Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.397
1.
Wei Sheng Yan Jiu ; 53(3): 382-388, 2024 May.
Article Zh | MEDLINE | ID: mdl-38839580

OBJECTIVE: To investigate the situation of women's dietary quality during pregnancy and explore the correlations between maternal dietary index and fetal immune function. METHODS: From September 2010 to February 2011, pregnant women who had routine physical examination in Yuexiu District and Baiyun District Maternal and Child Health Hospital of Guangzhou were recruited as study objects to use 3-day 24-hour dietary review to investigate diet during pregnancy, and general demographic information of pregnant women was collected through questionnaire, and the neonatal umbilical cord blood was collected during delivery. Laboratory detection of immunological indicators included IgG, IgA, IgM, IFN-γ and IL-6. The quality of diet during pregnancy was evaluated by diet quality index for pregnancy(DQI-P), dietary balance index for pregnancy(DBI-P) and alternate Medierranean diet score(aMED). Spearman correlation analysis and multiple linear regression analysis were used to explore the correlations between dietary quality during pregnancy and fetal immune function. RESULTS: The mean score of total DQI-P score of the study subjects was 55.8±10.0, and the mean score of overall food diversity and protein food source diversity was as high as 12.0±2.4 and 4.8±0.7. The mean score of nutrient energy ratio and fatty acid energy ratio was only 0.3±1.0 and 0.4±1.0, indicating that the population had good dietary diversity during pregnancy, but the dietary adequacy, suitability and balance were poor. The total score of DBI-P score was-19.2±9.4. The positive end score was 4.6±2.9, only 7.2% of the subjects had a high degree of dietary intake during pregnancy. The negative end score was 23.9±7.9, indicating the status of moderate dietary intake. Dietary quality was 28.5±7.1. Only 0.6% of the study population had a balanced dietary situation, and more than 67.9% of pregnant women had high intake imbalance. The mean total score of aMED score was 4.9±1.3, and the proportion of the food intake of beans and nuts was less than the median population was 62.5% and 79.1%, respectively, indicating that the food intake of beans and nuts was insufficient in this population. After adjusting for confounding factors such as maternal age, parity, parity, prepregnancy BMI, weight gain during pregnancy, and mode of delivery, multiple linear regression analysis showed DQI-P during pregnancy and negatively with IL-6(ß=0.143, ß=-0.155, P<0.05). DBI-P was negatively associated with IL-6(ß=-0.177, P<0.01) and aMED and IFN-γ(ß=-0.161, P<0.01). CONCLUSION: The dietary quality of women in late pregnancy in Guangzhou is low, the dietary structure is unbalanced. Higher dietary quality during pregnancy can promote the development of fetal immune system and improve fetal immune function.


Diet , Humans , Female , Pregnancy , China , Adult , Fetus/immunology , Surveys and Questionnaires , Fetal Blood/immunology , Fetal Blood/chemistry , Maternal Nutritional Physiological Phenomena , Diet Surveys , Interleukin-6/blood
2.
Nat Commun ; 15(1): 4711, 2024 Jun 03.
Article En | MEDLINE | ID: mdl-38830841

The fetal development of organs and functions is vulnerable to perturbation by maternal inflammation which may increase susceptibility to disorders after birth. Because it is not well understood how the placenta and fetus respond to acute lung- inflammation, we characterize the response to maternal pulmonary lipopolysaccharide exposure across 24 h in maternal and fetal organs using multi-omics, imaging and integrative analyses. Unlike maternal organs, which mount strong inflammatory immune responses, the placenta upregulates immuno-modulatory genes, in particular the IL-6 signaling suppressor Socs3. Similarly, we observe no immune response in the fetal liver, which instead displays metabolic changes, including increases in lipids containing docosahexaenoic acid, crucial for fetal brain development. The maternal liver and plasma display similar metabolic alterations, potentially increasing bioavailability of docosahexaenoic acid for the mother and fetus. Thus, our integrated temporal analysis shows that systemic inflammation in the mother leads to a metabolic perturbation in the fetus.


