Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Rheumatology (Oxford) ; 56(8): 1254-1263, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27686101

RESUMEN

RA is the most common chronic systemic autoimmune disease, with a higher prevalence in women, suggesting female hormonal factors play a role in the development of the disease. However, many controversies still exist. The aim of this review was to appraise data from recent research concerning female hormonal factors and their association with RA disease development. The study of female hormonal factors is challenging because serum levels may differ throughout a woman's lifetime and interact with various environmental, immunological, genetic and endocrine factors influencing the development of autoimmunity. As some female hormonal factors may be potentially modifiable, understanding their impact on RA development is clinically relevant and may result in specific preventive interventions in high-risk populations.


Asunto(s)
Artritis Reumatoide/inmunología , Estrógenos/inmunología , Hormonas Gonadales/inmunología , Progesterona/inmunología , Artritis Reumatoide/sangre , Autoinmunidad , Estrógenos/sangre , Femenino , Hormonas Gonadales/sangre , Humanos , Progesterona/sangre , Factores de Riesgo , Factores Sexuales
2.
FASEB J ; 25(12): 4326-37, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21885654

RESUMEN

Inflammation is intimately linked with naturally occurring remodeling events in the endometrium. Lipoxins comprise a group of short-lived, nonclassic eicosanoids possessing potent anti-inflammatory and proresolution properties. In the present study, we investigated the role of lipoxin A(4) (LXA(4)) in the endometrium and demonstrated that 15-LOX-2, an enzyme necessary for LX biosynthesis, is expressed in this tissue. Our results establish that LXA(4) possesses robust estrogenic activity through its capacity to alter ERE transcriptional activity, as well as expression of estrogen-regulated genes, alkaline phosphatase activity, and proliferation in human endometrial epithelial cells. Interestingly, LXA(4) also demonstrated antiestrogenic potential, significantly attenuating E2-induced activity. This estrogenic activity was directly mediated through estrogen receptors (ERs). Subsequent investigations determined that the actions of LXA(4) are exclusively mediated through ERα and closely mimic those of the potent estrogen 17ß-estradiol (E2). In binding assays, LXA(4) competed with E2 for ER binding, with an IC(50) of 46 nM. Furthermore, LXA(4) exhibited estrogenic activity in vivo, increasing uterine wet weight and modulating E2-regulated gene expression. These findings reveal a previously unappreciated facet of LXA(4) bioactions, implicating this lipid mediator in novel immunoendocrine crosstalk mechanisms.


Asunto(s)
Moduladores de los Receptores de Estrógeno/metabolismo , Lipoxinas/metabolismo , Fosfatasa Alcalina/metabolismo , Araquidonato 15-Lipooxigenasa/metabolismo , Secuencia de Bases , Unión Competitiva , Línea Celular , Proliferación Celular/efectos de los fármacos , Endometrio/citología , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Estradiol/metabolismo , Moduladores de los Receptores de Estrógeno/farmacología , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Lipoxinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Formil Péptido/antagonistas & inhibidores , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/antagonistas & inhibidores , Receptores de Lipoxina/genética , Receptores de Lipoxina/metabolismo , Transducción de Señal
4.
FASEB J ; 21(12): 3162-70, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17496159

RESUMEN

Migration of neutrophils (PMN) across epithelia is a pathological hallmark of numerous mucosal diseases. Whereas lesions at mucosal surfaces are generally self-limiting, endogenous mechanisms of resolution are incompletely understood. Previous studies revealed that resolvins directly act on PMN to attenuate transendothelial migration, less is known about the influence of resolvins on PMN-epithelial interactions and whether they act on epithelia. We studied the dynamics of resolvin E1 (RvE1) actions on leukocyte transepithelial migration. PMN exposure to RvE1 or chemerin (peptide agonist of ChemR23) reduced transepithelial migration in a concentration-dependent manner. Conversely, activation of epithelial ChemR23 promoted apical clearance of PMN. A nonbiased screen of known PMN ligands expressed on epithelial cells in response to RvE1 revealed selective induction of CD55, an apically expressed antiadhesive molecule. CD55 promoter analysis demonstrated that both RvE1 and chemerin activate the CD55 promoter. Inhibition of CD55 by neutralizing antibody prevented RvE1-dependent augmentation of apical PMN clearance. Taken together these findings implicate a "two-hit" model of inflammatory resolution, whereby activation of the PMN RvE1 receptor attenuates transepithelial migration and subsequent actions on the epithelium promote CD55-dependent clearance of PMN across the epithelial cell surface promoting active inflammatory resolution.


Asunto(s)
Movimiento Celular/fisiología , Ácido Eicosapentaenoico/análogos & derivados , Células Epiteliales , Inflamación/inmunología , Neutrófilos/inmunología , Animales , Antígenos CD55/genética , Antígenos CD55/metabolismo , Línea Celular , Quimiocinas/metabolismo , Ácido Eicosapentaenoico/metabolismo , Células Epiteliales/citología , Células Epiteliales/inmunología , Células Epiteliales/fisiología , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Membrana Mucosa/citología , Membrana Mucosa/inmunología , Neutrófilos/citología , Neutrófilos/fisiología , Regiones Promotoras Genéticas
5.
Reprod Sci ; 24(1): 67-76, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27217374

RESUMEN

The objective of the present study was to test the ability of OSU-03012 (2-amino-N-[4-[5-phenanthren-2-yl-3-(trifluoromethyl)pyrazol-1-yl]phenyl]acetamide), a novel and potent celecoxib-derivative, to impair endometriosis progression in in vitro and in vivo models based on its ability to indirectly block Y-box-binding protein 1 (YB-1) function. 12Z human endometriotic epithelial cells and sexually mature female C57BL/6J mice were treated with OSU-03012. Cellular proliferation was quantified by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromid assay. Expression of YB-1 and phosphorylated YB-1 in 12Z cells and endometriotic lesions was evaluated by Western blotting and immunohistochemistry (IHC). The IHC for proliferating cell nuclear antigen was performed. OSU-03012 treatment resulted in decreased YB-1 and its phosphorylated form in both in vitro and in vivo models. Endometriotic lesion size was significantly reduced in OSU-03012-treated mice (27.6 ± 4.0 mm3) compared to those from the control group (50.5 ± 6.9 mm3, P < .0001). A significant reduction in endometriotic epithelial cell proliferation was observed in endometriotic lesions exposed to OSU-03012 treatment ( P = .0346). In conclusion, targeting YB-1 via OSU-03012 showed a potent antiproliferative effect on endometriotic epithelial cells in vitro and in a mouse model of disease.

6.
Methods Mol Biol ; 341: 17-35, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16799186

RESUMEN

Epithelial cells at all mucosal surfaces are potentially apposed to bacteria, particularly in the intestine. It is established that intestinal epithelial cells (IECs) represent an important barrier between lamina propria cells and the potentially harmful lumenal contents. In addition, IECs are important immunoeffector cells with the capacity to release cytokines, chemokines, and other molecules involved in antigen presentation and immune defense. The interaction of IECs with intestinal bacteria can result in a decrease in barrier function and the development of inflammation, which is known to be an important factor in the development of intestinal pathology. The potential role of such crosstalk between bacteria and other intestinal cell types in normal physiology and/or pathophysiology is therefore a topic of intense investigation. In this chapter, we provide protocols for the identification of bacteria that are associated with the epithelium and mucosa in addition to functional assays examining the interactions of neutrophils with epithelial cells and epithelial cell-mediated killing of bacteria.


Asunto(s)
Presentación de Antígeno/inmunología , Bacterias/inmunología , Células Epiteliales/inmunología , Inmunidad Mucosa , Mucosa Intestinal/inmunología , Intestinos/inmunología , Animales , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/patología , Técnicas de Tipificación Bacteriana , Comunicación Celular , Línea Celular , Citocinas/inmunología , Células Epiteliales/patología , Humanos , Inflamación/inmunología , Inflamación/patología , Mucosa Intestinal/patología , Intestinos/patología , Membrana Mucosa/inmunología , Membrana Mucosa/patología
7.
Life Sci ; 78(9): 920-5, 2006 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-16185718

RESUMEN

Clostridium difficile infection of the intestinal epithelium and consequent pseudomembranous colitis is an important cause of morbidity and mortality. Pathogenesis has been ascribed exclusively to toxin production. Using in vitro models of human intestinal epithelial layers, we show that exposure to toxigenic C. difficile upregulates epithelial expression of IL-8 and ICAM-1, two molecules important in neutrophil chemoattraction and adhesion and subsequent inflammation. IL-8 production was also stimulated by toxin-containing supernatants. C difficile induced IL-8 release was inhibited by specific antiserum. Increased ICAM-1 expression only occurred after basolateral exposure to C. difficile while apical exposure had no effect on ICAM-1 expression. However, transepithelial electrical resistance was impaired by apical exposure to bacterial suspensions. We suggest that apical exposure to C. difficile induces changes in epithelial layer integrity which allows the bacteria and/or the toxin access to the basolateral compartment where pathogenic inflammatory mechanisms are activated.


Asunto(s)
Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/toxicidad , Clostridioides difficile/metabolismo , Enterotoxinas/toxicidad , Células Epiteliales/metabolismo , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Anticuerpos Bloqueadores/farmacología , Biomarcadores , Conductividad Eléctrica , Células Epiteliales/citología , Células HT29 , Humanos , Inmunoensayo , Inflamación/microbiología , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-8/metabolismo , Mucosa Intestinal/citología
8.
Artículo en Inglés | MEDLINE | ID: mdl-26779118

RESUMEN

Endometriosis affects approximately 10% of women of reproductive age. This chronic, gynecological inflammatory disease results in a decreased quality of life for patients, with the main symptoms including chronic pelvic pain and infertility. The steroid hormone 17-ß Estradiol (E2) plays a key role in the pathology. Our previous studies showed that the anti-inflammatory lipid Lipoxin A4 (LXA4) acts as an estrogen receptor-alpha agonist in endometrial epithelial cells, inhibiting certain E2-mediated effects. LXA4 also prevents the progression of endometriosis in a mouse model via anti-proliferative mechanisms and by impacting mediators downstream of ER signaling. The aim of the present study was therefore to examine global proteomic changes evoked by E2 and LXA4 in endometriotic epithelial cells. E2 impacted a greater number of proteins in endometriotic epithelial cells than LXA4. Interestingly, the combination of E2 and LXA4 resulted in a reduced number of regulated proteins, with LXA4 mediating a suppressive effect on E2-mediated signaling. These proteins are involved in diverse pathways of relevance to endometriosis pathology and metabolism, including mRNA translation, growth, proliferation, proteolysis, and immune responses. In summary, this study sheds light on novel pathways involved in endometriosis pathology and further understanding of signaling pathways activated by estrogenic molecules in endometriotic epithelial cells.

9.
PLoS One ; 9(2): e89742, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24587003

RESUMEN

Endometriosis, a leading cause of pelvic pain and infertility, is characterized by ectopic growth of endometrial-like tissue and affects approximately 176 million women worldwide. The pathophysiology involves inflammatory and angiogenic mediators as well as estrogen-mediated signaling and novel, improved therapeutics targeting these pathways are necessary. The aim of this study was to investigate mechanisms leading to the establishment and progression of endometriosis as well as the effect of local treatment with Lipoxin A4 (LXA4), an anti-inflammatory and pro-resolving lipid mediator that we have recently characterized as an estrogen receptor agonist. LXA4 treatment significantly reduced endometriotic lesion size and downregulated the pro-inflammatory cytokines IL-1ß and IL-6, as well as the angiogenic factor VEGF. LXA4 also inhibited COX-2 expression in both endometriotic lesions and peritoneal fluid cells, resulting in attenuated peritoneal fluid Prostaglandin E2 (PGE2) levels. Besides its anti-inflammatory effects, LXA4 differentially regulated the expression and activity of the matrix remodeling enzyme matrix metalloproteinase (MMP)-9 as well as modulating transforming growth factor (TGF)-ß isoform expression within endometriotic lesions and in peritoneal fluid cells. We also report for first time that LXA4 attenuated aromatase expression, estrogen signaling and estrogen-regulated genes implicated in cellular proliferation in a mouse model of disease. These effects were observed both when LXA4 was administered prior to disease induction and during established disease. Collectively, our findings highlight potential targets for the treatment of endometriosis and suggest a pleotropic effect of LXA4 on disease progression, by attenuating pro-inflammatory and angiogenic mediators, matrix remodeling enzymes, estrogen metabolism and signaling, as well as downstream proliferative pathways.


Asunto(s)
Vías Biosintéticas/efectos de los fármacos , Dinoprostona/biosíntesis , Endometriosis/prevención & control , Estrógenos/metabolismo , Lipoxinas/farmacología , Transducción de Señal/efectos de los fármacos , Análisis de Varianza , Animales , Aromatasa/metabolismo , Ciclooxigenasa 2/metabolismo , Cartilla de ADN/genética , Endometriosis/fisiopatología , Femenino , Inmunohistoquímica , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
PLoS One ; 8(12): e82512, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24324802

RESUMEN

BACKGROUND/AIMS: The neural cell adhesion molecule L1CAM is a transmembrane glycoprotein abnormally expressed in tumors and previously associated with cell proliferation, adhesion and invasion, as well as neurite outgrowth in endometriosis. Being an attractive target molecule for antibody-based therapy, the present study assessed the ability of the monoclonal anti-L1 antibody (anti-L1 mAb) to impair the development of endometriotic lesions in vivo and endometriosis-associated nerve fiber growth. METHODS AND RESULTS: Endometriosis was experimentally induced in sexually mature B6C3F1 (n=34) and CD-1 nude (n=21) mice by autologous and heterologous transplantation, respectively, of endometrial fragments into the peritoneal cavity. Transplantation was confirmed four weeks post-surgery by in vivo magnetic resonance imaging and laparotomy, respectively. Mice were then intraperitoneally injected with anti-L1 mAb or an IgG isotype control antibody twice weekly, over a period of four weeks. Upon treatment completion, mice were sacrificed and endometrial implants were excised, measured and fixed. Endometriosis was histologically confirmed and L1CAM was detected by immunohistochemistry. Endometriotic lesion size was significantly reduced in anti-L1-treated B6C3F1 and CD-1 nude mice compared to mice treated with control antibody (P<0.05). Accordingly, a decreased number of PCNA positive epithelial and stromal cells was detected in autologously and heterologously induced endometriotic lesions exposed to anti-L1 mAb treatment. Anti-L1-treated mice also presented a diminished number of intraperitoneal adhesions at implantation sites compared with controls. Furthermore, a double-blind counting of anti-neurofilament L stained nerves revealed significantly reduced nerve density within peritoneal lesions in anti-L1 treated B6C3F1 mice (P=0.0039). CONCLUSIONS: Local anti-L1 mAb treatment suppressed endometriosis growth in B6C3F1 and CD-1 nude mice and exerted a potent anti-neurogenic effect on induced endometriotic lesions in vivo. The findings of this preliminary study in mice provide a strong basis for further testing in in vivo models.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Endometriosis/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Adulto , Animales , Anticuerpos Monoclonales/administración & dosificación , Adhesión Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Endometriosis/tratamiento farmacológico , Endometriosis/patología , Femenino , Humanos , Ratones , Molécula L1 de Adhesión de Célula Nerviosa/antagonistas & inhibidores , Proteínas de Neurofilamentos/metabolismo , Adulto Joven
12.
Fertil Steril ; 99(7): 1965-73.e2, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23472950

RESUMEN

OBJECTIVE: To compare the expression of the prostaglandin (PG) E(2) transporter multidrug resistance-associated protein 4 (MRP4) in eutopic and ectopic endometrial tissue from endometriosis patients with that of control subjects and to examine whether MRP4 is regulated by the antiinflammatory lipid lipoxin A(4) (LXA(4)) in endometriotic epithelial cells. DESIGN: Molecular analysis in human samples and a cell line. SETTING: Two university hospitals and a private clinic. PATIENT(S): A total of 59 endometriosis patients and 32 age- and body mass index-matched control subjects undergoing laparoscopy or hysterectomy. INTERVENTION(S): Normal, eutopic, and ectopic endometrial biopsies as well as peritoneal fluid were obtained during surgery performed during the proliferative phase of the menstrual cycle. 12Z endometriotic epithelial cells were used for in vitro mechanistic studies. MAIN OUTCOME MEASURE(S): Tissue MRP4 mRNA levels were quantified by quantitative reverse-transcription polymerase chain reaction (qRT-PCR), and localization was analyzed with the use of immunohistochemistry. Cellular MRP4 mRNA and protein were quantified by qRT-PCR and Western blot, respectively. PGE(2) was measured in peritoneal fluid and cell supernatants using an enzyme immunoassay (EIA). RESULT(S): MRP4 was expressed in eutopic and ectopic endometrium, where it was overexpressed in peritoneal lesions and localized in the cytoplasm of glandular epithelial cells. LXA(4) attenuated MRP4 mRNA and protein levels in endometriotic epithelial cells in a dose-dependent manner, while not affecting the expression of enzymes involved in PGE(2) metabolism. Investigations employing receptor antagonists and small interfering RNA revealed that this occurred through estrogen receptor α. Accordingly, LXA(4) treatment inhibited extracellular PGE(2) release. CONCLUSION(S): We report for the first time that MRP4 is expressed in human endometrium, elevated in peritoneal endometriosis, and modulated by LXA(4) in endometriotic epithelial cells.


Asunto(s)
Endometriosis/metabolismo , Endometrio/metabolismo , Células Epiteliales/metabolismo , Lipoxinas/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Enfermedades Peritoneales/metabolismo , Adulto , Líquido Ascítico/metabolismo , Biopsia , Western Blotting , Estudios de Casos y Controles , Línea Celular , Dinoprostona/metabolismo , Endometriosis/genética , Endometriosis/cirugía , Endometrio/efectos de los fármacos , Endometrio/cirugía , Células Epiteliales/efectos de los fármacos , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Histerectomía , Técnicas para Inmunoenzimas , Inmunohistoquímica , Laparoscopía , Persona de Mediana Edad , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Enfermedades Peritoneales/genética , Enfermedades Peritoneales/cirugía , Interferencia de ARN , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Regulación hacia Arriba , Adulto Joven
13.
Fertil Steril ; 98(5): 1200-8, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22884659

RESUMEN

OBJECTIVE: To analyze the expression of estrogen receptors α and ß as well as their target genes implicated in proliferation, c-myc, cyclin D1, and GREB1, in the endometrium of women with or without endometriosis. DESIGN: Expression analysis in human tissue. SETTING: University hospitals and a clinic. PATIENT(S): Ninety-one premenopausal women (59 patients with endometriosis and 32 controls) undergoing laparoscopic surgery. INTERVENTION(S): Biopsies were obtained at time of surgery, performed during the proliferative phase of the cycle. MAIN OUTCOME MEASURE(S): Estrogen receptors α and ß as well as c-myc, cyclin D1, and GREB1 mRNA expression levels were determined by quantitative reverse transcriptase-polymerase chain reaction. Tissue localization of these estrogen-regulated genes was analyzed by immunohistochemistry. RESULT(S): Estrogen receptors α and ß as well as c-myc, cyclin D1, and GREB1 mRNA expression levels were increased in ectopic tissue in comparison with both normal and eutopic endometrium. Estrogen receptor mRNA levels also were upregulated in the eutopic peritoneal tissue of patients with endometriosis. Cyclin D1 and GREB1 expression was augmented in eutopic endometrium. c-myc, cyclin D1, and GREB1 proteins exhibited a nuclear localization in ectopic endometrial tissue. CONCLUSION(S): This constitutes the first report of increased expression of GREB1, as well as cyclin D1 and c-myc, in peritoneal endometriotic lesions, implicating these proteins in estrogen-dependent growth in this context.


Asunto(s)
Proliferación Celular , Coristoma/metabolismo , Ciclina D1/análisis , Endometriosis/metabolismo , Endometrio , Receptor alfa de Estrógeno/análisis , Receptor beta de Estrógeno/análisis , Proteínas de Neoplasias/análisis , Enfermedades Peritoneales/metabolismo , Proteínas Proto-Oncogénicas c-myc/análisis , Adulto , Biopsia , Estudios de Casos y Controles , Coristoma/genética , Coristoma/patología , Coristoma/cirugía , Ciclina D1/genética , Endometriosis/genética , Endometriosis/patología , Endometriosis/cirugía , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Femenino , Regulación de la Expresión Génica , Alemania , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Enfermedades Peritoneales/genética , Enfermedades Peritoneales/patología , Enfermedades Peritoneales/cirugía , Proteínas Proto-Oncogénicas c-myc/genética , ARN Mensajero/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Suiza , Adulto Joven
14.
Mol Cell Endocrinol ; 345(1-2): 27-37, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21784129

RESUMEN

The pro-inflammatory cytokine TNF-α and the female hormone estrogen have been implicated in the pathophysiology of two common gynecological diseases, endometriosis and endometrial adenocarcinoma. Here we describe a novel capacity of TNF-α to activate ER signaling in endometrial epithelial cells. TNF-α induced luciferase expression in the absence and presence of estradiol and also augmented expression of the estrogen-regulated genes c-fos, GREB1, and progesterone receptor. Furthermore, TNF-α mediated ER transcriptional activity is dependent on the Extracellular Regulated Kinase (ERK) 1/2 pathway. Co-treatment with a pure ER antagonist resulted in an inhibition of this TNF-α-induced ERE luciferase activity and gene expression, demonstrating that this cytokine signals through ERs. Additional investigations confirmed that TNF-α acts specifically via ERα. Taken together, these data provide a rationale for the potential use of inhibitors of TNF-α and estrogen production/activity in combination for the treatment of endometrial pathologies.


Asunto(s)
Endometrio/citología , Células Epiteliales/metabolismo , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Línea Celular , Activación Enzimática/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Estradiol/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Luciferasas/metabolismo , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptor Cross-Talk/efectos de los fármacos , Elementos de Respuesta/genética , Transducción de Señal/genética , Transcripción Genética
15.
J Reprod Immunol ; 92(1-2): 88-96, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21940052

RESUMEN

Immunity and hormonal responses in the reproductive tissues of postmenopausal women are poorly understood. Secretory leukocyte protease inhibitor (SLPI), a multifunctional antimicrobial protein expressed at mucosal surfaces, is thought to play a key role in infectious and inflammatory contexts. The aim of this study was to measure SLPI production along the female reproductive tract in postmenopausal women with and without hormonal treatment. We additionally quantified estrogen receptor alpha (ERα) and progesterone receptor A (PRA) in these tissues. Expression of SLPI was decreased in the vagina and ectocervix of women under hormonal treatment. Endocervical ERα mRNA expression was increased while this did not reach significance at the protein level. SLPI expression in the endometrium was not influenced by hormonal treatment. We observed attenuated ERα expression in the cervix and endometrium of hormonally treated women, whereas vaginal expression was increased. PRA expression was augmented in the cervix and endometrium and unchanged in the vagina. Taken together, our results indicate that hormonal responses and receptor expression are differentially regulated in vaginal tissue compared with the cervix and endometrium.


Asunto(s)
Genitales Femeninos/metabolismo , Hormonas/metabolismo , Inhibidor Secretorio de Peptidasas Leucocitarias/metabolismo , Anciano , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Terapia de Reemplazo de Estrógeno , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genitales Femeninos/inmunología , Genitales Femeninos/patología , Humanos , Inmunidad Mucosa , Persona de Mediana Edad , Posmenopausia , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Inhibidor Secretorio de Peptidasas Leucocitarias/genética
16.
Trends Immunol ; 29(11): 541-7, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18838299

RESUMEN

At mucosal surfaces, we must co-exist with a high density of diverse microorganisms; therefore, protection against these occurs on multiple levels. Leukocyte- and epithelial derived-antimicrobial peptides and proteins (AMPs) comprise an essential component of immune defense. These molecules possess antibacterial, antifungal and signalling properties and probably contribute to defence and maintenance of homeostasis between the host and commensal microorganisms. Among these AMPs is bactericidal/permeability-increasing protein (BPI), an antimicrobial protein with potent endotoxin-neutralising activity, and several homologs. This review explores the roles of BPI and and its homologs at the mucosal interface. Congeners of BPI are under biopharmaceutical development as novel anti-infective agents, highlighting the potential therapeutic relevance of this protein family.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/inmunología , Proteínas Sanguíneas/química , Proteínas Sanguíneas/inmunología , Membrana Mucosa/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Péptidos Catiónicos Antimicrobianos/uso terapéutico , Proteínas Sanguíneas/metabolismo , Proteínas Sanguíneas/uso terapéutico , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Membrana Mucosa/química , Membrana Mucosa/inmunología , Membrana Mucosa/microbiología , Fosfolípidos/inmunología , Fosfolípidos/metabolismo
17.
J Infect Dis ; 194(4): 498-502, 2006 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16845634

RESUMEN

Genital tract epithelia regularly encounter and adapt to the existence of bacterial pathogens. This study provides evidence that the endocervical and ectocervical epithelia of the human female genital tract express bactericidal/permeability-increasing protein (BPI). The constitutive expression of BPI was restricted to cell-bound protein and unaffected by human papillomavirus type 16/E6E7 immortalization and proinflammatory cytokine stimulation. Epithelial BPI was, in part, responsible for killing a commensal strain of Escherichia coli. The results of the present study suggest that BPI is tightly regulated and functionally expressed by epithelial cells in the female reproductive tract and may play a role in regulating bacterial colonization in the genital mucosa.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Proteínas Sanguíneas/metabolismo , Cuello del Útero/citología , Células Epiteliales/metabolismo , Proteínas de la Membrana/metabolismo , Péptidos Catiónicos Antimicrobianos/farmacología , Proteínas Sanguíneas/farmacología , Células Cultivadas , Escherichia coli/efectos de los fármacos , Femenino , Humanos , Inmunohistoquímica , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Proteínas de la Membrana/farmacología
18.
Am J Physiol Gastrointest Liver Physiol ; 290(3): G557-67, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16282362

RESUMEN

Epithelial cells of many mucosal organs have adapted to coexist with microbes and microbial products. In general, most studies suggest that epithelial cells benefit from interactions with commensal microorganisms present at the lumenal surface. However, potentially injurious molecules found in this microenvironment also have the capacity to elicit local inflammatory responses and even systemic disease. We have recently demonstrated that epithelia cells express the anti-infective molecule bactericidal/permeability-increasing protein (BPI). Here, we extend these findings to examine molecular mechanisms of intestinal epithelial cell (IEC) BPI expression and function. Initial experiments revealed a variance of BPI mRNA and protein expression among various IEC lines. Studies of BPI promoter expression in IECs identified regulatory regions of the BPI promoter and revealed a prominent role for CCAAT/enhancer binding protein and especially Sp1/Sp3 in the basal regulation of BPI. To assess the functional significance of this protein, we generated an IEC line stably transfected with full-length BPI. We demonstrated that, whereas epithelia express markedly less BPI protein than neutrophils, epithelial BPI contributes significantly to bacterial killing and attenuating bacterial-elicted proinflammatory signals. Additional studies in murine tissue ex vivo revealed that BPI is diffusely expressed along the crypt-villous axis and that epithelial BPI levels decrease along the length of the intestine. Taken together, these data confirm the transcriptional regulation of BPI in intestinal epithelia and provide insight into the relevance of BPI as an anti-infective molecule at intestinal surfaces.


Asunto(s)
Proteínas Sanguíneas/fisiología , Proteínas de la Membrana/fisiología , Animales , Péptidos Catiónicos Antimicrobianos , Actividad Bactericida de la Sangre , Proteínas Sanguíneas/genética , Proteínas Potenciadoras de Unión a CCAAT/fisiología , Células CACO-2 , Humanos , Mucosa Intestinal/metabolismo , Lipopolisacáridos/farmacología , Proteínas de la Membrana/genética , Ratones , Regiones Promotoras Genéticas/fisiología , Factores de Transcripción Sp/fisiología
19.
J Cell Biochem ; 99(6): 1616-27, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16823775

RESUMEN

Epithelial cells line mucosal surfaces (e.g., lung, intestine) and critically function as a semipermeable barrier to the outside world. Mucosal organs are highly vascular with extensive metabolic demands, and for this reason, are particularly susceptible to diminished blood flow and resultant tissue hypoxia. Here, we pursue the hypothesis that intestinal barrier function is regulated in a protective manner by hypoxia responsive genes. We demonstrate by PCR confirmation of microarray data and by avidin blotting of immunoprecipitated human Mucin 3 (MUC3), that surface MUC3 expression is induced in T84 intestinal epithelial cells following exposure to hypoxia. MUC3 RNA is minimally detectable while surface protein expression is absent under baseline normoxic conditions. There is a robust induction in both the mRNA (first evident by 8 h) and protein expression, first observed and maximally expressed following 24 h hypoxia. This is followed by a subsequent decline in protein expression, which remains well above baseline at 48 h of hypoxia. Further, we demonstrate that this induction of MUC3 protein is associated with a transient increase in the barrier restorative peptide, intestinal trefoil factor (ITF). ITF not only colocalizes with MUC3, by confocal microscopy, to the apical surface of T84 cells following exposure to hypoxia, but is also found, by co-immunoprecipitation, to be physically associated with MUC3, following 24 h of hypoxia. In exploration of the mechanism of hypoxic regulation of mucin 3 expression, we demonstrated by luciferase assay that the full-length promoter for mouse Mucin 3 (Muc3) is hypoxia-responsive with a 5.08 +/- 1.76-fold induction following 24 h of hypoxia. Furthermore, analysis of both the human (MUC3A) and mouse (Muc3) promoters revealed potential HIF-1 binding sites which were shown by chromatin immunoprecipitation to bind the pivotal hypoxia-regulating transcription factor HIF-1alpha. Taken together, these studies implicate the HIF-1alpha mediated hypoxic induced expression of mucin 3 and associated ITF in the maintenance of intestinal barrier function under hypoxic conditions.


Asunto(s)
Hipoxia de la Célula , Mucosa Intestinal/metabolismo , Mucinas/biosíntesis , Secuencia de Bases , Línea Celular , Cartilla de ADN , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mucosa Intestinal/citología , Mucinas/genética , Mucinas/metabolismo , Péptidos/metabolismo , Reacción en Cadena de la Polimerasa , Unión Proteica , ARN Mensajero/genética , Factor Trefoil-2
20.
Am J Physiol Gastrointest Liver Physiol ; 288(4): G593-7, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15764808

RESUMEN

Epithelial cells of many mucosal organs have adapted to coexist with microbes and microbial products. In general, most studies suggest that epithelial cells benefit from interactions with commensal microorganisms present at the lumenal surface. However, potentially injurious molecules found in this microenvironment also have the capacity to elicit local inflammatory responses and even systemic disease. In this environment, the epithelium has evolved effective mechanisms to cope with microbial products and to provide appropriate responses to potential pathogens. Although our understanding of these mechanisms is clearly in its infancy, a number of recent findings provide insight into phenotypic characteristics that allow for this discrimination. Here, we briefly review some of these mechanisms, with particular attention to epithelial expression of the anti-infective molecule bactericidal/permeability-increasing protein.


Asunto(s)
Adaptación Fisiológica , Proteínas Sanguíneas/fisiología , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/fisiología , Proteínas de la Membrana/fisiología , Animales , Péptidos Catiónicos Antimicrobianos , Tracto Gastrointestinal/metabolismo , Humanos , Mucosa Intestinal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA