Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Exp Med ; 221(2)2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38189779

ABSTRACT

The mechanisms whereby Eomes controls tissue accumulation of T cells and strengthens inflammation remain ill-defined. Here, we show that Eomes deletion in antigen-specific CD4+ T cells is sufficient to protect against central nervous system (CNS) inflammation. While Eomes is dispensable for the initial priming of CD4+ T cells, it is required for long-term maintenance of CNS-infiltrating CD4+ T cells. We reveal that the impact of Eomes on effector CD4+ T cell longevity is associated with sustained expression of multiple genes involved in mitochondrial organization and functions. Accordingly, epigenetic studies demonstrate that Eomes supports mitochondrial function by direct binding to either metabolism-associated genes or mitochondrial transcriptional modulators. Besides, the significance of these findings was confirmed in CD4+ T cells from healthy donors and multiple sclerosis patients. Together, our data reveal a new mechanism by which Eomes promotes severity and chronicity of inflammation via the enhancement of CD4+ T cell mitochondrial functions and resistance to stress-induced cell death.


Subject(s)
CD4-Positive T-Lymphocytes , Central Nervous System , T-Box Domain Proteins , Humans , Cell Death , Inflammation , Mitochondria , T-Box Domain Proteins/genetics
2.
Article in English | MEDLINE | ID: mdl-37280149

ABSTRACT

BACKGROUND: Intrinsic capacity (IC) is a concept related to functionality that reflects healthy aging. ATPase inhibitory factor 1 (IF1) is a multifaceted protein that regulates mitochondrial oxidative phosphorylation (OXPHOS), and may be involved in IC. The objective of this study is to investigate the association between plasma levels of IF1 and IC changes in community-dwelling older adults. METHODS: Community-dwelling older adults from the Multidomain Alzheimer Preventive Trial (MAPT Study) were enrolled in this study. A composite IC score was calculated based on 4 IC domains: locomotion, psychological dimension, cognition, and vitality (with data available annually over 4 years of follow-up). Secondary analyses were conducted on the sensory domain (with data available only for 1 year of follow-up). Mixed-model linear regression adjusted for confounders was conducted. RESULTS: A total of 1 090 participants with usable IF1 values were included in the study (75.3 ± 4.4 years; 64% females). Compared to the lowest quartile, both the low- and high-intermediate IF1 quartiles were found to be cross-sectionally associated with greater composite IC scores across 4 domains (ßlow-intermediate, 1.33; 95% confidence interval [CI] 0.06-2.60 and ßhigh-intermediate, 1.78; 95% CI 0.49-3.06). In the secondary analyses, the highest quartile was found to be associated with a slower decline in composite IC scores across 5 domains over 1 year (ßhigh 1.60; 95% CI 0.06-3.15). The low- and high-intermediate IF1 quartiles were also found to be cross-sectionally associated with greater locomotion (ßlow-intermediate, 2.72; 95% CI 0.36-5.08) and vitality scores (ßhigh-intermediate, 1.59; 95% CI 0.06-3.12), respectively. CONCLUSIONS: This study is the first to demonstrate that levels of circulating IF1, a mitochondrial-related biomarker, are associated with IC composite scores in both cross-sectional and prospective analyses among community-dwelling older adults. However, further research is needed to confirm these findings and elucidate the potential underlying mechanisms that may explain these associations.


Subject(s)
ATPase Inhibitory Protein , Alzheimer Disease , Independent Living , Aged , Female , Humans , Male , Cross-Sectional Studies , Prospective Studies , ATPase Inhibitory Protein/blood
3.
J Cell Biol ; 222(5)2023 05 01.
Article in English | MEDLINE | ID: mdl-36988579

ABSTRACT

Macrophages are essential for HIV-1 pathogenesis and represent major viral reservoirs. Therefore, it is critical to understand macrophage infection, especially in tissue macrophages, which are widely infected in vivo, but poorly permissive to cell-free infection. Although cell-to-cell transmission of HIV-1 is a determinant mode of macrophage infection in vivo, how HIV-1 transfers toward macrophages remains elusive. Here, we demonstrate that fusion of infected CD4+ T lymphocytes with human macrophages leads to their efficient and productive infection. Importantly, several tissue macrophage populations undergo this heterotypic cell fusion, including synovial, placental, lung alveolar, and tonsil macrophages. We also find that this mode of infection is modulated by the macrophage polarization state. This fusion process engages a specific short-lived adhesion structure and is controlled by the CD81 tetraspanin, which activates RhoA/ROCK-dependent actomyosin contractility in macrophages. Our study provides important insights into the mechanisms underlying infection of tissue-resident macrophages, and establishment of persistent cellular reservoirs in patients.