Fetus , Lipopolysaccharides , Liver , Lung , Placenta , Female , Pregnancy , Placenta/metabolism , Placenta/immunology , Animals , Fetus/immunology , Fetus/metabolism , Lung/immunology , Lung/metabolism , Liver/metabolism , Liver/immunology , Docosahexaenoic Acids/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Mice , Inflammation/immunology , Inflammation/metabolism , Mice, Inbred C57BL , Adaptation, Physiological/immunology , Fetal Development/immunology , Maternal-Fetal Exchange/immunology , Interleukin-6/metabolism , Interleukin-6/immunology
3.
Article En | MEDLINE | ID: mdl-38782369

Pregnancy is a remarkable event where the semi-allogeneic fetus develops in the mother's uterus, despite genetic and immunological differences. The antigen handling and processing at the maternal-fetal interface during pregnancy appear to be crucial for the adaptation of the maternal immune system and for tolerance to the developing fetus and placenta. Maternal antigen-presenting cells (APCs), such as macrophages (Mφs) and dendritic cells (DCs), are present at the maternal-fetal interface throughout pregnancy and are believed to play a crucial role in this process. Despite numerous studies focusing on the significance of Mφs, there is limited knowledge regarding the contribution of DCs in fetomaternal tolerance during pregnancy, making it a relatively new and growing field of research. This review focuses on how the behavior of DCs at the maternal-fetal interface adapts to pregnancy's unique demands. Moreover, it discusses how DCs interact with other cells in the decidual leukocyte network to regulate uterine and placental homeostasis and the local maternal immune responses to the fetus. The review particularly examines the different cell lineages of DCs with specific surface markers, which have not been critically reviewed in previous publications. Additionally, it emphasizes the impact that even minor disruptions in DC functions can have on pregnancy-related complications and proposes further research into the potential therapeutic benefits of targeting DCs to manage these complications.


Dendritic Cells , Immune Tolerance , Maternal-Fetal Exchange , Placenta , Humans , Pregnancy , Dendritic Cells/immunology , Female , Maternal-Fetal Exchange/immunology , Placenta/immunology , Fetus/immunology , Animals , Macrophages/immunology , Pregnancy Complications/immunology
4.
Int Immunopharmacol ; 133: 112070, 2024 May 30.
Article En | MEDLINE | ID: mdl-38640716

Skin, the largest organ of body, is a highly immunogenic tissue with a diverse collection of immune cells. Highly polymorphic human leukocyte antigen (HLA) molecules have a central role in coordinating immune responses as recognition molecules. Nevertheless, HLA gene expression patterns among diverse cell types within a specific organ, like the skin, have yet to be thoroughly investigated, with stromal cells attracting much less attention than immune cells. To illustrate HLA expression profiles across different cell types in the skin, we performed single-cell RNA sequencing (scRNA-seq) analyses on skin datasets, covering adult and fetal skin, and hair follicles as the skin appendages. We revealed the variation in HLA expression between different skin populations by examining normal adult skin datasets. Moreover, we evaluated the potential immunogenicity of multiple skin populations based on the expression of classical HLA class I genes, which were well represented in all cell types. Furthermore, we generated scRNA-seq data of developing skin from fetuses of 15 post conception weeks (PCW), 17 PCW, and 22 PCW, delineating the dynamic expression of HLA genes with cell type-dependent variation among various cell types during development. Notably, the pseudotime trajectory analysis unraveled the significant variance in HLA genes during the evolution of vascular endothelial cells. Moreover, we uncovered the immune-privileged properties of hair follicles at single-cell resolution. Our study presents a comprehensive single-cell transcriptomic landscape of HLA genes in the skin, which provides new insights into variation in HLA molecules and offers a clue for allogeneic skin transplantation.


Gene Expression Profiling , HLA Antigens , Single-Cell Analysis , Skin , Transcriptome , Humans , Skin/immunology , Skin/metabolism , HLA Antigens/genetics , HLA Antigens/immunology , Hair Follicle/immunology , Hair Follicle/metabolism , Fetus/immunology , Adult , Immune Privilege
5.
Science ; 381(6664): 1286, 2023 09 22.
Article En | MEDLINE | ID: mdl-37733842

Shifting pools of antigen can influence pregnancy-induced immune tolerance.


Chimerism , Fetus , Immune Tolerance , Maternal-Fetal Exchange , Child , Female , Humans , Pregnancy , Maternal-Fetal Exchange/immunology , Antigens/immunology , Fetus/cytology , Fetus/immunology
6.
Science ; 381(6664): 1324-1330, 2023 09 22.
Article En | MEDLINE | ID: mdl-37733857

Pregnancy confers partner-specific protection against complications in future pregnancy that parallel persistence of fetal microchimeric cells (FMcs) in mothers after parturition. We show that preexisting FMcs become displaced by new FMcs during pregnancy and that FMc tonic stimulation is essential for expansion of protective fetal-specific forkhead box P3 (FOXP3)-positive regulatory T cells (Treg cells). Maternal microchimeric cells and accumulation of Treg cells with noninherited maternal antigen (NIMA) specificity are similarly overturned in daughters after pregnancy, highlighting a fixed microchimeric cell niche. Whereas NIMA-specific tolerance is functionally erased by pregnancy, partner-specific resiliency against pregnancy complications persists in mothers despite paternity changes in intervening pregnancy. Persistent fetal tolerance reflects FOXP3 expression plasticity, which allows mothers to more durably remember their babies, whereas daughters forget their mothers with new pregnancy-imprinted immunological memories.


Chimerism , Fetus , Immune Tolerance , Immunologic Memory , Maternal-Fetal Exchange , Pregnancy , Animals , Female , Mice , Pregnancy/immunology , Antigens/immunology , Cell Plasticity , Fetus/cytology , Fetus/immunology , Forkhead Transcription Factors/immunology , Maternal-Fetal Exchange/immunology , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology
8.
Clin Lab ; 69(4)2023 Apr 01.
Article En | MEDLINE | ID: mdl-37057931

BACKGROUND: Anti-s is a rare alloantibody, and the reported cases of hemolytic disease of the fetus and newborn (HDFN) caused by anti-s are limited to non-Asian populations. METHODS: Here, we report the case of a Chinese woman with a history of multiple pregnancies who developed an alloantibody with anti-s specificity. RESULTS: Her newborn developed HDFN caused by anti-s but the clinical symptoms were not serious. After supportive treatment and bilirubin light phototherapy, the baby was discharged with a good prognosis. CONCLUSIONS: This is the first reported case of anti-s-induced HDFN in a Chinese patient, highlighting the need for further research in the Asian population.


Blood Group Antigens , East Asian People , Erythroblastosis, Fetal , Isoantibodies , Female , Humans , Infant, Newborn , Pregnancy , Erythroblastosis, Fetal/diagnosis , Erythroblastosis, Fetal/etiology , Erythroblastosis, Fetal/immunology , Erythroblastosis, Fetal/therapy , Fetus/immunology , Hemolysis/immunology , Isoantibodies/immunology , Blood Group Antigens/immunology , Phototherapy
9.
Nature ; 613(7945): 639-649, 2023 01.
Article En | MEDLINE | ID: mdl-36697862

Whether the human fetus and the prenatal intrauterine environment (amniotic fluid and placenta) are stably colonized by microbial communities in a healthy pregnancy remains a subject of debate. Here we evaluate recent studies that characterized microbial populations in human fetuses from the perspectives of reproductive biology, microbial ecology, bioinformatics, immunology, clinical microbiology and gnotobiology, and assess possible mechanisms by which the fetus might interact with microorganisms. Our analysis indicates that the detected microbial signals are likely the result of contamination during the clinical procedures to obtain fetal samples or during DNA extraction and DNA sequencing. Furthermore, the existence of live and replicating microbial populations in healthy fetal tissues is not compatible with fundamental concepts of immunology, clinical microbiology and the derivation of germ-free mammals. These conclusions are important to our understanding of human immune development and illustrate common pitfalls in the microbial analyses of many other low-biomass environments. The pursuit of a fetal microbiome serves as a cautionary example of the challenges of sequence-based microbiome studies when biomass is low or absent, and emphasizes the need for a trans-disciplinary approach that goes beyond contamination controls by also incorporating biological, ecological and mechanistic concepts.


Biomass , DNA Contamination , Fetus , Microbiota , Animals , Female , Humans , Pregnancy , Amniotic Fluid/immunology , Amniotic Fluid/microbiology , Mammals , Microbiota/genetics , Placenta/immunology , Placenta/microbiology , Fetus/immunology , Fetus/microbiology , Reproducibility of Results
10.
Proc Natl Acad Sci U S A ; 119(49): e2212548119, 2022 12 06.
Article En | MEDLINE | ID: mdl-36442114

Microbial exposure during development can elicit long-lasting effects on the health of an individual. However, how microbial exposure in early life leads to permanent changes in the immune system is unknown. Here, we show that the microbial environment alters the set point for immune susceptibility by altering the developmental architecture of the CD8+ T cell compartment. In particular, early microbial exposure results in the preferential expansion of highly responsive fetal-derived CD8+ T cells that persist into adulthood and provide the host with enhanced immune protection against intracellular pathogens. Interestingly, microbial education of fetal-derived CD8+ T cells occurs during thymic development rather than in the periphery and involves the acquisition of a more effector-like epigenetic program. Collectively, our results provide a conceptual framework for understanding how microbial colonization in early life leads to lifelong changes in the immune system.


CD8-Positive T-Lymphocytes , Fetus , Immunity , Cell Differentiation , Educational Status , Epigenomics , Fetus/immunology , Fetus/microbiology
11.
Proc Natl Acad Sci U S A ; 119(35): e2123267119, 2022 08 30.
Article En | MEDLINE | ID: mdl-35994660

The pregnant uterus is an immunologically rich organ, with dynamic changes in the inflammatory milieu and immune cell function underlying key stages of pregnancy. Recent studies have implicated dysregulated expression of the interleukin-1 (IL-1) family cytokine, IL-33, and its receptor, ST2, in poor pregnancy outcomes in women, including recurrent pregnancy loss, preeclampsia, and preterm labor. How IL-33 supports pregnancy progression in vivo is not well understood. Here, we demonstrate that maternal IL-33 signaling critically regulates uterine tissue remodeling and immune cell function during early pregnancy in mice. IL-33-deficient dams exhibit defects in implantation chamber formation and decidualization, and abnormal vascular remodeling during early pregnancy. These defects coincide with delays in early embryogenesis, increased resorptions, and impaired fetal and placental growth by late pregnancy. At a cellular level, myometrial fibroblasts, and decidual endothelial and stromal cells, are the main IL-33+ cell types in the uterus during decidualization and early placentation, whereas ST2 is expressed by uterine immune populations associated with type 2 immune responses, including ILC2s, Tregs, CD4+ T cells, M2- and cDC2-like myeloid cells, and mast cells. Early pregnancy defects in IL-33-deficient dams are associated with impaired type 2 cytokine responses by uterine lymphocytes and fewer Arginase-1+ macrophages in the uterine microenvironment. Collectively, our data highlight a regulatory network, involving crosstalk between IL-33-producing nonimmune cells and ST2+ immune cells at the maternal-fetal interface, that critically supports pregnancy progression in mice. This work has the potential to advance our understanding of how IL-33 signaling may support optimal pregnancy outcomes in women.


Interleukin-33 , Placenta , Placentation , Uterus , Animals , Decidua/blood supply , Decidua/cytology , Decidua/growth & development , Decidua/immunology , Female , Fetus/immunology , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/deficiency , Interleukin-33/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Placenta/immunology , Placenta/metabolism , Pregnancy , Uterus/blood supply , Uterus/growth & development , Uterus/immunology , Uterus/metabolism
12.
Development ; 149(8)2022 04 15.
Article En | MEDLINE | ID: mdl-35050308

Maintenance of a healthy pregnancy is reliant on a successful balance between the fetal and maternal immune systems. Although the maternal mechanisms responsible have been well studied, those used by the fetal immune system remain poorly understood. Using suspension mass cytometry and various imaging modalities, we report a complex immune system within the mid-gestation (17-23 weeks) human placental villi (PV). Consistent with recent reports in other fetal organs, T cells with memory phenotypes, although rare in abundance, were detected within the PV tissue and vasculature. Moreover, we determined that T cells isolated from PV samples may be more proliferative after T cell receptor stimulation than adult T cells at baseline. Collectively, we identified multiple subtypes of fetal immune cells within the PV and specifically highlight the enhanced proliferative capacity of fetal PV T cells.


Chorionic Villi/immunology , Placenta/immunology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Chorionic Villi/metabolism , Female , Fetus/immunology , Fetus/metabolism , Flow Cytometry , HLA-DR Antigens/genetics , HLA-DR Antigens/metabolism , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukocyte Common Antigens/metabolism , Lymphocyte Activation , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Memory T Cells/cytology , Memory T Cells/immunology , Memory T Cells/metabolism , Placenta/cytology , Placenta/metabolism , Pregnancy , Pregnancy Trimester, Second , Receptors, Cell Surface/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Single-Cell Analysis/methods , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
13.
Development ; 149(8)2022 04 15.
Article En | MEDLINE | ID: mdl-34604909

The adult human skin contains a vast number of T cells that are essential for skin homeostasis and pathogen defense. T cells are first observed in the skin at the early stages of gestation; however, our understanding of their contribution to early immunity has been limited by their low abundance and lack of comprehensive methodologies for their assessment. Here, we describe a new workflow for isolating and expanding significant amounts of T cells from fetal human skin. Using multiparametric flow cytometry and in situ immunofluorescence, we found a large population with a naive phenotype and small populations with a memory and regulatory phenotype. Their molecular state was characterized using single-cell transcriptomics and TCR repertoire profiling. Importantly, culture of total fetal skin biopsies facilitated T cell expansion without a substantial impact on their phenotype, a major prerequisite for subsequent functional assays. Collectively, our experimental approaches and data advance the understanding of fetal skin immunity and potential use in future therapeutic interventions.


Fetus , Flow Cytometry , Skin , T-Lymphocytes , Adult , Female , Fetus/cytology , Fetus/immunology , Humans , Male , Middle Aged , Skin/cytology , Skin/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
14.
Cells ; 10(12)2021 12 20.
Article En | MEDLINE | ID: mdl-34944105

Emerging evidence indicates that perinatal infection and inflammation can influence the developing immune system and may ultimately affect long-term health and disease outcomes in offspring by perturbing tissue and immune homeostasis. We posit that perinatal inflammation influences immune outcomes in offspring by perturbing (1) the development and function of fetal-derived immune cells that regulate tissue development and homeostasis, and (2) the establishment and function of developing hematopoietic stem cells (HSCs) that continually generate immune cells across the lifespan. To disentangle the complexities of these interlinked systems, we propose the cochlea as an ideal model tissue to investigate how perinatal infection affects immune, tissue, and stem cell development. The cochlea contains complex tissue architecture and a rich immune milieu that is established during early life. A wide range of congenital infections cause cochlea dysfunction and sensorineural hearing loss (SNHL), likely attributable to early life inflammation. Furthermore, we show that both immune cells and bone marrow hematopoietic progenitors can be simultaneously analyzed within neonatal cochlear samples. Future work investigating the pathogenesis of SNHL in the context of congenital infection will therefore provide critical information on how perinatal inflammation drives disease susceptibility in offspring.


Cochlea/pathology , Hematopoiesis , Immune System/growth & development , Inflammation/pathology , Animals , Fetus/immunology , Hearing Loss, Sensorineural/immunology , Humans
15.
Front Immunol ; 12: 758281, 2021.
Article En | MEDLINE | ID: mdl-34745133

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.


Fetus/immunology , Immune Tolerance/immunology , Macrophages/physiology , Pregnancy/immunology , Trophoblasts/physiology , Animals , Cell Communication , Cell Movement , Cellular Microenvironment , Cytokines/physiology , Extracellular Vesicles/physiology , Female , Humans , Macrophage Activation , Macrophages/classification , Macrophages/cytology , Macrophages/immunology , Mice , Trophoblasts/classification , Trophoblasts/cytology , Trophoblasts/immunology
16.
Front Immunol ; 12: 771054, 2021.
Article En | MEDLINE | ID: mdl-34745147

The placenta is a fetal-derived organ whose function is crucial for both maternal and fetal health. The human placenta contains a population of fetal macrophages termed Hofbauer cells. These macrophages play diverse roles, aiding in placental development, function and defence. The outer layer of the human placenta is formed by syncytiotrophoblast cells, that fuse to form the syncytium. Adhered to the syncytium at sites of damage, on the maternal side of the placenta, is a population of macrophages termed placenta associated maternal macrophages (PAMM1a). Here we discuss recent developments that have led to renewed insight into our understanding of the ontogeny, phenotype and function of placental macrophages. Finally, we discuss how the application of new technologies within placental research are helping us to further understand these cells.


Fetal Development/immunology , Fetus/immunology , Immunity, Innate/immunology , Macrophages/immunology , Placenta/immunology , Animals , Cell Movement/immunology , Cell Movement/physiology , Chorionic Villi/immunology , Chorionic Villi/metabolism , Female , Fetus/cytology , Fetus/physiology , Folate Receptor 2/immunology , Folate Receptor 2/metabolism , HLA-DR Antigens/immunology , HLA-DR Antigens/metabolism , Humans , Macrophages/metabolism , Macrophages/physiology , Phagocytosis/immunology , Phagocytosis/physiology , Placenta/cytology , Placenta/physiology , Pregnancy
17.
Front Immunol ; 12: 735564, 2021.
Article En | MEDLINE | ID: mdl-34777345

Pregnancy after renal transplantation is associated with an increased risk of complications. While a delicately balanced uterine immune system is essential for a successful pregnancy, little is known about the uterine immune environment of pregnant kidney transplant recipients. Moreover, children born to kidney transplant recipients are exposed in utero to immunosuppressive drugs, with possible consequences for neonatal outcomes. Here, we defined the effects of kidney transplantation on the immune cell composition during pregnancy with a cohort of kidney transplant recipients as well as healthy controls with uncomplicated pregnancies. Maternal immune cells from peripheral blood were collected during pregnancy as well as from decidua and cord blood obtained after delivery. Multiparameter flow cytometry was used to identify and characterize populations of cells. While systemic immune cell frequencies were altered in kidney transplant patients, immune cell dynamics over the course of pregnancy were largely similar to healthy women. In the decidua of women with a kidney transplant, we observed a decreased frequency of HLA-DR+ Treg, particularly in those treated with tacrolimus versus those that were treated with azathioprine next to tacrolimus, or with azathioprine alone. In addition, both the innate and adaptive neonatal immune system of children born to kidney transplant recipients was significantly altered compared to neonates born from uncomplicated pregnancies. Overall, our findings indicate a significant and distinct impact on the maternal systemic, uterine, and neonatal immune cell composition in pregnant kidney transplant recipients, which could have important consequences for the incidence of pregnancy complications, treatment decisions, and the offspring's health.


Decidua/drug effects , Fetus/drug effects , Immunosuppressive Agents/adverse effects , Kidney Transplantation/adverse effects , Lymphocyte Subsets/drug effects , Mothers , Transplant Recipients , Adult , Biomarkers/metabolism , Case-Control Studies , Cells, Cultured , Decidua/immunology , Decidua/metabolism , Female , Fetus/immunology , Fetus/metabolism , Flow Cytometry , Humans , Immunophenotyping , Infant, Newborn , Lymphocyte Activation/drug effects , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Phenotype , Pregnancy , Pregnancy Outcome , Young Adult
18.
Front Immunol ; 12: 744324, 2021.
Article En | MEDLINE | ID: mdl-34777357

Maternal-fetal immune-tolerance occurs throughout the whole gestational trimester, thus a mother can accept a genetically distinct fetus without immunological aggressive behavior. HLA-G, one of the non-classical HLA class I molecules, is restricted-expression at extravillous trophoblast. It can concordantly interact with various kinds of receptors mounted on maternally immune cells residing in the uterus (e.g. CD4+ T cells, CD8+ T cells, natural killer cells, macrophages, and dendritic cells) for maintaining immune homeostasis of the maternal-fetus interface. HLA-G is widely regarded as the pivotal protective factor for successful pregnancies. In the past 20 years, researches associated with HLA-G have been continually published. Indeed, HLA-G plays a mysterious role in the mechanism of maternal-fetal immune-tolerance. It can also be ectopically expressed on tumor cells, infected sites and other pathologic microenvironments to confer a significant local tolerance. Understanding the characteristics of HLA-G in immunologic tolerance is not only beneficial for pathological pregnancy, but also helpful to the therapy of other immune-related diseases, such as organ transplant rejection, tumor migration, and autoimmune disease. In this review, we describe the biological properties of HLA-G, then summarize our understanding of the mechanisms of fetomaternal immunologic tolerance and the difference from transplant tolerance. Furthermore, we will discuss how HLA-G contributes to the tolerogenic microenvironment during pregnancy. Finally, we hope to find some new aspects of HLA-G in fundamental research or clinical application for the future.


Fetus/immunology , HLA-G Antigens/immunology , Immune Privilege/immunology , Animals , Female , Humans , Pregnancy
19.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Article En | MEDLINE | ID: mdl-34785597

Zika virus (ZIKV) during pregnancy infects fetal trophoblasts and causes placental damage and birth defects including microcephaly. Little is known about the anti-ZIKV cellular immune response at the maternal-fetal interface. Decidual natural killer cells (dNK), which directly contact fetal trophoblasts, are the dominant maternal immune cells in the first-trimester placenta, when ZIKV infection is most hazardous. Although dNK express all the cytolytic molecules needed to kill, they usually do not kill infected fetal cells but promote placentation. Here, we show that dNK degranulate and kill ZIKV-infected placental trophoblasts. ZIKV infection of trophoblasts causes endoplasmic reticulum (ER) stress, which makes them dNK targets by down-regulating HLA-C/G, natural killer (NK) inhibitory receptor ligands that help maintain tolerance of the semiallogeneic fetus. ER stress also activates the NK activating receptor NKp46. ZIKV infection of Ifnar1 -/- pregnant mice results in high viral titers and severe intrauterine growth restriction, which are exacerbated by depletion of NK or CD8 T cells, indicating that killer lymphocytes, on balance, protect the fetus from ZIKV by eliminating infected cells and reducing the spread of infection.


Killer Cells, Natural/immunology , Trophoblasts/immunology , Zika Virus Infection/immunology , Zika Virus/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Fetus/immunology , HLA-C Antigens , Immune Tolerance , Mice , Placenta/immunology , Placentation , Pregnancy , Pregnancy Complications, Infectious/immunology , Receptors, KIR
20.
Front Immunol ; 12: 729742, 2021.
Article En | MEDLINE | ID: mdl-34764952

A wealth of innate and adaptive immune cells and hormones are involved in mounting tolerance towards the fetus, a key aspect of successful reproduction. We could recently show that the specific cross talk between the pregnancy hormone progesterone and dendritic cells (DCs) is significantly engaged in the generation of CD4+ FoxP3+ regulatory T (Treg) cells while a disruption led to placental alterations and intra-uterine growth restriction. Apart from progesterone, also glucocorticoids affect immune cell functions. However, their functional relevance in the context of pregnancy still needs clarification. We developed a mouse line with a selective knockout of the glucocorticoid receptor (GR) on DCs, utilizing the cre/flox system. Reproductive outcome and maternal immune and endocrine adaptation of Balb/c-mated C57Bl/6 GRflox/floxCD11ccre/wt (mutant) females was assessed on gestation days (gd) 13.5 and 18.5. Balb/c-mated C57Bl/6 GRwt/wtCD11ccre/wt (wt) females served as controls. The number of implantation and fetal loss rate did not differ between groups. However, we identified a significant increase in fetal weight in fetuses from mutant dams. While the frequencies of CD11c+ cells remained largely similar, a decreased expression of co-stimulatory molecules was observed on DCs of mutant females on gd 13.5, along with higher frequencies of CD4+ and CD8+ Treg cells. Histomorphological and gene expression analysis revealed an increased placental volume and an improved functional placental capacity in mice lacking the GR on CD11c+ DCs. In summary, we here demonstrate that the disrupted communication between GCs and DCs favors a tolerant immune microenvironment and improves placental function and fetal development.


CD11 Antigens/metabolism , Dendritic Cells/metabolism , Fetal Development , Fetus/metabolism , Glucocorticoids/metabolism , Receptors, Glucocorticoid/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , CD11 Antigens/genetics , Dendritic Cells/immunology , Female , Fetal Weight , Fetus/immunology , Gestational Age , Histocompatibility, Maternal-Fetal , Immune Tolerance , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Placentation , Pregnancy , Progesterone/metabolism , Receptors, Glucocorticoid/genetics , Signal Transduction , T-Lymphocytes, Regulatory/immunology
...