Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 8(11)2023 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-37191999

RESUMO

Progesterone (P4) is essential for embryo implantation, but the extent to which the pro-gestational effects of P4 depend on the maternal immune compartment is unknown. Here, we investigate whether regulatory T cells (Treg cells) act to mediate luteal phase P4 effects on uterine receptivity in mice. P4 antagonist RU486 administered to mice on days 0.5 and 2.5 postcoitum to model luteal phase P4 deficiency caused fewer CD4+Foxp3+ Treg cells and impaired Treg functional competence, along with dysfunctional uterine vascular remodeling and perturbed placental development in midgestation. These effects were linked with fetal loss and fetal growth restriction, accompanied by a Th1/CD8-skewed T cell profile. Adoptive transfer at implantation of Treg cells - but not conventional T cells - alleviated fetal loss and fetal growth restriction by mitigating adverse effects of reduced P4 signaling on uterine blood vessel remodeling and placental structure and by restoring maternal T cell imbalance. These findings demonstrate an essential role for Treg cells in mediating P4 effects at implantation and indicate that Treg cells are a sensitive and critical effector mechanism through which P4 drives uterine receptivity to support robust placental development and fetal growth.


Assuntos
Progesterona , Linfócitos T Reguladores , Humanos , Gravidez , Feminino , Animais , Camundongos , Progesterona/farmacologia , Placenta , Retardo do Crescimento Fetal , Implantação do Embrião/fisiologia , Desenvolvimento Fetal
2.
Endocrinology ; 163(9)2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35786711

RESUMO

Regulatory T (Treg) cells are a specialized CD4+ T cell subpopulation that are essential for immune homeostasis, immune tolerance, and protection against autoimmunity. There is evidence that sex-steroid hormones estrogen and progesterone modulate Treg cell abundance and phenotype in women. Since natural oscillations in these hormones are modified by hormonal contraceptives, we examined whether oral contraception (OC) use impacts Treg cells and related T cell populations. T cells were analyzed by multiparameter flow cytometry in peripheral blood collected across the menstrual cycle from healthy women either using OC or without hormonal contraception and from age-matched men. Compared to naturally cycling women, women using OC had fewer Treg cells and an altered Treg cell phenotype. Notably, Treg cells exhibiting a strongly suppressive phenotype, defined by high FOXP3, CD25, Helios, HLADR, CTLA4, and Ki67, comprised a lower proportion of total Treg cells, particularly in the early- and mid-cycle phases. The changes were moderate compared to more substantial differences in Treg cells between women and men, wherein women had fewer Treg cells-especially of the effector memory Treg cell subset-associated with more T helper type 1 (Th1) cells and CD8+ T cells and lower Treg:Th1 cell and Treg:CD8+ T cell ratios than men. These findings imply that OC can modulate the number and phenotype of peripheral blood Treg cells and raise the possibility that Treg cells contribute to the physiological changes and altered disease susceptibility linked with OC use.


Assuntos
Fatores de Transcrição Forkhead , Linfócitos T Reguladores , Anticoncepção , Feminino , Fatores de Transcrição Forkhead/metabolismo , Hormônios/metabolismo , Humanos , Fenótipo , Linfócitos T Reguladores/metabolismo
3.
Fertil Steril ; 117(6): 1107-1120, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35618356

RESUMO

Immune cells are essential for endometrial receptivity to embryo implantation and early placental development. They exert tissue-remodeling and immune regulatory roles-acting to promote epithelial attachment competence, regulate the differentiation of decidual cells, remodel the uterine vasculature, control and resolve inflammatory activation, and suppress destructive immunity to paternally inherited alloantigens. From a biological perspective, the endometrial immune response exerts a form of "quality control"-it promotes implantation success when conditions are favorable but constrains receptivity when physiological circumstances are not ideal. Women with recurrent implantation failure and recurrent miscarriage may exhibit altered numbers or disturbed function of certain uterine immune cell populations-most notably uterine natural killer cells and regulatory T cells. Preclinical and animal studies indicate that deficiencies or aberrant activation states in these cells can be causal in the pathophysiological mechanisms of infertility. Immune cells are, therefore, targets for diagnostic evaluation and therapeutic intervention. However, current diagnostic tests are overly simplistic and have limited clinical utility. To be more informative, they need to account for the full complexity and reflect the range of perturbations that can occur in uterine immune cell phenotypes and networks. Moreover, safe and effective interventions to modulate these cells are in their infancy, and personalized approaches matched to specific diagnostic criteria will be needed. Here we summarize current biological understanding and identify knowledge gaps to be resolved before the promise of therapies to target the uterine immune response can be fully realized.


Assuntos
Aborto Habitual , Placenta , Aborto Habitual/diagnóstico , Animais , Implantação do Embrião/fisiologia , Endométrio/fisiologia , Feminino , Humanos , Gravidez , Útero
4.
BMC Cancer ; 21(1): 736, 2021 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-34174867

RESUMO

BACKGROUND: The Oncotype DX 21-gene Recurrence Score is predictive of adjuvant chemotherapy benefit for women with early-stage, estrogen receptor (ER)-positive, HER2-negative breast cancer. In premenopausal women, fluctuations in estrogen and progesterone during the menstrual cycle impact gene expression in hormone-responsive cancers. However, the extent to which menstrual cycling affects the Oncotype DX 21-gene signature remains unclear. Here, we investigate the impact of ovarian cycle stage on the 21-gene signature using a naturally cycling mouse model of breast cancer. METHODS: ER-positive mammary tumours were dissected from naturally cycling Mmtv-Pymt mice at either the estrus or diestrus phase of the ovarian cycle. The Oncotype DX 21-gene signature was assessed through quantitative real time-PCR, and a 21-gene experimental recurrence score analogous to the Oncotype DX Recurrence Score was calculated. RESULTS: Tumours collected at diestrus exhibited significant differences in expression of 6 Oncotype DX signature genes (Ki67, Ccnb1, Esr1, Erbb2, Grb7, Bag1; p ≤ 0.05) and a significant increase in 21-gene recurrence score (21.8 ± 2.4; mean ± SEM) compared to tumours dissected at estrus (15.5 ± 1.9; p = 0.03). Clustering analysis revealed a subgroup of tumours collected at diestrus characterised by increased expression of proliferation- (p < 0.001) and invasion-group (p = 0.01) genes, and increased 21-gene recurrence score (p = 0.01). No correlation between ER, PR, HER2, and KI67 protein abundance measured by Western blot and abundance of mRNA for the corresponding gene was observed, suggesting that gene expression is more susceptible to hormone-induced fluctuation compared to protein expression. CONCLUSIONS: Ovarian cycle stage at the time of tissue collection critically affects the 21-gene signature in Mmtv-Pymt murine mammary tumours. Further studies are required to determine whether Oncotype DX Recurrence Scores in women are similarly affected by menstrual cycle stage.


Assuntos
Perfilação da Expressão Gênica/métodos , Genômica/métodos , Ciclo Menstrual/genética , Animais , Feminino , Neoplasias Mamárias Animais , Camundongos , Camundongos Transgênicos , Recidiva Local de Neoplasia
5.
Commun Biol ; 4(1): 572, 2021 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-33990675

RESUMO

Seminal fluid factors modulate the female immune response at conception to facilitate embryo implantation and reproductive success. Whether sperm affect this response has not been clear. We evaluated global gene expression by microarray in the mouse uterus after mating with intact or vasectomized males. Intact males induced greater changes in gene transcription, prominently affecting pro-inflammatory cytokine and immune regulatory genes, with TLR4 signaling identified as a top-ranked upstream driver. Recruitment of neutrophils and expansion of peripheral regulatory T cells were elevated by seminal fluid of intact males. In vitro, epididymal sperm induced IL6, CXCL2, and CSF3 in uterine epithelial cells of wild-type, but not Tlr4 null females. Collectively these experiments show that sperm assist in promoting female immune tolerance by eliciting uterine cytokine expression through TLR4-dependent signaling. The findings indicate a biological role for sperm beyond oocyte fertilization, in modulating immune mechanisms involved in female control of reproductive investment.


Assuntos
Implantação do Embrião/imunologia , Endométrio/imunologia , Tolerância Imunológica/imunologia , Reprodução , Espermatozoides/fisiologia , Linfócitos T Reguladores/imunologia , Útero/imunologia , Animais , Comunicação Celular , Citocinas/genética , Citocinas/metabolismo , Endométrio/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Transdução de Sinais , Linfócitos T Reguladores/metabolismo , Útero/metabolismo , Vasectomia
6.
J Immunol ; 203(3): 647-657, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31243091

RESUMO

Regulatory T cells (Tregs) are essential for maternal tolerance in allogeneic pregnancy. In preeclampsia, Tregs are fewer and display aberrant phenotypes, particularly in the thymic Treg (tTreg) compartment, potentially because of insufficient priming to male partner alloantigens before conception. To investigate how tTregs as well as peripheral Tregs (pTregs) respond to male partner seminal fluid, Foxp3+CD4+ Tregs were examined in the uterus and uterus-draining lymph nodes in virgin estrus mice and 3.5 d postcoitum. Mating elicited 5-fold increases in uterine Tregs accompanied by extensive Treg proliferation in the uterus-draining lymph nodes, comprising 70% neuropilin 1+ tTregs and 30% neuropilin 1- pTregs. Proliferation marker Ki67 and suppressive competence markers Foxp3 and CTLA4 were induced after mating in both subsets, and Ki67, CTLA4, CD25, and GITR were higher in tTregs than in pTregs. Analysis by t-stochastic neighbor embedding confirmed phenotypically distinct tTreg and pTreg clusters, with the proportion of tTregs but not pTregs among CD4+ T cells expanding in response to seminal fluid. Bisulphite sequencing revealed increased demethylation of the Treg-specific demethylation region in the Foxp3 locus in tTregs but not pTregs after mating. These data show that tTregs and pTregs with distinct phenotypes both respond to seminal fluid priming, but the Foxp3 epigenetic signature is uniquely increased in tTregs. We conclude that reproductive tract tTregs as well as pTregs are sensitive to local regulation by seminal fluid, providing a candidate mechanism warranting evaluation for the potential to influence preeclampsia susceptibility in women.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Sêmen/imunologia , Comportamento Sexual Animal , Linfócitos T Reguladores/imunologia , Útero/imunologia , Animais , Antígeno CTLA-4/metabolismo , Proliferação de Células/fisiologia , Epigênese Genética , Feminino , Fatores de Transcrição Forkhead/genética , Proteína Relacionada a TNFR Induzida por Glucocorticoide/metabolismo , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Linfonodos/citologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neuropilina-1/metabolismo , Pré-Eclâmpsia/imunologia , Pré-Eclâmpsia/patologia , Gravidez , Timo/citologia , Útero/citologia
7.
Hum Reprod ; 31(10): 2164-73, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27591233

RESUMO

There is ongoing interest in immune-suppressant corticosteroid drugs such as prednisolone to treat infertility in women with repeated IVF failure and recurrent miscarriage. The rationale draws on the pervasive but flawed view that immune activation is inconsistent with normal pregnancy. This ignores clear evidence that controlled inflammation and activation of the immune response is essential for embryo implantation. Generally, the immune response actively promotes reproductive success - by facilitating endometrial receptivity and tolerance of the foreign embryo, and promoting vascular adaptation to support placental morphogenesis. The peri-conception immune response also establishes developmental trajectories that can impact on fetal growth and gestational age at birth. Here, we describe immune changes accompanying conception that could be impeded by inappropriate corticosteroid administration. While women with specific clinical conditions may benefit from the anti-inflammatory and immune-deviating actions of prednisolone and related drugs, it is incorrect to assume a 'one-size-fits-all' approach. Better diagnostics and more preclinical studies are essential to define patient groups, build evidence for efficacy and fine-tune treatments so as not to inhibit essential actions of immune cells. We argue that unless overt immune pathology is evident, utilization of corticosteroids is not warranted and may be harmful. In most women, perturbing immune adaptation at implantation is expected to adversely influence placental development and impair immune-mediated quality control mechanisms, potentially elevating risk of altered fetal growth and developmental programming, congenital anomalies and preterm birth.


Assuntos
Aborto Habitual/imunologia , Endométrio/imunologia , Imunossupressores/uso terapêutico , Prednisolona/uso terapêutico , Técnicas de Reprodução Assistida , Implantação do Embrião/imunologia , Feminino , Humanos , Gravidez
8.
Stem Cell Res ; 14(3): 380-95, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25900163

RESUMO

Circulating endothelial progenitor cells (EPCs) provide revascularisation for cardiovascular disease and the expansion of these cells opens up the possibility of their use as a cell therapy. Herein we show that interleukin-3 (IL3) strongly expands a population of human non-adherent endothelial forming cells (EXnaEFCs) with low immunogenicity as well as pro-angiogenic capabilities in vivo, making their therapeutic utilisation a realistic option. Non-adherent CD133(+) EFCs isolated from human umbilical cord blood and cultured under different conditions were maximally expanded by day 12 in the presence of IL3 at which time a 350-fold increase in cell number was obtained. Cell surface marker phenotyping confirmed expression of the hematopoietic progenitor cell markers CD133, CD117 and CD34, vascular cell markers VEGFR2 and CD31, dim expression of CD45 and absence of myeloid markers CD14 and CD11b. Functional experiments revealed that EXnaEFCs exhibited classical properties of endothelial cells (ECs), namely binding of Ulex europaeus lectin, up-take of acetylated-low density lipoprotein and contribution to EC tube formation in vitro. These EXnaEFCs demonstrated a pro-angiogenic phenotype within two independent in vivo rodent models. Firstly, a Matrigel plug assay showed increased vascularisation in mice. Secondly, a rat model of acute myocardial infarction demonstrated reduced heart damage as determined by lower levels of serum creatinine and a modest increase in heart functionality. Taken together, these studies show IL3 as a potent growth factor for human CD133(+) cell expansion with clear pro-angiogenic properties (in vitro and in vivo) and thus may provide clinical utility for humans in the future.


Assuntos
Proliferação de Células/efeitos dos fármacos , Células Progenitoras Endoteliais/fisiologia , Interleucina-3/farmacologia , Neovascularização Fisiológica/fisiologia , Animais , Técnicas de Cultura de Células , Terapia Baseada em Transplante de Células e Tecidos , Células Cultivadas , Células Progenitoras Endoteliais/citologia , Células Progenitoras Endoteliais/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Infarto do Miocárdio/terapia , Miocárdio/patologia , Ratos , Regeneração
9.
Biol Reprod ; 91(3): 60, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25061095

RESUMO

Macrophages are abundant in the uterine stroma and are intimately juxtaposed with other cell lineages comprising the uterine epithelial and stromal compartments. We postulated that macrophages may participate in mediating or amplifying the effects of ovarian steroid hormones to facilitate the uterine remodeling that is a characteristic feature of every estrus cycle and is essential for pregnancy. Using the Cd11b-Dtr transgenic mouse model with an ovariectomy and hormone replacement strategy, we depleted macrophages to determine their role in hormone-driven proliferation of uterine epithelial and stromal cells and uterine vascular development. Following diphtheria toxin (DT) administration, approximately 85% of EMR1-positive (EMR1⁺) macrophages, as well as 70% of CD11C⁺ dendritic cells, were depleted from Cd11b-Dtr mice. There was no change in bromodeoxyuridine incorporation into epithelial cells induced to proliferate by administration of 17beta-estradiol (E2) to ovariectomized mice or into stromal cells induced to proliferate in response to E2 and progesterone (P4), and the resulting sizes and structures of the luminal epithelial and stromal cell compartments were not altered compared with those of leukocyte replete controls. Depletion of CD11B⁺ myeloid cells failed to alter the density or pattern of distribution of uterine blood vessels, as identified by staining PECAM1-positive endothelial cells in the uterine stroma of E2- or E2 combined with P4 (E2P4)-treated ovariectomized mice. These experiments support the interpretation that macrophages are dispensable to regulation of proliferative events induced by steroid hormones in the cycling and early pregnant mouse uterus to establish the epithelial, stromal, and vascular architecture which is critical for normal reproductive competence.


Assuntos
Endométrio/imunologia , Estradiol/metabolismo , Ciclo Estral , Macrófagos/imunologia , Progesterona/metabolismo , Útero/imunologia , Remodelação Vascular , Animais , Biomarcadores/metabolismo , Proliferação de Células , Toxina Diftérica , Endométrio/irrigação sanguínea , Endométrio/citologia , Endométrio/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Feminino , Terapia de Reposição Hormonal , Imuno-Histoquímica , Terapia de Imunossupressão , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos Transgênicos , Ovariectomia , Organismos Livres de Patógenos Específicos , Útero/irrigação sanguínea , Útero/citologia , Útero/metabolismo
10.
Int J Dev Biol ; 58(2-4): 205-17, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25023687

RESUMO

The capacity of the immune system to maintain the integrity of the individual requires recognition and control of entities identified as genetically distinct, or 'non-self'. In mammalian reproduction, the embryo and subsequent fetus and placenta are all recognized as non-self by the maternal immune system, and are vulnerable to immunological attack. An active system to prevent rejection must exist from when conceptus and maternal tissues first come into contact at implantation. Crucial mediators of immune protection are inducible regulatory T cells (Treg cells). Unless sufficient Treg cells are present in the endometrium, successful implantation and progression to pregnancy cannot ensue. This key role of Treg cells confers to the female immune system substantial capability to influence reproductive events, particularly around the time of conception and embryo implantation. While on the one hand this risks susceptibility to immune-based reproductive disorders, the potential evolutionary trade-off is the benefit of quality control to avoid poor reproductive outcomes. Here we summarize current knowledge of the factors required to establish a robust Treg cell response and an immune environment conducive to successful implantation and pregnancy. These factors include (a) appropriate cytokine balance; (b) correct phenotype of endometrial leukocytes to enable Treg cell activation; (c) sufficient estrogen and progesterone to stabilize and strengthen Treg cell phenotype, and (d) appropriate priming of Treg cell populations by male partner seminal fluid. Compromises in the quality of this immune adaptation at conception can influence the early embryo and either prevent implantation or impair placental morphogenesis. Failure to successfully establish Treg cell-mediated immune tolerance can result in poor fertility or impart long-term adverse consequences for the fetus and offspring.


Assuntos
Implantação do Embrião/imunologia , Embrião de Mamíferos/imunologia , Endométrio/imunologia , Fertilização/imunologia , Linfócitos T Reguladores/imunologia , Animais , Feminino , Humanos , Tolerância Imunológica , Masculino , Gravidez
11.
Am J Reprod Immunol ; 69(4): 315-30, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23480148

RESUMO

T regulatory (Treg) cells are essential mediators of the maternal immune adaptation necessary for embryo implantation. In mice, insufficient Treg cell activity results in implantation failure, or constrains placental function and fetal growth. In women, Treg cell deficiency is linked with unexplained infertility, miscarriage, and pre-eclampsia. To devise strategies to improve Treg cell function, it is essential to define the origin of the Treg cells in gestational tissues, and the regulators that control their functional competence and recruitment. Male seminal fluid is a potent source of the Treg cell-inducing agents TGFß and prostaglandin E, and coitus is one key factor involved in expanding the pool of inducible Treg cells that react with paternal alloantigens shared by conceptus tissues. In mice, coitus initiates a sequence of events whereby female dendritic cells cross-present seminal fluid antigens and activate T cells, which in turn circulate via the blood to be sequestered into the endometrium. Similar events may occur in the human genital tract, where seminal fluid induces immune cell changes that appear competent to prime Treg cells. Improved understanding of how seminal fluid influences Treg cells in women should ultimately assist in the development of new therapies for immune-mediated pathologies of pregnancy.


Assuntos
Implantação do Embrião/imunologia , Complicações na Gravidez/imunologia , Sêmen/imunologia , Proteínas de Plasma Seminal/imunologia , Linfócitos T Reguladores/imunologia , Aborto Espontâneo/imunologia , Animais , Células Dendríticas/imunologia , Endométrio/imunologia , Feminino , Humanos , Infertilidade/imunologia , Isoantígenos/imunologia , Masculino , Camundongos , Pré-Eclâmpsia/imunologia , Gravidez , Prostaglandinas E , Fator de Crescimento Transformador beta
12.
Development ; 137(24): 4229-38, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21068060

RESUMO

Each ovarian cycle, the mammary gland epithelium rotates through a sequence of hormonally regulated cell proliferation, differentiation and apoptosis. These studies investigate the role of macrophages in this cellular turnover. Macrophage populations and their spatial distribution were found to fluctuate across the cycle. The number of macrophages was highest at diestrus, and the greatest number of macrophages in direct contact with epithelial cells occurred at proestrus. The physiological necessity of macrophages in mammary gland morphogenesis during the estrous cycle was demonstrated in Cd11b-Dtr transgenic mice. Ovariectomised mice were treated with estradiol and progesterone to stimulate alveolar development, and with the progesterone receptor antagonist mifepristone to induce regression of the newly formed alveolar buds. Macrophage depletion during alveolar development resulted in a reduction in both ductal epithelial cell proliferation and the number of alveolar buds. Macrophage depletion during alveolar regression resulted in an increased number of branch points and an accumulation of TUNEL-positive cells. These studies show that macrophages have two roles in the cellular turnover of epithelial cells in the cycling mammary gland; following ovulation, they promote the development of alveolar buds in preparation for possible pregnancy, and they remodel the tissue back to its basic architecture in preparation for a new estrous cycle.


Assuntos
Epitélio/metabolismo , Ciclo Estral/fisiologia , Macrófagos/metabolismo , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Animais , Epitélio/efeitos dos fármacos , Estradiol/sangue , Estradiol/farmacologia , Estrogênios/farmacologia , Ciclo Estral/efeitos dos fármacos , Feminino , Citometria de Fluxo , Antagonistas de Hormônios/farmacologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Glândulas Mamárias Animais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Mifepristona/farmacologia , Progesterona/sangue , Progesterona/farmacologia , Progestinas/farmacologia , Receptores de Progesterona/antagonistas & inibidores
13.
J Immunol ; 185(11): 7085-96, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20974989

RESUMO

Uterine dendritic cells (DCs) are critical for activating the T cell response mediating maternal immune tolerance of the semiallogeneic fetus. GM-CSF (CSF2), a known regulator of DCs, is synthesized by uterine epithelial cells during induction of tolerance in early pregnancy. To investigate the role of GM-CSF in regulating uterine DCs and macrophages, Csf2-null mutant and wild-type mice were evaluated at estrus, and in the periconceptual and peri-implantation periods. Immunohistochemistry showed no effect of GM-CSF deficiency on numbers of uterine CD11c(+) cells and F4/80(+) macrophages at estrus or on days 0.5 and 3.5 postcoitum, but MHC class II(+) and class A scavenger receptor(+) cells were fewer. Flow cytometry revealed reduced CD80 and CD86 expression by uterine CD11c(+) cells and reduced MHC class II in both CD11c(+) and F4/80(+) cells from GM-CSF-deficient mice. CD80 and CD86 were induced in Csf2(-/-) uterine CD11c(+) cells by culture with GM-CSF. Substantially reduced ability to activate both CD4(+) and CD8(+) T cells in vivo was evident after delivery of OVA Ag by mating with Act-mOVA males or transcervical administration of OVA peptides. This study shows that GM-CSF regulates the efficiency with which uterine DCs and macrophages activate T cells, and it is essential for optimal MHC class II- and class I-mediated indirect presentation of reproductive Ags. Insufficient GM-CSF may impair generation of T cell-mediated immune tolerance at the outset of pregnancy and may contribute to the altered DC profile and dysregulated T cell tolerance evident in infertility, miscarriage, and preeclampsia.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Ativação Linfocitária/imunologia , Proteínas da Gravidez/fisiologia , Subpopulações de Linfócitos T/imunologia , Útero/imunologia , Útero/metabolismo , Sequência de Aminoácidos , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Endométrio/embriologia , Endométrio/imunologia , Endométrio/metabolismo , Epitopos de Linfócito T/imunologia , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/deficiência , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Especificidade de Órgãos/imunologia , Gravidez , Proteínas da Gravidez/deficiência , Proteínas da Gravidez/genética , Subpopulações de Linfócitos T/metabolismo , Útero/embriologia
14.
J Immunol ; 182(12): 8080-93, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19494334

RESUMO

The events that generate T cell-mediated immune tolerance in early pregnancy are ill-defined. To investigate the significance of seminal fluid Ags in activating maternal T cells, and define the underlying Ag presentation pathways, OVA-specific T cells were adoptively transferred to female mice inseminated by males ubiquitously expressing membrane-bound OVA. OVA-reactive CD8(+) OT-I and CD4(+) OT-II T cells transferred to mated recipients expressed activation markers CD25 and CD69 and proliferated vigorously in the para-aortic lymph nodes, but not in distal lymph nodes or spleen, and OT-I T cells expressed IFN-gamma and IL-2. In contrast, OT-I T cells transferred later in pregnancy or up to 10 days postpartum expressed CD25 and CD69 and proliferated in all peripheral lymphoid tissues examined. OVA Ag was present predominantly in the plasma fraction of seminal fluid, and seminal plasma, but not sperm, was necessary for T cell proliferation. Female H-2K(b) bone marrow-derived cells expressing TAP were essential for OT-I T cell proliferation, but responses were not elicited by OVA Ag presented by paternal MHC in seminal fluid or associated with placental cells. This study shows that at conception, seminal fluid drives activation and expansion of paternal Ag-reactive CD4(+) and CD8(+) T cell populations, and female APCs have an essential role in cross-presenting Ag to CD8(+) T cells via a TAP-dependent pathway. Delivery of paternal Ags and immune-deviating cytokines by seminal fluid at conception may activate Ag-dependent CD4(+) and CD8(+) regulatory T cells mediating tolerance of pregnancy.


Assuntos
Antígenos/imunologia , Apresentação Cruzada/imunologia , Imunidade Inata/imunologia , Ativação Linfocitária/imunologia , Gravidez/imunologia , Sêmen/imunologia , Linfócitos T/imunologia , Animais , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/imunologia , Medula Óssea/imunologia , Diferenciação Celular/imunologia , Proliferação de Células , Células Cultivadas , Citocinas/imunologia , Feminino , Cinética , Masculino , Camundongos , Camundongos Knockout , Comportamento Sexual Animal
15.
Exp Hematol ; 36(1): 51-60, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17949888

RESUMO

OBJECTIVE: Dendritic cells (DCs) play a pivotal role in the induction of immunity in response to pathogenic challenge or vaccination. As such, the fms-like tyrosine kinase 3-ligand (Flt-3L) has been used to increase DC populations in vivo, with contrasting outcomes, which include an increase in immunity, tolerance induction, or expansion of regulatory cells. This study examines the adjuvant role that human Flt-3L (hFL) administration has in generating immune responses upon immunization with a poorly immunogenic and soluble protein antigen. MATERIALS AND METHODS: Mice were immunized with the nominal antigen, ovalbumin, alone or with antigen emulsified in complete Freund's adjuvant (CFA), with or without prior hFL-mediated expansion of DC subsets. The maturation of DC subsets and activation status of antigen-specific T cells were analyzed by flow cytometry, with effector function assessed in cytolytic T-lymphocyte assays. RESULTS: hFL treatment expanded both conventional DC and plasmacytoid DC in vivo, resulting in increased antigen presentation by both direct and cross-presentation pathways. However, it was only in the context of CFA that antigen immunization could mature DCs and subsequently fully activate antigen-specific T cells with enhanced cytolytic activity. CONCLUSIONS: Our studies reveal that hFL essentially acts as a coadjuvant, as hFL augments the size of an immune response but requires further adjuvant activation to alter the quality of the response.


Assuntos
Apresentação de Antígeno/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Proteínas de Membrana/farmacologia , Adjuvantes Imunológicos , Transferência Adotiva , Sequência de Aminoácidos , Animais , Apresentação de Antígeno/fisiologia , Citotoxicidade Imunológica/efeitos dos fármacos , Células Dendríticas/classificação , Células Dendríticas/imunologia , Proteínas do Ovo/imunologia , Adjuvante de Freund/farmacologia , Humanos , Imunização , Linfonodos/citologia , Linfonodos/imunologia , Ativação Linfocitária , Proteínas de Membrana/imunologia , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Ovalbumina/imunologia , Fragmentos de Peptídeos/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Baço/citologia , Baço/imunologia , Subpopulações de Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA