Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
iScience ; 24(4): 102331, 2021 Apr 23.
Article in English | MEDLINE | ID: mdl-33889824

ABSTRACT

In order to ascertain the significance of transmembrane tumor necrosis factor (tmTNF) reverse signaling in vivo, we generated a triple transgenic mouse model (3TG, TNFR1-/-, TNFR2-/-, and tmTNFKI/KI) in which all canonical tumor necrosis factor (TNF) signaling was abolished. In bone-marrow-derived macrophages harvested from these mice, various anti-TNF biologics induced the expression of genes characteristic of alternative macrophages and also inhibited the expression of pro-inflammatory cytokines mainly through the upregulation of arginase-1. Injections of TNF inhibitors during arthritis increased pro-resolutive markers in bone marrow precursors and joint cells leading to a decrease in arthritis score. These results demonstrate that the binding of anti-TNF biologics to tmTNF results in decreased arthritis severity. Collectively, our data provide evidence for the significance of tmTNF reverse signaling in the modulation of arthritis. They suggest a complementary interpretation of anti-TNF biologics effects in the treatment of inflammatory diseases and pave the way to studies focused on new arginase-1-dependent therapeutic targets.

2.
Nat Immunol ; 21(9): 983-997, 2020 09.
Article in English | MEDLINE | ID: mdl-32690951

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are a major source of type I interferon (IFN-I). What other functions pDCs exert in vivo during viral infections is controversial, and more studies are needed to understand their orchestration. In the present study, we characterize in depth and link pDC activation states in animals infected by mouse cytomegalovirus by combining Ifnb1 reporter mice with flow cytometry, single-cell RNA sequencing, confocal microscopy and a cognate CD4 T cell activation assay. We show that IFN-I production and T cell activation were performed by the same pDC, but these occurred sequentially in time and in different micro-anatomical locations. In addition, we show that pDC commitment to IFN-I production was marked early on by their downregulation of leukemia inhibitory factor receptor and was promoted by cell-intrinsic tumor necrosis factor signaling. We propose a new model for how individual pDCs are endowed to exert different functions in vivo during a viral infection, in a manner tightly orchestrated in time and space.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Herpesviridae Infections/immunology , Muromegalovirus/physiology , Animals , Cells, Cultured , Interferon Type I/metabolism , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Sequence Analysis, RNA , Signal Transduction , Single-Cell Analysis , Tumor Necrosis Factor-alpha/metabolism
3.
J Bone Miner Res ; 35(4): 753-765, 2020 04.
Article in English | MEDLINE | ID: mdl-31834954

ABSTRACT

Increased osteoclastogenesis is a common feature of bone erosion, notably in osteoporosis but also in inflammatory diseases such as rheumatoid arthritis (RA) and osteoarticular infections. Human cytomegalovirus (HCMV) infection has been described to impair monocyte differentiation into macrophages and dendritic cells. However, its effect on monocyte-derived osteoclasts is yet to be determined. We showed here that in vitro HCMV infection is associated with an inhibition of osteoclastogenesis through decreased expression of colony stimulating factor 1 receptor (CSF-1R) and RANK in monocytes, which was mediated by an upregulation of quaking I-5 protein (QKI-5), a cellular RNA-interacting protein. We found that deliberate QKI5 overexpression in the absence of HCMV infection is able to decrease CSF-1R and RANK expression, leading to osteoclastogenesis inhibition. Finally, by using lentiviral vectors in a calvarial bone erosion mouse model, we showed that QKI5 inhibits bone degradation. This work identifies QKI5 as a strong inhibitor of bone resorption. Future research will point out whether QKI5 could be a target for bone pathologies. © 2019 American Society for Bone and Mineral Research.


Subject(s)
Bone Resorption , Osteogenesis , Cell Differentiation , Humans , Macrophage Colony-Stimulating Factor , Macrophages , Osteoclasts , RANK Ligand , RNA-Binding Proteins
5.
Front Immunol ; 10: 1482, 2019.
Article in English | MEDLINE | ID: mdl-31316519

