Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 172
Filter
1.
Int J Mol Sci ; 25(11)2024 May 29.
Article in English | MEDLINE | ID: mdl-38892139

ABSTRACT

Maternal obesity and over/undernutrition can have a long-lasting impact on offspring health during critical periods in the first 1000 days of life. Children born to mothers with obesity have reduced immune responses to stimuli which increase susceptibility to infections. Recently, maternal western-style diets (WSDs), high in fat and simple sugars, have been associated with skewing neonatal immune cell development, and recent evidence suggests that dysregulation of innate immunity in early life has long-term consequences on metabolic diseases and behavioral disorders in later life. Several factors contribute to abnormal innate immune tolerance or trained immunity, including changes in gut microbiota, metabolites, and epigenetic modifications. Critical knowledge gaps remain regarding the mechanisms whereby these factors impact fetal and postnatal immune cell development, especially in precursor stem cells in bone marrow and fetal liver. Components of the maternal microbiota that are transferred from mothers consuming a WSD to their offspring are understudied and identifying cause and effect on neonatal innate and adaptive immune development needs to be refined. Tools including single-cell RNA-sequencing, epigenetic analysis, and spatial location of specific immune cells in liver and bone marrow are critical for understanding immune system programming. Considering the vital role immune function plays in offspring health, it will be important to understand how maternal diets can control developmental programming of innate and adaptive immunity.


Subject(s)
Diet, Western , Fetal Development , Prenatal Exposure Delayed Effects , Humans , Female , Pregnancy , Diet, Western/adverse effects , Animals , Fetal Development/immunology , Prenatal Exposure Delayed Effects/immunology , Immune System/immunology , Immune System/metabolism , Epigenesis, Genetic , Gastrointestinal Microbiome/immunology , Immunity, Innate , Maternal Nutritional Physiological Phenomena , Fetus/immunology
3.
Nat Commun ; 15(1): 4711, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830841

ABSTRACT

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.


Subject(s)
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
4.
Front Immunol ; 13: 821542, 2022.
Article in English | MEDLINE | ID: mdl-35185911

ABSTRACT

CD49a+ natural killer (NK) cells play a critical role in promoting fetal development and maintaining immune tolerance at the maternal-fetal interface during the early stages of pregnancy. However, given their residency in human tissue, thorough studies and clinical applications are difficult to perform. It is still unclear as to how functional human CD49a+ NK cells can be induced to benefit pregnancy outcomes. In this study, we established three no-feeder cell induction systems to induce human CD49a+ NK cells from umbilical cord blood hematopoietic stem cells (HSCs), bone marrow HSCs, and peripheral blood NK cells in vitro. These induced NK cells (iNKs) from three cell induction systems display high levels of CD49a, CD9, CD39, CD151 expression, low levels of CD16 expression, and no obvious cytotoxic capability. They are phenotypically and functionally similar to decidual NK cells. Furthermore, these iNKs display a high expression of growth-promoting factors and proangiogenic factors and can promote fetal growth and improve uterine artery blood flow in a murine pregnancy model in vivo. This research demonstrates the ability of human-induced CD49a+ NK cells to promote fetal growth via three cell induction systems, which could eventually be used to treat patients experiencing adverse pregnancy outcomes.


Subject(s)
Fetal Development/immunology , Integrin alpha1/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Killer Cells, Natural/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Female , Gene Expression Regulation, Developmental , Humans , Integrin alpha1/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Mice , Pregnancy
5.
J Immunol ; 208(2): 221-226, 2022 01 15.
Article in English | MEDLINE | ID: mdl-35017211

ABSTRACT

Maternal infection during pregnancy is known to alter the development and function of offspring's immune system, leading to inappropriate immune responses to common childhood infections and immunizations. Although this is an expanding field, maternal parasitic infections remain understudied. Millions of women of reproductive age are currently at risk for parasitic infection, whereas many pregnant, chronically infected women are excluded from mass drug administration due partially to a lack of resources, as well as fear of unknown adverse fetal developmental outcomes. In areas endemic for multiple parasitic infections, such as sub-Saharan Africa, there are increased rates of morbidity and mortality for various infections during early childhood in comparison with nonendemic areas. Despite evidence supporting similar immunomodulatory effects between various parasite species, there is no clear mechanistic understanding of how maternal infection reprograms offspring immunity. This brief review will compare the effects of selected maternal parasitic infections on offspring immunity.


Subject(s)
Fetal Development/immunology , Helminthiasis/immunology , Malaria, Falciparum/immunology , Parasitic Diseases/transmission , Pregnancy Complications, Parasitic/epidemiology , Adult , Africa South of the Sahara/epidemiology , Animals , Female , Helminthiasis/parasitology , Helminthiasis/transmission , Helminths/pathogenicity , Humans , Infant, Newborn , Infectious Disease Transmission, Vertical , Malaria, Falciparum/parasitology , Malaria, Falciparum/transmission , Parasitic Diseases/epidemiology , Parasitic Diseases/immunology , Pregnancy , Soil/parasitology
6.
Front Immunol ; 12: 771054, 2021.
Article in English | MEDLINE | ID: mdl-34745147

ABSTRACT

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.


Subject(s)
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
7.
Immunology ; 164(4): 665-676, 2021 12.
Article in English | MEDLINE | ID: mdl-34287859

ABSTRACT

The immunology of pregnancy has been the focus of many studies to better understand how the mother is able to tolerate the presence of a semi-allogeneic fetus. Far from the initial view of pregnancy as a state of immunosuppression, successful fetal development from implantation to birth is now known to be under the control of an intricate balance of immune cells. The balance between pro-inflammatory functions used to promote embryo implantation and placental development and immunosuppressive activity to maintain maternal tolerance of the fetus is an immunological phenotype unique to pregnancy, which is dependent on the time of gestation. Neutrophils are one of a host of innate immune cells detected at the maternal-fetal interface, but very little is known of their function. In this review, we explore the emerging functions of neutrophils during pregnancy and their interactions with and regulation of T cells, a key adaptive immune cell population essential for the establishment of fetal-maternal tolerance.


Subject(s)
Adaptive Immunity , Fetal Development/immunology , Immune Tolerance , Immunity, Innate , Maternal-Fetal Exchange/immunology , Neutrophils/physiology , Animals , Cell Communication/immunology , Female , Humans , Immunomodulation , Phenotype , Placenta/immunology , Placenta/metabolism , Pregnancy , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
8.
Sci Immunol ; 6(58)2021 04 23.
Article in English | MEDLINE | ID: mdl-33893173

ABSTRACT

Accumulating evidence suggests that the mouse embryonic thymus produces distinct waves of innate effector γδ T cells. However, it is unclear whether this process occurs similarly in humans and whether it comprises a dedicated subset of innate-like type 3 effector γδ T cells. Here, we present a protocol for high-throughput sequencing of TRG and TRD pairs that comprise the clonal γδTCR. In combination with single-cell RNA sequencing, multiparameter flow cytometry, and TCR sequencing, we reveal a high heterogeneity of γδ T cells sorted from neonatal and adult blood that correlated with TCR usage. Immature γδ T cell clusters displayed mixed and diverse TCRs, but effector cell types segregated according to the expression of either highly expanded individual Vδ1+ TCRs or moderately expanded semi-invariant Vγ9Vδ2+ TCRs. The Vγ9Vδ2+ T cells shared expression of genes that mark innate-like T cells, including ZBTB16 (encoding PLZF), KLRB1, and KLRC1, but consisted of distinct clusters with unrelated Vγ9Vδ2+ TCR clones characterized either by TBX21, FCGR3A, and cytotoxicity-associated gene expression (type 1) or by CCR6, RORC, IL23R, and DPP4 expression (type 3). Effector γδ T cells with type 1 and type 3 innate T cell signatures were detected in a public dataset of early embryonic thymus organogenesis. Together, this study suggests that functionally distinct waves of human innate-like effector γδ T cells with semi-invariant Vγ9Vδ2+ TCR develop in the early fetal thymus and persist into adulthood.


Subject(s)
Fetal Blood/cytology , Fetal Development/immunology , Intraepithelial Lymphocytes/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/immunology , Adult , Cell Differentiation/immunology , Cells, Cultured , Female , Fetal Blood/immunology , Humans , Intraepithelial Lymphocytes/metabolism , Lymphocyte Activation , Male , RNA-Seq , Receptors, Antigen, T-Cell, gamma-delta/genetics , Single-Cell Analysis , T-Lymphocyte Subsets/metabolism
9.
Br J Cancer ; 124(12): 1897-1899, 2021 06.
Article in English | MEDLINE | ID: mdl-33767421
10.
Front Immunol ; 12: 637975, 2021.
Article in English | MEDLINE | ID: mdl-33679795

ABSTRACT

Human B-lymphopoiesis is a dynamic life-long process that starts in utero by around six post-conception weeks. A detailed understanding of human fetal B-lymphopoiesis and how it changes in postnatal life is vital for building a complete picture of normal B-lymphoid development through ontogeny, and its relevance in disease. B-cell acute lymphoblastic leukemia (B-ALL) is one of the most common cancers in children, with many of the leukemia-initiating events originating in utero. It is likely that the biology of B-ALL, including leukemia initiation, maintenance and progression depends on the developmental stage and type of B-lymphoid cell in which it originates. This is particularly important for early life leukemias, where specific characteristics of fetal B-cells might be key to determining how the disease behaves, including response to treatment. These cellular, molecular and/or epigenetic features are likely to change with age in a cell intrinsic and/or microenvironment directed manner. Most of our understanding of fetal B-lymphopoiesis has been based on murine data, but many recent studies have focussed on characterizing human fetal B-cell development, including functional and molecular assays at a single cell level. In this mini-review we will give a short overview of the recent advances in the understanding of human fetal B-lymphopoiesis, including its relevance to infant/childhood leukemia, and highlight future questions in the field.


Subject(s)
B-Lymphocytes/immunology , Fetal Development/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cells, B-Lymphoid/immunology , Carcinogenesis , Cell Differentiation , Humans , Lymphocyte Activation
11.
Nat Rev Endocrinol ; 17(4): 235-245, 2021 04.
Article in English | MEDLINE | ID: mdl-33526907

ABSTRACT

Intrauterine growth restriction (IUGR) is a common complication of pregnancy and increases the risk of the offspring developing type 2 diabetes mellitus (T2DM) later in life. Alterations in the immune system are implicated in the pathogenesis of IUGR-induced T2DM. The development of the fetal immune system is a delicate balance as it must remain tolerant of maternal antigens whilst also preparing for the post-birth environment. In addition, the fetal immune system is susceptible to an altered intrauterine milieu caused by maternal and placental inflammatory mediators or secondary to nutrient and oxygen deprivation. Pancreatic-resident macrophages populate the pancreas during fetal development, and their phenotype is dynamic through the neonatal period. Furthermore, macrophages in the islets are instrumental in islet development as they influence ß-cell proliferation and islet neogenesis. In addition, cytokines, derived from ß-cells and macrophages, are important to islet homeostasis in the fetus and adult and, when perturbed, can cause islet dysfunction. Several activated immune pathways have been identified in the islets of people who experienced IUGR, with alternations in the levels of IL-1ß and IL-4 as well as changes in TGFß signalling. Leptin levels are also altered. Immunomodulation has shown therapeutic benefit in T2DM and might be particularly useful in IUGR-induced T2DM.


Subject(s)
Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/immunology , Fetal Development/immunology , Fetal Growth Retardation/immunology , Animals , Humans , Immune System/immunology , Prenatal Injuries/immunology
12.
Int J Mol Sci ; 22(4)2021 Feb 09.
Article in English | MEDLINE | ID: mdl-33572203

ABSTRACT

Overweight and obesity during pregnancy have been associated with increased birth weight, childhood obesity, and noncommunicable diseases in the offspring, leading to a vicious transgenerational perpetuating of metabolic derangements. Key components in intrauterine developmental programming still remain to be identified. Obesity involves chronic low-grade systemic inflammation that, in addition to physiological adaptations to pregnancy, may potentially expand to the placental interface and lead to intrauterine derangements with a threshold effect. Animal models, where maternal inflammation is mimicked by single injections with lipopolysaccharide (LPS) resembling the obesity-induced immune profile, showed increased adiposity and impaired metabolic homeostasis in the offspring, similar to the phenotype observed after exposure to maternal obesity. Cytokine levels might be specifically important for the metabolic imprinting, as cytokines are transferable from maternal to fetal circulation and have the capability to modulate placental nutrient transfer. Maternal inflammation may induce metabolic reprogramming at several levels, starting from the periconceptional period with effects on the oocyte going through early stages of embryonic and placental development. Given the potential to reduce inflammation through inexpensive, widely available therapies, examinations of the impact of chronic inflammation on reproductive and pregnancy outcomes, as well as preventive interventions, are now needed.


Subject(s)
Child Development/physiology , Fetal Development/immunology , Obesity, Maternal/immunology , Pediatric Obesity/immunology , Prenatal Exposure Delayed Effects/immunology , Animals , Child , Disease Models, Animal , Female , Humans , Inflammation/immunology , Inflammation/metabolism , Maternal Nutritional Physiological Phenomena/immunology , Maternal-Fetal Exchange/immunology , Metabolic Networks and Pathways/immunology , Obesity, Maternal/complications , Obesity, Maternal/metabolism , Obesity, Maternal/therapy , Pediatric Obesity/metabolism , Pediatric Obesity/prevention & control , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/prevention & control
13.
Toxicol Lett ; 339: 1-11, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33301788

ABSTRACT

Despite numerous reports that ambient particulate matter is a key determinant for human health, toxicity data produced based on physicochemical properties of particulate matters is very lack, suggesting lack of scientific evidence for regulation. In this study, we sampled inhalable particulate matters (PM10) in northern Seoul, Korea. PM10 showed atypical- and fiber-type particles with the average size and the surface charge of 1,598.1 ± 128.7 nm and -27.5 ± 2.8, respectively, and various toxic elements were detected in the water extract. On day 90 after the first pulmonary exposure, total cell number dose-dependently increased in the lungs of both sexes of mice. PM10 induced Th1-dominant immune response with pathological changes in both sexes of mice. Meanwhile, composition of total cells and expression of proteins which functions in cell-to-cell communication showed different trends between sexes. Following, male and female mice were mated to identify effects of PM10 to the next generation. PM10 remained in the lung of dams until day 21 after birth, and the levels of IgA and IgE increased in the blood of dams exposed to the maximum dose compared to control. In addition, the interval between births of fetuses, the number of offspring, the neonatal survival rate (day 4 after birth) and the sex ratio seemed to be affected at the maximum dose, and particularly, all offspring from one dam were stillborn. In addition, expression of HIF-1α protein increased in the lung tissue of dams exposed to PM10, and level of hypoxia-related proteins was notably enhanced in PM10-exposed bronchial epithelial cells compared to control. Taken together, we suggest that inhaled PM10 may induce Th1-shifting immune response in the lung, and that it may affect reproduction (fetus development) by causing lung hypoxia. Additionally, we propose that further study is needed to identify particle-size-dependent effects on development of the next generation.


Subject(s)
Air Pollutants/toxicity , Fetal Development/drug effects , Fetal Development/immunology , Immunity/drug effects , Lung/drug effects , Lung/immunology , Particulate Matter/toxicity , Air Pollutants/immunology , Animals , Female , Humans , Male , Mice , Models, Animal , Particulate Matter/immunology , Republic of Korea
14.
Medicine (Baltimore) ; 99(46): e22722, 2020 Nov 13.
Article in English | MEDLINE | ID: mdl-33181648

ABSTRACT

In general terms, fetal growth restriction (FGR) is considered the impossibility of achieving the genetically determined potential size. In the vast majority of cases, it is related to uteroplacental insufficiency. Although its origin remains unknown and causes are only known in 30% of cases, it is believed to be related to an interaction of environmental and genetic factors with either a fetal or maternal origin. One hypothesis is that alterations in the gastrointestinal microbiota composition, and thus alteration in the immune response, could play a role in FGR development. We performed an observational, prospective study in a subpopulation affected with FGR to elucidate the implications of this microbiota on the FGR condition.A total of 63 fetuses with FGR diagnosed in the third trimester as defined by the Delphi consensus, and 63 fetuses with fetal growth appropriate for gestational age will be recruited. Obstetric and nutritional information will be registered by means of specific questionnaires. We will collect maternal fecal samples between 30 to 36 weeks, intrapartum samples (maternal feces, maternal and cord blood) and postpartum samples (meconium and new-born feces at 6 weeks of life). Samples will be analyzed in the Department of Biochemistry and Molecular Biology II, Nutrition and Food Technology Institute of the University of Granada (UGR), for the determination of the gastrointestinal microbiota composition and its relationship with inflammatory biomarkers.This study will contribute to a better understanding of the influence of gastrointestinal microbiota and related inflammatory biomarkers in the development of FGR.Trial registration: NCT04047966. Registered August 7, 2019, during the recruitment stage. Retrospectively registered. Ongoing research.


Subject(s)
Fetal Growth Retardation/immunology , Fetus/immunology , Microbiota/immunology , Pregnant Women , Adult , Biomarkers/analysis , Case-Control Studies , Cordocentesis/methods , Delphi Technique , Female , Fetal Development/immunology , Fetal Development/physiology , Fetus/physiopathology , Gestational Age , Humans , Microbiota/physiology , Pregnancy , Prospective Studies , Spain
15.
Sci Rep ; 10(1): 20288, 2020 11 20.
Article in English | MEDLINE | ID: mdl-33219314

ABSTRACT

Maternal stress during pregnancy is widespread and is associated with poor offspring outcomes, including long-term mental health issues. Prenatal stress-induced fetal neuroinflammation is thought to underlie aberrant neurodevelopment and to derive from a disruption in intrauterine immune homeostasis, though the exact origins are incompletely defined. We aimed to identify divergent immune and microbial metagenome profiles of stressed gestating mice that may trigger detrimental inflammatory signaling at the maternal-fetal interface. In response to stress, maternal glucocorticoid circuit activation corresponded with indicators of systemic immunosuppression. At the maternal-fetal interface, density of placental mononuclear leukocytes decreased with stress, yet maternal whole blood leukocyte analysis indicated monocytosis and classical M1 phenotypic shifts. Genome-resolved microbial metagenomic analyses revealed reductions in genes, microbial strains, and metabolic pathways in stressed dams that are primarily associated with pro-inflammatory function. In particular, disrupted Parasutterella excrementihominis appears to be integral to inflammatory and metabolic dysregulation during prenatal stress. Overall, these perturbations in maternal immunological and microbial regulation during pregnancy may displace immune equilibrium at the maternal-fetal interface. Notably, the absence of and reduction in overt maternal inflammation during stress indicates that the signaling patterns driving fetal outcomes in this context are more nuanced and complex than originally anticipated.


Subject(s)
Brain/embryology , Fetal Development/immunology , Gastrointestinal Microbiome/immunology , Pregnancy Complications/immunology , Stress, Psychological/immunology , Animals , Brain/immunology , Burkholderiales/genetics , Burkholderiales/immunology , Disease Models, Animal , Female , Gastrointestinal Microbiome/genetics , Glucocorticoids/metabolism , Humans , Leukocytes, Mononuclear/immunology , Maternal-Fetal Exchange/immunology , Mental Health , Metagenomics , Mice , Neuroimmunomodulation/immunology , Placenta/cytology , Placenta/immunology , Pregnancy , Pregnancy Complications/metabolism , Pregnancy Complications/psychology , Prenatal Exposure Delayed Effects/immunology , Stress, Psychological/metabolism , Stress, Psychological/psychology
16.
J Reprod Immunol ; 142: 103185, 2020 11.
Article in English | MEDLINE | ID: mdl-32853845

ABSTRACT

Preeclampsia (PE) and human immunodeficiency virus (HIV) have been linked with marked increases in maternal stress, resulting in a significant change in placental function ranging from alterations in placental structure to the precise and delicate transformations in placental gene expression. Such changes may lead to altered transport of essential signals to the fetus, which can have long-term impacts on offspring health and consequently affect fetal neurodevelopment. Therefore, this work investigated the role of placental 11ß-hydroxysteroid dehydrogenase types 2 (11ß-HSD2) in HIV associated preeclampsia. The placenta were obtained from 76 pregnant women, which were stratified based on pregnancy type and HIV status into; Normotensive HIV negative, normotensive HIV positive, PE HIV negative and PE HIV positive. The placental tissue was processed for immunocytochemistry and stained with rabbit polyclonal to 11ß-HSD2 Our results showed significant downregulation in the placental expression of 11ß-HSD2 in both the conducting and exchange villi of PE and HIV-positive patients when compared with Normotensive and HIV-negative individuals, respectively. Our results provide inferential evidence for comorbidity of PE and HIV in the downregulation of placental 11ß-HSD2 enzyme function, which mediates the programmed outcomes of an adverse maternal environment during pregnancy and long-term impacts on offspring health and consequently affects fetal neurodevelopment.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Fetal Development/immunology , HIV Infections/complications , Neurodevelopmental Disorders/immunology , Pre-Eclampsia/immunology , 11-beta-Hydroxysteroid Dehydrogenase Type 2/analysis , Adolescent , Adult , Brain/embryology , Case-Control Studies , Down-Regulation , Female , Glucocorticoids/immunology , Glucocorticoids/metabolism , HIV Infections/diagnosis , HIV Infections/immunology , HIV Infections/virology , HIV-1/isolation & purification , Humans , Maternal Age , Neurodevelopmental Disorders/pathology , Placenta/enzymology , Placenta/immunology , Placenta/pathology , Pre-Eclampsia/pathology , Pre-Eclampsia/virology , Pregnancy , Retrospective Studies , South Africa , Young Adult
17.
Front Immunol ; 11: 1672, 2020.
Article in English | MEDLINE | ID: mdl-32733490

ABSTRACT

Pregnancy comprises a unique immunological condition, to allow fetal development and to protect the host from pathogenic infections. Viral infections during pregnancy can disrupt immunological tolerance and may generate deleterious effects on the fetus. Despite these possible links between pregnancy and infection-induced morbidity, it is unclear how pregnancy interferes with maternal response to some viral pathogens. In this context, the novel coronavirus (SARS-CoV-2) can induce the coronavirus diseases-2019 (COVID-19) in pregnant women. The potential risk of vertical transmission is unclear, babies born from COVID-19-positive mothers seems to have no serious clinical symptoms, the possible mechanisms are discussed, which highlights that checking the children's outcome and more research is warranted. In this review, we investigate the reports concerning viral infections and COVID-19 during pregnancy, to establish a correlation and possible implications of COVID-19 during pregnancy and neonatal's health.


Subject(s)
Betacoronavirus , Coronavirus Infections/immunology , Coronavirus Infections/transmission , Pneumonia, Viral/immunology , Pneumonia, Viral/transmission , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/virology , COVID-19 , Child, Preschool , Coronavirus Infections/blood , Coronavirus Infections/virology , Cytokines/blood , Female , Fetal Development/immunology , Humans , Infant , Infant, Newborn , Infectious Disease Transmission, Vertical , Mothers , Pandemics , Pneumonia, Viral/blood , Pneumonia, Viral/virology , Pregnancy , Pregnancy Complications, Infectious/blood , SARS-CoV-2
18.
Front Immunol ; 11: 1681, 2020.
Article in English | MEDLINE | ID: mdl-32849586

ABSTRACT

The complement cascade was identified over 100 years ago, yet investigation of its role in pregnancy remains an area of intense research. Complement inhibitors at the maternal-fetal interface prevent inappropriate complement activation to protect the fetus. However, this versatile proteolytic cascade also favorably influences numerous stages of pregnancy, including implantation, fetal development, and labor. Inappropriate complement activation in pregnancy can have adverse lifelong sequelae for both mother and child. This review summarizes the current understanding of complement activation during all stages of pregnancy. In addition, consequences of complement dysregulation during adverse pregnancy outcomes from miscarriage, preeclampsia, and pre-term birth are examined. Finally, future research directions into complement activation during pregnancy are considered.


Subject(s)
Complement Activation/immunology , Embryo Implantation/immunology , Parturition/immunology , Pregnancy Complications/immunology , Pregnancy/immunology , Complement System Proteins/immunology , Female , Fetal Development/immunology , Humans
19.
Front Immunol ; 11: 1035, 2020.
Article in English | MEDLINE | ID: mdl-32582165

ABSTRACT

Preterm infants born before 32 weeks gestational age (GA) have high rates of late onset sepsis (LOS) and necrotizing enterocolitis (NEC) despite recent improvements in infection control and nutrition. Breast milk has a clear protective effect against both these outcomes likely due to multiple mechanisms which are not fully understood but may involve effects on both the infant's immune system and the developing gut microbiota. Congregating at the interface between the mucosal barrier and the microbiota, innate and adaptive T lymphocytes (T cells) participate in this interaction but few studies have explored their development after preterm delivery. We conducted a literature review of T cell development that focuses on fetal development, postnatal maturation and the influence of milk diet. The majority of circulating T cells in the preterm infant display a naïve phenotype but are still able to initiate functional responses similar to those seen in term infants. T cells from preterm infants display a skew toward a T-helper 2(Th2) phenotype and have an increased population of regulatory cells (Tregs). There are significant gaps in knowledge in this area, particularly in regards to innate-like T cells, but work is emerging: transcriptomics and mass cytometry are currently being used to map out T cell development, whilst microbiomic approaches may help improve understanding of events at mucosal surfaces. A rapid rise in organoid models will allow robust exploration of host-microbe interactions and may support the development of interventions that modulate T-cell responses for improved infant health.


Subject(s)
Infant, Premature/immunology , Milk, Human/immunology , T-Lymphocyte Subsets/immunology , Adaptive Immunity , Disease Susceptibility , Enterocolitis, Necrotizing/immunology , Female , Fetal Development/immunology , Gastrointestinal Microbiome/immunology , Host Microbial Interactions/immunology , Humans , Immunity, Innate , Immunity, Mucosal , Infant Formula , Infant Nutritional Physiological Phenomena/immunology , Infant, Newborn , Milk, Human/cytology , Models, Immunological , Pregnancy , Sepsis/immunology
20.
Science ; 368(6491): 608-612, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32381717

ABSTRACT

Viral infections during pregnancy can have devastating consequences on pregnancy outcomes, fetal development, and maternal health. In this review, we examine fetal and maternal immune defense mechanisms that mediate resistance against viral infections and discuss the range of syndromes that ensue when such mechanisms fail, from fetal developmental defects to establishment of chronic infection. Further, we highlight the role of maternal immune activation, or uncontrolled inflammation triggered by viral infections during pregnancy, and its potential downstream pathological effects, including tissue damage and fetal demise. Insights into the respective contributions of direct viral toxicity versus fetal and maternal immune responses that underlie the pathogenesis of congenital disease will guide future treatment strategies.


Subject(s)
Fetal Diseases/virology , Fetus/abnormalities , Maternal-Fetal Exchange/immunology , Pregnancy Complications, Infectious/virology , Virus Diseases/immunology , Female , Fetal Development/immunology , Fetal Diseases/immunology , Fetus/immunology , Fetus/virology , Humans , Immunity, Innate , Pregnancy , Pregnancy Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...