Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 182(5): 1125-1139.e18, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32822574

ABSTRACT

Maternal decidual NK (dNK) cells promote placentation, but how they protect against placental infection while maintaining fetal tolerance is unclear. Here we show that human dNK cells highly express the antimicrobial peptide granulysin (GNLY) and selectively transfer it via nanotubes to extravillous trophoblasts to kill intracellular Listeria monocytogenes (Lm) without killing the trophoblast. Transfer of GNLY, but not other cell death-inducing cytotoxic granule proteins, strongly inhibits Lm in human placental cultures and in mouse and human trophoblast cell lines. Placental and fetal Lm loads are lower and pregnancy success is greatly improved in pregnant Lm-infected GNLY-transgenic mice than in wild-type mice that lack GNLY. This immune defense is not restricted to pregnancy; peripheral NK (pNK) cells also transfer GNLY to kill bacteria in macrophages and dendritic cells without killing the host cell. Nanotube transfer of GNLY allows dNK to protect against infection while leaving the maternal-fetal barrier intact.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/immunology , Bacteria/immunology , Cell Movement/immunology , Killer Cells, Natural/immunology , Trophoblasts/immunology , Animals , Cell Line , Cell Line, Tumor , Dendritic Cells/immunology , Female , HeLa Cells , Humans , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Placenta/immunology , Placenta/microbiology , Pregnancy , Rats , THP-1 Cells , Trophoblasts/microbiology
2.
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
3.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Article in English | MEDLINE | ID: mdl-33622787

ABSTRACT

HLA-C arose during evolution of pregnancy in the great apes 10 to 15 million years ago. It has a dual function on placental extravillous trophoblasts (EVTs) as it contributes to both tolerance and immunity at the maternal-fetal interface. The mode of its regulation is of considerable interest in connection with the biology of pregnancy and pregnancy abnormalities. First-trimester primary EVTs in which HLA-C is highly expressed, as well as JEG3, an EVT model cell line, were employed. Single-cell RNA-seq data and quantitative PCR identified high expression of the transcription factor ELF3 in those cells. Chromatin immunoprecipitation (ChIP)-PCR confirmed that both ELF3 and MED1 bound to the proximal HLA-C promoter region. However, binding of RFX5 to this region was absent or severely reduced, and the adjacent HLA-B locus remained closed. Expression of HLA-C was inhibited by ELF3 small interfering RNAs (siRNAs) and by wrenchnolol treatment. Wrenchnolol is a cell-permeable synthetic organic molecule that mimics ELF3 and is relatively specific for binding to ELF3's coactivator, MED23, as our data also showed in JEG3. Moreover, the ELF3 gene is regulated by a superenhancer that spans more than 5 Mb, identified by assay for transposase-accessible chromatin using sequencing (ATAC-seq), as well as by its sensitivity to (+)-JQ1 (inhibitor of BRD4). ELF3 bound to its own promoter, thus creating an autoregulatory feedback loop that establishes expression of ELF3 and HLA-C in trophoblasts. Wrenchnolol blocked binding of MED23 to ELF3, thus disrupting the positive-feedback loop that drives ELF3 expression, with down-regulation of HLA-C expression as a consequence.


Subject(s)
DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Feedback, Physiological , HLA-C Antigens/genetics , Proto-Oncogene Proteins c-ets/genetics , Transcription Factors/genetics , Trophoblasts/immunology , Abortion, Legal , Adamantane/pharmacology , Azepines/pharmacology , Cell Line , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/immunology , Female , Gene Expression Regulation, Developmental/immunology , HLA-B Antigens/genetics , HLA-B Antigens/immunology , HLA-C Antigens/immunology , Humans , Immunity, Maternally-Acquired , Indoles/pharmacology , Mediator Complex/genetics , Mediator Complex/immunology , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/immunology , Pregnancy , Pregnancy Trimester, First , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-ets/antagonists & inhibitors , Proto-Oncogene Proteins c-ets/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Regulatory Factor X Transcription Factors/genetics , Regulatory Factor X Transcription Factors/immunology , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/immunology , Triazoles/pharmacology , Trophoblasts/cytology , Trophoblasts/drug effects
4.
Proc Natl Acad Sci U S A ; 117(27): 15772-15777, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32581122