ABSTRACT

Introduction: Adipose-derived mesenchymal stem cells (ADSC) have been shown to have remarkable immune-modulating effects. However, their efficacy in clinical trials has yet to be fully demonstrated. This could be due to a lack of a proper inflammatory environment in vivo that primes ADSC. Here, we define how the articular microenvironment of rheumatoid arthritis (RA) patients modulates the therapeutic efficiency of ADSC. Methods: Synovial fluids (SF) were collected from 8 RA patients, 2 Spondyloarthritis patients and one control synovial fluid from a patient undergoing traumatic-related surgery. SF inflammatory status was determined by routine analysis and quantification of pro-inflammatory cytokines. ADSC were first treated with SF and ADSC proliferation and gene expression of immunomodulatory factors was evaluated. In order to determine the mechanisms underlying the effect of SF on ADSC, tumor necrosis factor (TNF), interleukin-6 (IL-6), and NF-κB neutralization assays were performed. To evaluate the effect of SF on ADSC functions, ADSC were pre-treated with SF and then co-cultured with either macrophages or T cells. The modulation of their phenotype was assessed by flow cytometry. Results: Pro-inflammatory RASF maintained the proliferative capacity of ADSC and upregulated the gene expression of cyclooxygenase-2 (COX2), indoleamine-1,2-dioxygenase (IDO), interleukin-6 (IL-6), tumor-necrosis factor stimulated gene 6 (TSG6), intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), and programmed death-ligand 1 (PD-L1), all factors involved in ADSC immunomodulatory potential. The RASF-induced gene expression was mainly mediated by TNF alone or in combination with IL-6 and signaled through the NF-κB pathway. Conditioning ADSC with pro-inflammatory RASF enhanced their ability to induce CD4+Foxp3+CD25high regulatory T cells (Tregs) and inhibit pro-inflammatory markers CD40 and CD80 in activated macrophages. Conclusions: Inflammatory synovial fluids from RA patients had the capacity to modulate ADSC response, to induce Tregs and modulate the phenotype of macrophages. The clinical use of ADSC in affected joints should take into account the influence of the local articular environment on their potential. Having a sufficient pro-inflammatory microenvironment will determine whether optimal immunoregulatory response should be expected. Direct ADSC intra-articular delivery to patients could be a potential strategy to properly prime their immunomodulatory potential and enhance their clinical benefits.


Subject(s)
Adipose Tissue/cytology , Arthritis, Rheumatoid/immunology , Immunomodulation , Mesenchymal Stem Cells/immunology , NF-kappa B/immunology , Synovial Fluid/immunology , Tumor Necrosis Factor-alpha/immunology , Adipose Tissue/immunology , Adult , Aged , Child, Preschool , Humans , Infant , Infant, Newborn , Macrophages/immunology , Middle Aged
6.
Front Immunol ; 10: 3, 2019.
Article in English | MEDLINE | ID: mdl-30713533

ABSTRACT

Macrophages contribute to the pathogenesis of rheumatoid arthritis (RA). They can display different states of activation or "polarization," notably the so-called inflammatory "M1" and the various alternative "M2" polarizations, characterized by distinct functions. Data regarding the effects of RA anti-cytokine biological disease-modifying anti-rheumatic drugs (bDMARDs) on macrophage polarization are scarce. We aimed to assess in vitro modulation of macrophage polarization by bDMARDs targeting pro-inflammatory cytokines in RA. We generated monocyte derived macrophages using blood samples from 20 RA patients with active RA and 30 healthy controls. We evaluated in vitro the impact on M1 inflammatory macrophages of: etanercept (ETA), adalimumab (ADA), certolizumab (CZP), tocilizumab (TCZ), and rituximab (RTX). We assessed the impact on macrophage polarization using flow cytometry and RTqPCR to study the expression of surface markers and perform functional studies of cytokine production, phagocytosis, and negative feedback control of inflammation. Among evaluated bDMARDs, anti-TNF agents modulated the polarization of inflammatory macrophages by decreasing inflammatory surface markers (CD40, CD80) and favoring alternative markers (CD16, CD163, MerTK). Anti-TNF agents also induced alternative functions in macrophages activated in inflammatory condition with (i) the inhibition of inflammatory cytokines (TNF, IL-6, IL-12), (ii) an increase in phagocytosis. These findings were mechanistically related to an increase in early IL-10 production, responsible for higher negative feedback control of inflammation involving SOCS3 and Gas6. This IL-10 effect was STAT3-dependent. Anti-TNF agents not only inhibit in vitro inflammatory functions of macrophages, but also favor resolution of inflammation through polarization toward alternative features specifically involving the IL-10/STAT3 axis.


Subject(s)
Interleukin-10/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , STAT3 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Aged , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/therapy , Biomarkers , Cells, Cultured , Cytokines/metabolism , Female , Gene Expression Profiling , Humans , Immunophenotyping , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Macrophages/drug effects , Male , Middle Aged , Tumor Necrosis Factor-alpha/antagonists & inhibitors
7.
Arthritis Res Ther ; 20(1): 229, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30314507

ABSTRACT

Tumor necrosis factor (TNF) is a pleiotropic cytokine involved in many aspects of immune regulation. Anti-TNF biological therapy has been considered a breakthrough in the treatment of chronic autoimmune diseases, such as rheumatoid arthritis (RA). In this review, because of the major involvement of T cells in RA pathogenesis, we discuss the effects of anti-TNF biotherapy on T-cell responses in RA patients. We also outline the potential fields for future research in the area of anti-TNF therapy in RA.This could be useful to better understand the therapeutic efficiency and the side effects that are encountered in RA patients. Better targeting of T cells in RA could help set more specific anti-TNF strategies and develop prediction tools for response.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/metabolism , Biological Products/therapeutic use , T-Lymphocytes/physiology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Animals , Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/drug therapy , Biological Products/pharmacology , Biological Therapy/methods , Biological Therapy/trends , Humans , T-Lymphocytes/drug effects , Treatment Outcome
8.
Nucleic Acids Res ; 46(17): 8788-8802, 2018 09 28.
Article in English | MEDLINE | ID: mdl-29955894

ABSTRACT

During cell division, maintenance of chromatin features from the parental genome requires their proper establishment on its newly synthetized copy. The loss of epigenetic marks within heterochromatin, typically enriched in repetitive elements, endangers genome stability and permits chromosomal rearrangements via recombination. However, how histone modifications associated with heterochromatin are maintained across mitosis remains poorly understood. KAP1 is known to act as a scaffold for a repressor complex that mediates local heterochromatin formation, and was previously demonstrated to play an important role during DNA repair. Accordingly, we investigated a putative role for this protein in the replication of heterochromatic regions. We first found that KAP1 associates with several DNA replication factors including PCNA, MCM3 and MCM6. We then observed that these interactions are promoted by KAP1 phosphorylation on serine 473 during S phase. Finally, we could demonstrate that KAP1 forms a complex with PCNA and the histone-lysine methyltransferase Suv39h1 to reinstate heterochromatin after DNA replication.


Subject(s)
Chromatin Assembly and Disassembly/genetics , DNA Replication/physiology , Heterochromatin/metabolism , Tripartite Motif-Containing Protein 28/physiology , Animals , Cells, Cultured , HEK293 Cells , HeLa Cells , Humans , K562 Cells , Methyltransferases/metabolism , Mice , NIH 3T3 Cells , Phosphorylation , Proliferating Cell Nuclear Antigen/metabolism , Protein Processing, Post-Translational , Repressor Proteins/metabolism
9.
PLoS Pathog ; 12(4): e1005547, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27078877

ABSTRACT

Congenital infection by human cytomegalovirus (HCMV) is a leading cause of permanent sequelae of the central nervous system, including sensorineural deafness, cerebral palsies or devastating neurodevelopmental abnormalities (0.1% of all births). To gain insight on the impact of HCMV on neuronal development, we used both neural stem cells from human embryonic stem cells (NSC) and brain sections from infected fetuses and investigated the outcomes of infection on Peroxisome Proliferator-Activated Receptor gamma (PPARγ), a transcription factor critical in the developing brain. We observed that HCMV infection dramatically impaired the rate of neuronogenesis and strongly increased PPARγ levels and activity. Consistent with these findings, levels of 9-hydroxyoctadecadienoic acid (9-HODE), a known PPARγ agonist, were significantly increased in infected NSCs. Likewise, exposure of uninfected NSCs to 9-HODE recapitulated the effect of infection on PPARγ activity. It also increased the rate of cells expressing the IE antigen in HCMV-infected NSCs. Further, we demonstrated that (1) pharmacological activation of ectopically expressed PPARγ was sufficient to induce impaired neuronogenesis of uninfected NSCs, (2) treatment of uninfected NSCs with 9-HODE impaired NSC differentiation and (3) treatment of HCMV-infected NSCs with the PPARγ inhibitor T0070907 restored a normal rate of differentiation. The role of PPARγ in the disease phenotype was strongly supported by the immunodetection of nuclear PPARγ in brain germinative zones of congenitally infected fetuses (N = 20), but not in control samples. Altogether, our findings reveal a key role for PPARγ in neurogenesis and in the pathophysiology of HCMV congenital infection. They also pave the way to the identification of PPARγ gene targets in the infected brain.


Subject(s)
Cytomegalovirus Infections/congenital , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/metabolism , Neural Stem Cells/virology , Neurogenesis/physiology , PPAR gamma/metabolism , Blotting, Western , Cell Differentiation/physiology , Chromatin Immunoprecipitation , Chromatography, High Pressure Liquid , Fluorescent Antibody Technique , Humans , Microscopy, Electron, Transmission , Neural Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tandem Mass Spectrometry
10.
PLoS One ; 10(7): e0132627, 2015.
Article in English | MEDLINE | ID: mdl-26171612

ABSTRACT

INTRODUCTION: Congenital infection by human cytomegalovirus (HCMV) is a leading cause of congenital abnormalities of the central nervous system. Placenta infection by HCMV allows for viral spread to fetus and may result in intrauterine growth restriction, preeclampsia-like symptoms, or miscarriages. We previously reported that HCMV activates peroxisome proliferator-activated receptor gamma (PPARγ) for its own replication in cytotrophoblasts. Here, we investigated the molecular bases of PPARγ activation in infected cytotrophoblasts. RESULTS: We show that onboarded cPLA2 carried by HCMV particles is required for effective PPARγ activation in infected HIPEC cytotrophoblasts, and for the resulting inhibition of cell migration. Natural PPARγ agonists are generated by PLA2 driven oxidization of linoleic and arachidonic acids. Therefore, using HPLC coupled with mass spectrometry, we disclosed that cellular and secreted levels of 13-hydroxyoctadecadienoic acid (13-HODE) and 15-hydroxyeicosatetraenoic acid (15-HETE) were significantly increased in and from HIPEC cytotrophoblasts at soon as 6 hours post infection. 13-HODE treatment of uninfected HIPEC recapitulated the effect of infection (PPARγ activation, migration impairment). We found that infection of histocultures of normal, first-term, human placental explants resulted in significantly increased levels of secreted 15-HETE and 13-HODE. CONCLUSION: Our findings reveal that 15-HETE and 13-HODE could be new pathogenic effectors of HCMV congenital infection They provide a new insight about the pathogenesis of congenital infection by HCMV.


Subject(s)
Cytomegalovirus/physiology , Hydroxyeicosatetraenoic Acids/metabolism , Linoleic Acids/metabolism , PPAR gamma/agonists , Trophoblasts/metabolism , Trophoblasts/virology , Cell Movement , Female , Humans , PPAR gamma/metabolism , Phospholipases A2, Cytosolic/metabolism , Pregnancy , Tissue Culture Techniques , Trophoblasts/cytology
11.
Elife ; 42015 Apr 07.
Article in English | MEDLINE | ID: mdl-25846574

ABSTRACT

Human cytomegalovirus (HCMV) is a highly prevalent pathogen that induces life-long infections notably through the establishment of latency in hematopoietic stem cells (HSC). Bouts of reactivation are normally controlled by the immune system, but can be fatal in immuno-compromised individuals such as organ transplant recipients. Here, we reveal that HCMV latency in human CD34(+) HSC reflects the recruitment on the viral genome of KAP1, a master co-repressor, together with HP1 and the SETDB1 histone methyltransferase, which results in transcriptional silencing. During lytic infection, KAP1 is still associated with the viral genome, but its heterochromatin-inducing activity is suppressed by mTOR-mediated phosphorylation. Correspondingly, HCMV can be forced out of latency by KAP1 knockdown or pharmacological induction of KAP1 phosphorylation, and this process can be potentiated by activating NFkB with TNF-α. These results suggest new approaches both to curtail CMV infection and to purge the virus from organ transplants.


Subject(s)
Cytomegalovirus/physiology , Repressor Proteins/metabolism , Virus Latency/physiology , Antigens, CD34/metabolism , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , Cytomegalovirus/genetics , Genome, Viral , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/virology , Histone-Lysine N-Methyltransferase , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Monocytes/virology , NF-kappa B/metabolism , Phosphorylation , Protein Methyltransferases , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Tripartite Motif-Containing Protein 28 , Virus Replication
12.
Genome Res ; 24(8): 1251-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24879558

ABSTRACT

Endogenous retroelements (EREs) account for about half of the mouse or human genome, and their potential as insertional mutagens and transcriptional perturbators is suppressed by early embryonic epigenetic silencing. Here, we asked how ERE control is maintained during the generation of induced pluripotent stem cells (iPSCs), as this procedure involves profound epigenetic remodeling. We found that all EREs tested were markedly up-regulated during the reprogramming of either mouse embryonic fibroblasts, human CD34(+) cells, or human primary hepatocytes. At the iPSC stage, EREs of some classes were repressed, whereas others remained highly expressed, yielding a pattern somewhat reminiscent of that recorded in embryonic stem cells. However, variability persisted between individual iPSC clones in the control of specific ERE integrants. Both during reprogramming and in iPS cells, the up-regulation of specific EREs significantly impacted on the transcription of nearby cellular genes. While transcription triggered by specific ERE integrants at highly precise developmental stages may be an essential step toward obtaining pluripotent cells, the broad and unspecific unleashing of the repetitive genome observed here may contribute to the inefficiency of the reprogramming process and to the phenotypic heterogeneity of iPSCs.


Subject(s)
Endogenous Retroviruses/genetics , Induced Pluripotent Stem Cells/physiology , Transcriptome , Animals , Cells, Cultured , Cellular Reprogramming , Gene Silencing , Humans , Mice , Up-Regulation
14.
Science ; 340(6130): 350-3, 2013 Apr 19.
Article in English | MEDLINE | ID: mdl-23493425

ABSTRACT

During hematopoiesis, lineage- and stage-specific transcription factors work in concert with chromatin modifiers to direct the differentiation of all blood cells. We explored the role of KRAB-containing zinc finger proteins (KRAB-ZFPs) and their cofactor KAP1 in this process. In mice, hematopoietic-restricted deletion of Kap1 resulted in severe hypoproliferative anemia. Kap1-deleted erythroblasts failed to induce mitophagy-associated genes and retained mitochondria. This was due to persistent expression of microRNAs (miRNAs) targeting mitophagy transcripts, itself secondary to a lack of repression by stage-specific KRAB-ZFPs. The KRAB/KAP1-miRNA regulatory cascade is evolutionarily conserved, as it also controls mitophagy during human erythropoiesis. Thus, a multilayered transcription regulatory system is present, in which protein- and RNA-based repressors are superimposed in combinatorial fashion to govern the timely triggering of an important differentiation event.


Subject(s)
Autophagy/genetics , Erythroblasts/metabolism , Erythropoiesis/genetics , MicroRNAs/metabolism , Mitochondria/physiology , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Zinc Fingers , Anemia/genetics , Animals , Erythroblasts/ultrastructure , Female , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Mitochondria/genetics , Mitochondrial Proteins/metabolism , Nuclear Proteins/genetics , Repressor Proteins/genetics , Tripartite Motif-Containing Protein 28
15.
Blood ; 118(26): 6783-92, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22031867

ABSTRACT

A primary HCMV infection or virus reactivation may cause severe disease in hosts with a deficient immune system. The virus can disturb both innate and adaptive immunity by targeting dendritic cell (DC) functions. Monocytes, the precursors of DCs in vivo (MoDCs), are the primary targets of HCMV; they can also harbor latent virus. The DCs generated from infected monocytes (CMV-MoDCs) have an altered phenotype and functional defects. We have shown that CMV-MoDCs do not secrete IL-12 in response to lipopolysaccharide stimulation, cannot ingest dead cells, induce T(H)1 differentiation, or the proliferation of naive allogeneic CD4(+) T cells. We found that the GM-CSF signaling in an entire population of CMV-MoDCs was impaired, although only half of the cells were productively infected, and that IL-6 secretion and suppressors of cytokine signaling 3 induction contributed to this bystander effect. We also showed that MoDCs derived ex vivo from monocytes of viremic patients had the same altered phenotype as CMV-MoDCs, including decreased STAT5 phosphorylation, indicating defective GM-CSF signaling. We have thus described a new mechanism of HCMV-induced immunosupression, indicated how infection may disturb both GM-CSF-dependent physiologic processes and proposed GM-CSF-based therapeutic approaches.