Subject(s)
CD4-Positive T-Lymphocytes , Cell Fusion , HIV Infections , Macrophages , Humans , CD4-Positive T-Lymphocytes/metabolism , HIV Infections/metabolism , HIV-1/pathogenicity , Macrophages/metabolism , Macrophages/virology , Actomyosin/metabolism
4.
Front Immunol ; 13: 1000861, 2022.
Article in English | MEDLINE | ID: mdl-36483552

ABSTRACT

Unlike other Flaviviruses, Zika virus (ZIKV) infection during the first trimester of pregnancy causes severe pregnancy outcomes including the devastating microcephaly and diseases associated with placental dysfunctions. We have previously reported that the maternal decidua basalis, the major maternal-fetal interface, serves as a replication platform enabling virus amplification before dissemination to the fetal compartment. However, the rate of congenital infection is quite low, suggesting the presence of a natural barrier against viral infection. Using primary cells from first-trimester pregnancy samples, we investigated in this study how the maternal decidua can interfere with ZIKV infection. Our study reveals that whether through their interactions with dNK cells, the main immune cell population of the first-trimester decidua, or their production of proinflammatory cytokines, decidual stromal cells (DSCs) are the main regulators of ZIKV infection during pregnancy. We also validate the functional role of AXL as a crucial receptor for ZIKV entry in DSCs and demonstrate that targeted inhibition of ligand-receptor interaction at the early stage of the infection is effective in drastically reducing virus pathogenesis at the maternal-fetal interface. Collectively, our results provide insights into the mechanisms through which ZIKV infection and spreading can be limited. The strategy of circumventing viral entry at the maternal-fetus interface limits virus dissemination to fetal tissues, thereby preventing congenital abnormalities.


Subject(s)
Zika Virus Infection , Zika Virus , Pregnancy , Female , Humans , Placenta
5.
Methods Mol Biol ; 2407: 205-213, 2022.
Article in English | MEDLINE | ID: mdl-34985667

ABSTRACT

The human decidua basalis, main uterine mucosa during pregnancy, provides an ex vivo model for studying natural protection of macrophages against HIV-1 infection at the mucosal level. Beyond pregnancy, the decidua constitutes also a valuable tool to assess tissue-resident macrophage infection. Here, we provide a detailed protocol for decidual macrophage purification and tissue infection.


Subject(s)
HIV Infections , HIV-1 , Decidua , Female , Humans , Macrophages , Pregnancy
6.
Front Med (Lausanne) ; 8: 674645, 2021.
Article in English | MEDLINE | ID: mdl-34368184

ABSTRACT

The placenta, the first and largest organ to develop after conception, not only nurtures and promotes the development of the conceptus, but, it also functions as a barrier against invading pathogens. Early phases of pregnancy are associated with expansion of specific subsets of Natural Killer cells (dNK) and macrophages (dMφ) at the maternal uterine mucosa, the basal decidua. In concert with cells of fetal origin, dNK cells, and dMφ orchestrate all steps of placenta and fetus development, and provide the first line of defense to limit vertical transmission. However, some pathogens that infect the mother can overcome this protective barrier and jeopardize the fetus health. In this review, we will discuss how members of the classical TORCH family (Toxoplasma, Other, Rubella, Cytomegalovirus, and Herpes simplex virus) and some emerging viruses (Hepatitis E virus, Zika virus, and SARS-CoV2) can afford access to the placental fortress. We will also discuss how changes in the intrauterine environment as a consequence of maternal immune cell activation contribute to placental diseases and devastating pregnancy outcomes.

7.
PLoS Pathog ; 17(2): e1009367, 2021 02.
Article in English | MEDLINE | ID: mdl-33617602

ABSTRACT

Genotype 3 Hepatitis E virus (HEV-3) is an emerging threat for aging population. More than one third of older infected patients develops clinical symptoms with severe liver damage, while others remain asymptomatic. The origin of this discrepancy is still elusive although HEV-3 pathogenesis appears to be immune-mediated. Therefore, we investigated the role of CD8 T cells in the outcome of the infection in immunocompetent elderly subjects. We enrolled twenty two HEV-3-infected patients displaying similar viral determinants and fifteen healthy donors. Among the infected group, sixteen patients experienced clinical symptoms related to liver disease while six remained asymptomatic. Here we report that symptomatic infection is characterized by an expansion of highly activated effector memory CD8 T (EM) cells, regardless of antigen specificity. This robust activation is associated with key features of early T cell exhaustion including a loss in polyfunctional type-1 cytokine production and partial commitment to type-2 cells. In addition, we show that bystander activation of EM cells seems to be dependent on the inflammatory cytokines IL-15 and IL-18, and is supported by an upregulation of the activating receptor NKG2D and an exuberant expression of T-Bet and T-Bet-regulated genes including granzyme B and CXCR3. We also show that the inflammatory chemokines CXCL9-10 are increased in symptomatic patients thereby fostering the recruitment of highly cytotoxic EM cells into the liver in a CXCR3-dependent manner. Finally, we find that the EM-biased immune response returns to homeostasis following viral clearance and disease resolution, further linking the EM cells response to viral burden. Conversely, asymptomatic patients are endowed with low-to-moderate EM cell response. In summary, our findings define immune correlates that contribute to HEV-3 pathogenesis and emphasize the central role of EM cells in governing the outcome of the infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Hepatitis E virus/classification , Hepatitis E/pathology , Immunologic Memory/immunology , Receptors, CXCR3/metabolism , Aged , Case-Control Studies , Cytokines/metabolism , Female , Genotype , Hepatitis E/immunology , Hepatitis E/virology , Hepatitis E virus/genetics , Hepatitis E virus/immunology , Humans , Male , Middle Aged
8.
Nat Commun ; 11(1): 2967, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32528049

ABSTRACT

The recent outbreak of Zika virus (ZIKV) was associated with birth defects and pregnancy loss when maternal infection occurs in early pregnancy, but specific mechanisms driving placental insufficiency and subsequent ZIKV-mediated pathogenesis remain unclear. Here we show, using large scale metabolomics, that ZIKV infection reprograms placental lipidome by impairing the lipogenesis pathways. ZIKV-induced metabolic alterations provide building blocks for lipid droplet biogenesis and intracellular membrane rearrangements to support viral replication. Furthermore, lipidome reprogramming by ZIKV is paralleled by the mitochondrial dysfunction and inflammatory immune imbalance, which contribute to placental damage. In addition, we demonstrate the efficacy of a commercially available inhibitor in limiting ZIKV infection, provides a proof-of-concept for blocking congenital infection by targeting metabolic pathways. Collectively, our study provides mechanistic insights on how ZIKV targets essential hubs of the lipid metabolism that may lead to placental dysfunction and loss of barrier function.


Subject(s)
Placenta/virology , Zika Virus Infection/immunology , Zika Virus Infection/metabolism , Female , Humans , Infectious Disease Transmission, Vertical , Lipidomics/methods , Pregnancy , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/metabolism , Zika Virus/immunology , Zika Virus/pathogenicity
9.
Emerg Infect Dis ; 25(4): 823-825, 2019 04.
Article in English | MEDLINE | ID: mdl-30882325

ABSTRACT

We assessed Zika virus RNA and select cytokine levels in semen, blood, and plasma samples from an infected patient in South America. Viral RNA was detected in semen >2 months after viremia clearance; cytokine profiles differed in semen and plasma. After viremia, Zika virus appears to become compartmentalized in the male reproductive tract.


Subject(s)
Cytokines/metabolism , Semen/metabolism , Zika Virus Infection/metabolism , Zika Virus Infection/virology , Zika Virus , Biomarkers , Cytokines/blood , Host-Pathogen Interactions , Humans , Zika Virus Infection/blood
10.
Nat Commun ; 9(1): 4748, 2018 11 12.
Article in English | MEDLINE | ID: mdl-30420629

ABSTRACT

Hepatitis E virus (HEV) infection, particularly HEV genotype 1 (HEV-1), can result in fulminant hepatic failure and severe placental diseases, but mechanisms underlying genotype-specific pathogenicity are unclear and appropriate models are lacking. Here, we model HEV-1 infection ex vivo at the maternal-fetal interface using the decidua basalis and fetal placenta, and compare its effects to the less-pathogenic genotype 3 (HEV-3). We demonstrate that HEV-1 replicates more efficiently than HEV-3 both in tissue explants and stromal cells, produces more infectious progeny virions and causes severe tissue alterations. HEV-1 infection dysregulates the secretion of several soluble factors. These alterations to the cytokine microenvironment correlate with viral load and contribute to the tissue damage. Collectively, this study characterizes an ex vivo model for HEV infection and provides insights into HEV-1 pathogenesis during pregnancy that are linked to high viral replication, alteration of the local secretome and induction of tissue injuries.


Subject(s)
Hepatitis E virus/genetics , Hepatitis E virus/pathogenicity , Maternal-Fetal Exchange/physiology , Cells, Cultured , Decidua/pathology , Decidua/virology , Female , Genotype , Humans , Interferons/metabolism , Pregnancy , Stromal Cells/metabolism , Virus Replication , Interferon Lambda
11.
Virologie (Montrouge) ; 22(5): 239-250, 2018 10 01.
Article in French | MEDLINE | ID: mdl-33111685

ABSTRACT

Hepatitis E virus (HEV) presents a worldwide distribution. In developing countries, hepatitis E, related to HEV1 and HEV2, is a waterborne disease. In developed countries, hepatitis E is a zoonotic disease due to HEV3 and HEV4. It is mainly transmitted through meat consumption from animal reservoirs such as pig, boar, deer and rabbit. New clinical forms include neurological manifestations that are now clearly associated with HEV3 infection. Recent studies showed that ORF1 polyprotein was able to disrupt the innate immune response. It was also shown that ORF2 protein exists at least in two forms: a free, glycosylated form and a non-glycosylated form, which assembles to form the capsid. Lastly, it was shown that ORF3 protein, involved in the virus egress, acts as a viroporin. New culture systems and animal models have been developed recently, and will be very helpful to complete our understanding of HEV life cycle and pathogenesis.

12.
Front Immunol ; 8: 1413, 2017.
Article in English | MEDLINE | ID: mdl-29123530

ABSTRACT

Cardiac repair following MI relies on a finely regulated immune response involving sequential recruitment of monocytes to the injured tissue. Monocyte-derived cells are also critical for tissue homeostasis and healing process. Our previous findings demonstrated the interaction of T and natural killer cells with allogeneic human cardiac-derived stem/progenitor cells (hCPC) and suggested their beneficial effect in the context of cardiac repair. Therefore, we investigated here whether monocytes and their descendants could be also modulated by allogeneic hCPC toward a repair/anti-inflammatory phenotype. Through experimental in vitro assays, we assessed the impact of allogeneic hCPC on the recruitment, functions and differentiation of monocytes. We found that allogeneic hCPC at steady state or under inflammatory conditions can incite CCL-2/CCR2-dependent recruitment of circulating CD14+CD16- monocytes and fine-tune their activation toward an anti-inflammatory profile. Allogeneic hCPC also promoted CD14+CD16- monocyte polarization into anti-inflammatory/immune-regulatory macrophages with high phagocytic capacity and IL10 secretion. Moreover, hCPC bended the differentiation of CD14+CD16- monocytes to dendritic cells (DCs) toward anti-inflammatory macrophage-like features and impaired their antigen-presenting function in favor of immune-modulation. Collectively, our results demonstrate that allogeneic hCPC could reshape monocytes, macrophages as well as DCs responses by favoring their anti-inflammatory/tolerogenic activation/polarization. Thereby, therapeutic allogeneic hCPC might also contribute to post-infarct myocardial healing by modeling the activities of monocytes and their derived descendants.

13.
Emerg Infect Dis ; 23(7): 1191-1193, 2017 07.
Article in English | MEDLINE | ID: mdl-28628452

ABSTRACT

Hepatitis E virus (HEV) has been detected in rabbits, but whether rabbit HEV strains can be transmitted to humans is not known. Of 919 HEV-infected patients in France during 2015-2016, five were infected with a rabbit HEV strain. None of the patients had direct contact with rabbits, suggesting foodborne or waterborne infections.


Subject(s)
Hepatitis E virus/classification , Hepatitis E virus/genetics , Hepatitis E/transmission , Hepatitis E/virology , Zoonoses/transmission , Zoonoses/virology , Adult , Animals , France/epidemiology , Genotype , Hepatitis E/diagnosis , Hepatitis E/epidemiology , Humans , Immunocompromised Host , Male , Middle Aged , Phylogeny , RNA, Viral , Rabbits , Zoonoses/diagnosis , Zoonoses/epidemiology
14.
Sci Rep ; 6: 35296, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27759009

ABSTRACT

The outbreak of the Zika Virus (ZIKV) and its association with fetal abnormalities have raised worldwide concern. However, the cellular tropism and the mechanisms of ZIKV transmission to the fetus during early pregnancy are still largely unknown. Therefore, we ex vivo modeled the ZIKV transmission at the maternal-fetal interface using organ culture from first trimester pregnancy samples. Here, we provide evidence that ZIKV strain circulating in Brazil infects and damages tissue architecture of the maternal decidua basalis, the fetal placenta and umbilical cord. We also show that ZIKV replicates differentially in a wide range of maternal and fetal cells, including decidual fibroblasts and macrophages, trophoblasts, Hofbauer cells as well as umbilical cord mesenchymal stem cells. The striking cellular tropism of ZIKV and its cytopathic-induced tissue injury during the first trimester of pregnancy could provide an explanation for the irreversible congenital damages.


Subject(s)
Placenta/virology , Viral Tropism/genetics , Zika Virus Infection/transmission , Zika Virus/genetics , Adolescent , Adult , Brazil , Female , Humans , Maternal-Fetal Relations , Placenta/pathology , Pregnancy , Pregnancy Trimester, First/genetics , Zika Virus/pathogenicity , Zika Virus Infection/virology
15.
J Virol ; 89(2): 1329-39, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25392215

ABSTRACT

UNLABELLED: In order to develop strategies to prevent HIV-1 (human immunodeficiency virus type 1) transmission, it is crucial to better characterize HIV-1 target cells in the female reproductive tract (FRT) mucosae and to identify effective innate responses. Control of HIV-1 infection in the decidua (the uterine mucosa during pregnancy) can serve as a model to study natural mucosal protection. Macrophages are the main HIV-1 target cells in the decidua. Here we report that in vitro, macrophages and T cells are the main HIV-1 targets in the endometrium in nonpregnant women. As reported for decidual macrophages (dM), endometrial macrophages (eM) were found to have an M2-like phenotype (CD68+ CD163+ CD206+ IL-10high). However, eM and dM may belong to different subpopulations, as they differently express certain markers and secrete different amounts of proinflammatory and anti-inflammatory cytokines. We observed strong expression of the SAMHD1 restriction factor and weak expression of its inactive form (pSAMHD1, phosphorylated at residue Thr592) in both eM and dM. Infection of macrophages from both tissues was enhanced in the presence of the viral protein Vpx, suggesting a role for SAMHD1 in the restriction of HIV-1 infection. This study and further comparisons of the decidua with FRT mucosae in nonpregnant women should help to identify mechanisms of mucosal protection against HIV-1 infection. IMPORTANCE: The female reproductive tract mucosae are major portals of HIV-1 entry into the body. The decidua (uterine mucosa during pregnancy) can serve as a model for studying natural mucosal protection against HIV-1 transmission. A comparison of target cells and innate responses in the decidua versus the endometrium in nonpregnant women could help to identify protective mechanisms. Here, we report for the first time that macrophages are one of the main HIV-1 target cells in the endometrium and that infection of macrophages from both the endometrium and the decidua is restricted by SAMHD1. These findings might have implications for the development of vaccines to prevent HIV-1 mucosal transmission.


Subject(s)
Endometrium/immunology , HIV-1/growth & development , HIV-1/immunology , Macrophages/immunology , Macrophages/virology , Monomeric GTP-Binding Proteins/metabolism , Adult , Antigens, CD/analysis , Female , Humans , Immunophenotyping , Interleukin-10/analysis , Macrophages/chemistry , Middle Aged , Monomeric GTP-Binding Proteins/immunology , SAM Domain and HD Domain-Containing Protein 1 , T-Lymphocytes/immunology , T-Lymphocytes/virology , Young Adult
16.
Front Microbiol ; 5: 316, 2014.
Article in English | MEDLINE | ID: mdl-25071732

ABSTRACT

Maternofetal pathogen transmission is partially controlled at the level of the maternal uterine mucosa at the fetal implantation site (the decidua basalis), where maternal and fetal cells are in close contact. Toll-like receptors (TLRs) may play an important role in initiating rapid immune responses against pathogens in the decidua basalis, however the tolerant microenvironment should be preserved in order to allow fetal development. Here we investigated the expression and functionality of TLRs expressed by decidual macrophages (dMs) and NK cells (dNKs), the major decidual immune cell populations. We report for the first time that both human dMs and dNK cells express mRNAs encoding TLRs 1-9, albeit with a higher expression level in dMs. TLR2, TLR3, and TLR4 protein expression checked by flow cytometry was positive for both dMs and dNK cells. In vitro treatment of primary dMs and dNK cells with specific TLR2, TLR3, TLR4, TLR7/8, and TLR9 agonists enhanced their secretion of pro- and anti-inflammatory cytokines, as well as cytokines and chemokines involved in immune cell crosstalk. Only dNK cells released IFN-γ, whereas only dMs released IL-1ß, IL-10, and IL-12. TLR9 activation of dMs resulted in a distinct pattern of cytokine expression compared to the other TLRs. The cytokine profiles expressed by dMs and dNK cells upon TLR activation are compatible with maintenance of the fetotolerant immune environment during initiation of immune responses to pathogens at the maternofetal interface.

17.
PLoS Pathog ; 9(4): e1003257, 2013.
Article in English | MEDLINE | ID: mdl-23592985

ABSTRACT

During the first trimester of pregnancy the uterus is massively infiltrated by decidual natural killer cells (dNK). These cells are not killers, but they rather provide a microenvironment that is propitious to healthy placentation. Human cytomegalovirus (HCMV) is the most common cause of intrauterine viral infections and a known cause of severe birth defects or fetal death. The rate of HCMV congenital infection is often low in the first trimester of pregnancy. The mechanisms controlling HCMV spreading during pregnancy are not yet fully revealed, but evidence indicating that the innate immune system plays a role in controlling HCMV infection in healthy adults exists. In this study, we investigated whether dNK cells could be involved in controlling viral spreading and in protecting the fetus against congenital HCMV infection. We found that freshly isolated dNK cells acquire major functional and phenotypic changes when they are exposed to HCMV-infected decidual autologous fibroblasts. Functional studies revealed that dNK cells, which are mainly cytokines and chemokines producers during normal pregnancy, become cytotoxic effectors upon their exposure to HCMV-infected autologous decidual fibroblasts. Both the NKG2D and the CD94/NKG2C or 2E activating receptors are involved in the acquired cytotoxic function. Moreover, we demonstrate that CD56(pos) dNK cells are able to infiltrate HCMV-infected trophoblast organ culture ex-vivo and to co-localize with infected cells in situ in HCMV-infected placenta. Taken together, our results present the first evidence suggesting the involvement of dNK cells in controlling HCMV intrauterine infection and provide insights into the mechanisms through which these cells may operate to limit the spreading of viral infection to fetal tissues.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Decidua/immunology , Killer Cells, Natural/immunology , CD56 Antigen/metabolism , Cell Line , Cytomegalovirus Infections/congenital , Cytomegalovirus Infections/metabolism , Decidua/cytology , Decidua/virology , Fas Ligand Protein/metabolism , Female , HEK293 Cells , Humans , Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily C/immunology , NK Cell Lectin-Like Receptor Subfamily C/metabolism , NK Cell Lectin-Like Receptor Subfamily D/immunology , NK Cell Lectin-Like Receptor Subfamily D/metabolism , NK Cell Lectin-Like Receptor Subfamily K/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Placentation , Pregnancy , Pregnancy Trimester, First , TNF-Related Apoptosis-Inducing Ligand/metabolism , Uterus
18.
J Reprod Immunol ; 88(2): 170-5, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21277025

ABSTRACT

NK cells present in the peripheral blood respond rapidly to pathogens or pathogen-infected cells by various means including cytotoxicity and production of cytokines. Whether decidual NK (dNK) cells are able to play a similar role when the pregnant uterus is infected by viruses is still largely unknown. Decidual NK cells are generally considered as poorly cytotoxic when compared to their peripheral blood counterparts. However, we have recently demonstrated that freshly isolated dNK cells from healthy early pregnant uterus do have a cytotoxic potential mediated by the specific engagement of NKp46 activating receptor. We further found that the co-engagement of CD94/NKG2A inhibiting receptor drastically inhibits the cytolytic function of dNK. This latter observation suggests that in situ the CD94/NKG2A receptor interaction with its HLA-E specific ligand is a dominant negative regulatory mechanism that prevents unwanted dNK cell cytotoxicity in non-infected pregnant uterus. How do dNK cells behave when they are activated by virus-infected cells present at the maternal-fetal interface? Largely based on data obtained from circulating NK cells, this review briefly discusses the following questions: Does uterine viral infection promote decidual NK cell proliferative capacity in situ? Are dNK cells able to kill virus-infected autologous decidual target cells and thus limit the virus spreading to the fetus? Which viral-mediated signal(s) and molecular interactions may subvert inhibition of dNK cytotoxic potential? Does uterine viral infection promote decidual NK cell secretion of cytokines and chemokines that boost the anti-viral immune response?


Subject(s)
Cytotoxicity, Immunologic , Decidua/immunology , Killer Cells, Natural/immunology , Pregnancy Complications, Infectious/immunology , Virus Diseases/immunology , Animals , Antigen Presentation , Antigens, Viral/immunology , Blood Circulation , Decidua/virology , Female , Humans , Immune Evasion , Lymphocyte Activation , Placental Circulation , Pregnancy
19.
J Reprod Immunol ; 82(2): 142-7, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19615756

ABSTRACT

Natural killer (NK) cells represent the major lymphocyte population in the decidua basalis of the human uterus during healthy early pregnancy. The activity of decidual NK (dNK) cells and their activation status are different from those of peripheral blood (PB)-NK cells; i.e. dNK cells exhibit a unique phenotype. Decidual NK cells have been defined as CD56(bright), CD16(neg), and more recently CD160(neg). They express a unique repertoire of NK cell receptors, identical among all donors tested. Decidual NK cells express in particular NKp46-, NKp30- and NKp44-activating receptors, contrasting with PB-NK cells which are devoid of NKp44-activating receptors. Specific engagement of each of these three so-called natural cytotoxicity receptors in dNK cells has important functional consequences in terms of cytokine, chemokine and angiogenic factor secretion as well as cytotoxic potential. Strikingly, and in contrast with PB-NK cells, engagement of NKp46- but not NKp30-activating receptor on freshly isolated dNK cells triggers cytotoxicity. Such cytotoxic potential of dNK cells is negatively controlled by NKG2A inhibitory receptor co-engagement. This suggests that in situ, dNK cells cannot kill trophoblast cells during normal pregnancy. Whether such NKG2A-mediated inhibition is abolished during pregnancies complicated by pathologies including viral infection is an interesting hypothesis that remains to be tested.


Subject(s)
Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Receptors, Natural Cytotoxicity Triggering/metabolism , Antigens, CD/biosynthesis , Cells, Cultured , Cytotoxicity, Immunologic/immunology , Decidua/pathology , Feedback, Physiological , Female , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , NK Cell Lectin-Like Receptor Subfamily C/immunology , Pregnancy , Receptors, Natural Cytotoxicity Triggering/chemistry , Receptors, Natural Cytotoxicity Triggering/immunology
20.
Bull Acad Natl Med ; 193(5): 1029-41; discussion 1041-2, 1067-8, 2009 May.
Article in French | MEDLINE | ID: mdl-20120385

ABSTRACT

Pregnancy represents an immunological paradox, as underlined by the Nobel prize laureate Peter Medawar in the 1950s. This paradox is generating renewed interest with insights obtained in studies of pregnant mice and in ex vivo experiments performed with human cells and tissues. A number of molecular mechanisms have been discovered that prevent maternal placental immune effector cells located at the maternal-fetal interface from attacking fetus-derived cells. For example, maternal alloantibodies directed against paternal alloantigens expressed by the trophoblast are blocked by complement-inhibiting proteins, and maternal B cells specific for these paternal antigens are partially deleted Maternal antipaternal CD8+ cytotoxic T cells are inefficient, owing to the lack of HLA-A and HLA-B molecule expression on trophoblast target cells, together with the action of local immunosuppressive molecules, and transient tolerance of paternal alloantigens. NK cells present in the pregnant uterus and directed against fetus-derived trophoblast cells exhibit little if any cytotoxic potential. Interestingly, decidual NK cellltrophoblast interactions appear to play a physiological role in vascular uterine remodeling and in subsequent placental development. Most possible combinations of uterine NK KIR receptors and fetal HLA-C molecules expressed by the trophoblast are compatible with normal pregnancy, but the risk of severe preeclampsia appears to be far higher than normal when the mother's uterine NK cells do not express activating KIR (AA genotype) and when her fetus possesses group C2 HLA-C molecules.


Subject(s)
Pregnancy/immunology , Animals , Female , Humans , Killer Cells, Natural/immunology , T-Lymphocytes/immunology , Trophoblasts/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...