ABSTRACT

During pregnancy, invading HLA-G+ extravillous trophoblasts (EVT) play a key role in placental development, uterine spiral artery remodeling, and prevention of detrimental maternal immune responses to placental and fetal antigens. Failures of these processes are suggested to play a role in the development of pregnancy complications, but very little is known about the underlying mechanisms. Here we present validated methods to purify and culture primary HLA-G+ EVT from the placental disk and chorionic membrane from healthy term pregnancy. Characterization of HLA-G+ EVT from term pregnancy compared to first trimester revealed their unique phenotypes, gene expression profiles, and differing capacities to increase regulatory T cells (Treg) during coculture assays, features that cannot be captured by using surrogate cell lines or animal models. Furthermore, clinical variables including gestational age and fetal sex significantly influenced EVT biology and function. These methods and approaches form a solid basis for further investigation of the role of HLA-G+ EVT in the development of detrimental placental inflammatory responses associated with pregnancy complications, including spontaneous preterm delivery and preeclampsia.


Subject(s)
HLA-G Antigens/immunology , Immunity, Innate/genetics , Placentation/immunology , Pre-Eclampsia/immunology , Cell Line , Cell Movement/immunology , Female , Gene Expression Regulation, Developmental/immunology , Humans , Maternal-Fetal Relations , Placenta/immunology , Placenta/metabolism , Pre-Eclampsia/pathology , Pregnancy , Pregnancy Trimester, First , Trophoblasts/immunology
5.
J Immunol ; 204(12): 3149-3159, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32376646

ABSTRACT

Decidual NK cells (dNK) are the main lymphocyte population in early pregnancy decidual mucosa. Although dNK decrease during pregnancy, they remain present in decidual tissues at term. First trimester dNK facilitate trophoblast invasion, provide protection against infections, and were shown to have many differences in their expression of NKRs, cytokines, and cytolytic capacity compared with peripheral blood NK cells (pNK). However, only limited data are available on the phenotype and function of term pregnancy dNK. In this study, dNK from human term pregnancy decidua basalis and decidua parietalis tissues were compared with pNK and first trimester dNK. Profound differences were found, including: 1) term pregnancy dNK have an increased degranulation response to K562 and PMA/ionomycin but lower capacity to respond to human CMV-infected cells; 2) term pregnancy dNK are not skewed toward recognition of HLA-C, as was previously shown for first trimester dNK; and 3) protein and gene expression profiles identified multiple differences between pNK, first trimester, and term pregnancy dNK, suggesting term pregnancy dNK are a distinct type of NK cells. Understanding the role of dNK throughout pregnancy is of high clinical relevance for studies aiming to prevent placental inflammatory disorders as well as maternal-to-fetal transmission of pathogens.


Subject(s)
Decidua/immunology , Killer Cells, Natural/immunology , Cell Line, Tumor , Cell Movement/immunology , Cells, Cultured , Female , Gene Expression/immunology , HLA-C Antigens/immunology , Humans , K562 Cells , Pregnancy , Trophoblasts/immunology
6.
Proc Natl Acad Sci U S A ; 116(7): 2634-2639, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30683721

ABSTRACT

Random amino acid copolymers used in the treatment of multiple sclerosis in man or experimental autoimmune encephalomyelitis (EAE) in mice [poly(Y,E,A,K)n, known as Copaxone, and poly(Y,F,A,K)n] function at least in part by generation of IL-10-secreting regulatory T cells that mediate bystander immunosuppression. The mechanism through which these copolymers induce Tregs is unknown. To investigate this question, four previously described Vα3.2 Vß14 T cell receptor (TCR) cDNAs, the dominant clonotype generated in splenocytes after immunization of SJL mice, that differed only in their CDR3 sequences were utilized to generate retrogenic mice. The high-level production of IL-10 as well as IL-5 and small amounts of the related cytokines IL-4 and IL-13 by CD4+ T cells isolated from the splenocytes of these mice strongly suggests that the TCR itself encodes information for specific cytokine secretion. The proliferation and production of IL-10 by these Tregs was costimulated by activation of glucocorticoid-induced TNF receptor (GITR) (expressed at high levels by these cells) through its ligand GITRL. A mechanism for generation of cells with this specificity is proposed. Moreover, retrogenic mice expressing these Tregs were protected from induction of EAE by the appropriate autoantigen.


Subject(s)
Hematopoietic Stem Cells/cytology , Interleukin-10/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Animals , DNA, Complementary , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Genetic Vectors , Immune Tolerance , Interleukins/metabolism , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Tumor Necrosis Factors/metabolism
7.
Proc Natl Acad Sci U S A ; 116(21): 10441-10446, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31040209

ABSTRACT

Polymorphic HLAs form the primary immune barrier to cell therapy. In addition, innate immune surveillance impacts cell engraftment, yet a strategy to control both, adaptive and innate immunity, is lacking. Here we employed multiplex genome editing to specifically ablate the expression of the highly polymorphic HLA-A/-B/-C and HLA class II in human pluripotent stem cells. Furthermore, to prevent innate immune rejection and further suppress adaptive immune responses, we expressed the immunomodulatory factors PD-L1, HLA-G, and the macrophage "don't-eat me" signal CD47 from the AAVS1 safe harbor locus. Utilizing in vitro and in vivo immunoassays, we found that T cell responses were blunted. Moreover, NK cell killing and macrophage engulfment of our engineered cells were minimal. Our results describe an approach that effectively targets adaptive as well as innate immune responses and may therefore enable cell therapy on a broader scale.


Subject(s)
Genetic Engineering/methods , Pluripotent Stem Cells/immunology , CRISPR-Cas Systems , Cell Line , Gene Knockout Techniques , Genes, MHC Class I , Genes, MHC Class II , Humans
8.
Immunology ; 163(3): 338-343, 2021 07.
Article in English | MEDLINE | ID: mdl-33565605

ABSTRACT

Humanized double transgenic mice express both HLA-DR15 (the MHC gene linked to MS) and TCR.Ob1A12 from a multiple sclerosis patient (that recognizes MBP85-99 presented by HLA-DR15), yet they fail to develop autoimmune encephalomyelitis quickly, although 5-10% develop disease at 12 months. These mice were found to express large numbers of IL-10-secreting splenocytes as early as 4 weeks of age. These regulatory T cells appeared spontaneously without prior immunization with the autoantigen MBP85-99. They were of murine origin and had a cytokine secretion profile and surface phenotype similar to that reported for Tr1 cells. Notably, the frequency of disease appeared to increase at 14 months. The diseased mice had small spleens which averaged 47 mg, while the remaining non-diseased mice in our colony killed at ages 14-15 months had splenocytes that averaged 80 mg (ranging from 47-130 mg). Thus, the appearance of disease was associated with diminution in numbers of IL-10-secreting regulatory T cells with age.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , HLA-DR Serological Subtypes/metabolism , Interleukin-10/metabolism , Multiple Sclerosis/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Spleen/pathology , T-Lymphocytes, Regulatory/immunology , Animals , Cells, Cultured , Disease Models, Animal , HLA-DR Serological Subtypes/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Basic Protein/immunology , Peptide Fragments/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics
9.
Proc Natl Acad Sci U S A ; 115(2): 385-390, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29259116

ABSTRACT

Understanding how decidual CD8+ T cell (CD8+ dT) cytotoxicity is regulated and how these cells integrate the competing needs for maternal-fetal tolerance and immunity to infection is an important research and clinical goal. Gene-expression analysis of effector-memory CD8+ dT demonstrated a mixed transcriptional signature of T cell dysfunction, activation, and effector function. High protein expression of coinhibitory molecules PD1, CTLA4, and LAG3, accompanied by low expression of cytolytic molecules suggests that the decidual microenvironment reduces CD8+ dT effector responses to maintain tolerance to fetal antigens. However, CD8+ dT degranulated, proliferated, and produced IFN-γ, TNF-α, perforin, and granzymes upon in vitro stimulation, demonstrating that CD8+ dT are not permanently suppressed and retain the capacity to respond to proinflammatory events, such as infections. The balance between transient dysfunction of CD8+ dT that are permissive of placental and fetal development, and reversal of this dysfunctional state, is crucial in understanding the etiology of pregnancy complications and prevention of congenital infections.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Decidua/metabolism , Gene Expression Profiling/methods , Immune Tolerance/genetics , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus/immunology , Cytomegalovirus/physiology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Female , Granzymes/genetics , Granzymes/metabolism , Humans , Immune Tolerance/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Perforin/genetics , Perforin/metabolism , Time Factors
10.
Nat Immunol ; 9(8): 898-907, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18604210

ABSTRACT

The inhibitory signaling of natural killer (NK) cells is crucial in the regulation of innate immune responses. Here we show that the association of KIR2DL1, an inhibitory receptor of NK cells, with beta-arrestin 2 mediated recruitment of the tyrosine phosphatases SHP-1 and SHP-2 to KIR2DL1 and facilitated 'downstream' inhibitory signaling. Consequently, the cytotoxicity of NK cells was higher in beta-arrestin 2-deficient mice but was inhibited in beta-arrestin 2-transgenic mice. Moreover, beta-arrestin 2-deficient mice were less susceptible than wild-type mice to mouse cytomegalovirus infection, an effect that was abolished by depletion of NK cells. Our findings identify a previously unknown mechanism by which the inhibitory signaling in NK cells is regulated.


