sulfate, progesterone) in nipple aspirate fluid (NAF) were purified by HPLC and they along with serum steroids were quantified by immunoassays. Polymorphisms of the transporter SLCO2B1 and enzymes HSD3B1, CYP19A1, HSD17B12, AKR1C3, CYP1B1, and SRD5A1 were measured in white blood cell DNA. RESULTS: Steroid concentrations in NAF of subjects with homozygous minor genotypes differed from those with heterozygotes, i.e., SLCO2B1 (rs2851069) decreased DHEAS (p = 0.04), HSD17B12 (rs11555762) increased estradiol (p < 0.004), and CYP1B1 (rs1056836) decreased estradiol (p = 0.017) and increased progesterone (p = 0.05). Also, in serum, CYP19A1 (rs10046 and rs700518) both decreased testosterone (p = 0.02) and SRD5A1 increased androstenedione (p = 0.006). Steroids in subjects with major homozygotes did not differ from those with heterozygotes indicating recessive characteristics. CONCLUSIONS: In the breast, SNPs were associated with decreased uptake of DHEAS (SLCO2B1), increased estradiol concentrations through increased oxidoreductase activity (HSD17B12), or decreased estradiol concentrations by presumed formation of 4-hydroxyestradiol (CYP1B1). CYP19A1 was associated with decreased testosterone concentrations in serum but had no significant effect on estrogen or androgen concentrations within the breast. The hormone differences observed in NAF were not usually evident in serum, indicating the importance of assessing the effect of these SNPs within the breast.


Assuntos
17-Hidroxiesteroide Desidrogenases/genética , Aromatase/genética , Mama/metabolismo , Citocromo P-450 CYP1B1/genética , Transportadores de Ânions Orgânicos/genética , Polimorfismo Genético/genética , Esteroides/metabolismo , 17-Hidroxiesteroide Desidrogenases/metabolismo , Aromatase/metabolismo , Citocromo P-450 CYP1B1/metabolismo , Humanos , Transportadores de Ânions Orgânicos/metabolismo , Esteroides/sangue
3.
Breast Cancer Res ; 22(1): 125, 2020 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-33187540

RESUMO

BACKGROUND: CRIPTO is a multi-functional signaling protein that promotes stemness and oncogenesis. We previously developed a CRIPTO antagonist, ALK4L75A-Fc, and showed that it causes loss of the stem cell phenotype in normal mammary epithelia suggesting it may similarly inhibit CRIPTO-dependent plasticity in breast cancer cells. METHODS: We focused on two triple negative breast cancer cell lines (MDA-MB-231 and MDA-MB-468) to measure the effects of ALK4L75A-Fc on cancer cell behavior under nutrient deprivation and endoplasmic reticulum stress. We characterized the proliferation and migration of these cells in vitro using time-lapse microscopy and characterized stress-dependent changes in the levels and distribution of CRIPTO signaling mediators and cancer stem cell markers. We also assessed the effects of ALK4L75A-Fc on proliferation, EMT, and stem cell markers in vivo as well as on tumor growth and metastasis using inducible lentiviral delivery or systemic administration of purified ALK4L75A-Fc, which represents a candidate therapeutic approach. RESULTS: ALK4L75A-Fc inhibited adaptive responses of breast cancer cells under conditions of nutrient and ER stress and reduced their proliferation, migration, clonogenicity, and expression of EMT and cancer stem cell markers. ALK4L75A-Fc also inhibited proliferation of human breast cancer cells in stressed tumor microenvironments in xenografts and reduced both primary tumor size and metastatic burden. CONCLUSIONS: Cancer cell adaptation to stresses such as nutrient deprivation, hypoxia, and chemotherapy can critically contribute to dormancy, metastasis, therapy resistance, and recurrence. Identifying mechanisms that govern cellular adaptation, plasticity, and the emergence of stem-like cancer cells may be key to effective anticancer therapies. Results presented here indicate that targeting CRIPTO with ALK4L75A-Fc may have potential as such a therapy since it inhibits breast cancer cell adaptation to microenvironmental challenges and associated stem-like and EMT phenotypes.


Assuntos
Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas de Neoplasias/antagonistas & inibidores , Células-Tronco Neoplásicas/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Receptores de Ativinas Tipo I/genética , Animais , Linhagem Celular Tumoral , Plasticidade Celular/efeitos dos fármacos , Estresse do Retículo Endoplasmático , Feminino , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Recidiva Local de Neoplasia , Células-Tronco Neoplásicas/patologia , Mutação Puntual , Ligação Proteica/genética , Domínios Proteicos/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/uso terapêutico , Neoplasias de Mama Triplo Negativas/patologia , Hipóxia Tumoral , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Cell Rep ; 24(6): 1653-1666.e7, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30089273

RESUMO

The mammary gland consists of cells with gene expression patterns reflecting their cellular origins, function, and spatiotemporal context. However, knowledge of developmental kinetics and mechanisms of lineage specification is lacking. We address this significant knowledge gap by generating a single-cell transcriptome atlas encompassing embryonic, postnatal, and adult mouse mammary development. From these data, we map the chronology of transcriptionally and epigenetically distinct cell states and distinguish fetal mammary stem cells (fMaSCs) from their precursors and progeny. fMaSCs show balanced co-expression of factors associated with discrete adult lineages and a metabolic gene signature that subsides during maturation but reemerges in some human breast cancers and metastases. These data provide a useful resource for illuminating mammary cell heterogeneity, the kinetics of differentiation, and developmental correlates of tumorigenesis.


Assuntos
Glândulas Mamárias Animais/crescimento & desenvolvimento , Animais , Diferenciação Celular/fisiologia , Feminino , Humanos , Glândulas Mamárias Animais/citologia , Camundongos , Células-Tronco/metabolismo , Transcriptoma
5.
Cancer Prev Res (Phila) ; 11(9): 557-568, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29954758

RESUMO

Sex steroid hormones contribute to breast cancer development, but data on concentrations of these within breast tissue are limited. We performed simultaneous multiparameter measurement of breast sex steroids, breast epithelial cytology, and DNA methylation in 119 healthy women (54 pre- and 65 postmenopausal) without a history of breast cancer. Random fine-needle aspiration (rFNA) of the breast was performed simultaneously with blood collection. Breast samples were analyzed by LC/MS-MS for estrone, estradiol, progesterone, androstenedione, and testosterone. Blood samples were assayed for estradiol and progesterone by immunoassay. Cytomorphology was classified using the Masood Score, and DNA methylation of eight genes was analyzed using quantitative multiplexed methylation-specific PCR, and expressed as the cumulative methylation index (CMI). Serum and breast concentrations of estradiol and progesterone showed significant correlation (Spearman r = 0.34, Padj = 0.001 and r = 0.69, Padj < 0.0006, respectively). Progesterone concentration was significantly higher in the premenopausal breast (Padj < 0.0008), and showed a luteal surge. Breast estrone and estradiol concentrations did not differ significantly by menopause, but androstenedione concentration was higher in the breasts of postmenopausal women (P = 0.026 and Padj = 0.208). Breast androgens were significantly correlated with breast density (Spearman r = 0.27, Padj = 0.02 for testosterone) and CMI (Spearman r = 0.3, Padj = 0.038 for androstenedione). Our data indicate that future larger studies of breast steroid hormones along with other parameters are feasible. Significant associations of breast androgen concentrations with breast density and gene methylation warrant future study. Cancer Prev Res; 11(9); 557-68. ©2018 AACR.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias da Mama/diagnóstico , Mama/patologia , Metilação de DNA , Hormônios Esteroides Gonadais/análise , Biomarcadores Tumorais/metabolismo , Biópsia por Agulha Fina , Mama/metabolismo , Densidade da Mama/fisiologia , Neoplasias da Mama/sangue , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Estudos de Viabilidade , Feminino , Hormônios Esteroides Gonadais/metabolismo , Humanos , Pessoa de Meia-Idade , Pós-Menopausa/metabolismo , Pré-Menopausa/metabolismo , Estudos Prospectivos , Fatores de Risco
6.
Development ; 143(22): 4236-4248, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27729410

RESUMO

Profiling of RNA from mouse mammary epithelial cells (MECs) isolated on pregnancy day (P)14 and lactation day (L)2 revealed that the majority of differentially expressed microRNA declined precipitously between late pregnancy and lactation. The decline in miR-150, which exhibited the greatest fold-decrease, was verified quantitatively and qualitatively. To test the hypothesis that the decline in miR-150 is crucial for lactation, MEC-specific constitutive miR-150 was achieved by crossing ROSA26-lox-STOP-lox-miR-150 mice with WAP-driven Cre recombinase mice. Both biological and foster pups nursed by bitransgenic dams exhibited a dramatic decrease in survival compared with offspring nursed by littermate control dams. Protein products of predicted miR-150 targets Fasn, Olah, Acaca, and Stat5B were significantly suppressed in MECs of bitransgenic mice with constitutive miR-150 expression as compared with control mice at L2. Lipid profiling revealed a significant reduction in fatty acids synthesized by the de novo pathway in L2 MECs of bitransgenic versus control mice. Collectively, these data support the hypothesis that a synchronized decrease in miRNAs, such as miR-150, at late pregnancy serves to allow translation of targets crucial for lactation.


Assuntos
Lactação/genética , Lipogênese/genética , Glândulas Mamárias Animais/metabolismo , MicroRNAs/genética , Animais , Células Cultivadas , Regulação para Baixo/genética , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Lactação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , MicroRNAs/metabolismo , Análise em Microsséries , Gravidez/genética , Gravidez/metabolismo
7.
Horm Cancer ; 7(2): 127-36, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26902826

RESUMO

Prior reports identify higher serum concentrations of estrogens and androgens as risk factors for breast cancer, but steroids in nipple aspirate fluid (NAF) may be more related to risk. Incident breast cancer cases and mammography controls were recruited. Sex steroids were measured in NAF from the unaffected breasts of cases and one breast of controls. Menopausal status and menstrual cycle phase were determined. NAF steroids were purified by HPLC and quantified by immunoassays. Conditional logistic regression models were used to examine associations between NAF hormones and case-control status. NAF samples from 160 cases and 157 controls were evaluable for hormones. Except for progesterone and dehydroepiandrosterone (DHEA), the NAF and serum concentrations were not significantly correlated. NAF estradiol and estrone were not different between cases and controls. Higher NAF (but not serum) DHEA concentrations were associated with cases, particularly among estrogen receptor (ER)-positive cases (NAF odds ratio (OR) = 1.18, 95 % confidence interval (CI) 1.02, 1.36). NAF DHEA was highly correlated with NAF estradiol and estrone but not with androstenedione or testosterone. Higher progesterone concentrations in both NAF and serum were associated with a lower risk of ER-negative cancer (NAF OR = 0.69, 95 % CI 0.51, 0.92). However, this finding may be explained by case-control imbalance in the number of luteal phase subjects (2 cases and 19 controls). The significantly higher concentration of DHEA in NAF of cases and its correlation with NAF estradiol indicates a potentially important role of this steroid in breast cancer risk; however, the negative association of progesterone with risk is tentative.


Assuntos
Neoplasias da Mama/etiologia , Hormônios Esteroides Gonadais/análise , Hormônios Esteroides Gonadais/sangue , Fluido do Aspirado de Mamilo/química , Adulto , Idoso , Neoplasias da Mama/sangue , Neoplasias da Mama/metabolismo , Estudos de Casos e Controles , Feminino , Humanos , Modelos Logísticos , Pessoa de Meia-Idade , Fatores de Risco
8.
Mol Cancer Ther ; 14(3): 769-78, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25713333