Subject(s)
Cytomegalovirus/immunology , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Monocytes/immunology , Blotting, Western , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Line , Cells, Cultured , Cytomegalovirus/physiology , Dendritic Cells/metabolism , Dendritic Cells/virology , Flow Cytometry , Gene Expression/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Host-Pathogen Interactions/immunology , Humans , Immunophenotyping , Interleukin-13/immunology , Interleukin-13/metabolism , Interleukin-13/pharmacology , Interleukin-6/immunology , Interleukin-6/metabolism , Interleukin-6/pharmacology , Monocytes/metabolism , Monocytes/virology , Paracrine Communication/immunology , Phagocytosis/drug effects , Phagocytosis/immunology , Phosphorylation/drug effects , RNA Interference , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/immunology , STAT5 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/immunology , Suppressor of Cytokine Signaling Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors
16.
J Reprod Immunol ; 90(1): 41-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21704384

ABSTRACT

The peroxisome proliferator-activated receptor-γ (PPARγ) is a member of the nuclear receptor superfamily that controls in a ligand-dependent manner the expression of a large array of genes involved in the control of energy homeostasis and in cell differentiation, proliferation, apoptosis, and the inflammatory process. Unexpectedly, genetic studies performed in mice established that PPARγ is essential for placental development. In the human placenta, PPARγ is specifically expressed in the trophoblast, both endocrine villous and invasive extravillous cytotrophoblasts (EVCT). Activation of PPARγ induces accumulation of lipids, villous trophoblast differentiation and inhibits trophoblast invasiveness. Oxidized LDLs that contain potential PPARγ ligands, but not native LDL, induce PPARγ transcriptional activity and inhibit trophoblast invasion in vitro. Recently, human cytomegalovirus (HCMV) was shown to activate trophoblastic PPARγ for its own replication and consequently inhibits invasiveness of infected cytotrophoblasts. Analysis of PPARγ target genes revealed trophoblastic factors described to control trophoblast invasiveness and surprisingly chorionic gonadotropin hormone (hCG), known to be mainly produced by the endocrine villous trophoblast. Analysis of hCG gene expression revealed opposite regulation by PPARγ in the two trophoblast subtypes. Finally, a hyperglycosylated form of hCG (hCG-H) only produced by invasive EVCT was shown to promote trophoblast invasion. Together, these data underscore the major role of PPARγ and its target genes, such as hCG, in the control of human trophoblast differentiation and invasion, and suggest that over-activation of this nuclear receptor following HCMV infection or by excess of ligands at the maternal-fetal interface could impair implantation and placentation and therefore embryonic development.


Subject(s)
Cell Differentiation , PPAR gamma/metabolism , Trophoblasts/cytology , Trophoblasts/metabolism , Animals , Cells, Cultured , Chorionic Gonadotropin/biosynthesis , Cytomegalovirus/metabolism , Embryo Implantation , Female , Gene Expression Regulation, Developmental , Humans , Lipids/biosynthesis , Mice , Peroxisome Proliferator-Activated Receptors/metabolism , Placentation/physiology , Pregnancy
17.
J Virol ; 84(6): 2946-54, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20042507

ABSTRACT

Human cytomegalovirus (HCMV) contributes to pathogenic processes in immunosuppressed individuals, in fetuses, and in neonates. In the present report, by using reporter gene activation assays and confocal microscopy in the presence of a specific antagonist, we show for the first time that HCMV infection induces peroxisome proliferator-activated receptor gamma (PPARgamma) transcriptional activity in infected cells. We demonstrate that the PPARgamma antagonist dramatically impairs virus production and that the major immediate-early promoter contains PPAR response elements able to bind PPARgamma, as assessed by electrophoretic mobility shift and chromatin immunoprecipitation assays. Due to the key role of PPARgamma in placentation and its specific trophoblast expression within the human placenta, we then provided evidence that by activating PPARgamma human cytomegalovirus dramatically impaired early human trophoblast migration and invasiveness, as assessed by using well-established in vitro models of invasive trophoblast, i.e., primary cultures of extravillous cytotrophoblasts (EVCT) isolated from first-trimester placentas and the EVCT-derived cell line HIPEC. Our data provide new clues to explain how early infection during pregnancy could impair implantation and placentation and therefore embryonic development.


Subject(s)
Cell Movement/physiology , Cytomegalovirus/metabolism , PPAR gamma/metabolism , Placenta/cytology , Trophoblasts/physiology , Trophoblasts/virology , Virus Replication/physiology , Base Sequence , Cells, Cultured , Cytomegalovirus/genetics , Embryo Implantation/physiology , Female , Gestational Age , Humans , Molecular Sequence Data , PPAR gamma/genetics , Pregnancy , Transcription, Genetic , Trophoblasts/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...