Subject(s)
Arrestins/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Receptors, Immunologic/immunology , Signal Transduction/immunology , Animals , Cells, Cultured , Intracellular Signaling Peptides and Proteins , Mice , Receptors, Immunologic/chemistry , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , beta-Arrestin 2 , beta-Arrestins
11.
Trends Immunol ; 38(4): 272-286, 2017 04.
Article in English | MEDLINE | ID: mdl-28279591

ABSTRACT

During pregnancy, semiallogeneic fetal extravillous trophoblasts (EVT) invade the uterine mucosa without being rejected by the maternal immune system. Several mechanisms were initially proposed by Peter Medawar half a century ago to explain this apparent violation of the laws of transplantation. Then, three decades ago, an unusual human leukocyte antigen (HLA) molecule was identified: HLA-G. Uniquely expressed in EVT, HLA-G has since become the center of the present understanding of fetus-induced immune tolerance. Despite slow progress in the field, the last few years have seen an explosion in our knowledge of HLA-G biology. Here, we critically review new insights into the mechanisms controlling the expression and function of HLA-G at the maternal-fetal interface, and discuss their relevance for fetal tolerance.


Subject(s)
HLA-G Antigens/metabolism , Maternal-Fetal Relations , Pregnancy/immunology , Transplantation Tolerance , Trophoblasts/immunology , Animals , Female , HLA-G Antigens/immunology , Histocompatibility , Humans , Isoantigens/immunology
12.
Proc Natl Acad Sci U S A ; 113(52): 15072-15077, 2016 12 27.
Article in English | MEDLINE | ID: mdl-27956621

ABSTRACT

The combination of the activating killer cell Ig-like receptor 2DS1 (KIR2DS1) expressed by maternal decidual natural killer cells (dNK) and the presence of its ligand, the HLA-C allotype HLA-C2, expressed by fetal trophoblasts, reduces the risk of developing pregnancy complications. However, no molecular or cellular mechanism explains this genetic correlation. Here we demonstrate that KIR2DS1+ dNK acquired higher cytotoxic function than KIR2DS1- dNK when exposed to human cytomegalovirus (HCMV)-infected decidual stromal cells (DSC), particularly when DSCs express HLA-C2. Furthermore, dNK were unable to degranulate or secrete cytokines in response to HCMV-infected primary fetal extravillous trophoblasts. This emphasizes the immunological challenge to clear placental viral infections within the immune-privileged placenta. Activation of dNK through KIR2DS1/HLA-C2 interaction increases their ability to respond to placental HCMV infection and may limit subsequent virus-induced placental pathology. This mechanism is directly related to how KIR2DS1 expressed by dNK reduces development of severe pregnancy complications such as miscarriages and preterm delivery.


Subject(s)
Cytomegalovirus Infections/immunology , Decidua/immunology , Killer Cells, Natural/immunology , Placenta/immunology , Receptors, KIR/metabolism , Cell Separation , Cytomegalovirus , Decidua/virology , Female , Flow Cytometry , Gene Expression Profiling , HLA-C Antigens/analysis , Humans , Immune Tolerance , Immunoglobulins/metabolism , Placenta/virology , Placentation , Pregnancy , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/virology , Pregnancy Outcome , Receptors, KIR/genetics , Trophoblasts/virology
13.
Proc Natl Acad Sci U S A ; 113(19): 5364-9, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27078102

ABSTRACT

HLA-G, a nonclassical HLA molecule uniquely expressed in the placenta, is a central component of fetus-induced immune tolerance during pregnancy. The tissue-specific expression of HLA-G, however, remains poorly understood. Here, systematic interrogation of the HLA-G locus using massively parallel reporter assay (MPRA) uncovered a previously unidentified cis-regulatory element 12 kb upstream of HLA-G with enhancer activity, Enhancer L Strikingly, clustered regularly-interspaced short palindromic repeats (CRISPR)/Cas9-mediated deletion of this enhancer resulted in ablation of HLA-G expression in JEG3 cells and in primary human trophoblasts isolated from placenta. RNA-seq analysis demonstrated that Enhancer L specifically controls HLA-G expression. Moreover, DNase-seq and chromatin conformation capture (3C) defined Enhancer L as a cell type-specific enhancer that loops into the HLA-G promoter. Interestingly, MPRA-based saturation mutagenesis of Enhancer L identified motifs for transcription factors of the CEBP and GATA families essential for placentation. These factors associate with Enhancer L and regulate HLA-G expression. Our findings identify long-range chromatin looping mediated by core trophoblast transcription factors as the mechanism controlling tissue-specific HLA-G expression at the maternal-fetal interface. More broadly, these results establish the combination of MPRA and CRISPR/Cas9 deletion as a powerful strategy to investigate human immune gene regulation.


Subject(s)
Enhancer Elements, Genetic/immunology , Gene Expression Regulation, Developmental/immunology , HLA-G Antigens/immunology , Histocompatibility, Maternal-Fetal/immunology , Maternal-Fetal Exchange/immunology , Pregnancy/immunology , Trophoblasts/immunology , Enhancer Elements, Genetic/genetics , Female , Gene Expression Regulation, Developmental/genetics , HLA-G Antigens/genetics , Histocompatibility, Maternal-Fetal/genetics , Humans , Immunogenetic Phenomena/genetics , Maternal-Fetal Exchange/genetics , Placenta/immunology
14.
Proc Natl Acad Sci U S A ; 112(43): 13312-7, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26460007

ABSTRACT

The interaction of noncytotoxic decidual natural killer cells (dNK) and extravillous trophoblasts (EVT) at the maternal-fetal interface was studied. Confocal microscopy revealed that many dNK interact with a single large EVT. Filamentous projections from EVT enriched in HLA-G were shown to contact dNK, and may represent the initial stage of synapse formation. As isolated, 2.5% of dNK contained surface HLA-G. However, surface HLA-G-negative dNK contained internalized HLA-G. Activation of dNK resulted in the disappearance of internalized HLA-G in parallel with restoration of cytotoxicity. Surface HLA-G was reacquired by incubation with EVT. This HLA-G cycle of trogocytosis, endocytosis, degradation, and finally reacquisition provides a transient and localized acquisition of new functional properties by dNK upon interaction with EVT. Interruption of the cycle by activation of dNK by cytokines and/or viral products serves to ensure the NK control of virus infection at the interface, and is illustrated here by the response of dNK to human cytomegalo virus (HCMV)-infected decidual stromal cells. Thus, the HLA-G cycle in dNK can provide both for NK tolerance and antiviral immunity.


Subject(s)
Cytomegalovirus Infections/immunology , HLA-G Antigens/metabolism , Immune Tolerance/immunology , Killer Cells, Natural/immunology , Maternal-Fetal Exchange/immunology , Trophoblasts/immunology , Blotting, Western , Cytokines/metabolism , DNA Primers/genetics , Female , Flow Cytometry , Humans , Microscopy, Confocal , Pregnancy , Real-Time Polymerase Chain Reaction , Statistics, Nonparametric
15.
Proc Natl Acad Sci U S A ; 112(23): 7219-24, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26015573

ABSTRACT

Invading human leukocyte antigen-G+ (HLA-G+) extravillous trophoblasts (EVT) are rare cells that are believed to play a key role in the prevention of a maternal immune attack on foreign fetal tissues. Here highly purified HLA-G+ EVT and HLA-G- villous trophoblasts (VT) were isolated. Culture on fibronectin that EVT encounter on invading the uterus increased HLA-G, EGF-Receptor-2, and LIF-Receptor expression on EVT, presumably representing a further differentiation state. Microarray and functional gene set enrichment analysis revealed a striking immune-activating potential for EVT that was absent in VT. Cocultures of HLA-G+ EVT with sample matched decidual natural killer cells (dNK), macrophages, and CD4+ and CD8+ T cells were established. Interaction of EVT with CD4+ T cells resulted in increased numbers of CD4+CD25(HI)FOXP3+CD45RA+ resting regulatory T cells (Treg) and increased the expression level of the Treg-specific transcription factor FOXP3 in these cells. However, EVT did not enhance cytokine secretion in dNK, whereas stimulation of dNK with mitogens or classical natural killer targets confirmed the distinct cytokine secretion profiles of dNK and peripheral blood NK cells (pNK). EVT are specialized cells involved in maternal-fetal tolerance, the properties of which are not imitated by HLA-G-expressing surrogate cell lines.


Subject(s)
HLA-G Antigens/immunology , Leukocytes/immunology , Trophoblasts/immunology , Antigens, CD/immunology , Cells, Cultured , Gene Expression Profiling , Humans , Transcription, Genetic , Trophoblasts/metabolism , Up-Regulation
16.
Biol Reprod ; 96(4): 831-842, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28340094

ABSTRACT

During pregnancy, fetal extravillous trophoblasts (EVT) play a key role in the regulation of maternal T cell and NK cell responses. EVT display a unique combination of human leukocyte antigens (HLA); EVT do not express HLA-A and HLA-B, but do express HLA-C, HLA-E, and HLA-G. The mechanisms establishing this unique HLA expression pattern have not been fully elucidated. The major histocompatibility complex (MHC) class I and class II transcriptional activators NLRC5 and CIITA are expressed neither by EVT nor by the EVT model cell line JEG3, which has an MHC expression pattern identical to that of EVT. Therefore, other MHC regulators must be present to control HLA-C, HLA-E, and HLA-G expression in these cells. CIITA and NLRC5 are both members of the nucleotide-binding domain, leucine-rich repeat (NLR) family of proteins. Another member of this family, NLRP2, is highly expressed by EVT and JEG3, but not in maternal decidual stromal cells. In this study, transcription activator-like effector nuclease technology was used to delete NLRP2 in JEG3. Furthermore, lentiviral delivery of shRNA was used to knockdown NLRP2 in JEG3 and primary EVT. Upon NLRP2 deletion, Tumor Necrosis Factor-α (TNFα)-induced phosphorylation of NF-KB p65 increased in JEG3 and EVT, and more surprisingly a significant increase in constitutive HLA-C expression was observed in JEG3. These data suggest a broader role for NLR family members in the regulation of MHC expression during inflammation, thus forming a bridge between innate and adaptive immune responses. As suppressor of proinflammatory responses, NLRP2 may contribute to preventing unwanted antifetal responses.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Gene Expression Regulation, Developmental/physiology , HLA-C Antigens/metabolism , NF-kappa B/metabolism , Trophoblasts/metabolism , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins , Cell Line , Cytokines/genetics , Cytokines/metabolism , Gene Deletion , Genes, MHC Class I/genetics , HLA-C Antigens/genetics , Humans , NF-kappa B/genetics , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
17.
Proc Natl Acad Sci U S A ; 111(39): 14199-204, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25225399

ABSTRACT

The reciprocal activation of dendritic cells (DCs) and natural killer cells (NKs) plays a key role in both innate and adaptive immunity. The effect of aging on this cross-talk, a critical step in virus disease control and tumor immunology, has not been reported. Splenic DCs and NKs were purified from both young and old C57BL/6 mice and cocultured in the presence of polyinosinic:polycytidylic acid (poly I:C). The resulting activation of NKs was measured as expression of CD69 and secretion of IFN-γ. However, DCs from old mice could not activate NKs from either young or old mice in vitro or in vivo. In contrast, DCs from young mice efficiently activated NKs from both young and old mice. DCs from old mice were deficient in poly I:C-stimulated secretion of IL-15, IL-18, and IFN-α. Gene expression analysis revealed many other differences between DCs of old and young mice. Young mice strongly eradicated MHC class I-negative NK-sensitive RMA-S lymphoma mutant tumor cells, but old mice did not, in concert with the previous report that mousepox kills aged, but not young, C57BL/6 mice. Furthermore, a similar dysfunction of DC and its key role in NK activation was found in 27 out of 55 healthy human donors.


Subject(s)
Aging/immunology , Dendritic Cells/immunology , Killer Cells, Natural/immunology , Neoplasms, Experimental/immunology , Adult , Aged , Animals , Cell Line, Tumor , Coculture Techniques , Cytokines/biosynthesis , Humans , Interferon Inducers/administration & dosage , Interferon-gamma/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Poly I-C/administration & dosage
18.
Nat Rev Immunol ; 6(4): 271-82, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16557259

ABSTRACT

MHC class II molecules on the surface of antigen-presenting cells display a range of peptides for recognition by the T-cell receptors of CD4+ T helper cells. Therefore, MHC class II molecules are central to effective adaptive immune responses, but conversely, genetic and epidemiological data have implicated these molecules in the pathogenesis of autoimmune diseases. Indeed, the strength of the associations between particular MHC class II alleles and disease render them the main genetic risk factors for autoimmune disorders such as type 1 diabetes. Here, we discuss the insights that the crystal structures of MHC class II molecules provide into the molecular mechanisms by which sequence polymorphisms might contribute to disease susceptibility.


Subject(s)
Autoimmune Diseases/genetics , HLA-D Antigens/genetics , Autoimmune Diseases/immunology , Binding Sites/genetics , Binding Sites/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/metabolism , HLA-D Antigens/chemistry , HLA-D Antigens/metabolism , Humans , Models, Molecular , Protein Binding/genetics , Protein Binding/immunology , Protein Conformation
19.
Curr Opin Cell Biol ; 20(5): 597-605, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18639449

ABSTRACT

Natural killer (NK) cells comprise a subset of lymphocytes involved in protection against microbial pathogens and tumors. NK cells recognize host cells that are missing MHC class I molecules and eliminate them through localized delivery of lytic granules. The majority of NK cell effector functions require direct cell-to-cell contact. Binding to a target cell is accompanied by creation of complex structures at the cell-cell interface known as immunological synapses. Recent studies have contributed immensely to the characterization of several types of NK cell immunological synapses and understanding of the variety of processes originating at this intriguing place. The emerging picture illustrates NK cell immune synapses as the sites of highly complex regulation of NK cell activity.


Subject(s)
Cytotoxicity, Immunologic/immunology , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Genes, MHC Class I , Humans , Killer Cells, Natural/cytology , Lymphocyte Activation , Lymphocyte Subsets/cytology , Macrophages/cytology , Macrophages/immunology
20.
J Immunol ; 191(1): 208-16, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23740953

ABSTRACT

Multiple sclerosis (MS) is an autoimmune disease that affects the CNS. One approved treatment for relapsing forms of MS is YEAK, a random copolymer of the amino acids tyrosine, glutamic acid, alanine, and lysine. YFAK, a second-generation copolymer composed of tyrosine, phenylalanine, alanine, and lysine, is more successful in treating experimental autoimmune encephalomyelitis, a mouse model of MS. Although originally designed and optimized based on the autoantigen myelin basic protein (MBP) and the MBP-derived peptide MBP85-99 presented to the MS-associated class II MHC molecule HLA-DR2, YEAK and YFAK also stimulate cytokine and chemokine production in APCs that lack class II MHC products. How YEAK and YFAK copolymers interact with APCs remains enigmatic. We used biotinylated YFAK to affinity-purify YFAK-interacting proteins from RAW264.7 cells and tested APCs from mice deficient in several of the newly identified interactors for their capacity to secrete CCL22 in response to YEAK and YFAK. We propose that initial contact of YFAK with cells is mediated mainly by electrostatic interactions, and find that interaction of YFAK with host proteins is strongly dependent on ionic strength. Cells deficient in enzymes involved in sulfation of proteins and proteoglycans showed strongly reduced binding of biotinylated YFAK. Lastly, cells stimulated with YFAK in the presence of heparin, structurally similar to heparan sulfates, failed to produce CCL22. We conclude that charge-dependent interactions of copolymers that alleviate MS/experimental autoimmune encephalomyelitis are critical for their effects exerted on APCs and may well be the main initial mediators of these therapeutically active copolymers.


Subject(s)
Amino Acids/metabolism , Antigen-Presenting Cells/chemistry , Antigen-Presenting Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Heparitin Sulfate/metabolism , Membrane Glycoproteins/metabolism , Protein Interaction Mapping/methods , Amino Acids/chemistry , Amino Acids/pharmacology , Animals , Antigen-Presenting Cells/immunology , Biopolymers/chemistry , Biopolymers/metabolism , Biopolymers/pharmacology , Biotinylation , Cell Line , Encephalomyelitis, Autoimmune, Experimental/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Membrane Glycoproteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligopeptides/chemistry , Oligopeptides/metabolism , Oligopeptides/pharmacology , Primary Cell Culture , Random Allocation
SELECTION OF CITATIONS
SEARCH DETAIL