Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Annu Rev Immunol ; 31: 387-411, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23298207

RESUMEN

The immune cells that reside at the interface between the placenta and uterus are thought to play many important roles in pregnancy. Recent work has revealed that the composition and function of these cells are locally controlled by the specialized uterine stroma (the decidua) that surrounds the implanted conceptus. Here, I discuss how key immune cell types (natural killer cells, macrophages, dendritic cells, and T cells) are either enriched or excluded from the decidua, how their function is regulated within the decidua, and how they variously contribute to pregnancy success or failure. The discussion emphasizes the relationship between human and mouse studies. Deeper understanding of the immunology of the maternal-fetal interface promises to yield significant insight into the pathogenesis of many human pregnancy complications, including preeclampsia, intrauterine growth restriction, spontaneous abortion, preterm birth, and congenital infection.


Asunto(s)
Intercambio Materno-Fetal/inmunología , Animales , Diferenciación Celular/inmunología , Decidua/citología , Decidua/inmunología , Decidua/patología , Implantación del Embrión/inmunología , Femenino , Humanos , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Placenta/irrigación sanguínea , Placenta/inmunología , Embarazo
2.
Immunity ; 56(3): 606-619.e7, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36750100

RESUMEN

Although mice normally enter labor when their ovaries stop producing progesterone (luteolysis), parturition can also be triggered in this species through uterus-intrinsic pathways potentially analogous to the ones that trigger parturition in humans. Such pathways, however, remain largely undefined in both species. Here, we report that mice deficient in innate type 2 immunity experienced profound parturition delays when manipulated endocrinologically to circumvent luteolysis, thus obliging them to enter labor through uterus-intrinsic pathways. We found that these pathways were in part driven by the alarmin IL-33 produced by uterine interstitial fibroblasts. We also implicated important roles for uterine group 2 innate lymphoid cells, which demonstrated IL-33-dependent activation prior to labor onset, and eosinophils, which displayed evidence of elevated turnover in the prepartum uterus. These findings reveal a role for innate type 2 immunity in controlling the timing of labor onset through a cascade potentially relevant to human parturition.


Asunto(s)
Interleucina-33 , Luteólisis , Embarazo , Femenino , Ratones , Animales , Humanos , Interleucina-33/metabolismo , Inmunidad Innata , Miometrio/metabolismo , Linfocitos , Parto/metabolismo
4.
Infect Immun ; 85(8)2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28507070

RESUMEN

The bacterial pathogen Listeria monocytogenes causes foodborne systemic disease in pregnant women, which can lead to preterm labor, stillbirth, or severe neonatal disease. Colonization of the maternal decidua appears to be an initial step in the maternal component of the disease as well as bacterial transmission to the placenta and fetus. Host-pathogen interactions in the decidua during this early stage of infection remain poorly understood. Here, we assessed the dynamics of L. monocytogenes infection in primary human decidual organ cultures and in the murine decidua in vivo A high inoculum was necessary to infect both human and mouse deciduas, and the data support the existence of a barrier to initial colonization of the murine decidua. If successful, however, colonization in both species was followed by significant bacterial expansion associated with an inability of the decidua to mount appropriate innate cellular immune responses. The innate immune deficits included the failure of bacterial foci to attract macrophages and NK cells, cell types known to be important for early defenses against L. monocytogenes in the spleen, as well as a decrease in the tissue density of inflammatory Ly6Chi monocytes in vivo These results suggest that the infectivity of the decidua is not the result of an enhanced recruitment of L. monocytogenes to the gestational uterus but rather is due to compromised local innate cellular immune responses.


Asunto(s)
Decidua/microbiología , Interacciones Huésped-Patógeno , Listeria monocytogenes/inmunología , Listeria monocytogenes/fisiología , Listeriosis/inmunología , Animales , Antígenos Ly/inmunología , Decidua/inmunología , Femenino , Humanos , Inmunidad Innata , Células Asesinas Naturales/inmunología , Listeria monocytogenes/crecimiento & desarrollo , Listeria monocytogenes/patogenicidad , Listeriosis/microbiología , Listeriosis/transmisión , Macrófagos/inmunología , Ratones , Monocitos/microbiología , Técnicas de Cultivo de Órganos , Placenta/inmunología , Placenta/microbiología , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/microbiología , Bazo/inmunología , Bazo/microbiología
5.
Sci Am ; 317(4): 46-53, 2017 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-29565852
6.
J Exp Med ; 219(5)2022 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-35416936

RESUMEN

The paradox of fetomaternal tolerance has puzzled immunologists and reproductive biologists alike for almost 70 yr. Even the idea that the conceptus evokes a uniformly tolerogenic immune response in the mother is contradicted by the long-appreciated ability of pregnant women to mount robust antibody responses to paternal HLA molecules and RBC alloantigens such as Rh(D). Synthesizing these older observations with more recent work in mice, we discuss how the decision between tolerance or immunity to a given fetoplacental antigen appears to be a function of whether the antigen is trophoblast derived-and thus decorated with immunosuppressive glycans-or fetal blood cell derived.


Asunto(s)
Sangre Fetal , Trofoblastos , Animales , Femenino , Feto , Antígenos HLA , Humanos , Tolerancia Inmunológica , Isoantígenos , Ratones , Embarazo
7.
Cell Rep ; 38(5): 110329, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35108527

RESUMEN

A little-appreciated feature of early pregnancy is that embryo implantation and placental outgrowth do not evoke wound-healing responses in the decidua, the specialized endometrial tissue that surrounds the conceptus. Here, we provide evidence that this phenomenon is partly due to an active program of gene silencing mediated by EZH2, a histone methyltransferase that generates repressive histone 3 lysine 27 trimethyl (H3K27me3) histone marks. We find that pregnancies in mice with EZH2-deficient decidual stromal cells frequently fail by mid-gestation, with the decidua showing ectopic myofibroblast formation, peri-embryonic collagen deposition, and gene expression profiles associated with transforming growth factor ß (TGF-ß)-driven fibroblast activation and fibrogenic extracellular matrix (ECM) remodeling. Analogous responses are observed when the mutant decidua is surgically wounded, while blockade of TGF-ß receptor signaling inhibits the defects and improves reproductive outcomes. Together, these results highlight a critical feature of reproductive success and have implications for the context-specific control of TGF-ß-mediated wound-healing responses elsewhere in the body.


Asunto(s)
Implantación del Embrión/fisiología , Silenciador del Gen/fisiología , Placenta/metabolismo , Factor de Crecimiento Transformador beta/genética , Cicatrización de Heridas/fisiología , Animales , Decidua/metabolismo , Embrión de Mamíferos/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Femenino , Expresión Génica/fisiología , Histonas/metabolismo , Humanos , Ratones Endogámicos C57BL , Embarazo , Células del Estroma/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
8.
J Clin Invest ; 117(5): 1399-411, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17446933

RESUMEN

How the fetus escapes rejection by the maternal immune system remains one of the major unsolved questions in transplantation immunology. Using a system to visualize both CD4+ and CD8+ T cell responses during pregnancy in mice, we find that maternal T cells become aware of the fetal allograft exclusively through "indirect" antigen presentation, meaning that T cell engagement requires the uptake and processing of fetal/placental antigen by maternal APCs. This reliance on a relatively minor allorecognition pathway removes a major threat to fetal survival, since it avoids engaging the large number of T cells that typically drive acute transplant rejection through their ability to directly interact with foreign MHC molecules. Furthermore, CD8+ T cells that indirectly recognize fetal/placental antigen undergo clonal deletion without priming for cytotoxic effector function and cannot induce antigen-specific fetal demise even when artificially activated. Antigen presentation commenced only at mid-gestation, in association with the endovascular invasion of placental trophoblasts and the hematogenous release of placental debris. Our results suggest that limited pathways of antigen presentation, in conjunction with tandem mechanisms of immune evasion, contribute to the unique immunological status of the fetus. The pronounced degree of T cell ignorance of the fetus also has implications for the pathophysiology of immune-mediated early pregnancy loss.


Asunto(s)
Presentación de Antígeno/inmunología , Feto/inmunología , Antígenos H-2/inmunología , Antígenos H-2/metabolismo , Linfocitos T/inmunología , Animales , Proteínas del Huevo/inmunología , Proteínas del Huevo/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/inmunología , Ovalbúmina/metabolismo , Fragmentos de Péptidos , Embarazo , Linfocitos T/metabolismo
9.
J Clin Invest ; 130(1): 389-403, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31600172

RESUMEN

Polymorphonuclear neutrophils (PMNs) are increasingly recognized to influence solid tumor development, but why their effects are so context dependent and even frequently divergent remains poorly understood. Using an autochthonous mouse model of uterine cancer and the administration of respiratory hyperoxia as a means to improve tumor oxygenation, we provide in vivo evidence that hypoxia is a potent determinant of tumor-associated PMN phenotypes and direct PMN-tumor cell interactions. Upon relief of tumor hypoxia, PMNs were recruited less intensely to the tumor-bearing uterus, but the recruited cells much more effectively killed tumor cells, an activity our data moreover suggested was mediated via their production of NADPH oxidase-derived reactive oxygen species and MMP-9. Simultaneously, their ability to promote tumor cell proliferation, which appeared to be mediated via their production of neutrophil elastase, was rendered less effective. Relieving tumor hypoxia thus greatly improved net PMN-dependent tumor control, leading to a massive reduction in tumor burden. Remarkably, this outcome was T cell independent. Together, these findings identify key hypoxia-regulated molecular mechanisms through which PMNs directly induce tumor cell death and proliferation in vivo and suggest that the contrasting properties of PMNs in different tumor settings may in part reflect the effects of hypoxia on direct PMN-tumor cell interactions.


Asunto(s)
Comunicación Celular/inmunología , Neoplasias Experimentales/inmunología , Neutrófilos/inmunología , Neoplasias Uterinas/inmunología , Animales , Hipoxia de la Célula/inmunología , Femenino , Ratones , Ratones Noqueados , Neoplasias Experimentales/patología , Neutrófilos/patología , Neoplasias Uterinas/patología
10.
J Clin Invest ; 116(10): 2643-52, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16998586

RESUMEN

We studied the vascular effects of invasive human cytotrophoblasts in vivo by transplanting placental villi to the fifth mammary fat pads or beneath the kidney capsules of Scid mice. Over 3 weeks, robust cytotrophoblast invasion was observed in both locations. The architecture of the mammary fat pad allowed for detailed analysis of the cells' interactions with resident murine blood vessels, which revealed specific induction of apoptosis in the endothelial cells and smooth muscle walls of the arterioles. This finding, and confirmation of the results in an in vitro coculture model, suggests that a parallel process is important for enabling cytotrophoblast endovascular invasion during human pregnancy. Cytotrophoblast invasion of the kidney parenchyma was accompanied by a robust lymphangiogenic response, while in vitro, the cells stimulated lymphatic endothelial cell migration via the actions of VEGF family members, FGF, and TNF-alpha. Immunolocalization analyses revealed that human pregnancy is associated with lymphangiogenesis in the decidua since lymphatic vessels were not a prominent feature of the nonpregnant endometrium. Thus, the placenta triggers the development of a decidual lymphatic circulation, which we theorize plays an important role in maintaining fluid balance during pregnancy, with possible implications for maternal-fetal immune cell trafficking.


Asunto(s)
Apoptosis/fisiología , Arterias/citología , Linfangiogénesis/fisiología , Placentación/fisiología , Trofoblastos/fisiología , Animales , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Vellosidades Coriónicas/trasplante , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Decidua/citología , Decidua/crecimiento & desarrollo , Endometrio/citología , Endometrio/crecimiento & desarrollo , Células Endoteliales/citología , Femenino , Glicoproteínas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Vasos Linfáticos/metabolismo , Proteínas de Transporte de Membrana , Ratones , Ratones SCID , Modelos Animales , Embarazo , Trofoblastos/citología , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas de Transporte Vesicular
12.
J Clin Invest ; 128(1): 233-247, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29202469

RESUMEN

Uncovering the causes of pregnancy complications such as preterm labor requires greater insight into how the uterus remains in a noncontractile state until term and then surmounts this state to enter labor. Here, we show that dynamic generation and erasure of the repressive histone modification tri-methyl histone H3 lysine 27 (H3K27me3) in decidual stromal cells dictate both elements of pregnancy success in mice. In early gestation, H3K27me3-induced transcriptional silencing of select gene targets ensured uterine quiescence by preventing the decidua from expressing parturition-inducing hormone receptors, manifesting type 1 immunity, and most unexpectedly, generating myofibroblasts and associated wound-healing responses. In late gestation, genome-wide H3K27 demethylation allowed for target gene upregulation, decidual activation, and labor entry. Pharmacological inhibition of H3K27 demethylation in late gestation not only prevented term parturition, but also inhibited delivery while maintaining pup viability in a noninflammatory model of preterm parturition. Immunofluorescence analysis of human specimens suggested that similar regulatory events might occur in the human decidua. Together, these results reveal the centrality of regulated gene silencing in the uterine adaptation to pregnancy and suggest new areas in the study and treatment of pregnancy disorders.


Asunto(s)
Decidua/metabolismo , Histonas/metabolismo , Parto/metabolismo , Embarazo/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Femenino , Silenciador del Gen/fisiología , Humanos , Masculino , Metilación , Ratones , Regulación hacia Arriba/fisiología
13.
J Clin Invest ; 114(1): 39-48, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15232610

RESUMEN

We describe a murine model of early pregnancy failure induced by systemic activation of the CD40 immune costimulatory pathway. Although fetal loss involved an NK cell intermediate, it was not due to lymphocyte-mediated destruction of the fetus and placenta. Rather, pregnancy failure resulted from impaired progesterone synthesis by the corpus luteum of the ovary, an endocrine defect in turn associated with ovarian resistance to the gonadotropic effects of prolactin. Pregnancy failure also required the proinflammatory cytokine TNF-alpha and correlated with the luteal induction of the prolactin receptor signaling inhibitors suppressor of cytokine signaling 1 (Socs1) and Socs3. Such links between immune activation and reproductive endocrine dysfunction may be relevant to pregnancy loss and other clinical disorders of reproduction.


Asunto(s)
Aborto Espontáneo/inmunología , Insuficiencia Ovárica Primaria/inmunología , Animales , Antígenos CD40/inmunología , Cuerpo Lúteo/inmunología , Cuerpo Lúteo/fisiopatología , Modelos Animales de Enfermedad , Femenino , Muerte Fetal , Inflamación , Células Asesinas Naturales/inmunología , Ratones , Embarazo , Receptores de Prolactina/genética , Receptores de Prolactina/fisiología , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
15.
Trends Mol Med ; 23(12): 1070-1071, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29122491

RESUMEN

Maternal immune activation (MIA) during pregnancy is associated with an increased risk of behavioral disorders in the offspring of affected mothers. Two recent studies highlight how maternal inflammation disrupts inhibitory interneuron networks and suggest that the maternal gut microbiome may be a contributing risk factor for MIA-induced behavioral abnormalities.


Asunto(s)
Trastorno Autístico/etiología , Encéfalo/fisiopatología , Microbioma Gastrointestinal/fisiología , Inflamación/complicaciones , Animales , Trastorno Autístico/microbiología , Femenino , Inflamación/microbiología , Interneuronas/fisiología , Madres , Embarazo
16.
J Leukoc Biol ; 78(6): 1347-55, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16204645

RESUMEN

During human and rodent uterine decidualization, transient but abundant numbers of uterine natural killer (uNK) cells appear, proliferate, and differentiate. uNK cells share features with peripheral NK cells but are specialized to promote interferon-gamma (IFN-gamma)-mediated, pregnancy-associated, structural changes in maternal placental arteries. In CD8+ T cells and NK cells, the transcription factors T-bet and eomesodermin (Eomes) regulate maturation and effector functions, including IFN-gamma production. No studies are reported for uNK cells. Implantation sites in T-bet null mice, which have a defect in NK cell maturation, had uNK cells normal in morphology and number and normally modified spiral arteries. As Eomes null mice are not viable, real-time polymerase chain reaction comparisons between C57Bl/6J (B6) and alymphoid (Rag2(0/0)gammac0/0) mice were used to assess uNK cell expression of T-bet, Eomes, and the target genes IFN-gamma, granzyme A, and perforin. Gestation dated (gd) uterine tissues (mixed cell composition) and 200 morphologically homogeneous, laser-capture, microdissected uNK cells of different maturation stages were used. In uterus, Eomes transcripts greatly outnumbered those of T-bet, whether donors were nonpregnant or pregnant, and increased to gd10. In uNK cells, transcripts for T-bet, Eomes, and IFN-gamma were most abundant in mature stage cells, and transcripts for granzyme A and perforin were lower at this stage than in immature or senescent cells. Thus, Eomes dominance to T-bet discriminates regulation of the uNK cell subset from that observed for peripheral NK cells.


Asunto(s)
Diferenciación Celular/inmunología , Células Asesinas Naturales/inmunología , Proteínas de Dominio T Box/genética , Factores de Transcripción/genética , Útero/inmunología , Animales , Ciclo Estral/genética , Ciclo Estral/inmunología , Ciclo Estral/metabolismo , Femenino , Granzimas , Interferón gamma/genética , Células Asesinas Naturales/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Perforina , Proteínas Citotóxicas Formadoras de Poros , Embarazo , Elementos Reguladores de la Transcripción/genética , Elementos Reguladores de la Transcripción/fisiología , Serina Endopeptidasas/genética , Proteínas de Dominio T Box/inmunología , Factores de Transcripción/inmunología , Activación Transcripcional/genética , Útero/citología
17.
Cancer Cell ; 28(6): 785-799, 2015 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-26678340

RESUMEN

Polymorphonuclear neutrophils (PMNs) are largely considered to foster cancer development despite wielding an arsenal of cytotoxic agents. Using a mouse model of PTEN-deficient uterine cancer, we describe a surprising inhibitory role for PMNs in epithelial carcinogenesis. By inducing tumor cell detachment from the basement membrane, PMNs impeded early-stage tumor growth and retarded malignant progression. Unexpectedly, PMN recruitment and tumor growth control occurred independently of lymphocytes and cellular senescence and instead ensued as part of the tumor's intrinsic inflammatory response to hypoxia. In humans, a PMN gene signature correlated with improved survival in several cancer subtypes, including PTEN-deficient uterine cancer. These findings provide insight into tumor-associated PMNs and reveal a context-specific capacity for PMNs to directly combat tumorigenesis.


Asunto(s)
Carcinoma Endometrioide/prevención & control , Neutrófilos/inmunología , Neoplasias Ováricas/prevención & control , Fagocitosis , Microambiente Tumoral , Útero/inmunología , Animales , Trasplante de Médula Ósea , Carcinoma Endometrioide/enzimología , Carcinoma Endometrioide/genética , Carcinoma Endometrioide/inmunología , Carcinoma Endometrioide/mortalidad , Carcinoma Endometrioide/patología , Adhesión Celular , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular , Quimiotaxis , Biología Computacional , Bases de Datos Genéticas , Femenino , Perfilación de la Expresión Génica , Técnicas de Transferencia de Gen , Humanos , Mediadores de Inflamación/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Estadificación de Neoplasias , Activación Neutrófila , Infiltración Neutrófila , Neutrófilos/metabolismo , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Ovariectomía , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética , Receptores del Factor Estimulante de Colonias/deficiencia , Receptores del Factor Estimulante de Colonias/genética , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/metabolismo , Análisis de Supervivencia , Factores de Tiempo , Carga Tumoral , Útero/enzimología , Útero/patología
18.
Int J Dev Biol ; 58(2-4): 189-98, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25023685

RESUMEN

Understanding the function of T cells at the maternal-fetal interface remains one of the most difficult problems in reproductive immunology. A great deal of work over the last two decades has led to the view that the T cells that populate the decidua have important roles in both normal and pathological pregnancies, but the exact nature of these roles has remained unclear. Indeed, the old assumption that decidual T cells are uniformly threatening to fetal survival because the placenta is fundamentally an 'allograft' has given way to the idea that different T cell subsets contribute in different ways to pregnancy success or failure. Accordingly, some T cells are thought to protect the placenta from immune rejection and facilitate embryo implantation, while others are thought to contribute to pregnancy pathologies such as preeclampsia and spontaneous abortion. Here, we review the current state of information on the behavior of decidual T cells with a focus on both mouse and human studies, and with an emphasis on the many unresolved areas within this overall emerging framework.


Asunto(s)
Decidua/inmunología , Intercambio Materno-Fetal/inmunología , Placenta/inmunología , Linfocitos T/inmunología , Animales , Femenino , Humanos , Ratones , Embarazo
19.
Nat Rev Immunol ; 13(1): 23-33, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23237963

RESUMEN

Work on the mechanisms of fetomaternal tolerance has undergone a renaissance in recent years, and the general outlines of a solution to this long-standing paradox of 'transplantation' immunology have come into view. Here, we discuss several mechanisms, recently described in mice, that either minimize the activation of maternal T cells with fetal or placental specificity, or minimize the possibility that such T cells, if activated, are able to harm the fetus. The T cell response to antigens expressed by the conceptus serves as a paradigm for the study of tissue-specific immune tolerance and is relevant to the pathogenesis of immune-mediated pregnancy complications.


Asunto(s)
Feto/inmunología , Tolerancia Inmunológica , Linfocitos T/inmunología , Animales , Femenino , Histocompatibilidad Materno-Fetal/inmunología , Humanos , Isoantígenos , Activación de Linfocitos , Masculino , Intercambio Materno-Fetal/inmunología , Ratones , Modelos Inmunológicos , Embarazo , Linfocitos T Reguladores/inmunología , Útero/anatomía & histología , Útero/inmunología
20.
PLoS One ; 8(12): e84064, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24391885

RESUMEN

Maternal immune tolerance towards the fetus and placenta is thought to be established in part by pathways that attenuate T cell priming to antigens released from the placenta into maternal blood. These pathways remain largely undefined and their existence, at face value, seems incompatible with a mother's need to maintain a functional immune system during pregnancy. A particular conundrum is evident if we consider that maternal antigen presenting cells, activated in order to prime T cells to pathogen-derived antigens, would also have the capacity to prime T cells to co-ingested placental antigens. Here, we address this paradox using a transgenic system in which placental membranes are tagged with a strong surrogate antigen (ovalbumin). We find that although a remarkably large quantity of acellular ovalbumin-containing placental material is released into maternal blood, splenic CD8 T cells in pregnant mice bearing unmanipulated T cell repertoires are not primed to ovalbumin even if the mice are intravenously injected with adjuvants. This failure was largely independent of regulatory T cells, and instead was linked to the intrinsic characteristics of the released material that rendered it selectively non-immunogenic, potentially by sequestering it from CD8α(+) dendritic cells. The release of ovalbumin-containing placental material into maternal blood thus had no discernable impact on CD8 T cell priming to soluble ovalbumin injected intravenously during pregnancy, nor did it induce long-term tolerance to ovalbumin. Together, these results outline a major pathway governing the maternal immune response to the placenta, and suggest how tolerance to placental antigens can be maintained systemically without being detrimental to host defense.


Asunto(s)
Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/inmunología , Feto/inmunología , Tolerancia Inmunológica/inmunología , Ovalbúmina/inmunología , Placenta/inmunología , Receptores CCR7/fisiología , Linfocitos T Reguladores/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Células Dendríticas/inmunología , Femenino , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA