RESUMO
Neonates are highly susceptible to inflammation and infection. Here, we investigate how late fetal liver (FL) mouse hematopoietic stem and progenitor cells (HSPCs) respond to inflammation, testing the hypothesis that deficits in the engagement of emergency myelopoiesis (EM) pathways limit neutrophil output and contribute to perinatal neutropenia. We show that fetal HSPCs have limited production of myeloid cells at steady state and fail to activate a classical adult-like EM transcriptional program. Moreover, we find that fetal HSPCs can respond to EM-inducing inflammatory stimuli in vitro but are restricted by maternal anti-inflammatory factors, primarily interleukin-10 (IL-10), from activating EM pathways in utero. Accordingly, we demonstrate that the loss of maternal IL-10 restores EM activation in fetal HSPCs but at the cost of fetal demise. These results reveal the evolutionary trade-off inherent in maternal anti-inflammatory responses that maintain pregnancy but render the fetus unresponsive to EM activation signals and susceptible to infection.
Assuntos
Inflamação , Interleucina-10 , Mielopoese , Animais , Camundongos , Gravidez/imunologia , Feto , Hematopoese , Células-Tronco Hematopoéticas/citologia , Inflamação/imunologia , Interleucina-10/imunologia , Animais Recém-Nascidos , FemininoRESUMO
The juxtaposition of two seemingly disparate physiological phenomena within the human body-namely, cancer and pregnancy-may offer profound insights into the intricate interplay between malignancies and the immune system. Recent investigations have unveiled striking similarities between the pivotal processes underpinning fetal implantation and successful gestation and those governing tumor initiation and progression. Notably, a confluence of features has emerged, underscoring parallels between the microenvironment of tumors and the maternal-fetal interface. These shared attributes encompass establishing vascular networks, cellular mobilization, recruitment of auxiliary tissue components to facilitate continued growth, and, most significantly, the orchestration of immune-suppressive mechanisms.Our particular focus herein centers on the phenomenon of immune suppression and its protective utility in both of these contexts. In the context of pregnancy, immune suppression assumes a paramount role in shielding the semi-allogeneic fetus from the potentially hostile immune responses of the maternal host. In stark contrast, in the milieu of cancer, this very same immunological suppression fosters the transformation of the tumor microenvironment into a sanctuary personalized for the neoplastic cells.Thus, the striking parallels between the immunosuppressive strategies deployed during pregnancy and those co-opted by malignancies offer a tantalizing reservoir of insights. These insights promise to inform novel avenues in the realm of cancer immunotherapy. By harnessing our understanding of the immunological events that detrimentally impact fetal development, a knowledge grounded in the context of conditions such as preeclampsia or miscarriage, we may uncover innovative immunotherapeutic strategies to combat cancer.
Assuntos
Neoplasias , Gravidez , Animais , Feminino , Humanos , Gravidez/imunologia , Tolerância Imunológica , Imunoterapia , Neoplasias/imunologia , Complicações Neoplásicas na Gravidez/imunologia , Microambiente Tumoral/imunologiaRESUMO
Novel biomarkers of in utero infections are needed to help guide early therapy. The toll like receptors (TLRs) and retinoic acid-inducible gene 1 (RIG-1) are proteins involved in the initial reaction of the innate immune system to infectious diseases. This study tested the hypothesis that a panel of TLRs and RIG-1 in the placenta could serve as an early biomarker of in utero infections. The TLRs and RIG-1 expression as determined by immunohistochemistry was scored in 10 control placentas (normal delivery or neonatal damage from known non-infectious cause), 8 placentas from documented in utero bacterial infection, and 7 placentas from documented in utero viral infections blinded to the clinical information. The non-infected placentas showed the following profile: no expression (TLR1, TLR3, TLR4, TLR7, TLR8), moderate expression (TLR2), and strong expression (RIG-1). The bacterial and viral infection cases shared the following profile: no to mild expression (TLR 2, TLR7, and RIG1), moderate expression (TLR4), and strong expression (TLR1, TLR3, and TLR8). The histologic findings in the chorionic villi were equivalent in the infected cases and controls, underscoring the need for molecular testing by the surgical pathologist when in utero infection is suspected. The results suggest that a panel of TLRs/RIG-1 analyses can allow the pathologist and/or clinician to diagnose in utero infections soon after birth. Also, treatments to antagonize the effects of TLR1, 3, and 8 may help abrogate in utero neonatal damage.
Assuntos
Placenta , Complicações Infecciosas na Gravidez , Feminino , Humanos , Recém-Nascido , Gravidez/imunologia , Placenta/imunologia , Placenta/metabolismo , Receptor 1 Toll-Like/genética , Receptor 1 Toll-Like/metabolismo , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/metabolismo , Receptor 4 Toll-Like , Receptor 7 Toll-Like , Receptor 8 Toll-Like/genética , Receptor 8 Toll-Like/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/metabolismo , Complicações Infecciosas na Gravidez/genética , Complicações Infecciosas na Gravidez/metabolismoRESUMO
OBJECTIVE: To comprehensively characterize monocyte and neutrophil responses to E. coli and its product [lipopolysaccharide (LPS) or endotoxin] in vitro during pregnancy. MATERIAL OR SUBJECTS: Peripheral blood was collected from pregnant women during the third trimester (n = 20) and from non-pregnant women (n = 20). METHODS: The number, phagocytic activity, and reactive oxygen species (ROS) production of peripheral monocytes and neutrophils were investigated using flow cytometry. The phenotypes of peripheral monocytes and neutrophils after acute or chronic LPS stimulation were also determined using flow cytometry. Cytokine profiles were quantified for LPS-stimulated peripheral blood mononuclear cells (PBMCs) and a whole blood TruCulture® system using a multiplex immunoassay. RESULTS: Increased number, phagocytic activity, and ROS production capacity of monocytes and neutrophils were found in pregnant compared to non-pregnant women. Additionally, specific subsets of pro-inflammatory monocytes (IL-6+CD14+ or MIP-1α+CD14+ cells) and neutrophils (IL-1ß+CD15+ or MIP-1ß+CD15+ cells) were increased in pregnant women in response to acute LPS stimulation. Moreover, distinct subsets of intermediate-activated monocytes expressing CD142, IL-6, and IL-1RA were increased in pregnant women upon chronic LPS stimulation. Last, pregnant women displayed a different cytokine profile than non-pregnant women in LPS-stimulated PBMCs and in whole blood. CONCLUSIONS: Pregnancy tailors the immune responses of circulating monocytes and neutrophils to endotoxin, a Gram-negative bacterial product.
Assuntos
Endotoxinas , Monócitos , Neutrófilos , Gravidez , Endotoxinas/farmacologia , Escherichia coli , Feminino , Humanos , Interleucina-6 , Lipopolissacarídeos/farmacologia , Monócitos/imunologia , Monócitos/fisiologia , Neutrófilos/imunologia , Neutrófilos/fisiologia , Gravidez/sangue , Gravidez/imunologia , Gravidez/fisiologia , Espécies Reativas de OxigênioRESUMO
Discrimination of self from non-self is fundamental to a wide range of immunological processes1. During pregnancy, the mother does not recognize the placenta as immunologically foreign because antigens expressed by trophoblasts, the placental cells that interface with the maternal immune system, do not activate maternal T cells2. Currently, these activation defects are thought to reflect suppression by regulatory T cells3. By contrast, mechanisms of B cell tolerance to trophoblast antigens have not been identified. Here we provide evidence that glycan-mediated B cell suppression has a key role in establishing fetomaternal tolerance in mice. B cells specific for a model trophoblast antigen are strongly suppressed through CD22-LYN inhibitory signalling, which in turn implicates the sialylated glycans of the antigen as key suppressive determinants. Moreover, B cells mediate the MHC-class-II-restricted presentation of antigens to CD4+ T cells, which leads to T cell suppression, and trophoblast-derived sialoglycoproteins are released into the maternal circulation during pregnancy in mice and humans. How protein glycosylation promotes non-immunogenic placental self-recognition may have relevance to immune-mediated pregnancy complications and to tumour immune evasion. We also anticipate that our findings will bolster efforts to harness glycan biology to control antigen-specific immune responses in autoimmune disease.
Assuntos
Antígenos , Placenta , Trofoblastos , Animais , Doenças Autoimunes , Linfócitos B , Feminino , Tolerância Imunológica , Camundongos , Placenta/imunologia , Polissacarídeos/metabolismo , Gravidez/imunologiaRESUMO
As a non-classic major histocompatibility complex (MHC) class I molecule, human leukocyte antigen G (HLA-G) is expressed in fetal-maternal interface and immunoprivileged site only in healthy condition, and in pathological conditions such as cancer, it can be de novo expressed. It is now widely accepted that HLA-G is a key molecule in the process of immune escape of cancer cells, which is ubiquitously expressed in the tumor environment. This raises the possibility that it may play an adverse role in tumor immunity. The expression level of HLA-G has been demonstrated to be highly correlated with clinical parameters in many tumors, and its potential significance in the diagnosis and prognosis of cancer has been postulated. However, because HLA-G itself has up to seven different subtypes, and for some subtypes, detected antibodies are few or absent, it is hard to evaluate the actual expression of HLA-G in tumors. In the present work, we described (a) the structure and three main forms of HLA-G, (b) summarized the mechanism of HLA-G in the immune escape of tumor cells, (c) discussed the potential role of HLA-G as a tumor marker, and reviewed (d) the methods for detecting and quantifying HLA-G.
Assuntos
Biomarcadores Tumorais/metabolismo , Vesículas Extracelulares/metabolismo , Antígenos HLA-G/metabolismo , Neoplasias/imunologia , Gravidez/imunologia , Animais , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Privilégio Imunológico , Tolerância Imunológica , Neoplasias/diagnóstico , Circulação Placentária , Prognóstico , Evasão TumoralRESUMO
Pregnancy implies delicate immunological balance between two individuals, with constant changes and adaptions in response to maternal capacity and fetal demands. We performed cytokine profiling of 1149 longitudinal serum samples from 707 pregnant women to map immunological changes from first trimester to term and beyond. The serum levels of 22 cytokines and C-reactive protein (CRP) followed diverse but characteristic trajectories throughout pregnancy, consistent with staged immunological adaptions. Eotaxin showed a particularly robust decrease throughout pregnancy. A strong surge in cytokine levels developed when pregnancies progressed beyond term and the increase was amplified as labor approached. Maternal obesity, smoking and pregnancies with large fetuses showed sustained increase in distinct cytokines throughout pregnancy. Multiparous women had increased cytokine levels in the first trimester compared to nulliparous women with higher cytokine levels in the third trimester. Fetal sex affected first trimester cytokine levels with increased levels in pregnancies with a female fetus. These findings unravel important immunological dynamics of pregnancy, demonstrate how both maternal and fetal factors influence maternal systemic cytokines, and serve as a comprehensive reference for cytokine profiles in normal pregnancies.
Assuntos
Citocinas/sangue , Gravidez/imunologia , Feminino , Humanos , Primeiro Trimestre da Gravidez/imunologia , Segundo Trimestre da Gravidez/imunologia , Terceiro Trimestre da Gravidez/imunologiaRESUMO
The immune tolerance microenvironment is crucial for the establishment and maintenance of pregnancy at the maternal-fetal interface. The maternal-fetal interface is a complex system containing various cells, including lymphocytes, decidual stromal cells, and trophoblasts. Macrophages are the second-largest leukocytes at the maternal-fetal interface, which has been demonstrated to play essential roles in remodeling spiral arteries, maintaining maternal-fetal immune tolerance, and regulating trophoblast's biological behaviors. Many researchers, including us, have conducted a series of studies on the crosstalk between macrophages and trophoblasts at the maternal-fetal interface: on the one hand, macrophages can affect the invasion and migration of trophoblasts; on the other hand, trophoblasts can regulate macrophage polarization and influence the state of the maternal-fetal immune microenvironment. In this review, we systemically introduce the functions of macrophages and trophoblasts and the cell-cell interaction between them for the establishment and maintenance of pregnancy. Advances in this area will further accelerate the basic research and clinical translation of reproductive medicine.
Assuntos
Feto/imunologia , Tolerância Imunológica/imunologia , Macrófagos/fisiologia , Gravidez/imunologia , Trofoblastos/fisiologia , Animais , Comunicação Celular , Movimento Celular , Microambiente Celular , Citocinas/fisiologia , Vesículas Extracelulares/fisiologia , Feminino , Humanos , Ativação de Macrófagos , Macrófagos/classificação , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Trofoblastos/classificação , Trofoblastos/citologia , Trofoblastos/imunologiaRESUMO
Indoleamine 2, 3-dioxygenase (IDO), a tryptophan-catabolizing enzyme, is essential in physiological immunoregulation. The present research was conducted to elucidate the expression and roles of IDO in decidual macrophages (dMφ) during early pregnancy. Here, we observed a remarkable decrease of IDO+ dMφ from patients with unexplained recurrent spontaneous abortion (URSA). IDO+ dMφ displayed M2 phenotype with higher CD206, CD209 and CD163, and lower CD86. Interestingly, treatment with 1-methyl-d-tryptophan (1-MT, an IDO pathway inhibitor) led to the M1 bias of dMφ. Further analysis of the cytokine array and the qPCR showed decreased levels of trophoblast proliferation or invasion-related molecules (e.g., CXCL12 and BMP2) in 1-MT-treated dMφ. The data of co-culture system showed that 1-MT-pretreated dMφ decreased the proliferation and the expression of Ki-67 and Bcl-2, and increased cell apoptosis of HTR-8/Snveo cells. Additionally, the expression of IDO in U937 cells was up-regulated by decidual stromal cells (DSC) and HTR-8/Snveo cells in vitro, as well as estradiol and medroxyprogesterone. These data suggest that endocrine environment, DSC and trophoblasts should contribute to the high level of IDO in dMφ, and IDO+ dMφ with M2 dominant phenotype promote the survival of trophoblasts during early pregnancy. The abnormal lower level of IDO should trigger the dysfunction of dMφ, further suppress the survival of trophoblasts and increase the risk of miscarriage.
Assuntos
Aborto Espontâneo/imunologia , Decídua/imunologia , Macrófagos/imunologia , Gravidez/imunologia , Células Th2/imunologia , Trofoblastos/fisiologia , Apoptose , Diferenciação Celular , Proliferação de Células , Técnicas de Cocultura , Citocinas/metabolismo , Feminino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Recidiva , Células U937RESUMO
Las mujeres embarazadas o en período de lactancia han sido excluidas de los ensayos clínicos sobre vacunas contra SARS-CoV-2, evitando así la obtención de datos sólidos que permitan determinar la seguridad e inmunogenicidad de las vacunas en esta población, a la vez que se han asociado peores resultados maternos fetales. La evidencia acerca de la seguridad e inmunogenicidad en esta población es limitada, en base a estudios observacionales, con pocos casos y en mujeres vacunadas con plataformas ARNm, en las cuales no se ha descrito por ahora una mayor asociación a eventos adversos relacionados a vacunas, como tampoco, variaciones significativas en la respuesta inmunológica en comparación a la población no embarazada. También existen datos que documentan la adquisición de anticuerpos transplacentarios, considerándose de bajo riesgo la posibilidad de transmisión vertical. Se hacen necesarios ensayos clínicos que permitan precisar recomendaciones basadas en evidencia para esta población, en un contexto de utilización de emergencia de vacunas contra SARS-CoV-2. (AU)
Pregnant or breastfeeding women have been excluded from clinical trials on vaccines against SARSCoV-2, thus avoiding obtaining solid data to determine the safety and immunogenicity of vaccines in this population, as well as being associated worse maternal-fetal outcomes. The evidence about safety and immunogenicity in this population is limited, based on observational studies, with few cases and in women vaccinated with mRNA platforms, in which a greater association to adverse events related to vaccines has not been described or significant variations in the immune response compared to the non-pregnant population. There are also data that document the acquisition of transplacental antibodies, considering the possibility of vertical transmission as low risk. Clinical trials are necessary to evidence-based recommendations for this population, in a context of emergency use of vaccines against SARSCoV-2. (AU)
Assuntos
Humanos , Feminino , Gravidez , Aleitamento Materno , Gravidez/imunologia , Vacinas contra COVID-19/efeitos adversos , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , COVID-19/prevenção & controle , COVID-19/transmissãoRESUMO
Fetal antigen-specific tolerance is important for maintaining allogeneic pregnancies. Maternal conventional T cells recognize fetal antigens; however, regulatory T (Treg) cells suppress immune reactions against the fetus. Fetal antigen-specific Treg cells are induced in the decidua upon contact with antigen-presenting cells and extravillous trophoblasts (EVTs). Functional alteration of cytotoxic T cells (CTLs) in the decidua also contributes to maintaining the pregnancy. Reduced, dysfunctional, and imbalanced Treg cell distribution likely contributes to the pathogenesis of pregnancy complications, such as miscarriage and preeclampsia. Recent studies have revealed differences in Treg cell characteristics during preeclampsia and miscarriage. Treg cell reduction in the decidua is likely associated with miscarriage. Insufficient expansion of fetal antigen-specific Treg cells in the decidua probably plays a role in preeclampsia pathogenesis. In addition, the balance between Treg cell-mediated tolerance and functional alteration of CTLs is important. Further investigations of functional molecules in Treg cells will contribute to the development of immunotherapy for pregnancy complications.
Assuntos
Decídua/imunologia , Imunoterapia/métodos , Complicações na Gravidez/imunologia , Gravidez/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/imunologia , Animais , Citotoxicidade Imunológica , Feminino , Histocompatibilidade Materno-Fetal , Humanos , Tolerância Imunológica , Complicações na Gravidez/terapiaRESUMO
Successful pregnancy relies on maternal immunologic tolerance mechanisms limit maladaptive immune responses against the semi-allogeneic fetus and placenta and support fetal growth. Preeclampsia is a common disorder of pregnancy that affects 4-10% of pregnancies and is a leading cause of maternal and neonatal morbidity and mortality. Preeclampsia clinically manifests as maternal hypertension, proteinuria, and progressive multi-organ injury likely triggered by hypoxic injury to the placenta, resulting in local and systemic anti-angiogenic and inflammatory factor production. Despite the steady rising rates of preeclampsia in the United States, effective treatment options are limited to delivery, which improves maternal status often at the cost of prematurity in the newborn. Preeclampsia also increases the lifelong risk of cardiovascular disease for both mother and infant. Thus, identifying new therapeutic targets is a high priority area to improve maternal, fetal, and infant health outcomes. Immune abnormalities in the placenta and in the maternal circulation have been reported to precede the clinical onset of disease. In particular, excessive systemic and placental complement activation and impaired adaptive T cell tolerance with Th1/Th2/Th17/Treg imbalance has been reported in humans and in animal models of preeclampsia. In this review, we focus on the evidence for the immune origins of preeclampsia, discuss the promise of immune modulating therapy for prevention or treatment, and highlight key areas for future research.
Assuntos
Imunoterapia/tendências , Placenta/imunologia , Pré-Eclâmpsia/imunologia , Gravidez/imunologia , Linfócitos T/imunologia , Imunidade Adaptativa , Animais , Ativação do Complemento , Modelos Animais de Doenças , Feminino , Humanos , Tolerância Imunológica , Imunomodulação , Placentação , Pré-Eclâmpsia/terapiaRESUMO
OBJECTIVES: Uncontrolled TH17 differentiation has been suggested to play a role in the pathogenesis of pregnancy loss. We recently showed that menstrual blood stromal/stem cells (MenSCs) alter functional features of natural killer cells. Here, we hypothesized that MenSCs could modulate differentiation of TH17 cells. METHOD: MenSCs were collected from 18 apparently healthy women and characterized. Bone marrow mesenchymal stem cells (BMSCs) served as a control. TH17 polarization and proliferation of purified T CD4+ cells were assessed by flow cytometry in a well-defined co-culture system containing T CD4+ cells and MenSCs or BMSCs. Indoleamine 2,3-Dioxygenase (IDO) activity was evaluated in MenSC and BMSC culture supernatants by a colorimetric assay. The impact of MenSCs on expression of transcription factors, RORC, T-bet, Gata3, NRP-1 and Helios were studied by qPCR. RESULTS: MenSCs significantly inhibited TH17 differentiation (pâ¯=â¯0.0383) and percentage of the cells co-expressing IL-17 and IFN-γ (pâ¯=â¯0.0023). PGE2 blockade significantly reduced percentage and proliferation of T CD4+IL-17+ (pâ¯=â¯0.003, pâ¯=â¯0.0018), T CD4+ IFN-γ+ (pâ¯=â¯0.002, pâ¯=â¯0.0022) and T CD4+IL-17+ IFN-γ+ (pâ¯=â¯0.004, pâ¯=â¯0.02) cells. MenSCs produced a considerable activity of IDO (pâ¯=â¯0.0002), induced a significant rise in the Treg frequency (pâ¯=â¯0.0091) and a sharp increase in TH17/Tregs ratio (pâ¯=â¯0.0022). MenSCs increased expression of NRP1 (pâ¯=â¯0.001), while downregulated expression of RORC in T cells (pâ¯=â¯0.001). CONCLUSION: Our results suggest a supportive role for MenSCs in establishing a pregnancy-friendly microenvironment in the uterus and put forth the idea that inherent abnormalities of MenSCs may be a basis for dysregulated endometrial immune network leading to pregnancy loss.
Assuntos
Células-Tronco Adultas/imunologia , Menstruação/sangue , Gravidez/imunologia , Células Estromais/imunologia , Células Th17/imunologia , Adulto , Células-Tronco Adultas/enzimologia , Diferenciação Celular/imunologia , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Endométrio/citologia , Endométrio/imunologia , Feminino , Voluntários Saudáveis , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/análise , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Células Estromais/enzimologiaRESUMO
Predicted Indirectly ReCognizable Human Leukocyte Antigen (HLA) Epitopes (PIRCHE) are known to be a significant risk factor for the development of donor HLA-specific antibodies after organ transplantation. Most previous studies on PIRCHE limited their analyses on the presentation of the HLA-DRB1 locus, although HLA-DRB3/4/5, -DQ, and -DP are also known for presenting allopeptides to CD4+ T cells. In this study, we analyzed the impact of predicted allopeptides presented by these additional loci on the incidence of HLA-specific antibodies after an immunization event. We considered pregnancy as a model system of an HLA immunization and observed child-specific HLA antibody (CSA) development of 231 mothers during pregnancy by samples being taken at delivery. Our data confirm that PIRCHE presented by HLA-DRB1 along with HLA-DRB3/4/5, -DQ, and -DP are significant predictors for the development of CSA. Although there was limited peptidome overlap observed within the mothers' presenting HLA proteins, combining multiple presenting loci in a single predictor improved the model only marginally. Prediction performance of PIRCHE further improved when normalizing scores by the respective presenters' binding promiscuity. Immunogenicity analysis of specific allopeptides could not identify significant drivers of an immune response in this small cohort, suggesting confirmatory studies.
Assuntos
Antígenos HLA-DP/metabolismo , Antígenos HLA-DQ/metabolismo , Cadeias HLA-DRB1/metabolismo , Cadeias HLA-DRB4/metabolismo , Gravidez/imunologia , Adulto , Apresentação de Antígeno , Epitopos/imunologia , Epitopos/metabolismo , Feminino , Teste de Histocompatibilidade , Humanos , Isoanticorpos/metabolismo , Isoantígenos/imunologia , Isoantígenos/metabolismo , Masculino , Peptídeos/imunologia , Peptídeos/metabolismoRESUMO
Increased exposure to household air pollution and ambient air pollution has become one of the world's major environmental health threats. In developing and developed countries, environmental cigarette smoke (CS) exposure is one of the main sources of household air pollution (HAP). Moreover, results from different epidemiological and experimental studies indicate that there is a strong association between HAP, specifically CS exposure, and the development of allergic diseases that often persists into later life. Here, we investigated the impact of prenatal and postnatal CS exposure on offspring susceptibility to the development of allergic airway responses by using a preclinical mouse model. Pregnant BALB/c mice were exposed to either CS or air during pregnancy and lactation and in order to induce allergic asthma the offspring were sensitized and challenged with house dust mite (HDM). Decreased lung function parameters, like dynamic compliance and pleural pressure, were observed in PBS-treated offspring born to CS-exposed mothers compared to offspring from air-exposed mothers. Maternal CS exposure significantly increased the HDM-induced airway eosinophilia and neutrophilia in the offspring. Prenatal and postnatal CS exposure increased the frequency of Th2 cells in the lungs of HDM-treated offspring compared to offspring born to air-exposed mothers. Offspring born to CS-exposed mothers showed increased levels of IL-4, IL-5 and IL-13 in bronchoalveolar lavage fluid compared to offspring from air-exposed mothers. Ex-vivo restimulation of lung cells isolated from HDM-treated offspring born to CS-exposed mothers also resulted in increased IL-4 production. Finally, serum immunoglobulins levels of HDM-specific IgE and HDM-specific IgG1 were significantly increased upon a HDM challenge in offspring born to CS-exposed mothers compared to offspring from air-exposed mothers. In summary, our results reveal a biological plausibility for the epidemiological studies indicating that prenatal and postnatal CS exposure increases the susceptibility of offspring to allergic immune responses.
Assuntos
Fumar Cigarros/efeitos adversos , Hipersensibilidade/imunologia , Pulmão/imunologia , Gravidez/imunologia , Efeitos Tardios da Exposição Pré-Natal/imunologia , Hipersensibilidade Respiratória/imunologia , Células Th2/imunologia , Animais , Antígenos de Dermatophagoides/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Feminino , Humanos , Masculino , Exposição Materna/efeitos adversos , Camundongos , Camundongos Endogâmicos BALB C , Efeitos Tardios da Exposição Pré-Natal/etiologia , Pyroglyphidae/imunologia , RiscoRESUMO
Hofbauer cells (HBCs) are a population of macrophages found in high abundance within the stroma of the first-trimester human placenta. HBCs are the only fetal immune cell population within the stroma of healthy placenta. However, the functional properties of these cells are poorly described. Aligning with their predicted origin via primitive hematopoiesis, we find that HBCs are transcriptionally similar to yolk sac macrophages. Phenotypically, HBCs can be identified as HLA-DR-FOLR2+ macrophages. We identify a number of factors that HBCs secrete (including OPN and MMP-9) that could affect placental angiogenesis and remodeling. We determine that HBCs have the capacity to play a defensive role, where they are responsive to Toll-like receptor stimulation and are microbicidal. Finally, we also identify a population of placenta-associated maternal macrophages (PAMM1a) that adhere to the placental surface and express factors, such as fibronectin, that may aid in repair.
Assuntos
Macrófagos/imunologia , Placenta/imunologia , Primeiro Trimestre da Gravidez/imunologia , Gravidez/imunologia , Adulto , Feminino , Receptor 2 de Folato/imunologia , Antígenos HLA-DR/imunologia , Humanos , Metaloproteinase 9 da Matriz/imunologiaRESUMO
Circulating TFH (cTFH ) cells express CXCR5, PD-1, and, when activated, ICOS, and release IL-21. According to the production of IFN-γ, IL-4, and IL-17 and expression of FoxP3, these cells are also classified as cTFH 1, cTFH 2, cTFH 17, and cTFR cells, respectively. This CD4+ T-cell subset is pivotal to efficient humoral immunity, and pregnancy appears to favor IgG production. Here, not only pregnancy amplified the in vivo production of anti-HBsAg IgG in HBV immunized women, but the frequency of cTFH cells was directly correlated with estradiol levels. In vitro, pregnancy-related dose of 17-ß-estradiol (E2) directly increased the percentage of different cTFH subsets. While E2 and progesterone (P4) increased the proportion of differentiated TFH cells derived from naïve CD4+ T-cells, only E2 amplified the release of IL-21 in those cell cultures. In addition, E2 and P4 increased the proportion of memory B cells and plasma cells, respectively. In SEB-activated B/TFH cell co-cultures, E2, in the presence of P4, increased the production of total IgG. Finally, among the hormones, P4 was stronger in upregulating the percentage of IL-10+ TFR cells. Collectively, our findings suggested that E2 and P4 cooperate in the humoral immune response by favoring the expansion of different cTFH and B cell subsets.
Assuntos
Linfócitos B/imunologia , Estradiol/sangue , Estradiol/imunologia , Imunidade Humoral , Gravidez/sangue , Gravidez/imunologia , Progesterona/sangue , Progesterona/imunologia , Células T Auxiliares Foliculares/imunologia , Adulto , Subpopulações de Linfócitos B/efeitos dos fármacos , Subpopulações de Linfócitos B/imunologia , Diferenciação Celular , Citocinas/metabolismo , Estradiol/farmacologia , Feminino , Vacinas contra Hepatite B/imunologia , Humanos , Imunoglobulina G/biossíntese , Técnicas In Vitro , Interleucinas/biossíntese , Progesterona/farmacologia , Células T Auxiliares Foliculares/classificação , Células T Auxiliares Foliculares/citologia , Adulto JovemRESUMO
HLA-G immune modulatory genes and molecules are presently being studied by a widespread number of research groups. In the present study, we do not aim to be exhaustive since the number of manuscripts published every year is overwhelming. Instead, our aim is pointing out facts about HLA-G function, polymorphism and pathology that have been confirmed by several different researchers, together with exposing aspects that may have been overlooked or not sufficiently remarked in this productive field of study. On the other hand, we question whether performing mainly studies on HLA-G and disease associations is going to give a clear answer in the future, since 40 years of study of classical HLA molecules association with disease has still given no definite answer on this issue.
Assuntos
Antígenos HLA-G/imunologia , Regiões 3' não Traduzidas , Alelos , Animais , Doenças Autoimunes/imunologia , Feminino , Genes MHC Classe I , Antígenos HLA-G/genética , Humanos , Masculino , Proteínas de Membrana/imunologia , Neoplasias/imunologia , Peptídeos/imunologia , Polimorfismo Genético , Gravidez/imunologia , Primatas/genética , Primatas/imunologia , Isoformas de Proteínas/imunologia , Solubilidade , Imunologia de Transplantes , Viroses/imunologiaRESUMO
Implantation of the embryo is a rate-limiting step for a successful pregnancy, and it requires an intricate crosstalk between the embryo and the endometrium. Extracellular vesicles (EVs) are membrane-enclosed, nano-sized structures produced by cells to mediate cell to cell communication and modulate a diverse set of biological processes. Herein, we review the involvement of EVs in the process of embryo implantation and endometrial diseases. EVs have been isolated from uterine fluid, cultured endometrial epithelial/stromal cells and trophectodermal cells. The endometrial epithelial and stromal/decidual cell-derived EVs and its cargo are internalized bythe trophoblast cells, and they regulate a diverse set of genes involved in adhesion, invasion and migration. Conversely, the embryo-derived EVs and its cargo are internalized by epithelial and immune cells of the endometrium for biosensing and immunomodulation required for successful implantation. EVs have also been shown to play a role in infertility, recurrent implantation failure, endometriosis, endometritis and endometrial cancer. Further research should set a stage for EVs as non-invasive "liquid biopsy" tools for assessment of endometrial health.
Assuntos
Endométrio/patologia , Vesículas Extracelulares/imunologia , Placenta/imunologia , Complicações na Gravidez/imunologia , Gravidez/imunologia , Trofoblastos/imunologia , Animais , Implantação do Embrião , Embrião de Mamíferos , Feminino , Humanos , Imunomodulação , Nanoestruturas , Complicações na Gravidez/patologiaRESUMO
Cells produce cytoplasmic vesicles to facilitate the processing and transport of RNAs, proteins, and other signaling molecules among intracellular organelles. Moreover, most cells release a range of extracellular vesicles (EVs) that mediate intercellular communication in both physiological and pathological settings. In addition to a better understanding of their biological functions, the diagnostic and therapeutic prospects of EVs, particularly the nano-sized small EVs (sEVs, exosomes), are currently being rigorously pursued. While EVs and viruses such as retroviruses might have evolved independently, they share a number of similar characteristics, including biogenesis pathways, size distribution, cargo, and cell-targeting mechanisms. The interplay of EVs with viruses has profound effects on viral replication and infectivity. Our research indicates that sEVs, produced by primary human trophoblasts, can endow other non-placental cell types with antiviral response. Better insights into the interaction of EVs with viruses may illuminate new ways to attenuate viral infections during pregnancy, and perhaps develop new antiviral therapeutics to protect the feto-placental unit during critical times of human development.