Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Endocrinology ; 157(1): 258-67, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26485613

RESUMEN

Therapeutic strategies focused on restoring immune tolerance remain the main avenue to prevent type 1 diabetes (T1D). Because estrogens potentiate FoxP3+ regulatory T cells (Treg) and invariant natural killer T (iNKT) cells, two regulatory lymphocyte populations that are functionally deficient in nonobese diabetic (NOD) mice, we investigated whether estradiol (E2) therapy influences the course of T1D in this model. To this end, female NOD mice were sc implanted with E2- or placebo-delivering pellets to explore the course of spontaneous and cyclophosphamide-induced diabetes. Treg-depleted and iNKT-cell-deficient (Jα18(-/-)) NOD mice were used to assess the respective involvement of these lymphocyte populations in E2 effects. Early E2 administration (from 4 wk of age) was found to preserve NOD mice from both spontaneous and cyclophosphamide-induced diabetes, and a complete protection was also observed throughout treatment when E2 treatment was initiated after the onset of insulitis (from 12 wk of age). This delayed E2 treatment remained fully effective in Treg-depleted mice but failed to entirely protect Jα18(-/-) mice. Accordingly, E2 administration was shown to restore the cytokine production of iNKT cells in response to in vivo challenge with the cognate ligand α-galactosylceramide. Finally, transient E2 administration potentiated the previously described protective action of α-galactosylceramide treatment in NOD females. This study provides original evidence that E2 therapy strongly protects NOD mice from T1D and reveals the estrogen/iNKT cell axis as a new effective target to counteract diabetes onset at the stage of insulitis. Estrogen-based therapy should thus be considered for T1D prevention.


Asunto(s)
Enfermedades Autoinmunes/prevención & control , Diabetes Mellitus Tipo 1/prevención & control , Estradiol/uso terapéutico , Estrógenos/uso terapéutico , Células Asesinas Naturales/efectos de los fármacos , Activación de Linfocitos/efectos de los fármacos , Estado Prediabético/prevención & control , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Citocinas/sangre , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Implantes de Medicamentos , Estradiol/administración & dosificación , Terapia de Reemplazo de Estrógeno , Estrógenos/administración & dosificación , Femenino , Galactosilceramidas/agonistas , Galactosilceramidas/farmacología , Galactosilceramidas/uso terapéutico , Tolerancia Inmunológica/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Depleción Linfocítica/efectos adversos , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Mutantes , Ovariectomía/efectos adversos , Estado Prediabético/tratamiento farmacológico , Estado Prediabético/inmunología , Estado Prediabético/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
2.
PLoS One ; 10(5): e0128647, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26023782

RESUMEN

Human umbilical cord mesenchymal stromal cells (hUC-MSCs) are currently being used as novel therapeutic agents in numerous clinical trials. Previous works have shown that hUC-MSCs possess profound immunomodulatory capacities through IL-1 stimulation produced by peripheral blood mononuclear cells (PBMCs), their main cellular partner in most pathophysiological and therapeutic situations. The present study was designed to explore the role of TNF-α in these interactions. In these experiments, we demonstrated that TNF-α originated from PBMCs under the influence of IL-1. We also showed that TNF-α acted differently depending upon the concentrations reached. At low concentrations it clearly contributed to IL-6 and monocyte chemotactic protein 1 (MCP-1) production. At high concentrations, used alone or in association with the TNF-related apoptosis-inducing ligand, TNF-α also stimulated hUC-MSC IL-6 but, more intensely, MCP-1 production. This stimulation was associated but independent of apoptosis induction in a process involving Inhibitor of Apoptosis Proteins. Interferon gamma (IFN-γ), tested to stimulate PBMC and tissue activation, amplified IL-6 and MCP-1 production and cell death by, apparently, a different process involving necrosis. Our findings bring new insights into the complex interactions between hUC-MSCs and PBMCs, involving cytokines, chemokines and cell death, and are of fundamental importance for tissue homeostasis.


Asunto(s)
Apoptosis , Leucocitos Mononucleares/metabolismo , Células Madre Mesenquimatosas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Cordón Umbilical/metabolismo , Quimiocina CCL2/metabolismo , Técnicas de Cocultivo , Femenino , Humanos , Interferón gamma/metabolismo , Interleucina-6/metabolismo , Leucocitos Mononucleares/citología , Masculino , Células Madre Mesenquimatosas/citología , Cordón Umbilical/citología
3.
Cell Physiol Biochem ; 29(3-4): 551-60, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22508062

RESUMEN

BACKGROUND/AIMS: Human umbilical cord mesenchymal stem cells (hUC-MSCs) possess immunosuppressive activities but the mechanisms of such activities are not fully understood. Here, we investigated the role of IL-6, one of the characteristic factors of MSCs, in the immunoregulating effect of hUC-MSCs on CD4(+) T lymphocytes. METHODS: The condition media from human peripheral blood mononuclear cells (hPBMCs) or CD14+/- cell were tested if stimulating IL-6 production by hUC-MSCs. The related signaling pathway of IL-6, and the immunosuppressive activity of IL-6 on CD4(+) T lymphocytes were studied. RESULT: IL-6 production was dramatically increased by hUC-MSCs when co-culturing with resting or activated hPBMCs. CD14(+) monocytes-paracrined IL-1ß promoted the secretion of IL-6 by hUC-MSCs via JNK and NF-κB signaling pathway. Blocking of PGE2 synthesis did not affect the secretion of IL-6, anti-IL-6 antibody was not able to reverse hUC-MSCs-mediated inhibition on CD4(+) T lymphocytes. IL-6 did not mediate the suppressive activity of IL-1ß-hUC-MSCs- PGE2 on CD4(+) T cell. CONCLUSION: CD14(+) monocytes-paracrined IL-1ß promotes IL-6 secretion by hUC-MSCs through activating JNK and NF-κB signaling pathway. However, increased IL-6 production does not contribute to immunosuppressive activity of IL-1ß-hUC-MSCs- PGE2 on CD4(+) T cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Inmunosupresores/farmacología , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Leucocitos Mononucleares/inmunología , Células Madre Mesenquimatosas/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Dinoprostona/antagonistas & inhibidores , Dinoprostona/inmunología , Dinoprostona/farmacología , Humanos , Inmunofenotipificación , Inmunosupresores/inmunología , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Interleucina-6/metabolismo , Interleucina-6/farmacología , Receptores de Lipopolisacáridos/inmunología , Sistema de Señalización de MAP Quinasas , Activación de Macrófagos , Nitrobencenos/farmacología , Sulfonamidas/farmacología , Cordón Umbilical/citología , Cordón Umbilical/inmunología
4.
J Leukoc Biol ; 90(1): 145-54, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21498588

RESUMEN

Estrogens influence the incidence and the course of numerous immune or inflammatory diseases in humans and in experimental models. For instance, estrogens prevent the accumulation of granulocytes in acute inflammatory murine models, but the respective actions on neutrophil and eosinophil trafficking remain to be clarified. We demonstrate here that in a model of TGC-induced sterile peritonitis in ovx mice, chronic E2 administration electively and strongly inhibited peritoneal eosinophil accumulation. E2 decreased BM eosinophil number, contributing to a marked prevention of the TGC-induced eosinophil blood mobilization. These effects on eosinophil mobilization and peritoneal accumulation were abolished in ER-α(-/-) mice, demonstrating the crucial role of this nuclear receptor. Grafting ER-α(-/-) mice with ER-α(+/+) BM cells restored the suppressive effect of E2 on peritoneal eosinophilia, although the action on eosinophil blood mobilization was still abrogated. We therefore explored additional mechanisms and found that E2 reduced the peritoneal concentrations of key eosinophil prosurvival factors (IL-5, IL-9, and IL-25) and enhanced eosinophil apoptosis during the inflammatory process. Furthermore, this proapoptotic effect of E2 was abrogated in IL-5-overexpressing Tg mice. To conclude, we demonstrate for the first time that ER-α activation by exogenous E2 administration strongly inhibits eosinophil accumulation during acute inflammation in a nonreproductive target site for estrogen through combined actions on eosinophil mobilization and apoptosis. This specific, suppressive effect of chronic E2 replacement therapy on eosinophils has to be integrated to further understand the evolution of eosinophil-associated diseases in menopausal women.


Asunto(s)
Eosinofilia/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Peritonitis/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Separación Celular , Quimiotaxis de Leucocito/efectos de los fármacos , Quimiotaxis de Leucocito/fisiología , Modelos Animales de Enfermedad , Eosinófilos/citología , Eosinófilos/efectos de los fármacos , Eosinófilos/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/efectos de los fármacos , Estrógenos/farmacología , Femenino , Citometría de Flujo , Interleucina-5/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
J Immunol ; 185(2): 1169-76, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20554954

RESUMEN

17Beta-estradiol (E2) has been shown to promote the expression of inflammatory mediators by LPS-activated tissue resident macrophages through estrogen receptor alpha (ERalpha) signaling. However, it remained to be determined whether E2 similarly influences macrophages effector functions under inflammatory conditions in vivo, and whether this action of E2 resulted from a direct effect on macrophages. We show in this study that chronic E2 administration to ovariectomized mice significantly increased both cytokine (IL-1beta, IL-6, and TNF-alpha) and inducible NO synthase mRNA abundance in thioglycolate (TGC)-elicited macrophages. The proinflammatory action of E2 was also evidenced at the level of released IL-1beta and IL-6 by ex vivo LPS-activated macrophages. E2 concomitantly inhibited PI3K activity as well as Akt phosphorylation in TGC-elicited macrophages, suggesting that E2 promoted TLR-dependent macrophage activation by alleviating this suppressive signaling pathway. Indeed, this effect was abolished in the presence of the inhibitor wortmannin, demonstrating a key functional link between inhibition of PI3K activity and the E2 action on macrophage functions. Endogenous estrogens levels circulating in ovary-intact mice were sufficient to promote the above described actions. Finally, thanks to a CreLox strategy, targeted disruption of ERalpha gene in macrophages totally abolished the effect of E2 on the expression of inflammatory mediators by both resident and TGC-elicited peritoneal macrophages. In conclusion, we demonstrate that estrogens, through the activation of ERalpha in macrophages in vivo, enhance their ability to produce inflammatory mediators and cytokines upon subsequent TLR activation.


Asunto(s)
Citocinas/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Animales , Western Blotting , Células Cultivadas , Citocinas/genética , Estradiol/metabolismo , Receptor alfa de Estrógeno/genética , Femenino , Citometría de Flujo , Mediadores de Inflamación/metabolismo , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Tioglicolatos/farmacología , Receptor Toll-Like 4/genética
6.
Exp Cell Res ; 316(15): 2414-23, 2010 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-20420825

RESUMEN

Here, the effect of CD14(+) monocytes on human umbilical cord matrix stem cell (hUC-MSC)-mediated immunosuppression was studied in vitro. hUC-MSCs exerted a potent inhibitory effect on the proliferation and interferon-gamma (IFN-gamma) secretion capacities of CD4(+) and CD8(+) T cells in response to anti-CD3/CD28 stimulation. Transwell co-culture system revealed that the suppressive effect was primarily mediated by soluble factors. Addition of prostaglandin synthesis inhibitors (indomethacin or NS-398) almost completely abrogated the immunosuppression activity of hUC-MSCs, identifying prostaglandin E(2) (PGE(2)) as an important soluble mediator. CD14(+) monocytes were found to be able to enhance significantly the immunosuppressive effect of hUC-MSCs in a dose-dependent fashion. Moreover, the inflammatory cytokine IL-1beta, either exogenously added or produced by CD14(+) monocytes in culture, could trigger expression of high levels of PGE(2) by hUC-MSCs, whereas inclusion of the IL-1 receptor antagonist (IL-1RA) in the culture down-regulated not only PGE(2) expression, but also reversed the promotional effect of CD14(+) monocytes and partially restored CD4(+) and CD8(+) T cell proliferation and IFN-gamma secretion. Our data demonstrate an important role of monocytes in the hUC-MSC-induced immunomodulation, which may have important implications in future efforts to explore the clinical potentials of hUC-MSCs.


Asunto(s)
Tolerancia Inmunológica/fisiología , Receptores de Lipopolisacáridos/metabolismo , Células Madre Mesenquimatosas/fisiología , Monocitos/fisiología , Cordón Umbilical/inmunología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Dinoprostona/metabolismo , Dinoprostona/farmacología , Dinoprostona/fisiología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/inmunología , Endotelio Vascular/fisiología , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Factores Inmunológicos/química , Factores Inmunológicos/metabolismo , Factores Inmunológicos/farmacología , Mediadores de Inflamación/farmacología , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Interleucina-1beta/fisiología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Mitógenos/farmacología , Monocitos/inmunología , Monocitos/metabolismo , Solubilidad , Cordón Umbilical/citología
7.
Clin Immunol ; 135(3): 448-58, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20207200

RESUMEN

Human umbilical-cord-derived mesenchymal stem cells (hUC-MSCs) constitute an attractive alternative to bone-marrow-derived MSCs for potential clinical applications because of easy preparation and lower risk of viral contamination. In this study, both proliferation of human peripheral blood mononuclear cells (hPBMCs) and their IFN-gamma production in response to mitogenic or allogeneic stimulus were effectively inhibited by hUC-MSCs. Co-culture experiments in transwell systems indicated that the suppression was largely mediated by soluble factor(s). Blocking experiments identified prostaglandin E(2) (PGE(2)) as the major factor, because inhibition of PGE(2) synthesis almost completely mitigated the immunosuppressive effects, whereas neutralization of TGF-beta, IDO, and NO activities had little effects. Moreover, the inflammatory cytokines, IFN-gamma and IL-1beta, produced by hPBMCs upon activation notably upregulated the expression of cyclooxygenase-2 (COX-2) and the production of PGE(2) by hUC-MSCs. In conclusion, our data have demonstrated for the first time the PGE(2)-mediated mechanism by which hUC-MSCs exert their immunomodulatory effects.


Asunto(s)
Dinoprostona/inmunología , Tolerancia Inmunológica/inmunología , Leucocitos Mononucleares/inmunología , Células Madre Mesenquimatosas/inmunología , Proliferación Celular , Separación Celular , Técnicas de Cocultivo , Ciclooxigenasa 2/inmunología , Ciclooxigenasa 2/metabolismo , Citocinas/biosíntesis , Citocinas/inmunología , Dinoprostona/metabolismo , Ensayo de Inmunoadsorción Enzimática , Sangre Fetal/citología , Sangre Fetal/inmunología , Sangre Fetal/metabolismo , Citometría de Flujo , Expresión Génica/inmunología , Humanos , Inmunofenotipificación , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , ARN Mensajero/análisis , Cordón Umbilical/citología , Cordón Umbilical/inmunología , Cordón Umbilical/metabolismo
8.
J Immunol ; 180(12): 7980-8, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18523261

RESUMEN

Short-term exposure to 17beta-estradiol (E2) in vitro has been reported to decrease the production of proinflammatory cytokines by LPS-activated macrophages through estrogen receptor alpha (ERalpha)-dependent activation of the PI3K pathway. In the present study, we confirm that in vitro exposure of mouse peritoneal macrophages to E2 enhanced Akt phosphorylation and slightly decreased LPS-induced cytokine production. In striking contrast, we show that chronic administration of E2 to ovariectomized mice markedly increases the expression of IL-1beta, IL-6, IL-12p40, and inducible NO synthase by resident peritoneal macrophages in response to LPS ex vivo. These results clearly indicate that short-term E2 treatment in vitro does not predict the long-term effect of estrogens in vivo on peritoneal macrophage functions. We show that this in vivo proinflammatory effect of E2 was mediated through ERalpha. Although the expression of components of the LPS-recognition complex remained unchanged, we provided evidences for alterations of the TLR4 signaling pathway in macrophages from E2-treated mice. Indeed, E2 treatment resulted in the inhibition of PI3K activity and Akt phosphorylation in LPS-activated macrophages, whereas NF-kappaB p65 transcriptional activity was concomitantly increased. Incubation of macrophages with the PI3K inhibitor wortmanin enhanced proinflammatory cytokine gene expression in response to TLR4 activation, and abolishes the difference between cells from placebo- or E2-treated mice, demonstrating the pivotal role of the PI3K/Akt pathway. We conclude that the macrophage activation status is enhanced in vivo by E2 through ERalpha and, at least in part, by the down-modulation of the PI3K/Akt pathway, thereby alleviating this negative regulator of TLR4-signaling.


Asunto(s)
Citocinas/biosíntesis , Estradiol/administración & dosificación , Mediadores de Inflamación/administración & dosificación , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Fosfatidilinositol 3-Quinasas/fisiología , Transducción de Señal/inmunología , Receptor Toll-Like 4/metabolismo , Administración Cutánea , Animales , Células Cultivadas , Citocinas/fisiología , Preparaciones de Acción Retardada , Implantes de Medicamentos , Estradiol/farmacología , Receptor alfa de Estrógeno/deficiencia , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/fisiología , Femenino , Mediadores de Inflamación/farmacología , Lipopolisacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/inmunología , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidores de las Quinasa Fosfoinosítidos-3 , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Receptor Toll-Like 4/biosíntesis , Receptor Toll-Like 4/genética
9.
Arterioscler Thromb Vasc Biol ; 27(10): 2214-21, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17690314

RESUMEN

OBJECTIVE: Whereas estradiol prevents fatty streak deposit in immunocompetent apoE-/- or LDLr-/- mice, it is totally ineffective in immunodeficient mice, underlining the key role of immunoinflammation in this effect. In the present work, the role of several major pro- and antiinflammatory cytokines involved in the atheromatous process was evaluated in the effect of estradiol on fatty streak constitution. METHODS AND RESULTS: The preventive effect of estradiol was fully maintained in LDLr-/- mice grafted with bone marrow from either IFN-gamma or interleukin (IL)-12-deficient mice, showing that this beneficial effect was not mediated through a specific decrease in the production of these 2 proinflammatory cytokines. Furthermore, IL-10-/- apoE-/- mice remained protected by estradiol, excluding a significant contribution of this antiinflammatory cytokine. In contrast, the protective effect of estradiol was (1) associated with enhanced aortic expression of TGF-beta1 in apoE-/- mice during early steps of atherogenesis; (2) abolished and even reversed in apoE-/- mice administered with a neutralizing anti-TGF-beta antibody; (3) abolished in LDLr-/- mice grafted with bone marrow from Smad3-deficient mice. CONCLUSIONS: The status of the TGF-beta pathway crucially determines the antiatherogenic effect of estradiol in hypercholesterolemic mice, whereas neither IFN-gamma, IL-12, nor IL-10 are specifically involved in this protection.


Asunto(s)
Aorta/metabolismo , Aterosclerosis/metabolismo , Estradiol/metabolismo , Hipercolesterolemia/complicaciones , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Anticuerpos Monoclonales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerosis/etiología , Aterosclerosis/prevención & control , Trasplante de Médula Ósea , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Implantes de Medicamentos , Estradiol/administración & dosificación , Femenino , Hipercolesterolemia/genética , Hipercolesterolemia/metabolismo , Interferón gamma/deficiencia , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-10/deficiencia , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-12/deficiencia , Interleucina-12/genética , Interleucina-12/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovariectomía , Receptores de LDL/deficiencia , Receptores de LDL/genética , Receptores de LDL/metabolismo , Transducción de Señal , Proteína smad3/deficiencia , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/inmunología , Irradiación Corporal Total
10.
Am J Pathol ; 169(5): 1855-62, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17071606

RESUMEN

17beta-Estradiol (E2) accelerates reendothelialization and increases the number of circulating endothelial progenitor cells (EPCs), but whether fibroblast growth factor-2 (FGF2) is involved in these processes remains unknown. Here we explored the role of FGF2 in the effect of E2 on reendothelialization and EPC levels in a mouse model. As previously reported, E2 increased both the velocity of reendothelialization and the number of circulating EPCs in ovariectomized wild-type (Fgf2+/+) mice. In contrast, the effect of E2 on both parameters was abolished in FGF2-deficient mice (Fgf2-/-), demonstrating that FGF2 is absolutely required for these effects of E2. To test the implication of medullary and extramedullary FGF2, we developed chimeric mice by grafting Fgf2-/- bone marrow to Fgf2+/+ [Fgf2-/- bone marrow (BM) = > Fgf2+/+] mice and observed that the effect of E2 on both reendothelialization and EPC levels was abolished. In contrast, both effects of E2 in Fgf2+/+BM = >Fgf2-/- mice were similar to those observed in Fgf2+/+ mice, demonstrating that only BM-derived, but not extramedullary, FGF2 is required for both effects. Interestingly, E2 was found to markedly increase both FGF2(lmw) and FGF2(hmw) in bone marrow. In conclusion, FGF2, specifically medullary FGF2, is necessary and sufficient to mediate the accelerative effect of E2 on both reendothelialization and EPC mobilization.


Asunto(s)
Médula Ósea/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Estradiol/farmacología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Células Madre/citología , Animales , Aorta/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Trasplante de Médula Ósea , Arterias Carótidas/citología , Arterias Carótidas/patología , Factor 2 de Crecimiento de Fibroblastos/deficiencia , Factor 2 de Crecimiento de Fibroblastos/genética , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos/efectos de los fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
11.
FASEB J ; 20(3): 476-8, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16423876

RESUMEN

Spermatogenesis is a complex process involving cell proliferation, differentiation, and apoptosis. Fibroblast growth factor 2 (FGF-2) is involved in testicular function, but its role in spermatogenesis has not been fully documented. The control of FGF-2 expression particularly occurs at the translational level, by an internal ribosome entry site (IRES)-dependent mechanism driving the use of alternative initiation codons. To study IRES activity regulation in vivo, we have developed transgenic mice expressing a bicistronic construct coding for two luciferase genes. Here, we show that the FGF-2 IRES is age-dependently activated in mouse testis, whereas EMCV and c-myc IRESs are not. Real-time PCR confirms that this regulation is translational. By using immunohistological techniques, we demonstrate that FGF-2 IRES stimulation occurs in adult, but not in immature, type-A spermatogonias. This is correlated with activation of endogenous FGF-2 expression in spermatogonia; whereas FGF-2 mRNA transcription is known to decrease in adult testis. Interestingly, the FGF-2 IRES activation is triggered by testosterone and is partially inhibited by siRNA directed against the androgen receptor. Two-dimensional analysis of proteins bound to the FGF-2 mRNA 5'UTR after UV cross-linking reveals that testosterone treatment correlates with the binding of several proteins. These data suggest a paracrine loop where IRES-dependent FGF-2 expression, stimulated by Sertoli cells in response to testosterone produced by Leydig cells, would in turn activate Leydig function and testosterone production. In addition, nuclear FGF-2 isoforms could be involved in an intracrine function of FGF-2 in the start of spermatogenesis, mitosis, or meiosis initiation. This report demonstrates that mRNA translation regulation by an IRES-dependent mechanism participates in a physiological process.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/fisiología , Células Intersticiales del Testículo/fisiología , Biosíntesis de Proteínas , ARN Mensajero/genética , Secuencias Reguladoras de Ácidos Nucleicos , Células de Sertoli/fisiología , Espermatogénesis/fisiología , Testículo/fisiología , Testosterona/fisiología , Regiones no Traducidas 5' , Factores de Edad , Antagonistas de Receptores Androgénicos , Animales , Codón , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Factor 2 de Crecimiento de Fibroblastos/genética , Genes Reporteros , Genes Sintéticos , Luciferasas de Renilla/genética , Masculino , Meiosis , Ratones , Ratones Transgénicos , Mitosis , Comunicación Paracrina , Iniciación de la Cadena Peptídica Traduccional/fisiología , Isoformas de Proteínas/fisiología , ARN Mensajero/efectos de la radiación , ARN Interferente Pequeño/farmacología , Receptores Androgénicos/genética , Proteínas Recombinantes de Fusión/fisiología , Ribosomas/metabolismo , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Testosterona/metabolismo , Testosterona/farmacología , Rayos Ultravioleta
12.
Am J Pathol ; 167(1): 267-74, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15972970

RESUMEN

Estradiol prevents fatty streak formation in chow-fed atherosclerosis-prone apolipoprotein E (ApoE)-deficient mice. We previously reported that fatty streak development of immunodeficient ApoE(-/-)/recombination activating gene 2 (RAG-2(-/-)) double-deficient mice was insensitive to estradiol. In the present work, we demonstrate that the reconstitution of ApoE(-/-)/RAG-2(-/-) with bone marrow from immunocompetent ApoE(-/-)/RAG-2(+/+) mice restores the protective effect of estradiol on fatty streak constitution. We extended this demonstration to the model of low-density lipoprotein receptor-deficient mice, establishing the obligatory role of mature lymphocytes in this process. We then investigated whether the protective effect of estradiol was mediated by a specific lymphocyte subpopulation by studying the hormonal effect on fatty streak constitution in recently developed models of ApoE(-/-) mice deficient in selective T-lymphocyte subsets (either TCRalphabeta+, CD4+, CD8+, or TCRgammadelta+ lymphocytes) or B lymphocytes. In all these specifically immunodeficient mice, estradiol administration to ovariectomized mice conferred protection as in immunocompetent ApoE(-/-) mice, clearly demonstrating that no single lymphocyte subpopulation was specifically required for this effect. These results point to additional lymphocyte-dependent mechanisms such as modulating the interactions among lymphocytes and between lymphocytes and endothelial and/or antigen-presenting cells.


Asunto(s)
Arteriosclerosis/inmunología , Arteriosclerosis/prevención & control , Estradiol/farmacología , Subgrupos de Linfocitos T/inmunología , Animales , Aorta/efectos de los fármacos , Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Trasplante de Médula Ósea , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Femenino , Corazón/efectos de los fármacos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Ratones , Ratones Noqueados , Ratones Transgénicos , Miocardio/patología , Ovariectomía , Receptores de LDL/deficiencia , Receptores de LDL/genética
13.
Blood ; 105(6): 2415-20, 2005 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15383462

RESUMEN

Mechanisms accounting for gender dimorphism during immune responses are still poorly understood. Since invariant natural killer T (iNKT) cells exert important regulatory functions through their capacity to produce both T helper 1 (Th1) and Th2 cytokines, we addressed the question of whether these activities could be modulated by sexual hormones. We found that in vivo challenge with the specific ligand of iNKT cells, alpha-galactosylceramide (alpha-GalCer), induced significantly higher concentrations of interferon gamma (IFN-gamma) in the serum of female than in that of male mice, while interleukin 4 (IL-4) production was not modified. In support of a crucial role of ovarian hormones in this phenomenon, a significant decrease of serum IFN-gamma concentrations occurred in ovariectomized females, in response to treatment with alpha-GalCer, while orchidectomy affected neither IFN-gamma nor IL-4 serum concentrations in males. The implication of estrogens in this selective enhancement of IFN-gamma production by iNKT cells was demonstrated by (1) the increased alpha-GalCer-induced IFN-gamma synthesis by iNKT cells upon both in vitro and in vivo exposure to estradiol and (2) the abolition of the sex-linked difference in alpha-GalCer-induced IFN-gamma release in estrogen receptor alpha-deficient mice. These results provide the first evidence that estrogens influence iNKT cells leading to this gender dimorphism in their cytokine production profile.


Asunto(s)
Estradiol/farmacología , Interferón gamma/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/efectos de los fármacos , Caracteres Sexuales , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Células Cultivadas , Estradiol/inmunología , Estradiol/metabolismo , Receptor alfa de Estrógeno/deficiencia , Receptor alfa de Estrógeno/inmunología , Femenino , Galactosilceramidas/administración & dosificación , Galactosilceramidas/inmunología , Interferón gamma/sangre , Interleucina-4/sangre , Interleucina-4/inmunología , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Noqueados , Orquiectomía , Ovariectomía , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/metabolismo
14.
Am J Pathol ; 165(6): 2013-8, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15579444

RESUMEN

Recent studies have demonstrated the importance of lymphocytes, especially CD4(+) T cells, in early lesions of atherosclerosis in hypercholesterolemic mice. However, the role of other T cell subpopulations, like CD8(+) T cells or TCR gamma delta T lymphocytes, is not yet clear. We have therefore generated apolipoprotein E-deficient mice genetically deficient in specific T lymphocyte subpopulations and measured atherosclerotic lesions in the aortic sinus and en face whole aorta preparation at 18 weeks and at 1 year of age. Whereas TCR gamma delta(+) T lymphocytes appeared to play a modest role, TCR alpha beta(+) T lymphocytes played a major role as their deficiency significantly prevented early and late atherosclerosis at all arterial sites. However, neither CD4(+) nor CD8(+) T cells induced any significant decrease of the lesions at the aortic sinus, suggesting that compensatory proatherogenic mechanisms are operating at this site. Interestingly, the absence of CD4(+) T cells led to a dramatic increase in early lesion abundance at the level of the descending thoracic and abdominal aorta, which was still obvious at 1 year. In conclusion, whereas the TCR alpha beta(+) lymphocyte subset in its whole contribute to aggravate both early and late atherosclerosis, the CD4(+) T subpopulation appears to be critically protective at the level of the lower part of the aorta.


Asunto(s)
Apolipoproteínas E/deficiencia , Arteriosclerosis/etiología , Linfocitos T CD4-Positivos/fisiología , Receptores de Antígenos de Linfocitos T alfa-beta/fisiología , Animales , Aorta , Arteriosclerosis/metabolismo , Arteriosclerosis/patología , Linfocitos T CD8-positivos/fisiología , Cruzamientos Genéticos , Femenino , Eliminación de Gen , Genotipo , Lípidos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
15.
FASEB J ; 18(13): 1583-5, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15289445

RESUMEN

Fibroblast growth factor 2 (FGF-2) is normally synthesized at low levels but is elevated in various pathophysiological conditions including diabetes-associated vascular diseases. FGF-2 expression is regulated translationally through an internal ribosome entry site (IRES) located in its mRNA, which allows a nonclassical cap-independent translation. We addressed the pathophysiological regulation of the IRES in vivo by using a streptozotocin-induced hyperglycemic model known to suppress markedly overall translation. Evaluation of FGF-2 IRES-dependent translation was performed with transgenic mice expressing dual luciferase bicistronic mRNA containing the FGF-2 IRES. FGF-2 IRES-dependent reporter activity increased 240% of control in the diabetic aorta although the reporter mRNA levels significantly decreased. Expression of endogenous FGF-2 protein in the aorta closely correlated with the IRES activity but not with FGF-2 mRNA levels. Moreover, the biosynthesis of endogenous FGF-2 protein was stimulated in an IRES-dependent manner by high glucose that significantly suppressed global protein synthesis in aortic smooth muscle cells from the transgenic mice. These results suggest that IRES-dependent translational regulation could play a pathological role in FGF-2 expression in vivo, especially in the cardiovascular consequences of diabetes.


Asunto(s)
Aorta/metabolismo , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Factor 2 de Crecimiento de Fibroblastos/genética , Hiperglucemia/genética , Biosíntesis de Proteínas/genética , Ribosomas/metabolismo , Regulación hacia Arriba , Animales , Sitios de Unión , Glucosa/farmacología , Ratones , Especificidad de Órganos , Biosíntesis de Proteínas/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribosomas/efectos de los fármacos , Ribosomas/genética , Especificidad por Sustrato , Regulación hacia Arriba/efectos de los fármacos
16.
J Infect Dis ; 189(2): 339-45, 2004 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-14722900

RESUMEN

Q fever is a zoonosis caused by Coxiella burnetii and recently has been recognized as a potential agent of bioterrorism. In Q fever, men are symptomatic more often than women, despite equal seroprevalence. We hypothesized that sex hormones play a role in the pathogenesis of C. burnetii infection. When C57/BL6 mice were injected with C. burnetii, bacteria load and granuloma numbers were lower in females than in males. Ovarectomized mice showed increased bacteria load in the spleen and the liver, similar to that found in males. The granuloma number was also increased in ovarectomized mice and reached the levels found in males. Tissue infection and granulomatous response are largely under the control of estrogens: treatment of ovarectomized mice with 17beta-estradiol reduced both bacteria loads and granuloma numbers. These results show that sex hormones control host response to C. burnetii infection and may account for host-dependent clinical presentation of Q fever.


Asunto(s)
Estradiol/farmacología , Fiebre Q/inmunología , Animales , Femenino , Granuloma/etiología , Hígado/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ovariectomía , Fiebre Q/microbiología , Factores Sexuales , Bazo/microbiología
17.
Med Sci (Paris) ; 19(12): 1226-32, 2003 Dec.
Artículo en Francés | MEDLINE | ID: mdl-14691747

RESUMEN

Hormonal replacement therapy does not prevent cardiovascular events in postmenopausal women. In contrast, the incidence of cardiovascular diseases is higher in men than in premenopausal women but increases in postmenopausal women, and all animal studies demonstrate a prevention of fatty streak deposit by estradiol. Although estradiol improves the lipoprotein profile, this effect can account for only a minor part of the protective effect. Endothelium appears to be an important target for estradiol, because this hormone potentiates endothelial nitric oxide (NO) production, thus promoting the beneficial effects of NO, such as vasorelaxation and inhibition of platelet aggregation. Estradiol accelerates endothelial regrowth, thus favoring vascular healing, and prevents apoptosis of endothelial cells. Estradiol prevents fatty streak deposit through a mechanism which is clearly independent of NO. The immuno-inflammatory system appears to play a key role in the development of fatty streak deposit as well as in atherosclerotic plaque rupture. Mice deficient either in monocyte-macrophages or in lymphocytes are partially protected against fatty streak deposit. Interestingly, the atheroprotective effect of estradiol is absent in mice deficient in T and B lymphocytes. Most of these effects of estradiol are mediated by estrogen receptor alpha, and are independent of estrogen receptor beta. Thus, the inflammatory-immune system appears to be also a major target of estrogens. However, the effects of estrogens on the immuno-inflammatory system appear ambiguous, as in some models, estradiol rather promotes inflammation (by increasing interferon gamma which could elicit plaque destabilization). A better understanding of the mechanisms of estrogens on the normal and atheromatous arteries is required and should help to optimize the prevention of cardiovascular disease after menopause.


Asunto(s)
Enfermedades Cardiovasculares/fisiopatología , Células Endoteliales/fisiología , Estradiol/farmacología , Animales , Linfocitos B , Enfermedades Cardiovasculares/prevención & control , Modelos Animales de Enfermedad , Células Endoteliales/patología , Femenino , Humanos , Inflamación , Menopausia , Ratones , Linfocitos T
18.
Cardiovasc Res ; 59(1): 234-40, 2003 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12829194

RESUMEN

OBJECTIVE: Atherosclerosis is an inflammatory disease in which T helper 1 (Th1) immunity has been proposed to play an important role. Nai;ve CD4+ T cells differentiate into interferon-gamma (IFN-gamma) producing Th1 effector cells when stimulated by interleukin-18 (IL-18) and IL-12. We wanted to directly test whether the Th1 pathway is proatherogenic. METHODS: We bred IL-18(-/-) mice with apolipoprotein E(-/-) (apoE(-/-)) mice and assessed atherosclerosis in the aortic root of the offspring. RESULTS: 24-week-old IL-18 deficient apoE(-/-) mice exhibited substantially reduced lesion size (93,866+/-11273 vs. 144,019+/-9667 microm(2) in IL-18(+/+)xapoE(-/-) mice, P=0.005). Lesion cells in compound knockout mice displayed reduced I-A(b) expression, implying reduced local IFN-gamma stimulation. These mice also had an increased proportion of alpha-SM-actin+ smooth muscle cells, compatible with a more stable lesion phenotype. Immunoglobulin G (IgG) subclass analysis of antibodies to malondialdehyde-modified low density lipoprotein indicated increased Th2 and reduced Th1 helper to B cell antibody production. Surprisingly, serum cholesterol and triglyceride levels were significantly higher in IL-18(-/-)xapoE(-/-) mice in spite of their reduced atherosclerosis. However, no changes in lipoprotein cholesterol patterns were registered. CONCLUSION: These data show reduced atherosclerosis and Th1 activity in spite of increased serum cholesterol in IL-18 deficient apoE(-/-) mice. They support a proatherogenic role for IL-18.


Asunto(s)
Apolipoproteínas E/deficiencia , Arteriosclerosis/inmunología , Interleucina-18/genética , Músculo Liso Vascular/metabolismo , Células TH1/inmunología , Actinas/metabolismo , Animales , Apolipoproteínas E/genética , Arteriosclerosis/metabolismo , Autoanticuerpos/inmunología , Colesterol/sangre , Expresión Génica , Inmunoglobulina G/análisis , Interferón gamma/genética , Interferón gamma/inmunología , Lipoproteínas LDL/inmunología , Masculino , Ratones , Ratones Noqueados , Triglicéridos/sangre
19.
Eur J Immunol ; 33(2): 512-21, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12645950

RESUMEN

It is widely accepted that females have superior immune responses than males, but the ways by which sex hormones may enhance T cell responses are still poorly understood. In the present study, we analyzed the effect of estrogens on CD4 T cell activation and differentiation after immunization with exogenous antigens. We show that administration of low doses of 17beta-estradiol (E2) to castrated female mice results in a striking increase of antigen-specific CD4 T cell responses and in the selective development of IFN-gamma-producing cells. Quantitative assessment of the frequency of T cells bearing a public TCR beta chain CDR3 motif demonstrated that the clonal size of primary antigen-specific CD4 T cells was dramatically increased in immune lymph nodes from E2-treated mice. By using mice with disrupted estrogen receptor (ER) alpha or beta genes, we show that ERalpha, but not ERbeta, was necessary for the enhanced E2-driven Th1 cell responsiveness. Furthermore, ERalpha expression in hematopoietic cells was essential, since E2 effects on Th1 responses were only observed in mice reconstituted with bone marrow cells from ERalpha+/+, but not ERalpha-deficient mice. These results demonstrate that estrogen administration promotes strong antigen-specific Th1 cell responses in a mechanism that requires functional expression of ERalpha in hematopoietic cells.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Estradiol/farmacología , Células Madre Hematopoyéticas/inmunología , Receptores de Estrógenos/fisiología , Células TH1/efectos de los fármacos , Secuencias de Aminoácidos , Animales , Trasplante de Médula Ósea , Linfocitos T CD4-Positivos/citología , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Relación Dosis-Respuesta a Droga , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Femenino , Reordenamiento Génico de la Cadena beta de los Receptores de Antígenos de los Linfocitos T , Marcación de Gen , Interferón gamma/biosíntesis , Interleucina-4/biosíntesis , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ovariectomía , Quimera por Radiación , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Estrógenos/deficiencia , Receptores de Estrógenos/genética , Organismos Libres de Patógenos Específicos , Células TH1/citología
20.
Biochem Pharmacol ; 65(4): 629-36, 2003 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-12566092

RESUMEN

From the MCF-7 cell line we have developed, a human mammary cancer cell subline with the same karyotype as the mother strain and named MCF-7(SF), able to grow in serum-free chemically defined medium. This cell subline was firstly used to analyze the effect of basic fibroblast growth factor (FGF-2) in estrogen-receptor-positive human breast cancer cells. FGF-2 like estradiol is able to increase cell proliferation and pS2 expression but was also found to inhibit progesterone receptor (PR) expression. The anti-estrogen tamoxifen partly counteracts the effects of FGF-2 and to discriminate between its two main mediators (estrogen receptor vs. anti-estrogen binding site, AEBS) we compare the efficacies of pure anti-estrogen (ICI 182,780) and AEBS ligand (PBPE). It appears that pure anti-estrogen counteracts cell growth and pS2 effects of FGF-2 since AEBS ligand inhibits the cell growth but has no activity on pS2 expression. Secondly, adding insulin (10(-6)M) in the culture medium induces a strong increase in cell proliferation, which then elicits an inhibitory effect of FGF-2 and addition of anti-estrogens, are less efficient to further decrease growth, since the effects of FGF-2 and anti-estrogens on pS2 expression are conserved.


Asunto(s)
Antagonistas de Estrógenos/farmacología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Insulina/farmacología , Proteínas , Receptores de Estrógenos/metabolismo , Neoplasias de la Mama/patología , División Celular/efectos de los fármacos , Receptor alfa de Estrógeno , Expresión Génica/efectos de los fármacos , Humanos , Ligandos , Biosíntesis de Proteínas , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Receptores de Progesterona/biosíntesis , Factor Trefoil-1 , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...