Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Vis ; 28: 192-202, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36274817

RESUMEN

Purpose: Retinoblastoma (RB) is a malignant childhood intraocular tumor. Current treatment options for RB have undesirable side effects. A comprehensive understanding of gene expression in human RB is essential for the development of safe and effective new therapies. Methods: We reviewed published microarray and RNA sequencing studies in which gene expression profiles were compared between human RB and normal retina tissues. We investigated the expression of genes of interest using quantitative reverse transcription PCR. We examined the activities of cloned promoter DNA fragments with luciferase assay. Results: Dopachrome tautomerase (DCT) was among the most overexpressed genes in RB in published studies. We found that DCT was highly expressed in six of 13 samples microdissected from Thai RB tissues. Expression of DCT was absent or barely detected in retina tissues, various human ocular cells, and major organs. We also demonstrated that the -657 to +411 DCT promoter fragment efficiently directs RB cell-specific transcription of the luciferase reporter gene in cell lines. Conclusions: The present work highlights that DCT is one of the most RB-specific genes. The regulatory elements required for this cell-specific gene expression are likely located within its proximal promoter.


Asunto(s)
Oxidorreductasas Intramoleculares , Neoplasias de la Retina , Retinoblastoma , Niño , Humanos , Regulación Neoplásica de la Expresión Génica , Genes de Retinoblastoma/genética , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Neoplasias de la Retina/genética , Retinoblastoma/genética , Retinoblastoma/patología
2.
Adv Exp Med Biol ; 1329: 443-474, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34664251

RESUMEN

The tumor microenvironment (TME) is a complex infrastructure composed of stromal, epithelial, and immune cells embedded in a vasculature ECM. The microenvironment surrounding mammary epithelium plays a critical role during the development and differentiation of the mammary gland, enabling the coordination of the complex multihormones and growth factor signaling processes. Progesterone/progesterone receptor paracrine signaling interactions in the microenvironment play vital roles in stem/progenitor cell function during normal breast development. In breast cancer, the female sex hormones, estrogen and progesterone, and growth factor signals are altered in the TME. Progesterone signaling modulates not only breast tumors but also the breast TME, leading to the activation of a series of cross-communications that are implicated in the genesis of breast cancers. This chapter reviews the evidence that progesterone and PR signaling modulates not only breast epitheliums but also the breast TME. Furthermore, crosstalk between estrogen and progesterone signaling affecting different cell types within the TME is discussed. A better understanding of how PR and progesterone affect the TME of breast cancer may lead to novel drugs or a therapeutic approach for the treatment of breast cancer shortly.


Asunto(s)
Glándulas Mamarias Humanas , Receptores de Progesterona , Mama , Femenino , Humanos , Receptores de Progesterona/genética , Transducción de Señal , Microambiente Tumoral
3.
Nucleic Acids Res ; 41(19): 8872-85, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23935075

RESUMEN

A-repeats are the simplest form of tandem repeats and are found ubiquitously throughout genomes. These mononucleotide repeats have been widely believed to be non-functional 'junk' DNA. However, studies in yeasts suggest that A-repeats play crucial biological functions, and their role in humans remains largely unknown. Here, we showed a non-random pattern of distribution of sense A- and T-repeats within 20 kb around transcription start sites (TSSs) in the human genome. Different distributions of these repeats are observed upstream and downstream of TSSs. Sense A-repeats are enriched upstream, whereas sense T-repeats are enriched downstream of TSSs. This enrichment directly correlates with repeat size. Genes with different functions contain different lengths of repeats. In humans, tissue-specific genes are enriched for short repeats of <10 bp, whereas housekeeping genes are enriched for long repeats of ≥10 bp. We demonstrated that DICER1 and Argonaute proteins are required for the cis-regulatory role of A-repeats. Moreover, in the presence of a synthetic polymer that mimics an A-repeat, protein binding to A-repeats was blocked, resulting in a dramatic change in the expression of genes containing upstream A-repeats. Our findings suggest a length-dependent cis-regulatory function of A-repeats and that Argonaute proteins serve as trans-acting factors, binding to A-repeats.


Asunto(s)
Proteínas Argonautas/metabolismo , ARN Helicasas DEAD-box/metabolismo , Repeticiones de Microsatélite , Elementos Reguladores de la Transcripción , Ribonucleasa III/metabolismo , Animales , Células HEK293 , Humanos , Ratones , Ratas , Sitio de Iniciación de la Transcripción
4.
J Steroid Biochem Mol Biol ; 237: 106441, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38070754

RESUMEN

In addition to mediating nuclear transcription, PR mediates extranuclear functions mainly through the PR polyproline domain (PPD) interaction with the SH3 domain of cytoplasmic signaling molecules. PR-PPD-SH3 interaction inhibits EGF-mediated signaling and decreases lung cancer cell proliferation. Grb2 is an essential adaptor molecule with an SH2 domain flanked by two SH3 domains. In this study, we examined whether PR, through interaction between PR-PPD and Grb2-SH3, can interact with Grb2 in cells and breast cancer tissues. Our previous study shows that interaction between PR-PPD and Grb2 could interfere with cytoplasmic signaling and lead to inhibition of EGF-mediated signaling. GST-pulldown analysis shows that PR-PPD specifically interacts with the SH3 domains of Grb2. Immunofluorescence staining shows colocalization of PR and Grb2 in both the nucleus and cytoplasm in BT-474 breast cancer cells. Using Bimolecular Fluorescence Complementation (BiFC) analysis, we show that PR and Grb2 interact in breast cancer cells through the Grb2-SH3 domain. Proximity Ligation Assay (PLA) analysis of 43 breast cancer specimens shows that PR-Grb2 interaction is associated with low histological stage and negatively correlates with lymph node invasion and metastasis in breast cancer. These results, together with our previous findings, suggest that PR-PPD interaction with Grb2 plays an essential role in PR-mediated growth factor signaling inhibition and could contribute significantly to better prognosis in PR- and Grb2-positive breast cancer. Our finding provides a basis for additional studies to explore a novel therapeutic strategy for cancer treatment.


Asunto(s)
Neoplasias de la Mama , Receptores de Progesterona , Humanos , Femenino , Receptores de Progesterona/genética , Neoplasias de la Mama/genética , Factor de Crecimiento Epidérmico , Progesterona , Transducción de Señal/fisiología , Unión Proteica
5.
J Steroid Biochem Mol Biol ; 227: 106230, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36450315

RESUMEN

The role of sex steroid hormones (SSHs) has been shown to modulate cancer cytotoxic treatment sensitivity. Dysregulation of DNA repair associated with genomic instability, abnormal cell survival and not only promotes cancer progression but also resistance to cancer treatment. The three major SSHs, androgen, estrogen, and progesterone, have been shown to interact with several essential DNA repair components. The presence of androgens directly regulates key molecules in DNA double-strand break (DSB) repair. Estrogen can promote cell proliferation and DNA repair, allowing cancer cells to tolerate chemotherapy and radiotherapy. Information on the role of progesterone in DNA repair is limited: progesterone interaction with some DNA repair components has been identified, but the biological significance is still unknown. Here, we review the roles of how each SSH affects DNA repair regulation and modulates response to genotoxic therapies and discuss future research that can be beneficial when combining SSHs with cancer therapy. We also provide preliminary analysis from publicly available databases defining the link between progesterone/PR and DDRs & DNA repair regulation that plausibly contribute to chemotherapy response and breast cancer patient survival.


Asunto(s)
Neoplasias de la Mama , Progesterona , Humanos , Femenino , Reparación del ADN , Roturas del ADN de Doble Cadena , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Estrógenos , Andrógenos
6.
Endocr Relat Cancer ; 30(7)2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37130271

RESUMEN

The presence of progesterone receptor (PR) and PR isoform B (PRB) in breast cancer is generally correlated with better clinical outcomes. Additionally, the significance of hormone-independent effects of PR/PRB correlated with better prognosis has been reported in non-small cell lung cancer (NSCLC). However, the detailed mechanism of that still remains unclear. In this study, we examined how microRNAs (miRNAs) could contribute to tumor inhibition via PR/PRB expression, in order to find miRNAs that have tumor-agnostic effects between breast cancer and NSCLC. We obtained miRNA data using human tissues of breast cancer and NSCLC from The Cancer Genome Atlas (TCGA) database and PCR array from NSCLC patients of our cohort. Subsequently, we examined the function of the miRNA through in vitro study using breast cancer cell lines. As a result, only let-7b expression was significantly correlated with PR expression in both cancers. Additionally, the expression of let-7b significantly inhibited cell proliferation by inducing PR and PRB expression in breast cancer cell lines. However, the positive correlation of let-7b and PRB required a mediated factor, E2 promoter binding factor 1 (E2F1), obtained from TCGA database analysis. In vitro experiments showed that let-7b significantly inhibited E2F1, and E2F1 significantly inhibited PRB. This study revealed that PRB inhibits the proliferation of breast cancer cells by the let-7b-E2F1 interaction. In addition, the immunohistochemical analysis in NSCLC was also consistent with these in vitro data. Our results could contribute to developing novel therapeutic strategies for patients with PR/PRB-positive cancer by targeting let-7b or PRB expression in breast cancer and possibly NSCLC.


Asunto(s)
Neoplasias de la Mama , Carcinoma de Pulmón de Células no Pequeñas , Factor de Transcripción E2F1 , Neoplasias Pulmonares , MicroARNs , Receptores de Progesterona , Femenino , Humanos , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Proliferación Celular , Factor de Transcripción E2F1/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , MicroARNs/genética , Receptores de Progesterona/genética
7.
PLoS One ; 17(3): e0264717, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35235599

RESUMEN

Non-small cell lung cancer (NSCLC) accounts for the majority (80-85%) of all lung cancers. All current available treatments have limited efficacy. The epidermal growth factor receptor (EGFR) plays a critical role in the development and progression of NSCLC, with high EGFR expression associated with increased cell proliferation and poor prognosis. Thus, interfering with EGFR signaling has been shown to effectively reduce cell proliferation and help in the treatment of NSCLC. We previously demonstrated that the progesterone receptor (PR) contains a polyproline domain (PPD) that directly interacts with Src homology 3 (SH3) domain-containing molecules and expression of PR-PPD peptides inhibits NSCLC cell proliferation. In this study, we investigated whether the introduction of PR-PPD by cell-penetrating peptides (CPPs) could inhibit EGF-induced cell proliferation in NSCLC cells. PR-PPD was attached to a cancer-specific CPP, Buforin2 (BR2), to help deliver the PR-PPD into NSCLC cells. Interestingly, addition of BR2-2xPPD peptides containing two PR-PPD repeats was more effective in inhibiting NSCLC proliferation and significantly reduced EGF-induced phosphorylation of Erk1/2. BR2-2xPPD treatment induced cell cycle arrest by inhibiting the expression of cyclin D1 and CDK2 genes in EGFR-wild type A549 cells. Furthermore, the combination treatment of EGFR-tyrosine kinase inhibitors (TKIs), including Gefitinib or Erlotinib, with BR2-2xPPD peptides further suppressed the growth of NSCLC PC9 cells harboring EGFR mutations as compared to EGFR-TKIs treatment alone. Importantly, BR2-2xPPD peptides mediated growth inhibition in acquired Gefitinib- and Erlotinib- resistant lung adenocarcinoma cells. Our data suggests that PR-PPD is the minimal protein domain sufficient to inhibit NSCLC cell growth and has the potential to be developed as a novel NSCLC therapeutic agent.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Péptidos de Penetración Celular , Neoplasias Pulmonares , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular , Péptidos de Penetración Celular/farmacología , Péptidos de Penetración Celular/uso terapéutico , Resistencia a Antineoplásicos , Factor de Crecimiento Epidérmico/farmacología , Factor de Crecimiento Epidérmico/uso terapéutico , Receptores ErbB/genética , Clorhidrato de Erlotinib/uso terapéutico , Gefitinib/farmacología , Gefitinib/uso terapéutico , Humanos , Neoplasias Pulmonares/patología , Péptidos , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Progesterona
8.
Am J Vet Res ; 84(1)2022 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-36480332

RESUMEN

OBJECTIVE: To investigate an alternative treatment for bovine mastitis by using Pm11 antimicrobial peptide. SAMPLE: 5 bovine mastitis pathogens that were previously isolated from cows affected by either clinical or subclinical mastitis. PROCEDURES: The current study introduces Pm11 antimicrobial peptide as an alternative treatment for bovine mastitis. The antibacterial activity of Pm11 was tested against Escherichia coli strain SCM1249, Klebsiella spp strain SCM1282, Staphylococcus aureus strain CM967, Streptococcus agalactiae strain SCM1084, and Streptococcus uberis strain SCM1310 using minimum bactericidal concentrations (MBCs) and time-kill kinetics. The pathogens' morphological changes were demonstrated using a scanning electron microscope (SEM). The cytotoxicity of Pm11 was assessed using the minimum hemolytic concentration assay. RESULTS: MBCs ranged from 2.5 to 10 µM and IC50 ranged from 0.32 to 2.07 µM. Time-kill kinetics at MBC demonstrated that Pm11 reduced viable cell counts of S agalactiae strain SCM1084 and S uberis strain SCM1310 from 105 to 0 CFU/mL within 1 h. E coli strain SCM1249 and S aureus strain CM967 were reduced from 105 to 0 CFU/mL within 4 h. The average Pm11-induced hemolytic activity was < 10% for all Pm11 concentrations tested except at the maximum concentration tested (160 µM: 10.19 ± 2.29%). Based on SEM, Pm11 induced morphological and cellular changes in S aureus and E coli. CLINICAL RELEVANCE: Pm11 antimicrobial peptide demonstrated in vitro antibacterial activity against the common bovine mastitis pathogens E coli, S aureus, S agalactiae, and S uberis, except Klebsiella spp, and should be further investigated in vivo.


Asunto(s)
Enfermedades de los Bovinos , Infecciones por Escherichia coli , Mastitis Bovina , Infecciones Estafilocócicas , Animales , Bovinos , Femenino , Escherichia coli , Mastitis Bovina/tratamiento farmacológico , Mastitis Bovina/microbiología , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Infecciones por Escherichia coli/veterinaria , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/veterinaria , Staphylococcus aureus , Leche
9.
Sci Data ; 8(1): 100, 2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33846359

RESUMEN

Progesterone receptor (PR) isoforms, PRA and PRB, act in a progesterone-independent and dependent manner to differentially modulate the biology of breast cancer cells. Here we show that the differences in PRA and PRB structure facilitate the binding of common and distinct protein interacting partners affecting the downstream signaling events of each PR-isoform. Tet-inducible HA-tagged PRA or HA-tagged PRB constructs were expressed in T47DC42 (PR/ER negative) breast cancer cells. Affinity purification coupled with stable isotope labeling of amino acids in cell culture (SILAC) mass spectrometry technique was performed to comprehensively study PRA and PRB interacting partners in both unliganded and liganded conditions. To validate our findings, we applied both forward and reverse SILAC conditions to effectively minimize experimental errors. These datasets will be useful in investigating PRA- and PRB-specific molecular mechanisms and as a database for subsequent experiments to identify novel PRA and PRB interacting proteins that differentially mediated different biological functions in breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptores de Progesterona/metabolismo , Aminoácidos/química , Línea Celular Tumoral , Femenino , Humanos , Marcaje Isotópico/métodos , Espectrometría de Masas/métodos , Receptores de Progesterona/química , Técnicas del Sistema de Dos Híbridos
10.
J Steroid Biochem Mol Biol ; 198: 105560, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31809870

RESUMEN

Progesterone receptor isoforms A and B exert different biological effects in breast cancer cells. Alteration of PRA/PRB ratio is often observed during breast cancer progression. High PRA/PRB ratios in breast cancer patients are associated with resistance to chemotherapy and poor prognosis. While it is well accepted that PRA and PRB regulate different sets of genes, how the expression of PRA and PRB alters breast cancer proteomes has not been fully investigated. To directly investigate the effects of PR isoform expression on the breast cancer proteome, both in the presence and absence of progestin, PRA and PRB were independently stably expressed in T47DC42 PR-null breast cancer cells using a doxycycline (Dox)-regulated promoter. Dox induction dose-dependently increased PRA and PRB expression. Dox-induced PRA and PRB showed normal receptor localization and were transcriptionally active. Differential quantitative proteomic analysis by stable isotope dimethyl labeling was performed to quantitatively examine how PR isoforms altered global breast cancer proteomes. Cells expressing PRA in the absence of progestin were enriched in proteins involved in the TCA cycle and enriched in proteins involved in glycolysis in the presence of progestin, whilst cells expressing PRB in the absence and presence progestin were significantly enriched in proteins involved in the cell cycle and cell apoptosis pathways. This proteomic data revealed a link between PR isoform expression and alteration in cell metabolism, cell proliferation, and apoptosis. The enrichment of proteins involved in the glycolytic pathway in breast cancer cells expressing PRA is consistent with stem cell-like properties, previously reported in PRA-rich breast cancer cells. Moreover, compared to liganded PRB, liganded PRA differentially upregulated proteins involved in chromatin remodeling, such as linker histone H1.2. Silencing H1.2 gene expression suppressed PRA-mediated cell proliferation and promoted G2/M and S phase entry of the cell cycle. Additionally, liganded PRA upregulated the expression of cathepsin D (CTSD) protease, whose expression is associated with poor prognosis in breast cancer patients. Together, our data demonstrated that the expression of PRA or PRB dramatically and differentially altered breast cancer cell proteomes. These isoform-specific changes in the breast cancer proteome will help to explain the distinct phenotypic properties of breast cancer cells expressing different levels of PRA and PRB.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteómica/métodos , Receptores de Progesterona/metabolismo , Catepsina D/metabolismo , Ciclo Celular , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Femenino , Células HEK293 , Histonas/metabolismo , Humanos , Ligandos , Fenotipo , Pronóstico , Isoformas de Proteínas , Transducción de Señal
11.
J Steroid Biochem Mol Biol ; 199: 105599, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31991170

RESUMEN

Progesterone receptor (PR) is expressed in a wide variety of human tissues, including both reproductive and non-reproductive tissues. Upon binding to the PR, progesterone can display several non-reproductive functions, including neurosteroid activity in the central nervous system, inhibition of smooth muscle contractile activity in the gastrointestinal tract, and regulating the development and maturation of the lung. PR exists as two major isoforms, PRA and PRB. Differential expression of these PR isoforms reportedly contributes to different biological activities of the hormone. However, the distribution of the PR isoforms in human tissues has remained virtually unexplored. In this study, we immunolocalized PR expression in various human tissues using PR (1294) specific antibody, which is capable of detecting both PRA and PRB, and PRB (250H11) specific antibody. Tissues from the uterus, ovary, breast, placenta, prostate, testis, cerebrum, cerebellum, pituitary, spinal cord, esophagus, stomach, small intestine, colon, pancreas, liver, kidney, urinary bladder, lung, heart, aorta, thymus, adrenal gland, thyroid, spleen, skin, and bone were examined in four different age groups (fetal, pediatric, young, and old) in male and female subjects. PR and PRB were detected in the nuclei of cells in the female reproductive system, in both the nuclei and cytoplasm of pituitary gland and pancreatic acinar cells, and only in the cytoplasm of cells in the testis, stomach, small intestine, colon, liver, kidney, urinary bladder, lung, adrenal gland, and skin. Of particular interest, total PRB expression overlapped with that of total PR expression in most tissues but was negative in the female fetal reproductive system. The findings indicate that progesterone could affect diverse human organs differently than from reproductive organs. These findings provide new insights into the novel biological roles of progesterone in non-reproductive organs.


Asunto(s)
Receptores de Progesterona/metabolismo , Distribución Tisular , Adolescente , Adulto , Anciano , Niño , Preescolar , Femenino , Feto/metabolismo , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Embarazo , Progesterona/genética , Progesterona/metabolismo , Isoformas de Proteínas/metabolismo , Receptores de Progesterona/clasificación , Receptores de Progesterona/genética , Receptores de Progesterona/aislamiento & purificación , Reproducción/genética , Adulto Joven
12.
Cancer Res ; 67(18): 8942-51, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17875737

RESUMEN

Estrogen treatment of MCF-7 human breast cancer cells allows the reinitiation of synchronous cell cycle progression in antiestrogen-arrested cells. Here, we report that progestins also reinitiate cell cycle progression in this model. Using clonal cell lines derived from progesterone receptor (PR)-negative MCF-7M13 cells expressing wild-type or mutant forms of PRA and PRB, we show that this effect is mediated via PRB, not PRA. Cell cycle progression did not occur with a DNA-binding domain mutant of PRB but was unaffected by mutation in the NH(2)-terminal, SH3 domain interaction motif, which mediates rapid progestin activation of c-Src. Thus, the progestin-induced proliferative response in antiestrogen-inhibited cells is mediated primarily by the transcriptional activity of PRB. Analysis of selected cell cycle targets showed that progestin treatment induced levels of cyclin D1 expression and retinoblastoma protein (Rb) phosphorylation similar to those induced by estradiol. In contrast, progestin treatment resulted in only a 1.2-fold induction of c-Myc compared with a 10-fold induction by estradiol. These results support the conclusion that progestin, in a PRB-dependent manner, can overcome the growth-inhibitory effects of antiestrogens in estrogen receptor/PR-positive breast cancer cells by the induction of cyclin D1 expression. The mediation of this effect by PRB, but not PRA, further suggests a mechanism whereby abnormal regulation of the normal expression ratios of PR isoforms in breast cancer could lead to the attenuation of antiestrogen-mediated growth arrest.


Asunto(s)
Neoplasias de la Mama/patología , Moduladores de los Receptores de Estrógeno/farmacología , Progestinas/farmacología , Receptores de Progesterona/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Interacciones Farmacológicas , Estradiol/análogos & derivados , Estradiol/farmacología , Fulvestrant , Regulación Neoplásica de la Expresión Génica , Humanos , Pregnenodionas/farmacología , Proteínas Proto-Oncogénicas c-myc , Receptores de Progesterona/biosíntesis , Receptores de Progesterona/genética , Transcripción Genética
13.
J Steroid Biochem Mol Biol ; 193: 105440, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31386890

RESUMEN

Despite recent development in targeted therapies, lung cancer still remains the leading cause of cancer death. Therefore, a better understanding of its pathogenesis and progression could contribute to improving the eventual clinical outcome of the patients. Results of recently published several in vitro and clinical studies indicated the possible involvement of sex steroids in both development and progression of non-small cell lung carcinoma (NSCLC). Therefore we summarized the reported clinical relevant information of the sex steroids, their receptors and steroid metabolizing enzymes related to NSCLC in this mini-review. In addition, we also reviewed the potential "endocrine therapy", targeting sex steroid actions and/or metabolism in NSCLC patients.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Hormonas Esteroides Gonadales/metabolismo , Neoplasias Pulmonares/metabolismo , Animales , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Terapia de Reemplazo de Hormonas , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Receptores Androgénicos/metabolismo , Receptores de Progesterona/metabolismo
14.
J Steroid Biochem Mol Biol ; 190: 212-223, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30926428

RESUMEN

The progesterone receptor (PR) has been reported to play important roles in lung development and function, such as alveolarization, alveolar fluid clearance (AFC) and upper airway dilator muscle activity. In the lung, pulmonary neuroendocrine cells (PNECs) are important in the etiology and progression of lung neuroendocrine tumors (NETs). Women with lung NETs had significantly better survival rates than men, suggesting that sex steroids and their receptors, such as the PR, could be involved in the progression of lung NETs. The PR exists as two major isoforms, PRA and PRB. How the expression of different PR isoforms affects proliferation and the development of lung NETs is not well understood. To determine the role of the PR isoforms in PNECs, we constructed H727 lung NET cell models expressing PRB, PRA, Green Fluorescence Protein (GFP) (control). The expression of PRB significantly inhibited H727 cell proliferation better than that of PRA in the absence of progestin. The expression of the unrelated protein, GFP, had little to no effect on H727 cell proliferation. To better understand the role of the PR isoform in PNECs, we examined PR isoform expression in PNECs in lung tissues. A monoclonal antibody specific to the N-terminus of PRB (250H11 mAb) was developed to specifically recognize PRB, while a monoclonal antibody specific to a common N-terminus epitope present in both PRA and PRB (1294 mAb) was used to detect both PRA and PRB. Using these PR and PRB-specific antibodies, we demonstrated that PR (PRA&PRB) and PRB were expressed in the PNECs of the normal fetal and adult lung, with significantly higher PR expression in the fetal lung. Interestingly, PRB expression in the normal lung was associated with lower cell proliferation than PR expression, suggesting a distinct role of PRB in the PNECs. A better understanding of the molecular mechanism of PR and PR isoform signaling in lung NET cells may help in developing novel therapeutic strategies that will benefit lung NET patients in the future.


Asunto(s)
Proliferación Celular , Pulmón/citología , Células Neuroendocrinas/metabolismo , Receptores de Progesterona/genética , Adulto , Línea Celular , Femenino , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Pulmón/embriología , Pulmón/metabolismo , Neoplasias Pulmonares/genética , Masculino , Tumores Neuroendocrinos/genética , Receptores de Progesterona/análisis
15.
Steroids ; 73(9-10): 922-8, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18321550

RESUMEN

Human progesterone receptor (PR) contains a polyproline motif in the amino-terminal domain that interacts with the SH3 domain of Src and mediates rapid activation of c-Src and downstream MAPK (Erk-1/-2) independent of the transcriptional activity of PR. Forcedly target PR to different locations in the cell by use of mutations or tags for different cell compartments showed that progestin activation of Src/MAPK is mediated by PR outside the nucleus. No distinction could be made between the cytoplasm and cell membrane as the site of PR activation of Src. Therefore we can only conclude that this is an extra-nuclear action of PR. Interestingly, the B isoform of PR which is naturally distributed between cytoplasm and nucleus mediated progestin activation of Src/MAPK, whereas PR-A that is predominantly nuclear failed to do so indicating that the two PR isoforms have distinct abilities to mediate rapid activation of signaling pathways. Due to distinct cellular locations, progestin activation of Src/MAPK signaling can regulate selected target genes such as cyclin D1 (CCND1) that lack direct PR binding response elements (PREs). Progestin induction of CCND1 was observed in cells expressing PR-B but not PR-BDeltaSH3 or PR-A and induction in the presence of PR-B was dramatically reduced in the presence of inhibitors of Src or MAPK. In contrast progestin induction of Sgk (serum and glucocorticoid regulated kinase) gene, which contains a classical PRE, was observed with both PR isoforms as well as PR-BDeltaSH3 and was unaffected by Src and MAPK inhibitors. PR bound to enhancer region of Sgk in a progestin dependent manner as detected by chromatin co-immunoprecipitation (ChIP) whereas no PR binding to CCDN1 was observed. Consistent with CCND1 data, progestin stimulation of cell cycle progression was only observed in cells expressing PR-B but not cells expressing PR-BDeltaSH3 or PR-A. These results demonstrate the importance of PR activation of extra-nuclear signaling pathways in regulating selected target genes and cell cycle progression.


Asunto(s)
Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Receptores de Progesterona/metabolismo , Animales , Ciclo Celular , Ciclina D1/metabolismo , Humanos , Ratones , Modelos Biológicos , Progestinas/metabolismo , Isoformas de Proteínas , Estructura Terciaria de Proteína , Transducción de Señal , Proteína Elk-1 con Dominio ets/metabolismo , Familia-src Quinasas/metabolismo
16.
Mol Endocrinol ; 21(2): 359-75, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17138644

RESUMEN

Human progesterone receptor (PR) contains a motif that interacts with the SH3 domain of Src and mediates rapid activation of Src and downstream MAPK (Erk-1/-2) without relying on the transcriptional activity of the receptor. Here we investigated the role and intracellular location of this nontranscriptional activity of PR. Progestin activation of Src/MAPK occurred outside the nucleus with the B isoform of PR that was distributed between the cytoplasm and nucleus, but not with PR-A that was predominantly nuclear. Breast cancer cells stably expressing wild-type PR-B or PR-B with disrupting point mutations in the SH3 domain binding motif (PR-BDeltaSH3) that do not affect the transcriptional activity of PR, were compared for effects of progestin on endogenous target gene expression and cell proliferation. Progestin induction of the cyclin D1 gene, which lacks a progesterone response element, was dependent on PR activation of the Src/MAPK pathway, whereas induction of the Sgk (serum and glucocorticoid regulated kinase) gene that contains a functional progesterone response element was unaffected by mutations that interfere with PR activation of Src. Progestin induction of cell cycle progression was also abrogated in cells expressing PR-BDeltaSH3, and no effect of progestin on cyclin D1 expression and cell cycle was observed in the presence of PR-A. These results highlight the importance of PR activation of the Src/MAPK signaling pathway for progesterone-induced transcription of select target genes and cell cycle progression.


Asunto(s)
Ciclo Celular , Regulación Neoplásica de la Expresión Génica , Progesterona/fisiología , Receptores de Progesterona/fisiología , Animales , Línea Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Activación Enzimática/efectos de los fármacos , Femenino , Genes bcl-1 , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Congéneres de la Progesterona/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Progesterona/agonistas , Receptores de Progesterona/genética , Transducción de Señal , Dominios Homologos src , Familia-src Quinasas/metabolismo
17.
Mol Endocrinol ; 21(6): 1335-58, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17440047

RESUMEN

Accumulating evidence indicates that progestins are involved in controlling mammary gland tumorigenesis. Here, we assessed the molecular mechanisms of progestin action in breast cancer models with different phenotypes. We examined C4HD cells, an estrogen (ER) and progesterone (PR) receptor-positive murine breast cancer model in which progestins exert sustained proliferative response, the LM3 murine metastatic mammary tumor cell line, which lacks PR and ER expression, and human PR null T47D-Y breast cancer cells. In addition to acting as a transcription factor, PR can also function as an activator of signaling pathways. To explore which of these two functions were involved in progestin responses, reconstitution experiments in the PR-negative models were performed with wild-type PR-B, with a DNA binding mutant C587A-PR, and with mutant PR-BmPro, which lacks the ability to activate cytoplasm signaling pathways. We found that in a cell context either ER-positive or -negative, progestins induced cell growth and modulation of matrix metalloproteinases-9 (MMP-9) and -2 (MMP-2), and urokinase-type plasminogen activator (uPA) activities, via MAPK and phosphatidylinositol 3-kinase/Akt pathways, in cells expressing wild-type PR-B or DNA binding mutant C587A-PR. In contrast, in cells expressing mutant PR-BmPro, progestins did not induce growth. We also found that unliganded PR expression conferred breast cancer cells an in vitro less proliferative phenotype, as compared with cells lacking PR expression. Modulation of this behavior occurred when PR was functioning either as transcription factor or as signaling activator. Finally, we for the first time demonstrated that progestins favor development of breast tumor metastasis via PR function as activator of signaling pathways. Our present findings provide mechanistic support to the design of a novel therapeutic intervention in PR-positive breast tumors involving blockage of PR capacity to activate cytoplasmic signaling.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Péptido Hidrolasas/metabolismo , Progestinas/farmacología , Receptores de Progesterona/metabolismo , Animales , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citoplasma/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Metástasis de la Neoplasia , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Receptores de Progesterona/genética , Transducción de Señal , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
18.
Mol Cell Endocrinol ; 466: 51-72, 2018 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-29146555

RESUMEN

Estrogen and progesterone play essential roles in the development and progression of breast cancer. Over 70% of breast cancers express estrogen receptors (ER) and progesterone receptors (PR), emphasizing the need for better understanding of ER and PR signaling. ER and PR are traditionally viewed as transcription factors that directly bind DNA to regulate gene networks. In addition to nuclear signaling, ER and PR mediate hormone-induced, rapid extranuclear signaling at the cell membrane or in the cytoplasm which triggers downstream signaling to regulate rapid or extended cellular responses. Specialized membrane and cytoplasmic proteins may also initiate hormone-induced extranuclear signaling. Rapid extranuclear signaling converges with its nuclear counterpart to amplify ER/PR transcription and specify gene regulatory networks. This review summarizes current understanding and updates on ER and PR extranuclear signaling. Further investigation of ER/PR extranuclear signaling may lead to development of novel targeted therapeutics for breast cancer management.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Animales , Mama/patología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Membrana Celular/química , Membrana Celular/metabolismo , Femenino , Técnicas de Inactivación de Genes , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Ratones , Receptores de Estrógenos/química , Receptores de Estrógenos/genética , Receptores de Progesterona/química , Receptores de Progesterona/genética
19.
Semin Reprod Med ; 25(3): 139-53, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17447204

RESUMEN

Sex steroid hormones, including estrogen, progesterone, and androgen, mediate their biological effects on cell proliferation, differentiation, and homeostasis through their respective nuclear receptors. In addition to functioning as ligand-activated nuclear transcription factors to regulate gene transcription, these receptors also have been shown to mediate rapid activation of non-genomic signaling pathways independent of their transcriptional activity. Despite the fact that non-genomic effects of sex steroids have been observed since more than three decades ago, the receptor mechanisms mediating these rapid effects still are not well understood. A subpopulation of nuclear steroid receptors localized to the cell membrane or cytoplasm has been proposed to mediate steroid hormone activation of signaling pathways; however, novel membrane receptors unrelated to nuclear receptors have also been implicated. This review focuses on recent advances in our understanding of the nature of the receptors and mechanisms responsible for rapid non-genomic signaling actions of sex steroids, including novel membrane receptors and interactions of nuclear steroid receptors with membrane and cytoplasmic signaling molecules such as adapter proteins, G proteins, ion channels, and protein kinases. A better definition of receptor mechanisms involved in mediating activation of non-genomic signaling pathways is important to our overall understanding of the biology of steroid hormones.


Asunto(s)
Hormonas Esteroides Gonadales/metabolismo , Receptores de Esteroides/metabolismo , Transducción de Señal , Andrógenos/metabolismo , Animales , Estrógenos/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Canales Iónicos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Progesterona/metabolismo , Receptores Androgénicos/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Progesterona/metabolismo , Familia-src Quinasas/metabolismo
20.
Mol Cell Biol ; 22(15): 5451-66, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12101239

RESUMEN

The progesterone receptor (PR) contains two transcription activation function (AF) domains, constitutive AF-1 in the N terminus and AF-2 in the C terminus. AF-2 activity is mediated by a hormone-dependent interaction with a family of steroid receptor coactivators (SRCs). SRC-1 can also stimulate AF-1 activity through a secondary domain that interacts simultaneously with the primary AF-2 interaction site. Other protein interactions and mechanisms that mediate AF-1 activity are not well defined. By interaction cloning, we identified an AP-1 family member, Jun dimerization protein 2 (JDP-2), as a novel PR-interacting protein. JDP-2 was first defined as a c-Jun interacting protein that functions as an AP-1 repressor. PR and JDP-2 interact directly in vitro through the DNA binding domain (DBD) of PR and the basic leucine zipper (bZIP) region of JDP-2. The two proteins also physically associate in mammalian cells, as detected by coimmunoprecipitation, and are recruited in vivo to a progesterone-inducible target gene promoter, as detected by a chromatin immunoprecipitation (ChIP) assay. In cell transfection assays, JDP-2 substantially increased hormone-dependent PR-mediated transactivation and worked primarily by stimulating AF-1 activity. JDP-2 is a substantially stronger coactivator of AF-1 than SRC-1 and stimulates AF-1 independent of SRC-1 pathways. The PR DBD is necessary but not sufficient for JDP-2 stimulation of PR activity; the DBD and AF-1 are required together. JDP-2 lacks an intrinsic activation domain and makes direct protein interactions with other coactivators, including CBP and p300 CBP-associated factor (pCAF), but not with SRCs. These results indicate that JDP-2 stimulates AF-1 activity by the novel mechanism of docking to the DBD and recruiting or stabilizing N-terminal PR interactions with other general coactivators. JDP-2 has preferential activity on PR among the nuclear receptors tested and is expressed in progesterone target cells and tissues, suggesting that it has a physiological role in PR function.


Asunto(s)
Receptores de Progesterona/metabolismo , Proteínas Represoras/metabolismo , Animales , Células COS , Línea Celular , Cromatina/metabolismo , Genes Reporteros , Glutatión Transferasa/genética , Células HeLa , Histona Acetiltransferasas , Antagonistas de Hormonas/farmacología , Humanos , Leucina Zippers/fisiología , Sustancias Macromoleculares , Virus del Tumor Mamario del Ratón/genética , Datos de Secuencia Molecular , Coactivador 1 de Receptor Nuclear , Pruebas de Precipitina , Congéneres de la Progesterona/farmacología , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/fisiología , Unión Proteica/fisiología , Estructura Terciaria de Proteína/fisiología , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/farmacología , Homología de Secuencia de Aminoácido , Factores de Transcripción/farmacología , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/fisiología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA