Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.401
Filter
Add more filters

Publication year range
1.
Annu Rev Cell Dev Biol ; 36: 441-468, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32722920

ABSTRACT

Fetal neurodevelopment in utero is profoundly shaped by both systemic maternal immunity and local processes at the maternal-fetal interface. Immune pathways are a critical participant in the normal physiology of pregnancy and perturbations of maternal immunity due to infections during this period have been increasingly linked to a diverse array of poor neurological outcomes, including diseases that manifest much later in postnatal life. While experimental models of maternal immune activation (MIA) have provided groundbreaking characterizations of the maternal pathways underlying pathogenesis, less commonly examined are the immune factors that serve pathogen-independent developmental functions in the embryo and fetus. In this review, we explore what is known about the in vivo role of immune factors in fetal neurodevelopment during normal pregnancy and provide an overview of how MIA perturbs the proper orchestration of this sequence of events. Finally, we discuss how the dysregulation of immune factors may contribute to the manifestation of a variety of neurological disorders.


Subject(s)
Fetus/embryology , Fetus/immunology , Immunologic Factors/metabolism , Nervous System/embryology , Nervous System/immunology , Animals , Cytokines/metabolism , Female , Humans , Maternal-Fetal Exchange/immunology , Models, Biological , Pregnancy
2.
Nat Immunol ; 20(3): 301-312, 2019 03.
Article in English | MEDLINE | ID: mdl-30664737

ABSTRACT

The fetus is thought to be protected from exposure to foreign antigens, yet CD45RO+ T cells reside in the fetal intestine. Here we combined functional assays with mass cytometry, single-cell RNA sequencing and high-throughput T cell antigen receptor (TCR) sequencing to characterize the CD4+ T cell compartment in the human fetal intestine. We identified 22 CD4+ T cell clusters, including naive-like, regulatory-like and memory-like subpopulations, which were confirmed and further characterized at the transcriptional level. Memory-like CD4+ T cells had high expression of Ki-67, indicative of cell division, and CD5, a surrogate marker of TCR avidity, and produced the cytokines IFN-γ and IL-2. Pathway analysis revealed a differentiation trajectory associated with cellular activation and proinflammatory effector functions, and TCR repertoire analysis indicated clonal expansions, distinct repertoire characteristics and interconnections between subpopulations of memory-like CD4+ T cells. Imaging mass cytometry indicated that memory-like CD4+ T cells colocalized with antigen-presenting cells. Collectively, these results provide evidence for the generation of memory-like CD4+ T cells in the human fetal intestine that is consistent with exposure to foreign antigens.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Fetus/immunology , Immunologic Memory/immunology , Intestines/immunology , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD5 Antigens/genetics , CD5 Antigens/immunology , CD5 Antigens/metabolism , Cells, Cultured , Fetus/cytology , Fetus/metabolism , Flow Cytometry , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental/immunology , High-Throughput Nucleotide Sequencing , Humans , Immunologic Memory/genetics , Immunophenotyping , Intestines/cytology , Intestines/embryology , Ki-67 Antigen/genetics , Ki-67 Antigen/immunology , Ki-67 Antigen/metabolism
3.
Cell ; 162(3): 467-9, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26232215

ABSTRACT

Mendelian genetics presumes inheritance of fitness through DNA. Kinder et al. find that maternal microchimerism induces stable immune tolerance to non-inherited maternal antigens in offspring. Female offspring that share these antigens with their mate experience reduced fetal wasting, establishing a role for vertical transmission of non-genetic information in reproductive fitness.


Subject(s)
Fetus/immunology , Genetic Fitness , Immune Tolerance , Mammals/physiology , Pregnancy/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Female , Humans , Male
4.
Cell ; 162(3): 505-15, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26213383

ABSTRACT

Exposure to maternal tissue during in utero development imprints tolerance to immunologically foreign non-inherited maternal antigens (NIMA) that persists into adulthood. The biological advantage of this tolerance, conserved across mammalian species, remains unclear. Here, we show maternal cells that establish microchimerism in female offspring during development promote systemic accumulation of immune suppressive regulatory T cells (Tregs) with NIMA specificity. NIMA-specific Tregs expand during pregnancies sired by males expressing alloantigens with overlapping NIMA specificity, thereby averting fetal wastage triggered by prenatal infection and non-infectious disruptions of fetal tolerance. Therefore, exposure to NIMA selectively enhances reproductive success in second-generation females carrying embryos with overlapping paternally inherited antigens. These findings demonstrate that genetic fitness, canonically thought to be restricted to Mendelian inheritance, is enhanced in female placental mammals through vertically transferred maternal cells that promote conservation of NIMA and enforce cross-generational reproductive benefits.


Subject(s)
Fetus/immunology , Genetic Fitness , Immune Tolerance , Mammals/physiology , Pregnancy/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens/immunology , Chimerism , Female , Humans , Male , Mammals/immunology , Mice , Placenta/immunology
5.
Nat Immunol ; 18(11): 1261-1269, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28945244

ABSTRACT

The Zika virus (ZIKV) epidemic has resulted in congenital abnormalities in fetuses and neonates. Although some cross-reactive dengue virus (DENV)-specific antibodies can enhance ZIKV infection in mice, those recognizing the DENV E-dimer epitope (EDE) can neutralize ZIKV infection in cell culture. We evaluated the therapeutic activity of human monoclonal antibodies to DENV EDE for their ability to control ZIKV infection in the brains, testes, placentas, and fetuses of mice. A single dose of the EDE1-B10 antibody given 3 d after ZIKV infection protected against lethality, reduced ZIKV levels in brains and testes, and preserved sperm counts. In pregnant mice, wild-type or engineered LALA variants of EDE1-B10, which cannot engage Fcg receptors, diminished ZIKV burden in maternal and fetal tissues, and protected against fetal demise. Because neutralizing antibodies to EDE have therapeutic potential against ZIKV, in addition to their established inhibitory effects against DENV, it may be possible to develop therapies that control disease caused by both viruses.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Dengue Virus/immunology , Epitopes/immunology , Viral Envelope Proteins/immunology , Zika Virus Infection/immunology , Animals , Brain/immunology , Brain/virology , Chlorocebus aethiops , Cross Reactions/immunology , Dengue Virus/classification , Dengue Virus/metabolism , Female , Fetus/immunology , Fetus/virology , Host-Pathogen Interactions/immunology , Humans , Male , Mice , Neutralization Tests , Pregnancy , Protein Multimerization/immunology , Testis/immunology , Testis/virology , Vero Cells , Viral Envelope Proteins/chemistry , Viral Load/immunology , Zika Virus/immunology , Zika Virus/physiology , Zika Virus Infection/virology
6.
Nature ; 613(7945): 639-649, 2023 01.
Article in English | MEDLINE | ID: mdl-36697862

ABSTRACT

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.


Subject(s)
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
7.
Immunity ; 50(2): 462-476.e8, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30770246

ABSTRACT

Although the fetal immune system is considered tolerogenic, preterm infants can suffer from severe intestinal inflammation, including necrotizing enterocolitis (NEC). Here, we demonstrate that human fetal intestines predominantly contain tumor necrosis factor-α (TNF-α)+CD4+CD69+ T effector memory (Tem) cells. Single-cell RNA sequencing of fetal intestinal CD4+ T cells showed a T helper 1 phenotype and expression of genes mediating epithelial growth and cell cycling. Organoid co-cultures revealed a dose-dependent, TNF-α-mediated effect of fetal intestinal CD4+ T cells on intestinal stem cell (ISC) development, in which low T cell numbers supported epithelial development, whereas high numbers abrogated ISC proliferation. CD4+ Tem cell frequencies were higher in inflamed intestines from preterm infants with NEC than in healthy infant intestines and showed enhanced TNF signaling. These findings reveal a distinct population of TNF-α-producing CD4+ T cells that promote mucosal development in fetal intestines but can also mediate inflammation upon preterm birth.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Fetus/immunology , Immunologic Memory/immunology , Intestines/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Epithelial Cells/cytology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Female , Fetus/metabolism , Humans , Infant, Newborn , Intestinal Mucosa/embryology , Intestinal Mucosa/growth & development , Intestinal Mucosa/immunology , Intestines/embryology , Intestines/growth & development , Mice, Inbred C57BL , Pregnancy , Stem Cells/cytology , Stem Cells/immunology , Stem Cells/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Nat Immunol ; 16(2): 153-60, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25501629

ABSTRACT

Fetal lymphoid tissue inducer (LTi) cells are required for lymph node and Peyer's patch (PP) organogenesis, but where these specialized group 3 innate lymphoid cells (ILC3s) develop remains unclear. Here, we identify extrahepatic arginase-1(+) Id2(+) fetal ILC precursors that express a transitional developmental phenotype (ftILCPs) and differentiate into ILC1s, ILC2s and ILC3s in vitro. These cells populate the intestine by embryonic day (E) 13.5 and, before PP organogenesis (E14.5-15), are broadly dispersed in the proximal gut, correlating with regions where PPs first develop. At E16.5, after PP development begins, ftILCPs accumulate at PP anlagen in a lymphotoxin-α-dependent manner. Thus, ftILCPs reside in the intestine during PP development, where they aggregate at PP anlagen after stromal cell activation and become a localized source of ILC populations.


Subject(s)
Cell Differentiation , Immunity, Innate , Intestine, Small/cytology , Intestine, Small/embryology , Lymphoid Tissue/cytology , Lymphoid Tissue/embryology , Animals , Arginase/metabolism , Cells, Cultured , Fetus/cytology , Fetus/immunology , Flow Cytometry , Immunohistochemistry , Intestine, Small/immunology , Lymphoid Tissue/immunology , Mice
9.
Cell ; 150(1): 29-38, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22770213

ABSTRACT

Regulatory T (Treg) cells, whose differentiation and function are controlled by X chromosome-encoded transcription factor Foxp3, are generated in the thymus (tTreg) and extrathymically (peripheral, pTreg), and their deficiency results in fatal autoimmunity. Here, we demonstrate that a Foxp3 enhancer, conserved noncoding sequence 1 (CNS1), essential for pTreg but dispensable for tTreg cell generation, is present only in placental mammals. CNS1 is largely composed of mammalian-wide interspersed repeats (MIR) that have undergone retrotransposition during early mammalian radiation. During pregnancy, pTreg cells specific to a model paternal alloantigen were generated in a CNS1-dependent manner and accumulated in the placenta. Furthermore, when mated with allogeneic, but not syngeneic, males, CNS1-deficient females showed increased fetal resorption accompanied by increased immune cell infiltration and defective remodeling of spiral arteries. Our results suggest that, during evolution, a CNS1-dependent mechanism of extrathymic differentiation of Treg cells emerged in placental animals to enforce maternal-fetal tolerance.


Subject(s)
Immune Tolerance , Mammals/immunology , Placenta/cytology , Placenta/immunology , Pregnancy/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Enhancer Elements, Genetic , Female , Fetus/immunology , Forkhead Transcription Factors/genetics , Humans , Male , Mammals/genetics , Mice , Opossums
10.
Nature ; 589(7842): 442-447, 2021 01.
Article in English | MEDLINE | ID: mdl-33361811

ABSTRACT

Successful pregnancies rely on adaptations within the mother1, including marked changes within the immune system2. It has long been known that the thymus, the central lymphoid organ, changes markedly during pregnancy3. However, the molecular basis and importance of this process remain largely obscure. Here we show that the osteoclast differentiation receptor RANK4,5 couples female sex hormones to the rewiring of the thymus during pregnancy. Genetic deletion of Rank (also known as Tnfrsf11a) in thymic epithelial cells results in impaired thymic involution and blunted expansion of natural regulatory T (Treg) cells in pregnant female mice. Sex hormones, in particular progesterone, drive the development of thymic Treg cells through RANK in a manner that depends on AIRE+ medullary thymic epithelial cells. The depletion of Rank in the mouse thymic epithelium results in reduced accumulation of natural Treg cells in the placenta, and an increase in the number of miscarriages. Thymic deletion of Rank also results in impaired accumulation of Treg cells in visceral adipose tissue, and is associated with enlarged adipocyte size, tissue inflammation, enhanced maternal glucose intolerance, fetal macrosomia, and a long-lasting transgenerational alteration in glucose homeostasis, which are all key hallmarks of gestational diabetes. Transplantation of Treg cells rescued fetal loss, maternal glucose intolerance and fetal macrosomia. In human pregnancies, we found that gestational diabetes also correlates with a reduced number of Treg cells in the placenta. Our findings show that RANK promotes the hormone-mediated development of thymic Treg cells during pregnancy, and expand the functional role of maternal Treg cells to the development of gestational diabetes and the transgenerational metabolic rewiring of glucose homeostasis.


Subject(s)
Diabetes, Gestational/immunology , Fetal Death/etiology , Receptor Activator of Nuclear Factor-kappa B/metabolism , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Adipocytes/pathology , Animals , Cell Proliferation , Diabetes, Gestational/etiology , Diabetes, Gestational/metabolism , Diabetes, Gestational/pathology , Epithelial Cells/immunology , Female , Fetus/immunology , Fetus/metabolism , Fetus/pathology , Glucose/metabolism , Glucose Intolerance/genetics , Humans , Intra-Abdominal Fat/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Placenta/immunology , Placenta/pathology , Pregnancy , Receptor Activator of Nuclear Factor-kappa B/deficiency , Receptor Activator of Nuclear Factor-kappa B/genetics , T-Lymphocytes, Regulatory/cytology , Thymus Gland/cytology , Transcription Factors/metabolism , AIRE Protein
11.
Development ; 149(8)2022 04 15.
Article in English | MEDLINE | ID: mdl-34604909

ABSTRACT

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.


Subject(s)
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
12.
Development ; 149(8)2022 04 15.
Article in English | MEDLINE | ID: mdl-35050308

ABSTRACT

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.


Subject(s)
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.
Immunity ; 45(3): 527-539, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27566938

ABSTRACT

B cell development is often depicted as a linear process initiating in the fetus and continuing postnatally. Using a PU.1 hypomorphic mouse model, we found that B-1 and B-2 lymphopoiesis occurred in distinct fetal and adult waves differentially dependent on the Sfpi1 14 kB upstream regulatory element. The initial wave of fetal B-1 development was absent in PU.1 hypomorphic mice, while subsequent fetal and adult waves emerged. In contrast, B-2 lymphopoiesis occurred in distinct fetal and adult waves. Whole-transcriptome profiling of fetal and adult B cell progenitors supported the existence of three waves of B-1 and two waves of B-2 development and revealed that the network of transcription factors governing B lineage specification and commitment was highly divergent between B-1 and B-2 progenitors. These findings support the view that the B-1 and B-2 lineages are distinct and provide a genetic basis for layering of immune system development.


Subject(s)
B-Lymphocyte Subsets/immunology , Gene Regulatory Networks/immunology , Lymphopoiesis/immunology , Animals , Cell Lineage/immunology , Fetus/immunology , Gene Expression Profiling/methods , Mice , Precursor Cells, B-Lymphoid/immunology , Transcription Factors/immunology
14.
Proc Natl Acad Sci U S A ; 119(49): e2212548119, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36442114

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes , Fetus , Immunity , Cell Differentiation , Educational Status , Epigenomics , Fetus/immunology , Fetus/microbiology
15.
Proc Natl Acad Sci U S A ; 119(35): e2123267119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35994660

ABSTRACT

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.


Subject(s)
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
16.
EMBO J ; 39(13): e104159, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32627520

ABSTRACT

γδ T cells with distinct properties develop in the embryonic and adult thymus and have been identified as critical players in a broad range of infections, antitumor surveillance, autoimmune diseases, and tissue homeostasis. Despite their potential value for immunotherapy, differentiation of γδ T cells in the thymus is incompletely understood. Here, we establish a high-resolution map of γδ T-cell differentiation from the fetal and adult thymus using single-cell RNA sequencing. We reveal novel sub-types of immature and mature γδ T cells and identify an unpolarized thymic population which is expanded in the blood and lymph nodes. Our detailed comparative analysis reveals remarkable similarities between the gene networks active during fetal and adult γδ T-cell differentiation. By performing a combined single-cell analysis of Sox13, Maf, and Rorc knockout mice, we demonstrate sequential activation of these factors during IL-17-producing γδ T-cell (γδT17) differentiation. These findings substantially expand our understanding of γδ T-cell ontogeny in fetal and adult life. Our experimental and computational strategy provides a blueprint for comparing immune cell differentiation across developmental stages.


Subject(s)
Cell Differentiation/immunology , Fetus/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Animals , Autoantigens/genetics , Autoantigens/immunology , Cell Differentiation/genetics , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , T-Lymphocytes/cytology
17.
Nat Immunol ; 13(5): 511-8, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22473038

ABSTRACT

Innate γδ T cells function in the early phase of immune responses. Although innate γδ T cells have often been studied as one homogenous population, they can be functionally classified into effector subsets on the basis of the production of signature cytokines, analogous to adaptive helper T cell subsets. However, unlike the function of adaptive T cells, γδ effector T cell function correlates with genomically encoded T cell antigen receptor (TCR) chains, which suggests that clonal TCR selection is not the main determinant of the differentiation of γδ effector cells. A high-resolution transcriptome analysis of all emergent γδ thymocyte subsets segregated on the basis of use of the TCR γ-chain or δ-chain indicated the existence of three separate subtypes of γδ effector cells in the thymus. The immature γδ subsets were distinguished by unique transcription-factor modules that program effector function.


Subject(s)
Cell Differentiation/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , Transcriptome/immunology , Age Factors , Animals , CD24 Antigen/immunology , CD24 Antigen/metabolism , Cell Differentiation/genetics , Cell Lineage/immunology , Fetus/cytology , Fetus/immunology , Flow Cytometry , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Models, Immunological , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/metabolism , Principal Component Analysis , Receptors, Antigen, T-Cell, gamma-delta/classification , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism , Transcriptome/genetics
18.
Brain Behav Immun ; 120: 391-402, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38897330

ABSTRACT

Rodent models of maternal immune activation (MIA) are increasingly used as experimental tools in preclinical research of immune-mediated neurodevelopmental disorders and mental illnesses. Using a viral-like MIA model that is based on prenatal poly(I:C) exposure in mice, we have recently identified the existence of subgroups of MIA-exposed offspring that show dissociable behavioral, transcriptional, brain network and inflammatory profiles even under conditions of genetic homogeneity and identical MIA. Here, we tested the hypothesis that the intrauterine positions of fetuses, which are known to shape individual variability in litter-bearing mammals through variations in fetal hormone exposure, may contribute to the variable outcomes of MIA in mice. MIA was induced by maternal administration of poly(I:C) on gestation day 12 in C57BL/6N mice. Determining intrauterine positions using delivery by Cesarean section (C-section), we found that MIA-exposed offspring developing between female fetuses only (0M-MIA offspring) displayed significant deficits in sociability and sensorimotor gating at adult age, whereas MIA-exposed offspring developing between one or two males in utero (1/2M-MIA offspring) did not show the same deficits. These intrauterine position effects similarly emerged in male and female offspring. Furthermore, while MIA elevated fetal brain levels of pro- and anti-inflammatory cytokines independently of the precise intrauterine position and sex of adjacent fetuses during the acute phase, fetal brain levels of TNF-α remained elevated in 0M-MIA but not 1/2M-MIA offspring until the post-acute phase in late gestation. As expected, 1/2M offspring generally showed higher testosterone levels in the fetal brain during late gestation as compared to 0M offspring, confirming the transfer of testosterone from male fetuses to adjacent male or female fetuses. Taken together, our findings identify a novel source of within-litter variability contributing to heterogeneous outcomes of short- and long-term effects in a mouse model of MIA. In broader context, our findings highlight that individual differences in fetal exposure to hormonal and inflammatory signals may be a perinatal factor that shapes risk and resilience to MIA.


Subject(s)
Brain , Disease Models, Animal , Mice, Inbred C57BL , Poly I-C , Prenatal Exposure Delayed Effects , Animals , Female , Pregnancy , Mice , Male , Poly I-C/pharmacology , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/metabolism , Brain/metabolism , Brain/immunology , Cytokines/metabolism , Neurodevelopmental Disorders/immunology , Behavior, Animal/physiology , Fetus/immunology , Fetus/metabolism , Uterus/metabolism , Uterus/immunology
19.
Nature ; 563(7731): 347-353, 2018 11.
Article in English | MEDLINE | ID: mdl-30429548

ABSTRACT

During early human pregnancy the uterine mucosa transforms into the decidua, into which the fetal placenta implants and where placental trophoblast cells intermingle and communicate with maternal cells. Trophoblast-decidual interactions underlie common diseases of pregnancy, including pre-eclampsia and stillbirth. Here we profile the transcriptomes of about 70,000 single cells from first-trimester placentas with matched maternal blood and decidual cells. The cellular composition of human decidua reveals subsets of perivascular and stromal cells that are located in distinct decidual layers. There are three major subsets of decidual natural killer cells that have distinctive immunomodulatory and chemokine profiles. We develop a repository of ligand-receptor complexes and a statistical tool to predict the cell-type specificity of cell-cell communication via these molecular interactions. Our data identify many regulatory interactions that prevent harmful innate or adaptive immune responses in this environment. Our single-cell atlas of the maternal-fetal interface reveals the cellular organization of the decidua and placenta, and the interactions that are critical for placentation and reproductive success.


Subject(s)
Cell Communication , Fetus/cytology , Histocompatibility, Maternal-Fetal/immunology , Placenta/cytology , Placenta/metabolism , Pregnancy/immunology , Single-Cell Analysis , Cell Communication/immunology , Cell Differentiation/genetics , Decidua/cytology , Decidua/immunology , Decidua/metabolism , Female , Fetus/immunology , Fetus/metabolism , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Ligands , Placenta/immunology , RNA, Small Cytoplasmic/genetics , Sequence Analysis, RNA , Stromal Cells/cytology , Stromal Cells/metabolism , Transcriptome , Trophoblasts/cytology , Trophoblasts/immunology , Trophoblasts/metabolism
20.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Article in English | MEDLINE | ID: mdl-34785597

ABSTRACT

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.


Subject(s)
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
SELECTION OF CITATIONS
SEARCH DETAIL