Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.228
Filter
1.
Sci Rep ; 14(1): 10772, 2024 05 10.
Article in English | MEDLINE | ID: mdl-38730052

ABSTRACT

We aimed to determine SARS-CoV-2 antibody seropositivity among pregnant women and the transplacental transfer efficiency of SARS-CoV-2-specific antibodies relative to malaria antibodies among SARS-CoV-2 seropositive mother-cord pairs. This cross-sectional study was conducted in Accra, Ghana, from March to May 2022. Antigen- specific IgG antibodies against SARS-CoV-2 (nucleoprotein and spike-receptor binding domain) and malarial antigens (circumsporozoite protein and merozoite surface protein 3) in maternal and cord plasma were measured by ELISA. Plasma from both vaccinated and unvaccinated pregnant women were tested for neutralizing antibodies using commercial kit. Of the unvaccinated pregnant women tested, 58.12% at antenatal clinics and 55.56% at the delivery wards were seropositive for both SARS-CoV-2 nucleoprotein and RBD antibodies. Anti-SARS-CoV-2 antibodies in cord samples correlated with maternal antibody levels (N antigen rs = 0.7155, p < 0.001; RBD rs = 0.8693, p < 0.001). Transplacental transfer of SARS-CoV-2 nucleoprotein antibodies was comparable to circumsporozoite protein antibodies (p = 0.9999) but both were higher than transfer rates of merozoite surface protein 3 antibodies (p < 0.001). SARS-CoV-2 IgG seropositivity among pregnant women in Accra is high with a boost of SARS-CoV-2 RBD-specific IgG in vaccinated women. Transplacental transfer of anti-SARS-CoV-2 and malarial antibodies was efficient, supporting vaccination of mothers as a strategy to protect infants against SARS-CoV-2.


Subject(s)
Antibodies, Viral , COVID-19 , Immunoglobulin G , SARS-CoV-2 , Humans , Female , Pregnancy , Ghana , SARS-CoV-2/immunology , COVID-19/immunology , COVID-19/prevention & control , Antibodies, Viral/immunology , Antibodies, Viral/blood , Adult , Cross-Sectional Studies , Immunoglobulin G/blood , Immunoglobulin G/immunology , Maternal-Fetal Exchange/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Infant , Infant, Newborn , Spike Glycoprotein, Coronavirus/immunology , Immunity, Maternally-Acquired , Young Adult , Fetal Blood/immunology , Antibodies, Protozoan/immunology , Antibodies, Protozoan/blood
2.
Am J Reprod Immunol ; 91(5): e13859, 2024 May.
Article in English | MEDLINE | ID: mdl-38722063

ABSTRACT

Recurrent Spontaneous Abortion (RSA) is a common pregnancy complication, that has multifactorial causes, and currently, 40%-50% of cases remain unexplained, referred to as Unexplained RSA (URSA). Due to the elusive etiology and mechanisms, clinical management is exceedingly challenging. In recent years, with the progress in reproductive immunology, a growing body of evidence suggests a relationship between URSA and maternal-fetal immunology, offering hope for the development of tailored treatment strategies. This article provides an immunological perspective on the pathogenesis, diagnosis, and treatment of RSA. On one hand, it comprehensively reviews the immunological mechanisms underlying RSA, including abnormalities in maternal-fetal interface immune tolerance, maternal-fetal interface immune cell function, gut microbiota-mediated immune dysregulation, and vaginal microbiota-mediated immune anomalies. On the other hand, it presents the diagnosis and existing treatment modalities for RSA. This article offers a clear knowledge framework for understanding RSA from an immunological standpoint. In conclusion, while the "layers of the veil" regarding immunological factors in RSA are gradually being unveiled, our current research may only scratch the surface. In terms of immunological etiology, effective diagnostic tools for RSA are currently lacking, and the efficacy and safety of immunotherapies, primarily based on lymphocyte immunotherapy and intravenous immunoglobulin, remain contentious.


Subject(s)
Abortion, Habitual , Humans , Female , Pregnancy , Abortion, Habitual/immunology , Immune Tolerance , Maternal-Fetal Exchange/immunology , Gastrointestinal Microbiome/immunology , Immunotherapy/methods
3.
Science ; 381(6664): 1286, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37733842

ABSTRACT

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


Subject(s)
Chimerism , Fetus , Immune Tolerance , Maternal-Fetal Exchange , Child , Female , Humans , Pregnancy , Maternal-Fetal Exchange/immunology , Antigens/immunology , Fetus/cytology , Fetus/immunology
4.
Science ; 381(6664): 1324-1330, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37733857

ABSTRACT

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.


Subject(s)
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
5.
Nature ; 619(7970): 595-605, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37468587

ABSTRACT

Beginning in the first trimester, fetally derived extravillous trophoblasts (EVTs) invade the uterus and remodel its spiral arteries, transforming them into large, dilated blood vessels. Several mechanisms have been proposed to explain how EVTs coordinate with the maternal decidua to promote a tissue microenvironment conducive to spiral artery remodelling (SAR)1-3. However, it remains a matter of debate regarding which immune and stromal cells participate in these interactions and how this evolves with respect to gestational age. Here we used a multiomics approach, combining the strengths of spatial proteomics and transcriptomics, to construct a spatiotemporal atlas of the human maternal-fetal interface in the first half of pregnancy. We used multiplexed ion beam imaging by time-of-flight and a 37-plex antibody panel to analyse around 500,000 cells and 588 arteries within intact decidua from 66 individuals between 6 and 20 weeks of gestation, integrating this dataset with co-registered transcriptomics profiles. Gestational age substantially influenced the frequency of maternal immune and stromal cells, with tolerogenic subsets expressing CD206, CD163, TIM-3, galectin-9 and IDO-1 becoming increasingly enriched and colocalized at later time points. By contrast, SAR progression preferentially correlated with EVT invasion and was transcriptionally defined by 78 gene ontology pathways exhibiting distinct monotonic and biphasic trends. Last, we developed an integrated model of SAR whereby invasion is accompanied by the upregulation of pro-angiogenic, immunoregulatory EVT programmes that promote interactions with the vascular endothelium while avoiding the activation of maternal immune cells.


Subject(s)
Maternal-Fetal Exchange , Trophoblasts , Uterus , Female , Humans , Pregnancy , Arteries/physiology , Decidua/blood supply , Decidua/cytology , Decidua/immunology , Decidua/physiology , Pregnancy Trimester, First/genetics , Pregnancy Trimester, First/metabolism , Pregnancy Trimester, First/physiology , Trophoblasts/cytology , Trophoblasts/immunology , Trophoblasts/physiology , Uterus/blood supply , Uterus/cytology , Uterus/immunology , Uterus/physiology , Maternal-Fetal Exchange/genetics , Maternal-Fetal Exchange/immunology , Maternal-Fetal Exchange/physiology , Time Factors , Proteomics , Gene Expression Profiling , Datasets as Topic , Gestational Age
6.
J Clin Virol ; 165: 105495, 2023 08.
Article in English | MEDLINE | ID: mdl-37295035

ABSTRACT

BACKGROUNDS: Due to immaturity of their immune system, passive maternal immunization is essential for newborns during their first months of life. Therefore, in the current context of intense circulation of SARS-CoV-2, identifying factors influencing the transfer ratio (TR) of neutralizing antibodies against SARS-CoV-2 (NAb) appears important. METHODS: Our study nested in the COVIPREG cohort (NCT04355234), included mothers who had a SARS-CoV-2 PCR positive during their pregnancy and their newborns. Maternal and neonatal NAb levels were measured with the automated iFlash system. RESULTS: For the 173 mother-infant pairs included in our study, the median gestational age (GA) at delivery was 39.4 weeks of gestation (WG), and 29.7 WG at maternal SARS-CoV-2 infection. Using a multivariate logistic model, having a NAb TR above 1 was positively associated with a longer delay from maternal positive SARS-CoV-2 PCR to delivery (aOR 1.09, 95% CI: 1.03 - 1.17) and with a later GA at delivery (aOR = 1.58, 95% CI: 1.09 - 2.52). It was negatively associated with being a male newborn (aOR 0.21, 95% CI: 0.07 - 0.59). In 3rd trimester SARS-CoV-2 infected mothers, NAb TR was inferior to VZV, toxoplasmosis, CMV, measle and rubella's TR. However, in 1st or 2nd trimester infected mothers, only measle TR was different from NAb TR. CONCLUSION: Male newborn of mothers infected by SARS-CoV-2 during their pregnancy appear to have less protection against SARS-CoV-2 in their first months of life than female newborns. Measle TR was superior to NAb TR even in case of 1st or 2nd trimester maternal SARS-CoV-2 infection. Future studies are needed to investigate possible differences in transmission of NAb following infection vs vaccination and its impact on TR.


Subject(s)
Antibodies, Neutralizing , COVID-19 , Infant, Newborn, Diseases , Infectious Disease Transmission, Vertical , Maternal-Fetal Exchange , Pregnancy Complications , SARS-CoV-2 , SARS-CoV-2/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Maternal-Fetal Exchange/immunology , Gestational Age , Humans , Male , Female , COVID-19/blood , COVID-19/immunology , COVID-19/prevention & control , Delivery, Obstetric , Antibodies, Viral/blood , Antibodies, Viral/immunology , Pregnancy , Infant, Newborn , Sex Characteristics , COVID-19 Vaccines , Vaccination , Pregnancy Complications/blood , Pregnancy Complications/immunology , Infant, Newborn, Diseases/immunology , Infant, Newborn, Diseases/prevention & control , Infectious Disease Transmission, Vertical/prevention & control , Paris , Adult
7.
Proc Natl Acad Sci U S A ; 119(15): e2113310119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35377817

ABSTRACT

Gestational maternal immune activation (MIA) in mice induces persistent brain microglial activation and a range of neuropathologies in the adult offspring. Although long-term phenotypes are well documented, how MIA in utero leads to persistent brain inflammation is not well understood. Here, we found that offspring of mothers treated with polyriboinosinic­polyribocytidylic acid [poly(I:C)] to induce MIA at gestational day 13 exhibit blood­brain barrier (BBB) dysfunction throughout life. Live MRI in utero revealed fetal BBB hyperpermeability 2 d after MIA. Decreased pericyte­endothelium coupling in cerebral blood vessels and increased microglial activation were found in fetal and 1- and 6-mo-old offspring brains. The long-lasting disruptions result from abnormal prenatal BBB formation, driven by increased proliferation of cyclooxygenase-2 (COX2; Ptgs2)-expressing microglia in fetal brain parenchyma and perivascular spaces. Targeted deletion of the Ptgs2 gene in fetal myeloid cells or treatment with the inhibitor celecoxib 24 h after immune activation prevented microglial proliferation and disruption of BBB formation and function, showing that prenatal COX2 activation is a causal pathway of MIA effects. Thus, gestational MIA disrupts fetal BBB formation, inducing persistent BBB dysfunction, which promotes microglial overactivation and behavioral alterations across the offspring life span. Taken together, the data suggest that gestational MIA disruption of BBB formation could be an etiological contributor to neuropsychiatric disorders.


Subject(s)
Blood-Brain Barrier , Cyclooxygenase 2 , Encephalitis , Maternal-Fetal Exchange , Microglia , Prenatal Exposure Delayed Effects , Animals , Blood-Brain Barrier/abnormalities , Blood-Brain Barrier/physiopathology , Celecoxib/pharmacology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Encephalitis/immunology , Female , Gene Deletion , Maternal-Fetal Exchange/immunology , Mice , Microglia/enzymology , Poly I-C/immunology , Pregnancy , Prenatal Exposure Delayed Effects/immunology
9.
Placenta ; 119: 8-16, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35066308

ABSTRACT

INTRODUCTION: Placenta accreta spectrum (PAS) disorder is one of the major complications resulting in maternal death and serious adverse pregnancy outcomes. Uterine damage - principally that associated with cesarean section - is the leading risk factor for the development of PAS. However, the underlying pathogenesis of PAS related to uterine damage remains unclear. METHODS: For this study, we constructed a mouse PAS model using hysterotomy to simulate a cesarean section in humans. Pregnant mice were sacrificed on embryonic days 12.5 (E12.5) and E17.5. Trophoblast invasion and placental vascularization were analyzed using Hematoxylin-Eosin (H&E) staining and immunohistochemistry (IHC), and the proportions of immune cells at the maternal-fetal interface were analyzed using flow cytometry. We analyzed the expressions of genes in the decidua and placenta using RNA sequencing and subsequent validation by QPCR, and measured serum angiogenic factors by ELISA. RESULTS: Uterine damage led to increased trophoblast invasion and placental vascularization, with extensive changes to the immune-cell profiles at the maternal-fetal interface. The proportions of T and NK cells in the deciduas diminished significantly, with the decidual NK cells and M - 2 macrophages showing the greatest decline. The expression of TNF-α and IL4 was upregulated in the deciduas, while that of IFN-γ and IL10 was downregulated significantly. The expression of Mmp2, Mmp9, Mmp3, and Dock4 was significantly elevated in the placenta, and the serum levels of anti-angiogenic factors were significantly attenuated. DISCUSSION: Uterine damage can cause immune imbalance at the maternal-fetal interface, which may contribute to abnormal trophoblast invasion and enhanced vascularization of the mouse placenta.


Subject(s)
Maternal-Fetal Exchange/immunology , Neovascularization, Physiologic , Obstetric Surgical Procedures/adverse effects , Placenta Accreta/etiology , Trophoblasts/physiology , Animals , Female , Mice, Inbred C57BL , Pregnancy , Random Allocation
10.
J Dev Orig Health Dis ; 13(1): 3-8, 2022 02.
Article in English | MEDLINE | ID: mdl-33526164

ABSTRACT

Little is known about the consequences of viral infection for pregnant woman or for the fetus. This issue became important with the appearance of the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). The infection with SARS-CoV-2 causes a respiratory syndrome known as COVID-19. The fast spreading around the world and the fact that without a treatment or vaccine humans are completely exposed, converts emerging viral diseases in a significant risk for pregnant women and their infants. At this time, during SARS-CoV-2 pandemics pregnant women are not considered as a risk population and little is known about the effects of viral infections over the offspring although the amount of emerging evidence showing detrimental effects for the mother and the fetus. This issue highlights the importance to understand the effects of viral infections during pregnancy. In this work, we analyze the effects of viral infections, like SARS-CoV-2 and other related viruses during pregnancy over the mother and the consequences for the offspring.


Subject(s)
COVID-19/complications , Pregnancy Complications, Infectious/virology , SARS-CoV-2 , COVID-19/immunology , Coronavirus Infections/complications , Female , Humans , Infant, Newborn/immunology , Infectious Disease Transmission, Vertical , Influenza A Virus, H1N1 Subtype , Influenza, Human/complications , Maternal-Fetal Exchange/immunology , Pregnancy , Severe Acute Respiratory Syndrome/complications
11.
J Immunol Res ; 2021: 7507459, 2021.
Article in English | MEDLINE | ID: mdl-34950737

ABSTRACT

The search for common mechanisms underlying the pathogenesis of chronic inflammatory conditions has crystalized the concept of continuous dual resetting of the immune repertoire (CDR) as a basic principle of the immune system function. Consequently, outlined was the first dynamic comprehensive picture of the immune system function. The goal of this study is to elaborate on regulation of immune responses and mechanisms of tolerance, particularly focusing on adaptive immunity. It is well established that the T/B cell repertoire is selected and maintained based on interactions with self. However, their activation also requires interaction with a self-specific major histocompatibility complex (MHC) "code," i.e., the context of MHC molecules. Therefore, not only repertoire selection and maintenance but also the T/B cell activation and function are self-centered. Thus, adaptive effectors may be primarily focused on the state of self and maintenance of integrity of the self, and only to a certain degree on elimination of the foreign. As examples of such function are used immunologically poorly understood MHC-disparate settings typical for transplantation and pregnancy. Transplantation represents an extreme setting of strong systemic compartment-level adaptive/MHC-restricted immune responses. Described are clinically identified conditions for operational tolerance of MHC-disparate tissues/living systems in allotransplantation, which are in line with the CDR-proposed self-centered regulatory role of T/B cells. In contrast, normal pregnancy is coexistence of semiallogeneic or entirely allogeneic mother and fetus, but without alloreactivity akin to transplantation settings. Presented data support the notion that maintenance of pregnancy is a process that relies predominantly on innate/MHC-independent immune mechanisms. By the inception of hemotrophic stage of pregnancy (second and third trimester), both mother and child are individual living systems, with established adaptive immune repertoires. Although mother-fetus interactions at that point become indirect systemic compartment-level communications, their interactions throughout gestation remain within the innate realm of molecular-level adaptations.


Subject(s)
Adaptive Immunity , Immune Tolerance , Immunomodulation , Self Tolerance/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Female , Humans , Immune System Phenomena , Immunity, Innate , Male , Maternal-Fetal Exchange/immunology , Pregnancy , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thyroid Gland/immunology , Thyroid Gland/metabolism , Transplantation Immunology
12.
PLoS One ; 16(12): e0261357, 2021.
Article in English | MEDLINE | ID: mdl-34941916

ABSTRACT

During pregnancy in placental mammals, small numbers of maternal cells (maternal microchimeric cells, or MMc cells) migrate into the fetus and persist decades, or perhaps for the rest of their lives, and higher frequencies of MMc cells are reported to correlate with variety of phenomena, such as immune tolerance, tissue repair, and autoimmune diseases. While detection of these MMc cells is considered in all pregnancies, their frequency differs largely according to tissue type and disease cases, and it remains unclear whether the number of MMc cells differs significantly among embryos in normal pregnancies. Here, for the first time, we developed a whole embryonic detection method for MMc cells using transgenic mice and counted live MMc cells in each individual embryo. Using this technique, we found that the number of MMc cells was comparable in most of the analyzed embryos; however, around 500 times higher number of MMc cells was detected in one embryo at the latest stage. This result suggests that the number of MMc cells could largely differ in rare cases with unknown underlying mechanisms. Our methodology provides a basis for testing differences in the numbers of MMc cells among individual embryos and for analyzing differences in MMc cell type repertoires in future studies. These data could provide a hint toward understanding the mechanisms underlying the variety of apparently inconsistent MMc-related phenomena.


Subject(s)
Chimerism/embryology , Animals , Chimerism/statistics & numerical data , Embryo, Mammalian/immunology , Embryo, Mammalian/metabolism , Eutheria/metabolism , Female , Fetus , Immune Tolerance/immunology , Maternal-Fetal Exchange/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Placenta , Pregnancy
13.
EBioMedicine ; 74: 103721, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34844192

ABSTRACT

BACKGROUND: During pregnancy a feto-maternal exchange of cells through the placenta conducts to maternal microchimerism (Mc) in the child and fetal Mc in the mother. Because of this bidirectional traffic of cells, pregnant women have also acquired maternal cells in utero from their mother and could transfer grandmaternal (GdM) cells to their child through the maternal bloodstream during pregnancy. Thus, cord blood (CB) samples could theoretically carry GdMMc. Nevertheless this has never been demonstrated. METHODS: Using Human Leukocyte Antigen (HLA)-specific quantitative PCR assays on three-generation families, we were able to test 28 CB samples from healthy primigravid women for GdMMc in whole blood (WB) and isolated cells (PBMC, T, B, granulocytes, stem cells). FINDINGS: Five CB samples (18%) had GdMMc which could not be confounded with maternal source, with quantities 100 fold lower than maternal Mc in WB and PBMC. Risk of aneuploidies and/or related invasive prenatal procedures significantly correlated with the presence of GdMMc in CB (p=0.024). Significantly decreased HLA compatibility was observed in three-generation families from CB samples carrying GdMMc (p=0.019). INTERPRETATION: Transgenerational transfer of cells could have implications in immunology and evolution. Further analyses will be necessary to evaluate whether GdMMc in CB is a passive or immunologically active transfer and whether invasive prenatal procedures could trigger GdMMc. FUNDING: Provence-Alpes-Côte d'Azur APEX grant # 2012_06549E, 2012_11786F and 2014_03978) and the Foundation for Medical Research (FRM Grant #ING20140129045).


Subject(s)
Fetal Blood/immunology , HLA Antigens/genetics , Maternal-Fetal Exchange/immunology , Adult , Aneuploidy , Chimerism , Female , France , Grandparents , Healthy Volunteers , Humans , Maternal Age , Maternal Inheritance , Maternal-Fetal Exchange/genetics , Pedigree , Pregnancy
14.
Int J Mol Sci ; 22(21)2021 Oct 26.
Article in English | MEDLINE | ID: mdl-34768946

ABSTRACT

Autism spectrum disorder (ASD) is a neurodevelopmental disease that is characterized by a deficit in social interactions and communication, as well as repetitive and restrictive behaviors. Increasing lines of evidence suggest an important role for immune dysregulation and/or inflammation in the development of ASD. Recently, a relationship between inflammation, oxidative stress, and mitochondrial dysfunction has been reported in the brain tissue of individuals with ASD. Some recent studies have also reported oxidative stress and mitochondrial abnormalities in animal models of maternal immune activation (MIA). This review is focused on the hypothesis that MIA induces microglial activation, oxidative stress, and mitochondrial dysfunction, a deleterious trio in the brain that can lead to neuroinflammation and neurodevelopmental pathologies in offspring. Infection during pregnancy activates the mother's immune system to release proinflammatory cytokines, such as IL-6, TNF-α, and others. Furthermore, these cytokines can directly cross the placenta and enter the fetal circulation, or activate resident immune cells, resulting in an increased production of proinflammatory cytokines, including IL-6. Proinflammatory cytokines that cross the blood-brain barrier (BBB) may initiate a neuroinflammation cascade, starting with the activation of the microglia. Inflammatory processes induce oxidative stress and mitochondrial dysfunction that, in turn, may exacerbate oxidative stress in a self-perpetuating vicious cycle that can lead to downstream abnormalities in brain development and behavior.


Subject(s)
Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/immunology , Prenatal Exposure Delayed Effects/immunology , Animals , Autism Spectrum Disorder/therapy , Cytokines/immunology , Disease Models, Animal , Female , Humans , Inflammation Mediators/immunology , Maternal-Fetal Exchange/immunology , Microglia/immunology , Mitochondria/immunology , Models, Immunological , Neuroimmunomodulation , Oxidative Stress/immunology , Pregnancy
16.
J Cell Mol Med ; 25(22): 10362-10375, 2021 11.
Article in English | MEDLINE | ID: mdl-34708495

ABSTRACT

The foetus can be regarded as a half-allograft implanted into the maternal body. In a successful pregnancy, the mother does not reject the foetus because of the immune tolerance mechanism at the maternal-foetal interface. The innate immune cells are a large part of the decidual leukocytes contributing significantly to a successful pregnancy. Although the contributions have been recognized, their role in human pregnancy has not been completely elucidated. Additionally, the accumulated evidence demonstrates that the immune checkpoint molecules expressed on the immune cells are co-inhibitory receptors regulating their activation and biological function. Therefore, it is critical to understand the immune microenvironment and explore the function of the innate immune cells during pregnancy. This review summarizes the classic immune checkpoints such as PD-1, CTLA-4 and some novel molecules recently identified, including TIM-3, CD200, TIGIT and the Siglecs family on the decidual and peripheral innate immune cells during pregnancy. Furthermore, it emphasizes the role of the immune checkpoint molecules in pregnancy-associated complications and reproductive immunotherapy.


Subject(s)
Immune Checkpoint Proteins/genetics , Immunity, Innate , Immunomodulation , Reproduction/immunology , Biomarkers , Disease Management , Disease Susceptibility , Female , Gene Expression Regulation , Humans , Immune Checkpoint Proteins/metabolism , Immune Tolerance , Immunotherapy , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Maternal-Fetal Exchange/immunology , Placenta/immunology , Placenta/metabolism , Pregnancy , Reproduction/genetics
17.
Biol Futur ; 72(1): 69-74, 2021 Mar.
Article in English | MEDLINE | ID: mdl-34554496

ABSTRACT

Paternal antigens expressed by the foetus are recognized as foreign. Therefore,-according to the rules of transplantation immunity-the foetus ought to be "rejected". However, during normal gestation, maternal immune functions are re-adjusted, in order to create a favourable environment for the developing foetus. Some of the mechanisms that contribute to the altered immunological environment, for example, the cytokine balance and NK cell function, with special emphasis on the role of progesterone and the progesterone-induced blocking factor (PIBF) will be reviewed.


Subject(s)
Embryo, Mammalian/immunology , Extracellular Vesicles/immunology , Immune Tolerance/immunology , Maternal-Fetal Exchange/immunology , Pregnancy Proteins/immunology , Suppressor Factors, Immunologic/immunology , Cytokines/immunology , Cytokines/metabolism , Embryo, Mammalian/embryology , Extracellular Vesicles/metabolism , Female , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Pregnancy , Pregnancy Proteins/metabolism , Suppressor Factors, Immunologic/metabolism
18.
Front Immunol ; 12: 717014, 2021.
Article in English | MEDLINE | ID: mdl-34566973

ABSTRACT

Immunity and metabolism are interdependent and coordinated, which are the core mechanisms for the body to maintain homeostasis. In tumor immunology research, immunometabolism has been a research hotspot and has achieved groundbreaking changes in recent years. However, in the field of maternal-fetal medicine, research on immunometabolism is still lagging. Reports directly investigating the roles of immunometabolism in the endometrial microenvironment and regulation of maternal-fetal immune tolerance are relatively few. This review highlights the leading techniques used to study immunometabolism and their development, the immune cells at the maternal-fetal interface and their metabolic features required for the implementation of their functions, explores the interaction between immunometabolism and pregnancy regulation based on little evidence and clues, and attempts to propose some new research directions and perspectives.


Subject(s)
Energy Metabolism , Immune Tolerance , Immunity , Maternal-Fetal Exchange/immunology , Decidua/immunology , Decidua/metabolism , Endometrium/immunology , Endometrium/metabolism , Female , Homeostasis , Humans , Immune System , Immunomodulation , Killer Cells, Natural/metabolism , Macrophages/immunology , Macrophages/metabolism , Metabolic Networks and Pathways , Metabolomics/methods , Pregnancy , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
19.
Cytokine ; 148: 155707, 2021 12.
Article in English | MEDLINE | ID: mdl-34560611

ABSTRACT

BACKGROUND: Elevated cytokines, like IL-1ßand IL-6, are known to contribute to the pathogenesis of labor. However, the change of inflammatory mediators in maternal-fetal interface to fetal circulation is obscure. STUDY DESIGN AND METHODS: We investigated the changes of inflammatory cytokines, chemokines and macrophage in maternal-fetal interface tissues and fetal circulation of women in labor vs. non-labor. Human myometrium, placenta, decidua, fetal membrane and umbilical blood were obtained from in-labor and non-in-labor women who eventually delivered live, singleton infants at term (>37 weeks gestation) by elective caesarean section. Luminex was used to measure the level of cytokines (TNF-α, IL-1ß, IL-6, IL-8) and chemokines (MCP-1, GM-CSF, MIP-1α, MIP-1ß) in each sample (tissue and umbilical blood). Macrophage infiltration was demonstrated by immunohistochemistry. RESULTS: During labor, the level of cytokines TNF-α, IL-1ß, IL-6 and IL-8 and chemokine MCP-1 and MIP-1ß in myometrium is significantly higher (p < 0.05), than those obtained from non-laboring patients. This increase coincides with the influx of macrophage into the myometrium. In addition, IL-1ß and IL-8 (p < 0.05) are also up regulated in fetal membrane during labor compared to non-labor. The cytokines do not change significantly in placenta and decidua tissue. In fetal circulation, IL-6 (p < 0.05) is up regulated in umbilical vein blood in labor group. IL-8 (p = 0.08) in umbilical vein also show an increasing trend during labor. CONCLUSIONS: There are markedly elevated inflammatory mediators in maternal-fetal interface during labor. The increased maternal inflammatory factors released into the fetal circulation through placenta circulation at the time of labor. This increase coincides with the influx of macrophage into the pregnancy tissue, suggesting that the inflammatory response might play an important role in the onset of labor.


Subject(s)
Blood Circulation/physiology , Fetus/physiology , Inflammation Mediators/blood , Labor, Obstetric , Maternal-Fetal Exchange/physiology , Adult , Female , Humans , Maternal-Fetal Exchange/immunology , Pregnancy
20.
Front Immunol ; 12: 717808, 2021.
Article in English | MEDLINE | ID: mdl-34394125

ABSTRACT

Its semi-allogeneic nature renders the conceptus vulnerable to attack by the maternal immune system. Several protective mechanisms operate during gestation to correct the harmful effects of anti-fetal immunity and to support a healthy pregnancy outcome. Pregnancy is characterized by gross alterations in endocrine functions. Progesterone is indispensable for pregnancy and humans, and it affects immune functions both directly and via mediators. The progesterone-induced mediator - PIBF - acts in favor of Th2-type immunity, by increasing Th2 type cytokines production. Except for implantation and parturition, pregnancy is characterized by a Th2-dominant cytokine pattern. Progesterone and the orally-administered progestogen dydrogesterone upregulate the production of Th2-type cytokines and suppress the production of Th1 and Th17 cytokine production in vitro. This is particularly relevant to the fact that the Th1-type cytokines TNF-α and IFN-γ and the Th17 cytokine IL-17 have embryotoxic and anti-trophoblast activities. These cytokine-modulating effects and the PIBF-inducing capabilities of dydrogesterone may contribute to the demonstrated beneficial effects of dydrogesterone in recurrent spontaneous miscarriage and threatened miscarriage. IL-17 and IL-22 produced by T helper cells are involved in allograft rejection, and therefore could account for the rejection of paternal HLA-C-expressing trophoblast. Th17 cells (producing IL-17 and IL-22) and Th22 cells (producing IL-22) exhibit plasticity and could produce IL-22 and IL-17 in association with Th2-type cytokines or with Th1-type cytokines. IL-17 and IL-22 producing Th cells are not harmful for the conceptus, if they also produce IL-4. Another important protective mechanism is connected with the expansion and action of regulatory T cells, which play a major role in the induction of tolerance both in pregnant women and in tumour-bearing patients. Clonally-expanded Treg cells increase at the feto-maternal interface and in tumour-infiltrating regions. While in cancer patients, clonally-expanded Treg cells are present in peripheral blood, they are scarce in pregnancy blood, suggesting that fetal antigen-specific tolerance is restricted to the foeto-maternal interface. The significance of Treg cells in maintaining a normal materno-foetal interaction is underlined by the fact that miscarriage is characterized by a decreased number of total effector Treg cells, and the number of clonally-expanded effector Treg cells is markedly reduced in preeclampsia. In this review we present an overview of the above mechanisms, attempt to show how they are connected, how they operate during normal gestation and how their failure might lead to pregnancy pathologies.


Subject(s)
Cytokines/metabolism , Hormones/metabolism , Reproduction/physiology , Animals , Cytokines/genetics , Dietary Supplements , Dydrogesterone/administration & dosage , Female , Gene Expression Regulation , Hormones/genetics , Humans , Immunomodulation , Maternal-Fetal Exchange/immunology , Pregnancy , Progesterone/genetics , Progesterone/metabolism , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...