RESUMO

Triple-negative breast cancer (TNBC) has the lowest 5-year survival rate of invasive breast carcinomas, and currently there are no approved targeted therapies for this aggressive form of the disease. The androgen receptor (AR) is expressed in up to one third of TNBC and we find that all AR(+) TNBC primary tumors tested display nuclear localization of AR, indicative of transcriptionally active receptors. While AR is most abundant in the "luminal AR (LAR)" molecular subtype of TNBC, here, for the first time, we use both the new-generation anti-androgen enzalutamide and AR knockdown to demonstrate that the other non-LAR molecular subtypes of TNBC are critically dependent on AR protein. Indeed, AR inhibition significantly reduces baseline proliferation, anchorage-independent growth, migration, and invasion and increases apoptosis in four TNBC lines (SUM159PT, HCC1806, BT549, and MDA-MB-231), representing three non-LAR TNBC molecular subtypes (mesenchymal-like, mesenchymal stem-like, and basal-like 2). In vivo, enzalutamide significantly decreases viability of SUM159PT and HCC1806 xenografts. Furthermore, mechanistic analysis reveals that AR activation upregulates secretion of the EGFR ligand amphiregulin (AREG), an effect abrogated by enzalutamide in vitro and in vivo. Exogenous AREG partially rescues the effects of AR knockdown on proliferation, migration, and invasion, demonstrating that upregulation of AREG is one mechanism by which AR influences tumorigenicity. Together, our findings indicate that non-LAR subtypes of TNBC are AR dependent and, moreover, that enzalutamide is a promising targeted therapy for multiple molecular subtypes of AR(+) TNBC.


Assuntos
Feniltioidantoína/análogos & derivados , Receptores Androgênicos/genética , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Antagonistas de Androgênios/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Benzamidas , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Camundongos , Camundongos Nus , Nitrilas , Feniltioidantoína/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
9.
Cancer Epidemiol Biomarkers Prev ; 22(12): 2277-84, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24049126

RESUMO

BACKGROUND: Nipple aspiration fluid (NAF) use as a biosample is limited by the variable yield across studies. We investigated the endocrine determinants of yield in an ongoing breast cancer case-control study. METHODS: One-hundred and eighteen women yielding ≥2 µL NAF and 120 non-yielders were included; serum hormones were measured; differences in median hormones were assessed using the Wilcoxon rank-sum test. ORs and 95% confidence intervals (95% CI) for yielder status relative to hormone levels were estimated using logistic regression, adjusting for parity and lactation, and, in premenopausal women, menstrual cycle phase (MCP). RESULTS: Prolactin concentrations were higher in yielders than non-yielders (premenopausal: 7.6 and 2.5 ng/mL, P < 0.01; postmenopausal 5.3 and 2.2 ng/mL; P < 0.01). Among premenopausal-yielders, estradiol was lower (64.3 vs. 90.5 pg/mL, MCP-adjusted P = 0.02). In separate menopausal status and parity-adjusted models, significant case-control differences persisted in prolactin: case OR 1.93 (95% CI, 1.35-2.77), control OR 1.64 (95% CI, 1.17-2.29). Premenopausal control yielders had higher progesterone (OR, 1.70; 95% CI, 1.18-2.46) and sex-hormone binding-globulin (OR, 2.09; 95% CI, 1.08-4.05) than non-yielders. Among parous women, further adjustment for lactation suggested a stronger positive association of serum prolactin with yield in cases than controls. CONCLUSION: NAF-yielders show higher prolactin than non-yielders, regardless of menopause and parity; implications of this and other endocrine differences on NAF biomarkers of breast cancer risk deserve further study. IMPACT: NAF yield is associated with a distinct endocrine environment that must be considered in studies of NAF-based breast cancer risk markers.


Assuntos
Neoplasias da Mama/metabolismo , Hormônios/análise , Fluido do Aspirado de Mamilo/química , Adulto , Idoso , Neoplasias da Mama/sangue , Estudos de Casos e Controles , Detecção Precoce de Câncer/métodos , Feminino , Hormônios/sangue , Humanos , Pessoa de Meia-Idade , Fluido do Aspirado de Mamilo/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA