73 results on '"Jean-Charles, Guéry"'
Search Results
2. Cav1.4 calcium channels control cytokine production by human peripheral TH17 cells and psoriatic skin-infiltrating T cells
- Author
-
Daniel Redoules, Eléonore Gravier, Stéphanie Bosch, Magali Savignac, Marie Tauber, Fabrice Lestienne, Marie-Dominique Thouvenin, Marion Mars, Christian Rouvière, Simon Lachambre, Carle Paul, Alexia Brocario, Catherine Leclerc, Marine Babin, Marc Moreau, Clara Douzal, Lucette Pelletier, Jean-Charles Guéry, Hélène Duplan, and Isabelle Néant
- Subjects
business.industry ,medicine.medical_treatment ,Calcium channel ,Immunology ,Human skin ,Inflammation ,medicine.disease ,Proinflammatory cytokine ,medicine.anatomical_structure ,Cytokine ,RAR-related orphan receptor gamma ,Psoriasis ,Cancer research ,Immunology and Allergy ,Medicine ,medicine.symptom ,business ,Keratinocyte - Abstract
Background Type-17 inflammation characterizes psoriasis, a chronic skin disease. Because several inflammatory cytokines contribute to psoriasis pathogenesis, inhibiting the simultaneous production of these cytokines in TH17 cells may be beneficial in psoriasis. We found that Cav1.4, encoded by CACNA1F, was the only Cav1 calcium channel expressed in TH17 cells. Objective We sought to investigate the role of Cav1.4 expression in early TH17-activation events and effector functions, as well as its association with TH17 signature genes in lesional psoriatic (LP) skins. Methods Transcriptional gene signatures associated with CACNA1F expression were examined in LP skins by RT-PCR and in situ hybridization. Cav1 inhibitor and/or shRNA lentivectors were used to assess the contribution of Cav1.4 in TH17 activation and effector functions in a 3-dimensional skin reconstruction model. Results CACNA1F expression correlated with inflammatory cytokine expression that characterizes LP skins and was preferentially associated with RORC expression in CD4+ and CD4− cells from LP biopsies. Nicardipine, a Cav1 channel antagonist, markedly reduced inflammatory cytokine production by TH17 cells from blood or LP skin. This was associated with decreased TCR-induced early calcium events at cell membrane and proximal signaling events. The knockdown of Cav1.4 in TH17 cells impaired cytokine production. Finally, Cav1 inhibition reduced the expression of the keratinocyte genes characteristic of TH17-mediated psoriasis inflammation in human skin equivalents. Conclusions Cav1.4 channels promote TH17-cell functions both at the periphery and in inflammatory psoriatic skin.
- Published
- 2022
- Full Text
- View/download PDF
3. Influence of X chromosome in sex-biased autoimmune diseases
- Author
-
Charles-Henry Miquel, Berenice Faz-Lopez, and Jean-Charles Guéry
- Subjects
Immunology ,Immunology and Allergy - Published
- 2023
- Full Text
- View/download PDF
4. Review for 'Sex differences regulate immune responses in experimental autoimmune encephalomyelitis and multiple sclerosis'
- Author
-
Jean-Charles Guéry
- Subjects
Immune system ,Multiple sclerosis ,Experimental autoimmune encephalomyelitis ,Immunology ,medicine ,Biology ,medicine.disease - Published
- 2021
- Full Text
- View/download PDF
5. Hydroxychloroquine inhibits proteolytic processing of endogenous TLR7 protein in human primary plasmacytoid dendritic cells
- Author
-
Jean-Charles Guéry, Mariette F. Ducatez, and Claire Cenac
- Subjects
Endosome ,Immunology ,Endogeny ,Endosomes ,Cleavage (embryo) ,Peripheral blood mononuclear cell ,Cell Line ,Immune system ,Immunology and Allergy ,Humans ,Receptor ,biology ,SARS-CoV-2 ,Chemistry ,COVID-19 ,virus diseases ,RNA ,hemic and immune systems ,Dendritic Cells ,TLR7 ,COVID-19 Drug Treatment ,Cell biology ,Toll-Like Receptor 7 ,Proteolysis ,biology.protein ,Antibody ,Hydroxychloroquine - Abstract
Toll-like receptor 7 (TLR7) triggers antiviral immune responses through its capacity to recognize single-stranded RNA. Proteolytic cleavage of TLR7 protein is required for its functional maturation in the endosomal compartment. Structural studies demonstrated that the N- and C-terminal domains of TLR7 are connected and involved in ligand binding after cleavage. Hydroxychloroquine (HCQ), an antimalarial drug, has been studied for its antiviral effects. HCQ increases pH in acidic organelles and has been reported to potently inhibit endosomal TLR activation. Whether HCQ can prevent endogenous TLR7 cleavage in primary immune cells, such as plasmacytoid dendritic cells (pDCs), had never been examined. Here, using a validated anti-TLR7 antibody suitable for biochemical detection of native TLR7 protein, we show that HCQ-treatment of fresh PBMCs, CAL-1 leukemic and primary human pDCs inhibits TLR7 cleavage and results in accumulation of full-length protein. As a consequence, we observe an inhibition of pDC activation in response to TLR7 stimulation with synthetic ligands and viruses including inactivated SARS-CoV2, which we show herein activates pDCs through TLR7-signaling. Together, our finding suggests that the major pathway by which HCQ inhibits ssRNA-sensing by pDCs may rely on its capacity to inhibit endosomal acidification and the functional maturation of TLR7 protein.
- Published
- 2021
- Full Text
- View/download PDF
6. Sex Differences in Primary HIV Infection: Revisiting the Role of TLR7-Driven Type 1 IFN Production by Plasmacytoid Dendritic Cells in Women
- Author
-
Jean-Charles Guéry
- Subjects
Male ,Mini Review ,Immunology ,HIV Infections ,Polymorphism, Single Nucleotide ,Sex Factors ,Risk Factors ,sex bias ,Animals ,Humans ,innate immunity ,Sex Characteristics ,virus diseases ,HIV ,Dendritic Cells ,RC581-607 ,Protective Factors ,Viral Load ,Immunity, Innate ,Toll-like receptor 7 ,plasmacytoid dendritic cells ,Host-Pathogen Interactions ,Interferon Type I ,HIV-1 ,Female ,Immunologic diseases. Allergy ,type I IFN ,Signal Transduction - Abstract
Plasmacytoid dendritic cells (pDCs) produce type I interferon (IFN-I) during HIV-1 infection in response to TLR7 stimulation. However, IFN-I-signaling has been shown to play opposite effects in HIV-1 and SIV infection. TLR7-driven type I interferon production in pDCs is higher in women than in men due to the cell-intrinsic actions of estrogen and X-chromosome complement. Indeed, TLR7 is encoded on the X-chromosome, and the TLR7 gene escapes the X-chromosome inactivation in immune cells of women which express significantly higher levels of TLR7 protein than male cells. Following HIV infection, women have a lower viremia during acute infection and exhibit stronger antiviral responses than men, which has been attributed to the increased capacity of female pDCs to produce IFN-α upon TLR7-stimulation. However, a deleterious functional impact of an excessive TLR7 response on acute viremia in women has been recently revealed by the analysis of the frequent rs179008 c.32A>T SNP of TLR7. This SNP was identified as a sex-specific protein abundance quantitative trait locus (pQTL) causing a difference in the TLR7 protein dosage and effector function in females only. T allele expression was associated with a lower TLR7 protein synthesis, blunted production of IFN-α by pDCs upon TLR7 stimulation, and an unexpectedly lower viral load during primary HIV-1 infection in women. In the present review, the author will revisit the role of TLR7-driven pDC innate function in the context of HIV-1 infection to discuss at what stage of primary HIV-1 infection the TLR7 rs179008 T allele is likely to be protective in women.
- Published
- 2021
7. Separation of the Ca V 1.2‐Ca V 1.3 calcium channel duo prevents type 2 allergic airway inflammation
- Author
-
Joerg Striessnig, Marion Mars, Brice Ronsin, Magali Savignac, Marc Moreau, Jean-Charles Guéry, Lucette Pelletier, Marine Michelet, Nicolas Giang, Antoine Magnan, Geoffrey G. Murphy, José E. Mejía, Grégory Bouchaud, Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre de Physiopathologie Toulouse Purpan (CPTP), Centre de biologie du développement (CBD), Centre National de la Recherche Scientifique (CNRS)-Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Centre de Biologie Intégrative (CBI), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Centre National de la Recherche Scientifique (CNRS)-Centre National de la Recherche Scientifique (CNRS), Unité de recherche sur les Biopolymères, Interactions Assemblages (BIA), Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Institut du thorax, Université de Nantes (UN)-IFR26-Institut National de la Santé et de la Recherche Médicale (INSERM), CHU Toulouse, Hôpital des Enfants, Unité de Gastroentérologie, Hépatologie et Nutrition, Département de Pédiatrie, Hôpital Purpan [Toulouse], CHU Toulouse [Toulouse]-CHU Toulouse [Toulouse], Eastern Michigan University, Universität Innsbruck [Innsbruck], Leopold Franzens Universität Innsbruck - University of Innsbruck, Austrian Science Fund (FWF) : P27809, Foundation for Medical Research : DEQ20180339187, and French Society of Allergology : A11013BS.
- Subjects
Calcium metabolism ,Th2 lymphocytes ,Voltage-dependent calcium channel ,Calcium channel ,T cell ,Immunology ,chemistry.chemical_element ,Tyrosine phosphorylation ,[SDV.MHEP.HEM]Life Sciences [q-bio]/Human health and pathology/Hematology ,Calcium ,asthma ,Ca(v)1 ,cytokines ,Cell biology ,chemistry.chemical_compound ,Calcium imaging ,medicine.anatomical_structure ,chemistry ,Transcription (biology) ,calcium channels ,cardiovascular system ,medicine ,Immunology and Allergy ,[SDV.IMM.ALL]Life Sciences [q-bio]/Immunology/Allergology ,signaling - Abstract
International audience; Background Voltage-gated calcium (Ca(v)1) channels contribute to T-lymphocyte activation. Ca(v)1.2 and Ca(v)1.3 channels are expressed in Th2 cells but their respective roles are unknown, which is investigated herein. Methods We generated mice deleted for Ca(v)1.2 in T cells or Ca(v)1.3 and analyzed TCR-driven signaling. In this line, we developed original fast calcium imaging to measure early elementary calcium events (ECE). We also tested the impact of Ca(v)1.2 or Ca(v)1.3 deletion in models of type 2 airway inflammation. Finally, we checked whether the expression of both Ca(v)1.2 and Ca(v)1.3 in T cells from asthmatic children correlates with Th2-cytokine expression. Results We demonstrated non-redundant and synergistic functions of Ca(v)1.2 and Ca(v)1.3 in Th2 cells. Indeed, the deficiency of only one channel in Th2 cells triggers TCR-driven hyporesponsiveness with weakened tyrosine phosphorylation profile, a strong decrease in initial ECE and subsequent reduction in the global calcium response. Moreover, Ca(v)1.3 has a particular role in calcium homeostasis. In accordance with the singular roles of Ca(v)1.2 and Ca(v)1.3 in Th2 cells, deficiency in either one of these channels was sufficient to inhibit cardinal features of type 2 airway inflammation. Furthermore, Ca(v)1.2 and Ca(v)1.3 must be co-expressed within the same CD4(+) T cell to trigger allergic airway inflammation. Accordingly with the concerted roles of Ca(v)1.2 and Ca(v)1.3, the expression of both channels by activated CD4(+) T cells from asthmatic children was associated with increased Th2-cytokine transcription. Conclusions Thus, Ca(v)1.2 and Ca(v)1.3 act as a duo, and targeting only one of these channels would be efficient in allergy treatment.
- Published
- 2021
- Full Text
- View/download PDF
8. TLR7 dosage polymorphism shapes interferogenesis and HIV-1 acute viremia in women
- Author
-
Jean-Charles Guéry, Jacques Izopet, Asma Essat, Arnoo Shaiykova, Caroline Passaes, Sophie Laffont, Ali Youness, Pascal Azar, Michaela Müller-Trutwin, José E. Mejía, Pierre Delobel, Laurence Meyer, Claire Cenac, Centre de Physiopathologie Toulouse Purpan ex IFR 30 et IFR 150 (CPTP), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre de Physiopathologie Toulouse Purpan (CPTP), Centre de recherche en épidémiologie et santé des populations (CESP), Université de Versailles Saint-Quentin-en-Yvelines (UVSQ)-Université Paris-Sud - Paris 11 (UP11)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Paul Brousse-Institut National de la Santé et de la Recherche Médicale (INSERM), Laboratoire de Virologie [Purpan], CHU Toulouse [Toulouse]-Institut Fédératif de Biologie (IFB) - Hôpital Purpan, Hôpital Purpan [Toulouse], CHU Toulouse [Toulouse]-CHU Toulouse [Toulouse]-Hôpital Purpan [Toulouse], CHU Toulouse [Toulouse], HIV, Inflammation et persistance, Institut Pasteur [Paris], Service maladies infectieuses et tropicales [CHU Purpan], CHU Toulouse [Toulouse]-CHU Toulouse [Toulouse], his work was supported by grants from the French National Agency for Research on AIDS and Viral Hepatitis (ANRS, EP-53 study), Fondation pour la Recherche Médicale (DEQ20180339187), and Conseil Régional Occitanie-Midi-Pyrénées. PA was supported by a fellowship from SIDACTION. MMT received grants from the ANRS and the Fondation Beytout., We gratefully acknowledge support from F. L’Faqihi, A.L. Iscache, and V. Duplan at the flow cytometry facility (CPTP), E. Lhuillier, C. Naylies, A. Emile, and Y. Lippi for DNA sequencing and RNA analyses at the GeT-Purpan and GeT-TRiX facilities, and the technical assistance of M. Requena, M. Cazabat, and R. Carcénac. We also thank Anaïs Bellin-Robert and Guilhem Vazzoler for assistance during plasmid cloning, G. von Heijne and H. Nielsen for comments on SignalP, H. Blanché-Koch for curating the CEPH sample panel, S. Lupold for the Metridia luciferase vector, and W. Chowdhury and S. Chavanas for advice on luciferase assays., Université de Toulouse (UT)-Université de Toulouse (UT)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Laboratoire Virologie [CHU Toulouse], Institut Fédératif de Biologie (IFB), Centre Hospitalier Universitaire de Toulouse (CHU Toulouse)-Centre Hospitalier Universitaire de Toulouse (CHU Toulouse)-Pôle Biologie [CHU Toulouse], Centre Hospitalier Universitaire de Toulouse (CHU Toulouse), HIV, Inflammation et persistance - HIV, Inflammation and Persistence, Institut Pasteur [Paris] (IP), Service Maladies infectieuses et tropicales [CHU Toulouse], Pôle Inflammation, infection, immunologie et loco-moteur [CHU Toulouse] (Pôle I3LM Toulouse), Centre Hospitalier Universitaire de Toulouse (CHU Toulouse)-Centre Hospitalier Universitaire de Toulouse (CHU Toulouse), and Passaes, Caroline
- Subjects
0301 basic medicine ,Adult ,CD4-Positive T-Lymphocytes ,[SDV.IMM] Life Sciences [q-bio]/Immunology ,Viremia ,HIV Infections ,03 medical and health sciences ,0302 clinical medicine ,medicine ,CXCL10 ,Humans ,Allele ,ComputingMilieux_MISCELLANEOUS ,Innate immune system ,business.industry ,virus diseases ,Interferon-alpha ,General Medicine ,TLR7 ,Dendritic Cells ,Middle Aged ,medicine.disease ,Phenotype ,3. Good health ,030104 developmental biology ,Toll-Like Receptor 7 ,030220 oncology & carcinogenesis ,Cohort ,Immunology ,HIV-1 ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Female ,business ,Viral load ,Research Article - Abstract
International audience; Type I IFN (IFN-I) production by plasmacytoid DCs (pDCs) occurs during acute HIV-1 infection in response to TLR7 stimulation, but the role of pDC-derived IFN-I in controlling or promoting HIV-1 infection is ambiguous. We report here a sex-biased interferogenic phenotype for a frequent single-nucleotide polymorphism of human TLR7, rs179008, displaying an impact on key parameters of acute HIV-1 infection. We show allele rs179008 T to determine lower TLR7 protein abundance in cells from women, specifically — likely by diminishing TLR7 mRNA translation efficiency through codon usage. The hypomorphic TLR7 phenotype is mirrored by decreased TLR7-driven IFN-I production by female pDCs. Among women from the French ANRS PRIMO cohort of acute HIV-1 patients, carriage of allele rs179008 T associated with lower viremia, cell-associated HIV-1 DNA, and CXCL10 (IP-10) plasma concentrations. RNA viral load was decreased by 0.85 log10 (95% CI, −1.51 to −0.18) among T/T homozygotes, who also exhibited a lower frequency of acute symptoms. TLR7 emerges as an important control locus for acute HIV-1 viremia, and the clinical phenotype for allele rs179008 T, carried by 30%–50% of European women, supports a beneficial effect of toning down TLR7-driven IFN-I production by pDCs during acute HIV-1 infection.
- Published
- 2020
- Full Text
- View/download PDF
9. Female predisposition to TLR7-driven autoimmunity: gene dosage and the escape from X chromosome inactivation
- Author
-
José E. Mejía, Jean-Charles Guéry, Mélanie Souyris, and Julie Chaumeil
- Subjects
Male ,0301 basic medicine ,Immunology ,Gene Dosage ,Biology ,medicine.disease_cause ,Gene dosage ,X-inactivation ,Autoimmunity ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,X Chromosome Inactivation ,medicine ,Animals ,Humans ,Lupus Erythematosus, Systemic ,Immunology and Allergy ,Genetic Predisposition to Disease ,Gene ,X chromosome ,Chromosomes, Human, X ,Sex Characteristics ,Membrane Glycoproteins ,virus diseases ,TLR7 ,030104 developmental biology ,Toll-Like Receptor 7 ,biology.protein ,Female ,Antibody ,030215 immunology - Abstract
Women develop stronger immune responses than men, with positive effects on the resistance to viral or bacterial infections but magnifying also the susceptibility to autoimmune diseases like systemic lupus erythematosus (SLE). In SLE, the dosage of the endosomal Toll-like receptor 7 (TLR7) is crucial. Murine models have shown that TLR7 overexpression suffices to induce spontaneous lupus-like disease. Conversely, suppressing TLR7 in lupus-prone mice abolishes SLE development. TLR7 is encoded by a gene on the X chromosome gene, denoted TLR7 in humans and Tlr7 in the mouse, and expressed in plasmacytoid dendritic cells (pDC), monocytes/macrophages, and B cells. The receptor recognizes single-stranded RNA, and its engagement promotes B cell maturation and the production of pro-inflammatory cytokines and antibodies. In female mammals, each cell randomly inactivates one of its two X chromosomes to equalize gene dosage with XY males. However, 15 to 23% of X-linked human genes escape X chromosome inactivation so that both alleles can be expressed simultaneously. It has been hypothesized that biallelic expression of X-linked genes could occur in female immune cells, hence fostering harmful autoreactive and inflammatory responses. We review here the current knowledge of the role of TLR7 in SLE, and recent evidence demonstrating that TLR7 escapes from X chromosome inactivation in pDCs, monocytes, and B lymphocytes from women and Klinefelter syndrome men. Female B cells where TLR7 is thus biallelically expressed display higher TLR7-driven functional responses, connecting the presence of two X chromosomes with the enhanced immunity of women and their increased susceptibility to TLR7-dependent autoimmune syndromes.
- Published
- 2018
- Full Text
- View/download PDF
10. Biais de sexe dans l’asthme allergique
- Author
-
Eve Blanquart, Jean-Charles Guéry, and Sophie Laffont
- Subjects
0301 basic medicine ,medicine.medical_specialty ,business.industry ,Innate lymphoid cell ,Allergic asthma ,General Medicine ,Disease ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,respiratory tract diseases ,Allergic inflammation ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030228 respiratory system ,Immunity ,Epidemiology ,Immunology ,medicine ,business ,Asthma ,Hormone - Abstract
Allergic asthma is a chronic pulmonary inflammatory disease initiated by exposure to normally harmless allergens and marked by bronchial hyperreactivity. It affects more than 300 million people worldwide. Asthma often starts in childhood. Epidemiological studies show that there are sexual disparities in the prevalence and severity of asthma. Before the age of 10, the disease is more common in boys. This tendency reverses at puberty suggesting a regulating role of the sex hormones. In this synthesis, we summarize current knowledge on the role of sex hormones in allergic inflammation, with a particular focus on the impact of androgens on the development and function of recently introduced group 2 innate lymphoid cell subsets (ILC2) as critical actors in the initiation of allergic responses.♢.
- Published
- 2018
- Full Text
- View/download PDF
11. Why Is Systemic Lupus Erythematosus More Common in Women?
- Author
-
Jean-Charles Guéry
- Subjects
Regulation of gene expression ,Systemic blood ,Lupus erythematosus ,Rheumatology ,business.industry ,Sex factors ,Immunology ,Severity of illness ,Medicine ,In situ hybridization ,business ,medicine.disease ,X chromosome - Published
- 2019
- Full Text
- View/download PDF
12. Sex hormone regulation of innate lymphoid cells
- Author
-
Sophie Laffont, Jean-Charles Guéry, Eve Blanquart, PINIER, CHRISTINE, Centre de Physiopathologie Toulouse Purpan (CPTP), Université Toulouse III - Paul Sabatier (UT3), and Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Medicine (General) ,QH301-705.5 ,Sex steroid hormones ,[SDV]Life Sciences [q-bio] ,Allergic asthma ,Estrogen receptors ,NK cells ,Biology ,03 medical and health sciences ,R5-920 ,0302 clinical medicine ,Sex hormone-binding globulin ,Immunity ,[SDV.MHEP.MI]Life Sciences [q-bio]/Human health and pathology/Infectious diseases ,Sex differences ,Homeostasis ,Humans ,Lymphocytes ,Biology (General) ,Gonadal Steroid Hormones ,skin and connective tissue diseases ,Tissue homeostasis ,MESH: Gonadal Steroid Hormones ,MESH: Cytokines ,MESH: Humans ,Group 2 innate lymphoid cells ,Innate lymphoid cell ,General Medicine ,Immunity, Innate ,3. Good health ,Androgen receptor ,body regions ,[SDV] Life Sciences [q-bio] ,030104 developmental biology ,Drug development ,Sex steroid ,MESH: Homeostasis ,030220 oncology & carcinogenesis ,Review Article: Special Edition ,Immunology ,biology.protein ,[SDV.MHEP.MI] Life Sciences [q-bio]/Human health and pathology/Infectious diseases ,Cytokines ,MESH: Immunity, Innate ,MESH: Lymphocytes ,Hormone - Abstract
International audience; Innate lymphoid cell (ILC) subsets at barrier surfaces contribute to maintain tissue homeostasis and appropriate responses to infection. ILCs respond to environmental factors produced by non-hematopoietic cells within tissues, but also circulating cytokines or dietary compounds which allow them to adapt to organ milieu. Among these extrinsic signals, evidence is emerging that sex steroid hormones may act in a cell-intrinsic manner to regulate the development, maintenance in tissues and effector functions of specific subsets of ILCs. Understanding the nature and molecular mechanisms of sex steroid hormone actions on ILCs is important to unravel the cause of sexual disparity in human diseases and could lead to new drug development for the treatment of chronic inflammatory diseases or cancers. This review discusses the recent development in our understanding of the cell-intrinsic actions of sex steroid hormones on ILCs and their consequences on tissue-specific immunity with a particular focus on group 2 innate lymphoid cells and NK cells.
- Published
- 2020
- Full Text
- View/download PDF
13. Androgen signaling negatively controls group 2 innate lymphoid cells
- Author
-
Daniel Metzger, Claire Cenac, Gilles Laverny, Eve Blanquart, Jean-Charles Guéry, Sophie Laffont, Magali Savignac, Lucette Pelletier, Cyril Seillet, Jean-Philippe Girard, Gabrielle T. Belz, Centre de Physiopathologie Toulouse Purpan (CPTP), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg (UNISTRA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Université de Strasbourg (UNISTRA), Institut de pharmacologie et de biologie structurale (IPBS), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Centre National de la Recherche Scientifique (CNRS), University of Melbourne, and metzger, daniel
- Subjects
0301 basic medicine ,Male ,[SDV]Life Sciences [q-bio] ,Immunology and Allergy ,Orchiectomy ,Lymphocytes ,Lung ,Research Articles ,Innate lymphoid cell ,Pyroglyphidae ,[SDV] Life Sciences [q-bio] ,Receptors, Androgen ,innate lymphoid cell ,Androgens ,Female ,Disease Susceptibility ,medicine.symptom ,Signal transduction ,Signal Transduction ,medicine.medical_specialty ,medicine.drug_class ,sex difference ,Immunology ,hormone ,Sexism ,Inflammation ,Biology ,03 medical and health sciences ,Internal medicine ,medicine ,Hypersensitivity ,Animals ,Castration ,Lymphocyte Count ,Cell Proliferation ,immune protection ,lung inflammation ,Brief Definitive Report ,Pneumonia ,Androgen ,Interleukin-33 ,Asthma ,Immunity, Innate ,Androgen receptor ,Interleukin 33 ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,IL-33 ,Homeostasis - Abstract
At the onset of adolescence, asthma becomes less prevalent in males than in females, suggesting a protective role of male sex hormones. Here, Laffont et al. show that androgens negatively control ILC2 development and ILC2-driven lung inflammation in male mice., Prevalence of asthma is higher in women than in men, but the mechanisms underlying this sex bias are unknown. Group 2 innate lymphoid cells (ILC2s) are key regulators of type 2 inflammatory responses. Here, we show that ILC2 development is greatly influenced by male sex hormones. Male mice have reduced numbers of ILC2 progenitors (ILC2Ps) and mature ILC2s in peripheral tissues compared with females. In consequence, males exhibit reduced susceptibility to allergic airway inflammation in response to environmental allergens and less severe IL-33–driven lung inflammation, correlating with an impaired expansion of lung ILC2s. Importantly, orchiectomy, but not ovariectomy, abolishes the sex differences in ILC2 development and restores IL-33–mediated lung inflammation. ILC2Ps express the androgen receptor (AR), and AR signaling inhibits their differentiation into mature ILC2s. Finally, we show that hematopoietic AR expression limits IL-33–driven lung inflammation through a cell-intrinsic inhibition of ILC2 expansion. Thus, androgens play a crucial protective role in type 2 airway inflammation by negatively regulating ILC2 homeostasis, thereby limiting their capacity to expand locally in response to IL-33.
- Published
- 2017
- Full Text
- View/download PDF
14. CD49d/CD29-integrin controls the accumulation of plasmacytoid dendritic cells into the CNS during neuroinflammation
- Author
-
Britta Engelhardt, Sophie Laffont, Jean-Charles Guéry, Urban Deutsch, Laure Garnier, Arnaud Garnier, Elisa Kaba, Imperial College London, CRLCC Eugène Marquis (CRLCC), Centre de Physiopathologie Toulouse Purpan (CPTP), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Theodor Kocher Institute, University of Bern, and Theodor Kocher Institut
- Subjects
0301 basic medicine ,Adoptive cell transfer ,Encephalomyelitis, Autoimmune, Experimental ,Integrin alpha4 ,Interferon Regulatory Factor-7 ,Immunology ,Biology ,CD49d ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Movement ,medicine ,Immunology and Allergy ,Animals ,610 Medicine & health ,Neuroinflammation ,ComputingMilieux_MISCELLANEOUS ,Innate immune system ,Interleukin-12 Subunit p40 ,Multiple sclerosis ,Integrin beta1 ,Experimental autoimmune encephalomyelitis ,VLA-4 ,Brain ,hemic and immune systems ,Dendritic Cells ,medicine.disease ,Adoptive Transfer ,Immunity, Innate ,Peptide Fragments ,Mice, Inbred C57BL ,030104 developmental biology ,Gene Expression Regulation ,Pertussis Toxin ,Spinal Cord ,Toll-Like Receptor 9 ,Interferon Type I ,570 Life sciences ,biology ,IRF7 ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Female ,Myelin-Oligodendrocyte Glycoprotein ,030215 immunology ,Signal Transduction - Abstract
Plasmacytoid dendritic cells (pDCs) are found in the CNS during neuroinflammation and have been reported to exert regulatory functions in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). However, the mechanisms of entry of pDCs into the CNS as well as their phenotype and innate functional properties, once recruited into the CNS, have not been thoroughly examined. Herein, we show that pDCs rapidly accumulate into the brain and spinal cord during the acute phase of EAE, and maintain the expression of numerous phenotypic markers typical of peripheral pDCs. Functionally, CNS-pDCs constitutively expressed IRF7 and were able to rapidly produce type I IFNs and IL-12p40 upon ex vivo TLR-9 stimulation. Using adoptive transfer experiments, we provide evidence that CNS-pDC are recruited from the blood and accumulate into the CNS during the acute phase of EAE. Accumulation of pDCs into the CNS was strongly inhibited in the absence of CD29, but not CD18, suggesting a major role for ß1 but not ß2 integrins. Indeed, blocking the CD49d α4-integrins during acute EAE drastically diminished CNS-pDC numbers. Together, our results demonstrate that circulating pDCs are actively recruited into the CNS during acute EAE through a mechanism largely dependent on CD49d/CD29-integrins.
- Published
- 2019
- Full Text
- View/download PDF
15. Deconstructing the sex bias in allergy and autoimmunity: From sex hormones and beyond
- Author
-
Sophie Laffont, Jean-Charles Guéry, Centre de Physiopathologie Toulouse Purpan (CPTP), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), and Centre de Physiopathologie Toulouse Purpan ex IFR 30 et IFR 150 (CPTP)
- Subjects
Toll-like receptor ,Allergy ,TLR7 ,Biology ,medicine.disease_cause ,medicine.disease ,3. Good health ,Autoimmunity ,Sexual dimorphism ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Immunity ,Immunology ,medicine ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,ComputingMilieux_MISCELLANEOUS ,030215 immunology ,Hormone - Abstract
Men and women differ in their susceptibility to develop autoimmunity and allergy but also in their capacity to cope with infections. Mechanisms responsible for this sexual dimorphism are still poorly documented and probably multifactorial. This review discusses the recent development in our understanding of the cell-intrinsic actions of biological factors linked to sex, sex hormones and sex chromosome complement, on immune cells, which may account for the sex differences in the enhanced susceptibility of women to develop immunological disorders, such as allergic asthma or systemic lupus erythematosus (SLE). We choose to more specifically discuss the impact of sex hormones on the development and function of immune cell populations directly involved in type-2 immunity, and the role of the X-linked Toll like receptor 7 (TLR7) in anti-viral immunity and in SLE. We will also elaborate on the recent evidence demonstrating that TLR7 escapes from X chromosome inactivation in the immune cells of women, and how this may contribute to endow woman immune system with enhanced responsiveness to RNA-virus and susceptibility to SLE.
- Published
- 2019
- Full Text
- View/download PDF
16. Estrogen Signaling in Bystander Foxp3 neg CD4 + T Cells Suppresses Cognate Th17 Differentiation in Trans and Protects from Central Nervous System Autoimmunity
- Author
-
Sophie Laffont, Jean-Charles Guéry, Ari Waisman, Karine Lélu, Laure Garnier, Nir Yogev, Institut de Génomique Fonctionnelle (IGF), Université de Montpellier (UM)-Université Montpellier 1 (UM1)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Montpellier 2 - Sciences et Techniques (UM2)-Centre National de la Recherche Scientifique (CNRS), Centre de Physiopathologie Toulouse Purpan (CPTP), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Department of Internal Medicine, and Johannes Gutenberg - Universität Mainz (JGU)
- Subjects
0301 basic medicine ,Adoptive cell transfer ,Cell growth ,Chemistry ,Cellular differentiation ,[SDV]Life Sciences [q-bio] ,Immunology ,Experimental autoimmune encephalomyelitis ,Estrogen receptor ,FOXP3 ,medicine.disease ,Oligodendrocyte ,3. Good health ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Bystander effect ,medicine ,Immunology and Allergy ,ComputingMilieux_MISCELLANEOUS ,030215 immunology - Abstract
17β-Estradiol (E2) suppresses the development of experimental autoimmune encephalomyelitis (EAE) through estrogen receptor (ER) α, yet the cellular targets remain elusive. We have used an adoptive transfer model of myelin oligodendrocyte glycoprotein–specific CD4+ T cells from 2D2 TCR transgenic mice. We show that in the recipient mice, ERα expression in bystander CD4+ T cells, rather than in cognate 2D2 T cells, is required for the inhibition of Th17 cell differentiation by E2. Coadministration of estrogen-primed WT, but not ERα-deficient CD4+ T cells, with naive 2D2 T cells lacking ERα inhibited the development of Th17 cell–mediated EAE. Suppression of Th17 cells and protection from EAE were maintained when ERα was deleted in Foxp3+ regulatory T cells. We showed that in vivo PD-L1 blockade alleviated the anti-inflammatory action of E2 and that PD-1 expression on cognate but not bystander T cells was required for the E2-dependent inhibition of Th17 differentiation. In cotransfer experiments, we found that only WT but not PD-1KO 2D2 T cells were amenable to E2-dependent inhibition of Th17 differentiation. These results support the conclusion that the restriction of Th17 cell development by E2-primed bystander CD4+ T cells requires cell-intrinsic PD-1 signaling within cognate T cells rather than induction of regulatory 2D2 T cells through PD-1 engagement. Altogether, our results indicate that pregnancy-level concentrations of estrogen signal in conventional Foxp3neg CD4+ T cells to limit the differentiation of cognate Th17 cells through a trans-acting mechanism of suppression that requires a functional PD-1/PD-L1 regulatory axis.
- Published
- 2018
- Full Text
- View/download PDF
17. Eomesodermin Expression in CD4+ T Cells Restricts Peripheral Foxp3 Induction
- Author
-
Sebastian J. Arnold, Laure Garnier, Katharina S. Rauch, Kristina Schachtrup, Sophie Laffont, Maria Brack, Ana Izcue, Jean-Charles Guéry, and Ekaterina Lupar
- Subjects
CD4-Positive T-Lymphocytes ,Aging ,Encephalomyelitis, Autoimmune, Experimental ,genetic structures ,Regulatory T cell ,T cell ,Cellular differentiation ,Immunology ,Eomesodermin ,Biology ,T-Lymphocytes, Regulatory ,Interferon-gamma ,Mice ,Interleukin 21 ,Th2 Cells ,medicine ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,Interferon gamma ,Mice, Knockout ,FOXP3 ,Cell Differentiation ,Forkhead Transcription Factors ,Th1 Cells ,eye diseases ,Cell biology ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,Th17 Cells ,sense organs ,T-Box Domain Proteins ,medicine.drug - Abstract
CD4+ T cells polarize into effector Th subsets characterized by signature transcription factors and cytokines. Although T-bet drives Th1 responses and represses the alternative Th2, Th17, and Foxp3+ regulatory T cell fates, the role of the T-bet–related transcription factor eomesodermin (Eomes) in CD4+ T cells is less well understood. In this study, we analyze the expression and effects of Eomes in mouse CD4+ T lymphocytes. We find that Eomes is readily expressed in activated CD4+ Th1 T cells in vivo. Eomes+ CD4+ T cells accumulated in old mice, under lymphopenic conditions in a T cell transfer model of colitis, and upon oral Ag administration. However, despite its expression, genetic deletion of Eomes in CD4+ T cells did not impact on IFN-γ production nor increase Th2 or Th17 responses. In contrast, Eomes deficiency favored the accumulation of Foxp3+ cells in old mice, after in vivo differentiation of Eomes-deficient naive CD4+ T cells, and in response to oral Ag in a cell-intrinsic way. Enforced Eomes expression during in vitro regulatory T cell induction also reduced Foxp3 transcription. Likewise, bystander Eomes-deficient CD4+ T cells were more efficient at protecting from experimental autoimmune encephalitis compared with wild-type CD4+ T cells. This enhanced capacity of Eomes-deficient CD4+ T cells to inhibit EAE in trans was associated with an enhanced frequency of Foxp3+ cells. Our data identify a novel role for Eomes in CD4+ T cells and indicate that Eomes expression may act by limiting Foxp3 induction, which may contribute to the association of EOMES to susceptibility to multiple sclerosis.
- Published
- 2015
- Full Text
- View/download PDF
18. TLR7 escapes X chromosome inactivation in immune cells
- Author
-
Solange Grunenwald, Jean-Charles Guéry, Astrid Canivet, Pascal Azar, Julie Chaumeil, Claire Cenac, José E. Mejía, Catherine Pienkowski, Danièle Daviaud, and Mélanie Souyris
- Subjects
0301 basic medicine ,Lupus erythematosus ,biology ,Immunology ,virus diseases ,General Medicine ,TLR7 ,medicine.disease ,Immunoglobulin G ,X-inactivation ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,medicine.anatomical_structure ,medicine ,biology.protein ,Klinefelter syndrome ,X chromosome ,B cell ,030215 immunology - Abstract
Toll-like receptor 7 (TLR7) is critical to the induction of antiviral immunity, but TLR7 dosage is also a key pathogenic factor in systemic lupus erythematosus (SLE), an autoimmune disease with strong female bias. SLE prevalence is also elevated in individuals with Klinefelter syndrome, who carry one or more supernumerary X chromosomes, suggesting that the X chromosome complement contributes to SLE susceptibility. TLR7 is encoded by an X chromosome locus, and we examined here whether the TLR7 gene evades silencing by X chromosome inactivation in immune cells from women and Klinefelter syndrome males. Single-cell analyses of TLR7 allelic expression demonstrated that substantial fractions of primary B lymphocytes, monocytes, and plasmacytoid dendritic cells not only in women but also in Klinefelter syndrome males express TLR7 on both X chromosomes. Biallelic B lymphocytes from women displayed greater TLR7 transcriptional expression than the monoallelic cells, correlated with higher TLR7 protein expression in female than in male leukocyte populations. Biallelic B cells were preferentially enriched during the TLR7-driven proliferation of CD27+ plasma cells. In addition, biallelic cells showed a greater than twofold increase over monoallelic cells in the propensity to immunoglobulin G class switch during the TLR7-driven, T cell-dependent differentiation of naive B lymphocytes into immunoglobulin-secreting cells. TLR7 escape from X inactivation endows the B cell compartment with added responsiveness to TLR7 ligands. This finding supports the hypothesis that enhanced TLR7 expression owing to biallelism contributes to the higher risk of developing SLE and other autoimmune disorders in women and in men with Klinefelter syndrome.
- Published
- 2018
- Full Text
- View/download PDF
19. The β and α2δ auxiliary subunits of voltage-gated calcium channel 1 (Cav1) are required for Th2-lymphocyte function and acute allergic airway inflammation
- Author
-
Antoine Magnan, Jean-Charles Guéry, Grégory Bouchaud, Nicolas Rosa, Marion Mars, Magali Savignac, Lucette Pelletier, Astrid Canivet, Martin Klein, Virginie Robert, Emily Triffaux, Centre de Physiopathologie Toulouse Purpan ex IFR 30 et IFR 150 (CPTP), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Centre National de la Recherche Scientifique (CNRS), unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN), Unité de recherche sur les Biopolymères, Interactions Assemblages (BIA), Institut National de la Recherche Agronomique (INRA), Centre de Physiopathologie Toulouse Purpan (CPTP), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), French National Institute for Health and Medical research (INSERM), French Society of Allergology, Conseil Regional Midi-Pyrenees, Conseil Regional of Midi-Pyrenees, and INSERM
- Subjects
0301 basic medicine ,medicine.medical_specialty ,medicine.medical_treatment ,Lymphocyte ,Ca(v)1 calcium channel ,[SDV]Life Sciences [q-bio] ,Immunology ,chemistry.chemical_element ,Calcium ,Biology ,03 medical and health sciences ,Th2 ,Internal medicine ,medicine ,Immunology and Allergy ,calcium ,Voltage-dependent calcium channel ,ORAI1 ,Calcium channel ,T-cell receptor ,asthma ,cytokines ,3. Good health ,Cell biology ,030104 developmental biology ,Cytokine ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Proteasome inhibitor ,medicine.drug - Abstract
International audience; BACKGROUND: T-lymphocytes express not only the cell membrane calcium ORAI1 but also voltage-dependent Cav1 channels. In excitable cells, these channels are composed of the ion forming pore α1 and auxiliary subunits (β and α2δ) needed for proper trafficking and activation of the channel. We previously disclosed the role of Cav1.2 α1 in mouse and human Th2- but not Th1-cell functions and showed that knocking-down Cav1 α1 prevents experimental asthma OBJECTIVE: We investigated the role of β and α2δ auxiliary subunits on Cav1 α1 function in Th2 lymphocytes and on the development of acute allergic airway inflammation. METHODS: We used antisense oligonucleotides (CavβAS) to knockdown Cavβ and gabapentin, a drug that binds to and inhibits α2δ1 and α2δ2, to test their effects on Th2 functions and their capacity to reduce allergic airway inflammation. RESULTS: Mouse and human Th2-cells express mainly Cavβ1, β3 and α2δ2 subunits. CavβAS reduces TCR-driven calcium responses and cytokine production by mouse and human Th2, with no effect on Th1-cells. Cavβ is mainly involved in restraining Cav1.2 α1 degradation through the proteasome as a proteasome inhibitor partially restores the α1 protein level. Gabapentin impairs TCR-driven calcium response and cytokine production associated with the loss of α2δ2 protein in Th2-cells. CONCLUSIONS: These results stress the role of Cavβ and α2δ2 auxiliary subunits in the stability and activation of Cav1.2 channels in Th2 lymphocytes both in vitro and in vivo as demonstrated by the beneficial effect of CavβAS and gabapentin in allergic airway inflammation. CLINICAL IMPLICATIONS: The demonstration that auxiliary subunits are involved in calcium signaling through Cav1 channels and function of mouse and human Th2-lymphocytes supports their potential beneficial effect on allergic asthma.
- Published
- 2017
- Full Text
- View/download PDF
20. Sex Differences in Asthma: A Key Role of Androgen-Signaling in Group 2 Innate Lymphoid Cells
- Author
-
Sophie Laffont, Eve Blanquart, and Jean-Charles Guéry
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,sex differences ,Allergy ,group 2 innate lymphoid cells ,Mini Review ,Immunology ,Inflammation ,Biology ,medicine.disease_cause ,Autoimmunity ,Allergic inflammation ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Immunity ,medicine ,Immunology and Allergy ,Asthma ,Innate lymphoid cell ,androgens ,medicine.disease ,androgen receptors ,030104 developmental biology ,030228 respiratory system ,sex steroid hormones ,medicine.symptom ,lcsh:RC581-607 ,allergic asthma - Abstract
Infectious diseases, autoimmune diseases, and also allergy differentially affect women and men. In general, women develop strongest immune responses and thus the proportion of infected individuals and the severity of many viral, bacterial, or parasitic infections are increased in men. However, heightened immunity in women makes them more susceptible than men to autoimmunity and allergy. While sex differences in immunity are well documented, little is known about the cellular and molecular mechanisms underlying these immunological differences, particularly in allergic asthma. Asthma is a chronic inflammation of the airways mediated by exacerbated type 2 immune responses. Sex differences have been reported in the incidence, prevalence, and severity of asthma. While during childhood, males are more susceptible to asthma than females, there is a switch at the onset of puberty as for many other allergic diseases. This decrease of asthma incidence around puberty in males suggests that hormonal mediators could play a protective role in the susceptibility to allergic responses in male. Group 2 innate lymphoid cells (ILC2s) have recently emerged as critical players in the initiation of allergic responses, but also in the resolution of parasitic infection, through their capacity to rapidly and potently produce type 2 cytokines. This review will cover the current understanding of the impact of sex-linked factors in allergic inflammation, with a particular focus on the role of sex hormones on the development and function of tissue-resident ILC2s.
- Published
- 2017
- Full Text
- View/download PDF
21. Estrogen Receptor-Dependent Regulation of Dendritic Cell Development and Function
- Author
-
Jean-Charles Guéry, Cyril Seillet, and Sophie Laffont
- Subjects
0301 basic medicine ,sex differences ,CD40 ,Innate immune system ,biology ,medicine.medical_treatment ,Immunology ,Estrogen receptor ,estrogen receptors ,Review ,Dendritic cell ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,Cytokine ,Immune system ,toll-like receptors ,biology.protein ,medicine ,Immunology and Allergy ,dendritic cells ,Estrogen receptor alpha ,Transcription factor ,estrogens - Abstract
Autoimmunity, infectious diseases and cancer affect women and men differently. Because they tend to develop more vigorous adaptive immune responses than men, women are less susceptible to some infectious diseases but also at higher risk of autoimmunity. The regulation of immune responses by sex-dependent factors probably involves several non-redundant mechanisms. A privileged area of study, however, concerns the role of sex steroid hormones in the biology of innate immune cells, especially dendritic cells (DCs). In recent years, our understanding of the lineage origin of DC populations has expanded, and the lineage-committing transcription factors shaping peripheral DC subsets have been identified. Both progenitor cells and mature DC subsets express estrogen receptors (ERs), which are ligand-dependent transcription factors. This suggests that estrogens may contribute to the reported sex differences in immunity by regulating DC biology. Here, we review the recent literature and highlight evidence that estrogen-dependent activation of ERα regulates the development or the functional responses of particular DC subsets. The in vitro model of GM-CSF-induced DC differentiation shows that CD11c+ CD11bint Ly6cneg cells depend on ERα activation by estrogen for their development, and for the acquisition of competence to activate naive CD4+ T lymphocytes and mount a robust pro-inflammatory cytokine response to CD40 stimulation. In this model, estrogen signaling in conjunction with GM-CSF is necessary to promote early interferon regulatory factor (Irf)-4 expression in macrophage-DC progenitors and their subsequent differentiation into IRF-4hi CD11c+ CD11bint Ly6cneg cells, closely related to the cDC2 subset. The Flt3L-induced model of DC differentiation in turn shows that ERα signaling promotes the development of conventional DC (cDC) and plasmacytoid DC (pDC) with higher capability of pro-inflammatory cytokine production in response to TLR stimulation. Likewise, cell-intrinsic ER signaling positively regulates the TLR-driven production of type I interferons (IFNs) in mouse pDCs in vivo. This effect of estrogens likely contributes to the greater proficiency of women’s pDCs than men’s as regards the production of type I IFNs elicited by TLR7 ligands. In summary, evidence is emerging in support of the notion that estrogen signaling regulates important aspects of cDC and pDC development and/or effector functions, in both mice and humans.
- Published
- 2017
- Full Text
- View/download PDF
22. X-Chromosome Complement and Estrogen Receptor Signaling Independently Contribute to the Enhanced TLR7-Mediated IFN-α Production of Plasmacytoid Dendritic Cells from Women
- Author
-
Nelly Rouquié, Jean-Charles Guéry, Sophie Laffont, Caroline Aspord, Joel Plumas, Pascal Azar, and Cyril Seillet
- Subjects
Male ,medicine.medical_specialty ,Cellular differentiation ,Immunology ,Gene Dosage ,Estrogen receptor ,Mice, SCID ,Biology ,Mice ,Genes, X-Linked ,Internal medicine ,medicine ,Animals ,Estrogen Receptor beta ,Humans ,Immunology and Allergy ,Cells, Cultured ,X chromosome ,Innate immune system ,Tumor Necrosis Factor-alpha ,Estrogen Receptor alpha ,Hematopoietic Stem Cell Transplantation ,Interferon-alpha ,Cell Differentiation ,hemic and immune systems ,Dendritic Cells ,TLR7 ,Hematopoietic Stem Cells ,Immunity, Innate ,Endocrinology ,Toll-Like Receptor 7 ,Humanized mouse ,Female ,Estrogen receptor alpha ,Signal Transduction ,Hormone - Abstract
Human plasmacytoid dendritic cells (pDCs) play a major role in innate immunity through the production of type I IFNs after TLR engagement by pathogens. Sex-based differences in the innate function of human pDCs have been established, with pDCs from women exhibiting enhanced TLR7-mediated IFN-α production as compared with pDCs from males. In mice, we recently provided evidence for a role of estrogens as a positive regulator of pDC innate functions through cell-intrinsic estrogen receptor α signaling, but did not exclude a role for other X-linked factors, particularly in human pDCs. In this study, we investigated the respective contribution of X chromosome dosage and sex hormones using a humanized mouse model in which male or female NOD-SCID-β2m−/− were transplanted with human progenitor cells purified from either male or female cord blood cells. We showed that, in response to TLR7 ligands, the frequency of IFN-α– and TNF-α–producing pDCs from either sex was greater in female than in male host mice, suggesting a positive role for estrogens. Indeed, blockade of estrogen receptor signaling during pDC development in vitro inhibited TLR7-mediated IFN-α production by human pDCs, which expressed both ESR1 and ESR2 genes. Interestingly, we also found that X chromosome dosage contributed to this sex bias as female pDCs have an enhanced TLR7-mediated IFN-α response as compared with male ones, irrespective of the sex of the recipient mice. Together, these results indicate that female sex hormones, estrogens, and X chromosome complement independently contribute to the enhanced TLR7-mediated IFN-α response of pDCs in women.
- Published
- 2014
- Full Text
- View/download PDF
23. Bispecificity for Myelin and Neuronal Self-Antigens Is a Common Feature of CD4 T Cells in C57BL/6 Mice
- Author
-
Lennart T. Mars, Jean-Charles Guéry, Abdulraouf Ramadan, Miriam Eisenstein, Roland S. Liblau, Liliana E. Lucca, Phuong Nguyen, Alessandro Sette, Nadège Carrié, Avraham Ben-Nun, Sabine Desbois, and Terrence L. Geiger
- Subjects
CD4-Positive T-Lymphocytes ,T cell ,Immunology ,Context (language use) ,Cross Reactions ,Biology ,medicine.disease_cause ,Autoantigens ,Epitope ,Autoimmunity ,Myelin oligodendrocyte glycoprotein ,Mice ,Myelin ,Autoimmune Diseases of the Nervous System ,Neurofilament Proteins ,medicine ,Animals ,Immunology and Allergy ,Receptor ,Myelin Sheath ,Mice, Knockout ,T-cell receptor ,Molecular biology ,Peptide Fragments ,Receptors, Antigen ,medicine.anatomical_structure ,biology.protein ,Myelin-Oligodendrocyte Glycoprotein - Abstract
The recognition of multiple ligands by a single TCR is an intrinsic feature of T cell biology, with important consequences for physiological and pathological processes. Polyspecific T cells targeting distinct self-antigens have been identified in healthy individuals as well as in the context of autoimmunity. We have previously shown that the 2D2 TCR recognizes the myelin oligodendrocyte glycoprotein epitope (MOG)35–55 as well as an epitope within the axonal protein neurofilament medium (NF-M15–35) in H-2b mice. In this study, we assess whether this cross-reactivity is a common feature of the MOG35–55-specific T cell response. To this end, we analyzed the CD4 T cell response of MOG35–55-immunized C57BL/6 mice for cross-reactivity with NF-M15–35. Using Ag recall responses, we established that an important proportion of MOG35–55-specific CD4 T cells also responded to NF-M15–35 in all mice tested. To study the clonality of this response, we analyzed 22 MOG35–55-specific T cell hybridomas expressing distinct TCR. Seven hybridomas were found to cross-react with NF-M15–35. Using an alanine scan of NF-M18–30 and an in silico predictive model, we dissected the molecular basis of cross-reactivity between MOG35–55 and NF-M15–35. We established that NF-M F24, R26, and V27 proved important TCR contacts. Strikingly, the identified TCR contacts are conserved within MOG38–50. Our data indicate that due to linear sequence homology, part of the MOG35–55-specific T cell repertoire of all C57BL/6 mice also recognizes NF-M15–35, with potential implications for CNS autoimmunity.
- Published
- 2014
- Full Text
- View/download PDF
24. Effect of the thiol group on experimental gold-induced autoimmunity
- Author
-
Jean-Charles Guéry, Lucette Pelletier, Philippe Druet, M. C. Vial, E. Druet, Chantal Mandet, H. Tournade, Patrick M. Dansette, and Régine Pasquier
- Subjects
Propanols ,Immunology ,Autoimmunity ,Systemic autoimmunity ,medicine.disease_cause ,Thiol group ,Autoimmune Diseases ,Rheumatology ,Rats, Inbred BN ,Organometallic Compounds ,Animals ,Immunology and Allergy ,Medicine ,Pharmacology (medical) ,Sulfhydryl Compounds ,chemistry.chemical_classification ,Autoimmune disease ,Experimental model ,business.industry ,BROWN NORWAY ,medicine.disease ,Rats ,Sodium salt ,Disease Models, Animal ,chemistry ,Antirheumatic Agents ,Thiol ,Dimercaprol ,Female ,business ,Organogold Compounds - Abstract
Brown Norway rats injected with aurothiopro-panolsulfonate sodium salt develop systemic autoimmunity. The aim of this study was to assess the influence of the sulfur-containing group in this experimental model of gold-induced autoimmunity. It was shown that the sulfur-containing group does not induce autoimmunity of itself, but potentiates the immunotoxic effects of gold.
- Published
- 2010
- Full Text
- View/download PDF
25. Knocking Down Cav1 Calcium Channels Implicated in Th2 Cell Activation Prevents Experimental Asthma
- Author
-
Marie-Laure Renoud, Virginie Robert, Catherine Leclerc, Marilena Djata Cabral, Antoine Magnan, Bruno Gomes, Pierre-Emmanuel Paulet, David Lair, Jean-Charles Guéry, Hans Yssel, Bernard Mariamé, Marc Moreau, Magali Savignac, Lucette Pelletier, and Marie Langelot
- Subjects
Pulmonary and Respiratory Medicine ,Cell signaling ,Ovalbumin ,medicine.medical_treatment ,Blotting, Western ,Caveolin 1 ,Cell ,chemistry.chemical_element ,Calcium ,Critical Care and Intensive Care Medicine ,Mice ,Th2 Cells ,Intensive care ,Animals ,Medicine ,Administration, Intranasal ,Cell Proliferation ,Mice, Inbred BALB C ,Voltage-dependent calcium channel ,Reverse Transcriptase Polymerase Chain Reaction ,business.industry ,Transfection ,Calcium Channel Blockers ,Asthma ,Up-Regulation ,Cell biology ,Blot ,Disease Models, Animal ,Cytokine ,medicine.anatomical_structure ,chemistry ,Immunology ,Female ,Calcium Channels ,business - Abstract
Th2 cells orchestrate allergic asthma and the cytokines they produce (IL-4, IL-5, and IL-13) are deleterious in allergy. Therefore, it is important to identify key signaling molecules expressed by Th2 cells that are essential for their function. We have previously shown that dihydropyridines selectively modulate Th2 cell functions.Because dihydropyridines bind to and modulate voltage-dependent calcium (Ca(v)1) channel in excitable cells, we aimed at showing that Th2 cells selectively express functional Ca(v)1-related channels, the inhibition of which may prevent asthma.We looked for Ca(v)1 channel expression in Th2 and Th1 cells by real-time polymerase chain reaction and Western blotting. We sequenced the isoforms expressed by Th2 cells and tested whether Ca(v)1 antisense oligodeoxynucleotides (Ca(v)1AS) affected Ca(2+) signaling and cytokine production. Finally, we tested the effect of Ca(v)1AS in the passive asthma model by injection of ovalbumin-specific Th2 cells transfected with Ca(v)1AS into BALB/c mice challenged with intranasal ovalbumin and in the active model of asthma by intranasal delivery of Ca(v)1AS together with soluble ovalbumin in BALB/c mice previously immunized with ovalbumin in alum.We show that mouse Th2 but not Th1 cells expressed Ca(v)1.2 and Ca(v)1.3 channels. Th2 cells transfected with Ca(v)1AS had impaired Ca(2+) signaling and cytokine production, and lost their ability to induce airway inflammation on adoptive transfer. Furthermore, intranasal administration of Ca(v)1AS suppressed airway inflammation and hyperreactivity in an active model of asthma.These results indicate that Th2 cells selectively express Ca(v)1 channels that may be efficiently targeted in T lymphocytes to prevent experimental asthma.
- Published
- 2010
- Full Text
- View/download PDF
26. Estrogen Receptor α, but Not β, Is Required for Optimal Dendritic Cell Differentiation and CD40-Induced Cytokine Production
- Author
-
Sophie Laffont, Jean-Charles Guéry, Cyril Seillet, Pierre Chambon, Jean-François Arnal, Andrée Krust, Pierre Gourdy, Laurent Delpy, and Victorine Douin-Echinard
- Subjects
CD86 ,CD40 ,biology ,Cellular differentiation ,medicine.medical_treatment ,Immunology ,Estrogen receptor ,Dendritic cell differentiation ,Proinflammatory cytokine ,Cell biology ,Cytokine ,biology.protein ,medicine ,Immunology and Allergy ,Estrogen receptor alpha ,hormones, hormone substitutes, and hormone antagonists - Abstract
Dendritic cells (DC) are critical actors in the initiation of primary immune responses and regulation of self-tolerance. The steroid sex hormone 17β-estradiol (E2) has been shown to promote the differentiation of DCs from bone marrow (BM) precursors in vitro. However, the estrogen receptor (ER) involved in this effect has not yet been characterized. Using recently generated ERα- or ERβ-deficient mice, we investigated the role of ER isotypes in DC differentiation and acquisition of effector functions. We report that estrogen-dependent activation of ERα, but not ERβ, is required for normal DC development from BM precursors cultured with GM-CSF. We show that reduced numbers of DCs were generated in the absence of ERα activation and provide evidence for a cell-autonomous function of ERα signaling in DC differentiation. ERα-deficient DCs were phenotypically and functionally distinct from wild-type DCs generated in the presence of estrogens. In response to microbial components, ERα-deficient DCs failed to up-regulate MHC class II and CD86 molecules, which could account for their reduced capacity to prime naive CD4+ T lymphocytes. Although they retained the ability to express CD40 and to produce proinflammatory cytokines (e.g., IL-12, IL-6) upon TLR engagement, ERα-deficient DCs were defective in their ability to secrete such cytokines in response to CD40–CD40L interactions. Taken together, these results provide the first genetic evidence that ERα is the main receptor regulating estrogen-dependent DC differentiation in vitro and acquisition of their effector functions.
- Published
- 2008
- Full Text
- View/download PDF
27. Sex Differences in Plasmacytoid Dendritic Cell Levels of IRF5 Drive Higher IFN-α Production in Women
- Author
-
Heike Hildebrandt, Jean-Philippe Herbeuval, Susanne Ziegler, Claudia Beisel, Marcus Altfeld, Sophie Laffont, Michael Sirignano, Brigitte Autran, Nikaïa Smith, Jean-Charles Guéry, Armon Sharei, Sylvie Le Gall, Lise Chauveau, Meghan G. Hart, Olivier Schwartz, Thomas J. Diefenbach, Phillip Tomezsko, Georgio Kourjian, Filippos Porichis, Christine D. Palmer, Morgane Griesbeck, J. Judy Chang, Alexandra-Chloé Villani, Claire Cenac, Centre d'Immunologie et de Maladies Infectieuses (CIMI), Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Ragon Institute of MGH, MIT and Harvard, Heinrich-Pette-Institut, Leibniz Institute for Experimental Virology, Centre de Physiopathologie Toulouse Purpan (CPTP), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques (LCBPT - UMR 8601), Université Paris Descartes - Paris 5 (UPD5)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS), Virus et Immunité, Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Medical Department [Hamburg], Universitaetsklinikum Hamburg-Eppendorf = University Medical Center Hamburg-Eppendorf [Hamburg] (UKE), Broad Institute of MIT and Harvard (BROAD INSTITUTE), Harvard Medical School [Boston] (HMS)-Massachusetts Institute of Technology (MIT)-Massachusetts General Hospital [Boston], Department of Infectious Diseases [Monash], Monash University [Melbourne], This work was supported by National Institutes of Health/National Institute of Allergy and Infectious Diseases Grants R01 AI078784 and P01 AI078897, fellowships from the National Health and Medical Research Council of Australia (Grant 519578 to J.J.C.), a Ragon Fellowship from The Phillip T. and Susan M. Ragon Foundation (to J.J.C.), a fellowship from the French National Agency for Research on AIDS and Viral Hepatitis (Grant 2013-219 to M.G.), the Fondation pour la Recherche Médicale (Grant DEQ2000329169 to J.-C.G.), the Conseil Régional Midi-Pyrénées (J.-C.G.), the Arthritis Fondation Courtin (J.-C.G.), and the Fondation ARC pour la recherche sur le cancer (J.-C.G.), Centre d'Immunologie et de Maladies Infectieuses ( CIMI ), Centre National de la Recherche Scientifique ( CNRS ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Université Pierre et Marie Curie - Paris 6 ( UPMC ), Centre de Physiopathologie de Toulouse Purpan ( CPTP ), Université Toulouse III - Paul Sabatier ( UPS ), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Centre National de la Recherche Scientifique ( CNRS ), Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques ( LCBPT - UMR 8601 ), Université Paris Descartes - Paris 5 ( UPD5 ) -Centre National de la Recherche Scientifique ( CNRS ), Institut Pasteur [Paris]-Centre National de la Recherche Scientifique ( CNRS ), Massachusetts Institute of Technology ( MIT ), Universitaetsklinikum Hamburg-Eppendorf = University Medical Center Hamburg-Eppendorf [Hamburg] ( UKE ), Broad Institute of MIT and Harvard ( BROAD INSTITUTE ), Harvard Medical School [Boston] ( HMS ) -Massachusetts General Hospital [Boston] ( MGH ) -Massachusetts Institute of Technology ( MIT ), Université de Toulouse (UT)-Université de Toulouse (UT)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Koch Institute for Integrative Cancer Research at MIT [Cambridge, MA], Centre de Physiopathologie Toulouse Purpan ex IFR 30 et IFR 150 (CPTP), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Centre National de la Recherche Scientifique (CNRS), and Université Paris Descartes - Paris 5 (UPD5)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Male ,medicine.medical_specialty ,[SDV]Life Sciences [q-bio] ,Innate Immunity and Inflammation ,Immunology ,Estrogen receptor ,Mice, Transgenic ,Plasmacytoid dendritic cell ,Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Internal medicine ,medicine ,Animals ,Humans ,Immunology and Allergy ,RNA, Messenger ,Cells, Cultured ,030304 developmental biology ,Regulation of gene expression ,Sex Characteristics ,0303 health sciences ,[ SDV ] Life Sciences [q-bio] ,Estrogen Receptor alpha ,Interferon-alpha ,hemic and immune systems ,Dendritic Cells ,TLR7 ,Recombinant Proteins ,Endocrinology ,Gene Expression Regulation ,Toll-Like Receptor 7 ,Interferon Regulatory Factors ,Female ,Estrogen receptor alpha ,IRF5 ,Signal Transduction ,030215 immunology ,Interferon regulatory factors - Abstract
Increased IFN-α production contributes to the pathogenesis of infectious and autoimmune diseases. Plasmacytoid dendritic cells (pDCs) from females produce more IFN-α upon TLR7 stimulation than pDCs from males, yet the mechanisms underlying this difference remain unclear. In this article, we show that basal levels of IFN regulatory factor (IRF) 5 in pDCs were significantly higher in females compared with males and positively correlated with the percentage of IFN-α–secreting pDCs. Delivery of recombinant IRF5 protein into human primary pDCs increased TLR7-mediated IFN-α secretion. In mice, genetic ablation of the estrogen receptor 1 (Esr1) gene in the hematopoietic compartment or DC lineage reduced Irf5 mRNA expression in pDCs and IFN-α production. IRF5 mRNA levels furthermore correlated with ESR1 mRNA levels in human pDCs, consistent with IRF5 regulation at the transcriptional level by ESR1. Taken together, these data demonstrate a critical mechanism by which sex differences in basal pDC IRF5 expression lead to higher IFN-α production upon TLR7 stimulation in females and provide novel targets for the modulation of immune responses and inflammation.
- Published
- 2015
- Full Text
- View/download PDF
28. Estrogen receptor-dependent modulation of dendritic cell biology: of mice and women
- Author
-
Jean-Charles Guéry and Sophie Laffont
- Subjects
Innate immune system ,Immune system ,Estrogen ,medicine.drug_class ,Immunity ,Humanized mouse ,Immunology ,medicine ,Estrogen receptor ,General Medicine ,Dendritic cell ,Progenitor cell ,Biology - Abstract
Autoimmune and infectious diseases differentially affect women from men. Women tend to develop stronger immune responses and thus in general men are more susceptible to infectious diseases whereas women are more likely to develop autoimmune diseases. These differences could be in part attributable to the pro-inflammatory role of the female sex hormone estrogen on immunity and particularly on dendritic cells (DCs), a key subset of innate immune cells. For several years now, we have undertaken studies to understand how estrogens influence the biology of murine and human DCs. We and others have demonstrated that estradiol (E2) was required for the optimal in vitro differentiation of murine DCs and acquisition of their effector functions. These effects on DC biology were dependent on the activation of the estrogen receptor a (ERa). More recently, we focused our interest on plasmacytoid dendritic cells (pDCs). Indeed, this subset that produces large amounts of IFN-a/b in response to viral or endogenous nucleic acids through activation of their TLR-7 and TLR-9 show gender differences with enhanced IFN-a production by pDCs from women, compared to men. We could establish, in Human and in mice, that in vivo treatment with E2 enhanced the TLR-dependent production of IFNa by pDCs. In mice, we demonstrated that the amplifying effect of endogenous and exogenous estrogens is dependent on the intrinsic activation of ERα by hormone in the pDCs. To further characterize the mechanisms underlying this sex-based difference in pDC innate functions, we investigated the respective contribution of X chromosome dosage versus sex hormones using a humanized mouse model in which male or female NOD-SCID-s2m -/- mice were transplanted with human progenitor cells (HPCs) purified from either male (XY) or female (XX) donors. We could show that cell-intrinsic ER-signaling and X chromosome complement both independently contribute to the enhanced TLR-7-mediated response of pDCs in women, which may account for the sex-based differences in autoimmune and infectious diseases. Altogether, our work demonstrates that estrogen-mediated activation of ER signaling is a key regulator of DC biology both in Human and in mouse.
- Published
- 2015
- Full Text
- View/download PDF
29. Estrogen Receptor α Signaling in Inflammatory Leukocytes Is Dispensable for 17β-Estradiol-Mediated Inhibition of Experimental Autoimmune Encephalomyelitis
- Author
-
Christiane Coureau, Pierre Chambon, Sophie Laffont, Andrée Krust, Victorine Douin-Echinard, Lucile Garidou, and Jean-Charles Guéry
- Subjects
Encephalomyelitis, Autoimmune, Experimental ,medicine.drug_class ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,Immunology ,Estrogen receptor ,Biology ,Polymerase Chain Reaction ,Mice ,Immune system ,Leukocytes ,medicine ,Animals ,Immunology and Allergy ,Inflammation ,Autoimmune disease ,Estradiol ,Microglia ,Experimental autoimmune encephalomyelitis ,Estrogen Receptor alpha ,Flow Cytometry ,medicine.disease ,Mice, Mutant Strains ,Cytokine ,medicine.anatomical_structure ,Receptors, Estrogen ,Spinal Cord ,Estrogen ,Radiation Chimera ,Cancer research ,Signal Transduction - Abstract
Estrogen treatment has been shown to exert a protective effect on experimental autoimmune encephalomyelitis (EAE), and is under clinical trial for multiple sclerosis. Although it is commonly assumed that estrogens exert their effect by modulating immune functions, we show in this study that 17β-estradiol (E2) treatment can inhibit mouse EAE without affecting autoantigen-specific T cell responsiveness and type 1 cytokine production. Using mutant mice in which estrogen receptor α (ERα) has been unambiguously inactivated, we found that ERα was responsible for the E2-mediated inhibition of EAE. We next generated irradiation bone marrow chimeras in which ERα expression was selectively impaired in inflammatory T lymphocytes or was limited to the radiosensitive hemopoietic compartment. Our data show that the protective effect of E2 on clinical EAE and CNS inflammation was not dependent on ERα signaling in inflammatory T cells. Likewise, EAE development was not prevented by E2 treatment in chimeric mice that selectively expressed ERα in the systemic immune compartment. In conclusion, our data demonstrate that the beneficial effect of E2 on this autoimmune disease does not involve ERα signaling in blood-derived inflammatory cells, and indicate that ERα expressed in other tissues, such as CNS-resident microglia or endothelial cells, mediates this effect.
- Published
- 2004
- Full Text
- View/download PDF
30. Estrogens and inflammatory autoimmune diseases
- Author
-
Jean-Charles Guéry
- Subjects
Male ,Multiple Sclerosis ,MEDLINE ,Estrogen receptor ,Arthritis ,Hashimoto Disease ,medicine.disease_cause ,Autoimmune Diseases ,Autoimmunity ,Arthritis, Rheumatoid ,Mice ,Rheumatology ,Pregnancy ,medicine ,Animals ,Humans ,Lupus Erythematosus, Systemic ,Inflammation ,Brain Diseases ,Lupus erythematosus ,business.industry ,Estrogens ,medicine.disease ,Disease Models, Animal ,Immunology ,Encephalitis ,Female ,business ,Signal Transduction - Published
- 2012
- Full Text
- View/download PDF
31. Skin Graft Rejection Elicited by β2-Microglobulin as a Minor Transplantation Antigen Involves Multiple Effector Pathways: Role of Fas-Fas Ligand Interactions and Th2-Dependent Graft Eosinophil Infiltrates
- Author
-
Jean-Charles Guéry, Murielle Surquin, Nathalie Nagy, Patrick Stordeur, Michel Goldman, Alain Le Moine, François-Xavier Demoor, Daniel Abramowicz, Isabelle Salmon, Véronique Flamand, and Katia Rombaut
- Subjects
CD4-Positive T-Lymphocytes ,Cytotoxicity, Immunologic ,Graft Rejection ,Fas Ligand Protein ,Transplantation Conditioning ,CD8 Antigens ,Immunology ,CD8-Positive T-Lymphocytes ,Biology ,Ligands ,Fas ligand ,Minor Histocompatibility Antigens ,Mice ,Th2 Cells ,Antigen ,Cell Movement ,MHC class I ,medicine ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,RNA, Messenger ,fas Receptor ,Mice, Knockout ,Membrane Glycoproteins ,Beta-2 microglobulin ,Antibodies, Monoclonal ,Skin Transplantation ,Eosinophil ,Molecular biology ,Eosinophils ,Mice, Inbred C57BL ,Transplantation ,medicine.anatomical_structure ,biology.protein ,Cytokines ,Female ,Lymph Nodes ,beta 2-Microglobulin ,Injections, Intraperitoneal ,Spleen ,CD8 ,Signal Transduction - Abstract
Beta(2)-microglobulin (beta(2)m)-derived peptides are minor transplantation Ags in mice as beta(2)m-positive skin grafts (beta(2)m(+/+)) are rejected by genetically beta(2)m-deficient recipient mice (beta(2)m(-/-)). We studied the effector pathways responsible for the rejection induced by beta(2)-microglobulin-derived minor transplantation Ags. The rejection of beta(2)m(+/+) skin grafts by naive beta(2)m(-/-) mice was dependent on both CD4 and CD8 T cells as shown by administration of depleting mAbs. Experiments performed with beta(2)m(-/-)CD8(-/-) double knockout mice grafted with a beta(2)m(+/+) MHC class I-deficient skin showed that sensitized CD4 T cells directed at beta(2)m peptides-MHC class II complexes are sufficient to trigger rapid rejection. Rejection of beta(2)m(+/+) grafts was associated with the production of IL-5 in vitro, the expression of IL-4 and IL-5 mRNAs in the grafted tissue, and the presence within rejected grafts of a considerable eosinophil infiltrate. Blocking IL-4 and IL-5 in vivo and depleting eosinophils with an anti-CCR3 mAb prevented graft eosinophil infiltration and prolonged beta(2)m(+/+) skin graft survival. Lymphocytes from rejecting beta(2)m(-/-) mice also displayed an increased production of IFN-gamma after culture with beta(2)m(+/+) minor alloantigens. In vivo neutralization of IFN-gamma inhibited skin graft rejection. Finally, beta(2)m(+/+) skin grafts harvested from B6(lpr/lpr) donor mice, which lack a functional Fas molecule, survived longer than wild-type beta(2)m(+/+) skin grafts, showing that Fas-Fas ligand interactions are involved in the rejection process. We conclude that IL-4- and IL-5-dependent eosinophilic rejection, IFN-gamma-dependent mechanisms, and Fas-Fas ligand interactions are effector pathways in the acute rejection of minor transplantation Ags.
- Published
- 2002
- Full Text
- View/download PDF
32. Chronic Soluble Antigen Sensitization Primes a Unique Memory/Effector T Cell Repertoire Associated with Th2 Phenotype Acquisition In Vivo
- Author
-
Jean-Charles Guéry, Gilles Foucras, Christiane Coureau, Jean-M. Kanellopoulos, and Alexandra Gallard
- Subjects
Time Factors ,Receptors, Antigen, T-Cell, alpha-beta ,Molecular Sequence Data ,Immunology ,Cell ,Epitopes, T-Lymphocyte ,chemical and pharmacologic phenomena ,Biology ,Gene Rearrangement, T-Lymphocyte ,Lymphocyte Activation ,Conserved sequence ,Mice ,Th2 Cells ,medicine ,Animals ,Immunology and Allergy ,Amino Acid Sequence ,Antigens ,Cells, Cultured ,Mice, Inbred BALB C ,Base Sequence ,Sequence Homology, Amino Acid ,Cell growth ,Effector ,Repertoire ,T-cell receptor ,Histocompatibility Antigens Class II ,hemic and immune systems ,Phenotype ,medicine.anatomical_structure ,Genes, T-Cell Receptor beta ,Female ,Muramidase ,Interleukin-4 ,Signal transduction ,Immunologic Memory - Abstract
Although much progress has been made in characterization of the signaling pathways that control Th cell commitment, little is known about the early events that govern differentiation of IL-4-producing T lymphocytes in vivo. We have previously shown that chronic administration of low dose, soluble hen egg white lysozyme (HEL) induced the selective development of Ag-specific Th2 in genetically predisposed BALB/c mice. Here, we show that these memory/effector Th2 cells express a unique TCR Vβ repertoire, different from the TCR Vβ profile of primary effector cells from HEL-adjuvant-primed mice. This Th2-associated repertoire contains a highly frequent public clonotype characterized by preferred TCR AV and BV gene segment usage along with conserved sequences in the third hypervariable regions of both TCR chains. This Th2 clonotype, which is not recruited in primary effector T cells from HEL-adjuvant-immunized mice, recognized an IAd-restricted HEL determinant, preferentially processed by dendritic cells, but not by B cells. Thus, IL-4-producing CD4 T cells that expand following chronic Ag sensitization emerge from a distinct pool of precursors, supporting the hypothesis that ligand-TCR interactions play a crucial role in the regulation of Ag-specific Th2 cell development in vivo.
- Published
- 2002
- Full Text
- View/download PDF
33. Weak TCR stimulation induces a calcium signal that triggers IL-4 synthesis, stronger TCR stimulation induces MAP kinases that control IFN-γ production
- Author
-
Jean-Charles Guéry, Catherine Leclerc, Dominique Lagrange, Gilles Foucras, Pierre Paulet, Abdallah Badou, Marc Moreau, George Cassar, Magali Savignac, Lucette Pelletier, and Philippe Druet
- Subjects
MAPK/ERK pathway ,Cellular differentiation ,T cell ,Immunology ,T-cell receptor ,chemistry.chemical_element ,chemical and pharmacologic phenomena ,Stimulation ,Biology ,Calcium ,Cell biology ,medicine.anatomical_structure ,chemistry ,medicine ,Immunology and Allergy ,Signal transduction ,Calcium signaling - Abstract
Th1 and Th2 cells produce different cytokines and have distinct functions. Th1/Th2 cell differentiation is influenced, among other factors, by the nature of TCR-MHC interactions. However, how the TCR transduces a signal resulting in IFN-gamma or IL-4 production is a matter of debate. For example, some authors reported a loss of calcium signaling pathway in Th2 cells. We used a T cell hybridoma producing IL-4 upon weak TCR stimulation and both IL-4 and IFN-gamma for strong TCR engagement as a model to study how TCR signaling pathways are differentially activated in both conditions of stimulation and how this influences the production of cytokines. We show that: (1) the calcium response is identical following weak and strong TCR stimulation; (2) mitogen-activated protein kinase(MAPK) activation is a gradual phenomenon depending upon the strength of TCR activation; (3) a calcium response, even weak, triggers IL-4 expression; (4) IFN-gamma synthesis requires not only a calcium response but also MAPK activation. The MAPK pathway is dispensable for IL-4 production, although it amplifies IL-4 synthesis upon strong TCR stimulation; (5) TCR-induced IL-4 production also depends on calcium signaling in Th2 cells, while IFN-gamma synthesis is dependent, in addition, on MAPK activation in Th1 cells.
- Published
- 2001
- Full Text
- View/download PDF
34. Dendritic Cells Prime In Vivo Alloreactive CD4 T Lymphocytes Toward Type 2 Cytokine- and TGF-β-Producing Cells in the Absence of CD8 T Cell Activation
- Author
-
Jean-Charles Guéry, Jérôme D. Coudert, Christiane Coureau, and Gilles Foucras
- Subjects
CD4-Positive T-Lymphocytes ,Isoantigens ,Injections, Subcutaneous ,Immunology ,CD1 ,Priming (immunology) ,CD8-Positive T-Lymphocytes ,Biology ,Lymphocyte Activation ,Mice ,Th2 Cells ,Species Specificity ,Transforming Growth Factor beta ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,IL-2 receptor ,Antigen-presenting cell ,Cells, Cultured ,Mice, Knockout ,Mice, Inbred BALB C ,CD28 ,Cell Differentiation ,Dendritic Cells ,Th1 Cells ,MHC restriction ,Natural killer T cell ,Adoptive Transfer ,Cell biology ,Mice, Inbred C57BL ,Cytokines ,Immunization ,Interleukin-4 ,beta 2-Microglobulin ,Cell Division ,Spleen - Abstract
The mechanisms that influence the polarization of CD4 T cells specific for allogeneic MHC class II molecules in vivo are still poorly understood. We have examined the pathway of alloreactive CD4 T cell differentiation in a situation in which only CD4 T cells could be activated in vivo. In this report we show that priming of adult mice with allogeneic APC, in the absence of MHC class I-T cell interactions, induces a strong expansion of type 2 cytokine-producing allohelper T cells. These alloantigen-specific CD4 T cells directly recognize native allogeneic MHC class II molecules on APC and secrete, in addition to the prototypic Th2 cytokines IL-4, IL-5, and IL-10, large amounts of TGF-β. The default Th2-phenotype acquisition is not genetically controlled and occurred both in BALB/c and C57BL/6 mice. CD8 T cells are the principal cell type that controls CD4 T cell differentiation in vivo. Furthermore, we demonstrate that strong Th2 priming can be induced not only with allogeneic splenocytes but also with a low number of bone marrow-derived dendritic cells. Finally, using a passive transfer system, we provide direct evidence that CD8 T cell expansion in situ promotes alloreactive Th1 cell development principally by preventing their default development to the Th2 pathway in a mechanism that is largely IFN-γ independent. Therefore, this work demonstrates that type 2 cytokine production represents a dominant pathway of alloreactive CD4 T cell differentiation in adult mice, a phenomenon that was initially thought to occur only during the neonatal period.
- Published
- 2000
- Full Text
- View/download PDF
35. β2-Microglobulin-Dependent T Cells Are Not Necessary for Alloantigen-Induced Th2 Responses After Neonatal Induction of Lymphoid Chimerism in Mice
- Author
-
Gilles Foucras, Christiane Coureau, Leo Beijleveld, Philippe Druet, Abdelhadi Saoudi, and Jean-Charles Guéry
- Subjects
Immunology ,Immunology and Allergy - Abstract
We have analyzed the requirement for β2-microglobulin (β2m)-dependent T cells in the generation of allogeneic Th2 responses in vivo. A neonatal injection of semiallogeneic cells in BALB/c mice induces a state of chimerism that promotes the differentiation of donor-specific CD4+ T cells toward the Th2 phenotype. Polyclonal T-B cell interactions occur in this model between host Th2 and donor B cells, resulting in the production of IgE Abs. IgE production and Th2-priming are critically dependent upon the early production of IL-4. Our data in the present paper demonstrate that: 1) IgE synthesis and the up-regulation of MHC class II and CD23 molecules on B cells are independent of β2m expression in the host, 2) no difference in the induction of CD4 alloreactive Th2 cells could be observed between β2m−/− and their wild-type control littermates when Th2-priming was measured in adult mice, and 3) the Th2 response and IgE production is induced in the complete absence of β2m-dependent T cells both in the host and in the inoculum. Therefore, using a variety of assays, we could not demonstrate diminished responses in mice with a disrupted β2m gene in this model of Th2-mediated allogeneic interaction, indicating that β2m-dependent NK1.1+ and CD8+ T cells are not required for the generation of alloreactive Th2 responses in vivo.
- Published
- 1998
- Full Text
- View/download PDF
36. Regulation of the IL-12 receptor β2 subunit by soluble antigen and IL-12in vivo
- Author
-
Luciano Adorini, Jean-Charles Guéry, Francesca Galbiati, Lars Rogge, and Simona Smiroldo
- Subjects
Cell growth ,Cellular differentiation ,Immunology ,Priming (immunology) ,Biology ,Molecular biology ,Antigen ,Interferon ,In vivo ,medicine ,Interleukin 12 ,Immunology and Allergy ,Receptor ,medicine.drug - Abstract
Continuous administration of soluble protein antigen to BALB/c mice inhibits the development of Th1 and induces selective differentiation of Th2 cells. Here we show that interleukin (IL)-12, administered together with soluble protein through a mini-osmotic pump implanted subcutaneously, not only prevents the inhibition of Th1 cell development, but stimulates higher interferon (IFN)-gamma production than in mice receiving IL-12 alone. In parallel to co-stimulation of Th1 cell development, co-administration of IL-12 blocks the Th2 response induced by soluble protein. IL-12 administered in adjuvant with antigen or intraperitoneally 2 days after the immunization does not break the inhibition of Th1 but can still decrease the Th2 response induced by pretreatment with soluble protein antigen. In contrast to IL-12, co-administration of IL-2 or IFN-gamma does not affect the diversion to Th2 induced by soluble antigen. Thus IL-12, but not IL-2 nor IFN-gamma, converts in vivo the inhibitory signal for Th1 cell development delivered by soluble antigen into an immunogenic one, while blocking a positive signal for Th2 cell differentiation. A molecular basis for the co-stimulation of Th1 priming and the prevention of Th2 differentiation by IL-12 in vivo is provided by the observation that transcripts encoding the IL-12 receptor beta2 chain, which is required for IL-12 signaling and Th1 cell development, are selectively inhibited by soluble antigen but are enhanced by IL-12 co-administration.
- Published
- 1998
- Full Text
- View/download PDF
37. Impaired antigen presentation by murine I-Ad class II MHC molecules expressed in normal and HLA-DM-defective human B cell lines
- Author
-
James McCluskey, Jean-Charles Guéry, Tanya Boston, Sarah Weenink, Luciano Adorini, Elizabeth D. Mellins, Holger Averdunk, Vicki Boswarva, and Anand Gautam
- Subjects
T-Lymphocytes ,Immunology ,Antigen presentation ,CD1 ,HLA-DM ,Biology ,Cell Line ,Mice ,MHC class I ,Animals ,Humans ,Immunology and Allergy ,Antigen-presenting cell ,Antigen Presentation ,B-Lymphocytes ,HLA-D Antigens ,MHC class II ,Hybridomas ,Antigen processing ,Histocompatibility Antigens Class II ,General Medicine ,MHC restriction ,Molecular biology ,Antigens, Differentiation, B-Lymphocyte ,Mutagenesis ,Mutation ,biology.protein ,Dimerization - Abstract
The inability of certain antigen processing mutant cell lines to present intact proteins to T cells and to form SDS-stable MHC class II dimers has been shown to result from defective expression of HLA-encoded DMA and DMB genes. We have utilized some of these mutants to determine species compatibility of antigen presentation components. Mouse MHC class II I-A d cDNA was transfected into the human B cell lymphoblastoid cell lines 8.1.6, 7.9.6 (a mutant cell line derived from 8.1.6) and an independent deletion mutant T2 (called 8.1.6d, 7.9.6d and T2.d respectively). These cells were then examined for various functions in antigen presentation. Interestingly, none of the cells transfected with I-A d presented peptides derived from intact proteins to specific T cell hybridomas. However, presentation of synthetic peptides by these cells was normal. The ability to form SDS-stable dimers was dramatically reduced in the transfectants. In addition, I-A d molecules at the cell surface appeared loaded predominantly with the invariant chain peptides, CLIP. These properties of the I-A d transfectants are identical to those described for HLA class II molecules expressed in HLA-DM mutants. Perhaps the most interesting finding was the inability of I-A d in 8.1.6 to present protein antigens. Since 8.1.6 cells present antigens to HLA-DR, DP, DQ-restricted T cells and also have intact HLA-DM and invariant chain (Ii) functions, these results argue that some component of human antigen processing machinery is incompatible with I-A d molecules.
- Published
- 1997
- Full Text
- View/download PDF
38. The mode of protein antigen administration determines preferential presentation of peptide-class II complexes by lymph node dendritic or B cells
- Author
-
Jean-Charles Guéry, Simona Smiroldo, Francesca Galbiati, Francesco Ria, and Luciano Adorini
- Subjects
medicine.medical_treatment ,Molecular Sequence Data ,Immunology ,Population ,Antigen presentation ,chemical and pharmacologic phenomena ,Cell Separation ,Mice ,Antigen ,Lymph node stromal cell ,medicine ,Animals ,Immunology and Allergy ,Amino Acid Sequence ,Antigens ,education ,Antigen-presenting cell ,Lymph node ,Antigen Presentation ,B-Lymphocytes ,Mice, Inbred BALB C ,education.field_of_study ,Chemistry ,Histocompatibility Antigens Class II ,hemic and immune systems ,Dendritic Cells ,General Medicine ,Virology ,Molecular biology ,medicine.anatomical_structure ,Solubility ,Female ,Muramidase ,Lymph Nodes ,Lymph ,Peptides ,Adjuvant - Abstract
We have compared the capacity of dendritic cells (DC) and B cells to present peptide-class II complexes following administration of protein in adjuvant or in soluble form. Three different antigen-presenting cell (APC) populations were separated from draining lymph node cells from mice immunized s.c. with hen egg-white lysozyme (HEL) in adjuvant or with adjuvant only followed by soluble HEL: DC (N418+, class II+, B220-, low buoyant density), large B cells (B220+, low buoyant density) and small B cells (B220+, high buoyant density). HEL peptide-class II complexes displayed by these APC were evaluated by their capacity to activate HEL-specific T hybridoma cells. Following immunization with HEL in adjuvant, DC are the only lymph node APC population expressing detectable HEL peptide-class II complexes. Conversely, after i.v. administration of soluble HEL in mice previously injected with adjuvant only, lymph node B cells are much more efficient than DC in presenting peptide-class II complexes to T cells. Therefore, different modes of protein antigen administration lead to selective expression of antigenic complexes by different APC populations. These data correlate with the observation that, unlike B cells, DC recruited in lymph nodes of mice injected with adjuvant only present in vitro processed protein antigen much less efficiently than synthetic peptides, probably as a consequence of their maturation in vivo.
- Published
- 1997
- Full Text
- View/download PDF
39. Estradiol promotes functional responses in inflammatory and steady-state dendritic cells through differential requirement for activation function-1 of estrogen receptor α
- Author
-
Sophie Laffont, Cyril Seillet, Pierre Chambon, Andrée Krust, Nelly Rouquié, Victorine Douin-Echinard, Jean-Charles Guéry, Jean-François Arnal, Eliane Foulon, Simon, Marie Francoise, Centre de Physiopathologie Toulouse Purpan (CPTP), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), and Université de Strasbourg (UNISTRA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Male ,MESH: Signal Transduction ,Myeloid ,Estrogen receptor ,Mice ,0302 clinical medicine ,Transcription (biology) ,Immunology and Allergy ,MESH: Animals ,MESH: Estrogen Receptor alpha ,0303 health sciences ,Estradiol ,biology ,MESH: Dendritic Cells ,Toll-Like Receptors ,Cell Differentiation ,hemic and immune systems ,MESH: Antigens, CD11 ,MESH: Granulocyte-Macrophage Colony-Stimulating Factor ,Cell biology ,Phenotype ,medicine.anatomical_structure ,Integrin alpha M ,Interferon Regulatory Factors ,Female ,MESH: Membrane Proteins ,MESH: Estradiol ,MESH: Interferon Regulatory Factors ,MESH: Toll-Like Receptors ,Signal Transduction ,MESH: Cell Differentiation ,Cell type ,MESH: Mice, Transgenic ,Immunology ,Mice, Transgenic ,MESH: Phenotype ,03 medical and health sciences ,Downregulation and upregulation ,medicine ,[SDV.BBM] Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Animals ,Protein Interaction Domains and Motifs ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Progenitor cell ,MESH: Mice ,030304 developmental biology ,MESH: Protein Interaction Domains and Motifs ,CD11 Antigens ,Estrogen Receptor alpha ,Granulocyte-Macrophage Colony-Stimulating Factor ,Membrane Proteins ,Dendritic Cells ,Dendritic cell ,MESH: Male ,biology.protein ,MESH: Female ,030215 immunology - Abstract
17β-Estradiol (E2) has been shown to regulate GM-CSF– or Flt3 ligand–driven dendritic cell (DC) development through estrogen receptor (ER) α signaling in myeloid progenitors. ERα regulates transcription of target genes through two distinct activation functions (AFs), AF-1 and AF-2, whose respective involvement varies in a cell type– or tissue-specific manner. In this study, we investigated the role of ERα AFs in the development and effector functions of inflammatory DCs, steady-state conventional DCs, and plasmacytoid DCs (pDC), using mouse lacking either AF-1 or AF-2. In agreement with previous works, we showed that E2 fostered the differentiation and effector functions of inflammatory DCs through ERα-dependent upregulation of IFN regulatory factor (IRF)-4 in GM-CSF–stimulated myeloid progenitors. Interestingly, whereas AF-1 was required for early IRF-4 upregulation in DC precursors, it was dispensable to enhance IRF-4 expression in differentiated DCs to a level compatible with the development of the more functional Ly6C− CD11b+ DC subset. Presence of E2 had no effect on progenitors from either knock-in mice with 7-aa deletion in helix 12 of ERα, lacking AF-2, or ERα−/− mice. By contrast, in Flt3 ligand–driven DC differentiation, activation of AF-1 domain was required to promote the development of more functionally competent conventional DCs and pDCs. Moreover, lack of ERα AF-1 blunted the TLR7-mediated IFN-α response of female pDCs in vivo. Thus, our study demonstrates that ERα uses AF-1 differently in steady-state and inflammatory DC lineages to regulate their innate functions, suggesting that selective ER modulators could be used to target specific DC subsets.
- Published
- 2013
- Full Text
- View/download PDF
40. DRα:Eβ heterodimers in DRA transgenic mice hinder expression of Eα:Eβ molecules and are more efficient in antigen presentation
- Author
-
Alessandro Sette, Ettore Appella, Sylvie Trembleau, Luciano Adorini, Patrizio Giacomini, Juergen Hammer, Andrea Setini, and Jean-Charles Guéry
- Subjects
Chemistry ,Immunology ,T-cell receptor ,Antigen presentation ,Immunology and Allergy ,Alpha (ethology) ,Peptide binding ,General Medicine ,Immunodominance ,MHC restriction ,Beta (finance) ,Molecular biology ,Alpha chain - Abstract
HLA-DRA transgenic (tg) mice on H-2d background were constructed to study assembly, expression and function of DR alpha: E beta class II heterodimers when an alternate E alpha chain is available. Cytofluorimetric analysis and immunoprecipitation studies demonstrate that the majority (90%) of E beta d molecules on class II-positive splenocytes from DRA-tg mice are associated with DR alpha rather than E alpha chains. To characterize the functional role of the interspecies as compared with the wild-type I-E molecules, MHC restriction and T cell epitope immunodominance of synthetic peptides spanning the entire sequence of 65 kDa heat shock protein (hsp) from Mycobacterium tuberculosis were determined in hsp-primed DRA-tg and DBA/2 mice. A similar pattern of responsiveness was observed in both strains, but hsp epitopes recalled a higher response in DRA-tg as compared with DBA/2 mice. A panel of T cell hybridomas specific for two hsp peptides or a hen egg white lysozyme peptide presented by both DR alpha: E beta d and E alpha d: E beta d was studied in detail. Surprisingly, DR alpha: E beta d dimers present these peptides more efficiently than E alpha d: E beta d, even when the TCR was selected in mice expressing only E alpha d: E beta d molecules. The higher efficiency of antigen presentation by DR alpha: E beta d dimers does not appear to depend on increased binding affinity for peptides, as demonstrated by competition for antigen presentation, nor on increased efficiency in the interaction with CD4 molecules. Rather, the higher efficiency of antigen presentation could be explained by a more effective ligand-TCR interaction. This is consistent with molecular modeling based on the class II structure, indicating that 16 out of 17 substitutions between the first domain of E alpha d and DR alpha chains ile outside the peptide binding groove and are potentially available for interaction with the TCR.
- Published
- 1995
- Full Text
- View/download PDF
41. The TLR-mediated response of plasmacytoid dendritic cells is positively regulated by estradiol in vivo through cell-intrinsic estrogen receptor α signaling
- Author
-
Sophie Laffont, Victorine Douin-Echinard, F. Tremollieres, Jean-Charles Guéry, Cyril Seillet, Pierre Gourdy, Claude Ribot, Nelly Rouquié, Jean-François Arnal, Simon, Marie Francoise, Centre de Physiopathologie Toulouse Purpan (CPTP), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre de Ménopause [CHU Toulouse], Centre Hospitalier Universitaire de Toulouse (CHU Toulouse), Centre de Ménopause, CHU Toulouse [Toulouse]-Hôpital Paule de Viguier, and CHU Toulouse [Toulouse]
- Subjects
MESH: Interferon Type I ,MESH: Signal Transduction ,Male ,MESH: Mice, Inbred NOD ,medicine.medical_treatment ,Estrogen receptor ,[SDV.GEN] Life Sciences [q-bio]/Genetics ,Mice, SCID ,Biochemistry ,Mice ,0302 clinical medicine ,Mice, Inbred NOD ,MESH: Animals ,MESH: Mice, SCID ,Receptor ,MESH: Estrogen Receptor alpha ,0303 health sciences ,Toll-like receptor ,MESH: Toll-Like Receptor 9 ,MESH: Cytokines ,MESH: Middle Aged ,MESH: Dendritic Cells ,Estradiol ,hemic and immune systems ,Hematology ,Middle Aged ,MESH: Case-Control Studies ,3. Good health ,Cell biology ,MESH: Toll-Like Receptor 7 ,Postmenopause ,Cytokine ,MESH: Young Adult ,Interferon Type I ,Cytokines ,Female ,MESH: Estradiol ,Signal transduction ,MESH: Interferon-alpha ,MESH: Postmenopause ,Signal Transduction ,Adult ,Adolescent ,medicine.drug_class ,Immunology ,Biology ,03 medical and health sciences ,Young Adult ,MESH: Mice, Inbred C57BL ,medicine ,Animals ,Humans ,MESH: Mice ,030304 developmental biology ,MESH: Adolescent ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,MESH: Humans ,Estrogen Receptor alpha ,Interferon-alpha ,MESH: Adult ,Cell Biology ,Dendritic Cells ,MESH: Male ,Mice, Inbred C57BL ,Toll-Like Receptor 7 ,Estrogen ,Case-Control Studies ,Toll-Like Receptor 9 ,Cytokine secretion ,Estrogen receptor alpha ,MESH: Female ,030215 immunology - Abstract
International audience; Plasmacytoid dendritic cells (pDCs) produce large amounts of type I interferons (IFN-α/β) in response to viral or endogenous nucleic acids through activation of their endosomal Toll-like receptors (TLR-7 and TLR-9). Enhanced TLR-7-mediated IFN-α production by pDCs in women, compared with men, has been reported, but whether sex hormones, such as estrogens, are involved in this sex-based difference is unknown. Here we show, in humanized mice, that the TLR-7-mediated response of human pDCs is increased in female host mice relative to male. In a clinical trial, we establish that treatment of postmenopausal women with 17β-estradiol markedly enhances TLR-7- and TLR-9-dependent production of IFN-α by pDCs stimulated by synthetic ligands or by nucleic acid-containing immune complexes. In mice, we found exogenous and endogenous estrogens to promote the TLR-mediated cytokine secretion by pDCs through hematopoietic expression of estrogen receptor (ER) α. Genetic ablation of ERα gene in the DC lineage abrogated the enhancing effect of 17β-estradiol on their TLR-mediated production of IFN-α, showing that estrogens directly target pDCs in vivo. Our results uncover a previously unappreciated role for estrogens in regulating the innate functions of pDCs, which may account for sex-based differences in autoimmune and infectious diseases.
- Published
- 2011
- Full Text
- View/download PDF
42. Estradiol administration controls eosinophilia through estrogen receptor-alpha activation during acute peritoneal inflammation
- Author
-
Jean-Charles Guéry, Pierre Gourdy, Victorine Douin-Echinard, Jean-François Arnal, Françoise Lenfant, Audrey Billon-Galés, Florence Trémollières, Bertrand Calippe, Francis Bayard, Coralie Fontaine, Institut de médecine moléculaire de Rangueil (I2MR), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-IFR150-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre de Physiopathologie Toulouse Purpan (CPTP), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Simon, Marie Francoise, and Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées- Institut Fédératif de Recherche Bio-médicale Institution (IFR150)-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
medicine.medical_specialty ,medicine.drug_class ,Immunology ,Inflammation ,Apoptosis ,Cell Separation ,Peritonitis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Internal medicine ,Eosinophilia ,medicine ,Immunology and Allergy ,Animals ,Interleukin 5 ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Estradiol ,business.industry ,Reverse Transcriptase Polymerase Chain Reaction ,Estrogen Receptor alpha ,Estrogens ,Cell Biology ,Eosinophil ,respiratory system ,Flow Cytometry ,Eosinophils ,Mice, Inbred C57BL ,Chemotaxis, Leukocyte ,Disease Models, Animal ,medicine.anatomical_structure ,Endocrinology ,Estrogen ,Female ,medicine.symptom ,Interleukin-5 ,business ,Estrogen receptor alpha ,030215 immunology - Abstract
Estrogens influence the incidence and the course of numerous immune or inflammatory diseases in humans and in experimental models. For instance, estrogens prevent the accumulation of granulocytes in acute inflammatory murine models, but the respective actions on neutrophil and eosinophil trafficking remain to be clarified. We demonstrate here that in a model of TGC-induced sterile peritonitis in ovx mice, chronic E2 administration electively and strongly inhibited peritoneal eosinophil accumulation. E2 decreased BM eosinophil number, contributing to a marked prevention of the TGC-induced eosinophil blood mobilization. These effects on eosinophil mobilization and peritoneal accumulation were abolished in ER-α−/− mice, demonstrating the crucial role of this nuclear receptor. Grafting ER-α−/− mice with ER-α+/+ BM cells restored the suppressive effect of E2 on peritoneal eosinophilia, although the action on eosinophil blood mobilization was still abrogated. We therefore explored additional mechanisms and found that E2 reduced the peritoneal concentrations of key eosinophil prosurvival factors (IL-5, IL-9, and IL-25) and enhanced eosinophil apoptosis during the inflammatory process. Furthermore, this proapoptotic effect of E2 was abrogated in IL-5-overexpressing Tg mice. To conclude, we demonstrate for the first time that ER-α activation by exogenous E2 administration strongly inhibits eosinophil accumulation during acute inflammation in a nonreproductive target site for estrogen through combined actions on eosinophil mobilization and apoptosis. This specific, suppressive effect of chronic E2 replacement therapy on eosinophils has to be integrated to further understand the evolution of eosinophil-associated diseases in menopausal women.
- Published
- 2011
- Full Text
- View/download PDF
43. Endogenous estrogens, through estrogen receptor α, constrain autoimmune inflammation in female mice by limiting CD4+ T-cell homing into the CNS
- Author
-
Lucette Pelletier, Jean-Charles Guéry, Karine Lélu, Virginie Robert, Britta Engelhardt, Sophie Laffont, Laurent Delpy, Eliane Foulon, Contrôle de la Réponse Immune B et des Lymphoproliférations (CRIBL), Université de Limoges (UNILIM)-Génomique, Environnement, Immunité, Santé, Thérapeutique (GEIST FR CNRS 3503)-Centre National de la Recherche Scientifique (CNRS), Interactions hôtes-agents pathogènes [Toulouse] (IHAP), Institut National de la Recherche Agronomique (INRA)-Ecole Nationale Vétérinaire de Toulouse (ENVT), Institut National Polytechnique (Toulouse) (Toulouse INP), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National Polytechnique (Toulouse) (Toulouse INP), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées, and Centre National de la Recherche Scientifique (CNRS)-Université de Limoges (UNILIM)-Génomique, Environnement, Immunité, Santé, Thérapeutique (GEIST FR CNRS 3503)
- Subjects
CD4-Positive T-Lymphocytes ,Adoptive cell transfer ,Estrogen receptor ,Inbred C57BL ,MESH: Estrogens ,MESH: Mice, Knockout ,Mice ,0302 clinical medicine ,Cell Movement ,Immunology and Allergy ,MESH: Animals ,Encephalomyelitis ,MESH: Cell Movement ,MESH: Estrogen Receptor alpha ,Cells, Cultured ,Mice, Knockout ,Cultured ,biology ,Brain ,MESH: CD4-Positive T-Lymphocytes ,Adoptive Transfer ,Myelin-Associated Glycoprotein ,medicine.anatomical_structure ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Female ,medicine.symptom ,Inflammation Mediators ,Myelin Proteins ,MESH: Cells, Cultured ,medicine.medical_specialty ,Encephalomyelitis, Autoimmune, Experimental ,Multiple Sclerosis ,medicine.drug_class ,Knockout ,Ovariectomy ,Cells ,Immunology ,MESH: Inflammation Mediators ,MESH: Ovariectomy ,Inflammation ,Myelin oligodendrocyte glycoprotein ,03 medical and health sciences ,Experimental ,MESH: Brain ,Immune system ,MESH: Mice, Inbred C57BL ,Internal medicine ,medicine ,Animals ,Humans ,MESH: Myelin Proteins ,MESH: Myelin-Associated Glycoprotein ,MESH: Encephalomyelitis, Autoimmune, Experimental ,MESH: Mice ,MESH: Humans ,Animal ,Estrogen Receptor alpha ,Estrogens ,MESH: Multiple Sclerosis ,Oligodendrocyte ,Mice, Inbred C57BL ,Disease Models, Animal ,MESH: Adoptive Transfer ,MESH: Cytoprotection ,Endocrinology ,Estrogen ,Cytoprotection ,Disease Models ,biology.protein ,Myelin-Oligodendrocyte Glycoprotein ,MESH: Disease Models, Animal ,MESH: Female ,030217 neurology & neurosurgery ,030215 immunology ,Hormone ,Autoimmune - Abstract
International audience; Sex hormones influence immune responses and the development of autoimmune diseases including MS and its animal model, EAE. Although it has been previously reported that ovariectomy could worsen EAE, the mechanisms implicated in the protective action of endogenous ovarian hormones have not been addressed. In this report, we now show that endogenous estrogens limit EAE development and CNS inflammation in adult female mice through estrogen receptor α expression in the host non-hematopoietic tissues. We provide evidence that the enhancing effect of gonadectomy on EAE development was due to quantitative rather than qualitative changes in effector Th1 or Th17 cell recruitment into the CNS. Consistent with this observation, adoptive transfer of myelin oligodendrocyte glycoprotein-specific encephalitogenic CD4(+) T lymphocytes induced more severe EAE in ovariectomized mice as compared to normal female mice. Finally, we show that gonadectomy accelerated the early recruitment of inflammatory cells into the CNS upon adoptive transfer of encephalitogenic CD4(+) T cells. Altogether, these data show that endogenous estrogens, through estrogen receptor α, exert a protective effect on EAE by limiting the recruitment of blood-derived inflammatory cells into the CNS.
- Published
- 2010
- Full Text
- View/download PDF
44. 17Beta-estradiol promotes TLR4-triggered proinflammatory mediator production through direct estrogen receptor alpha signaling in macrophages in vivo
- Author
-
Jean-Charles Guéry, Andrée Krust, Bernard Pipy, Muriel Laffargue, Jean-François Arnal, Karine Lélu, Victorine Douin-Echinard, Francis Bayard, Laurent Delpy, Pierre Gourdy, Bertrand Calippe, Institut de médecine moléculaire de Rangueil (I2MR), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-IFR150-Institut National de la Santé et de la Recherche Médicale (INSERM), Physiologie Moléculaire de la Réponse Immune et des Lymphoproliférations (PMRIL), Université de Limoges (UNILIM)-Génomique, Environnement, Immunité, Santé, Thérapeutique (GEIST FR CNRS 3503)-Centre National de la Recherche Scientifique (CNRS), Centre de Physiopathologie Toulouse Purpan (CPTP), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut Clinique de la Souris (ICS), Université de Strasbourg (UNISTRA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Unité Propre de Recherche de l'Enseignement Supérieur Associée (EA 2405), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-IFR150, Service de Diabétologie, Pôle CardioVasculaire et Métabolique, CHU Toulouse [Toulouse], Simon, Marie Francoise, Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées- Institut Fédératif de Recherche Bio-médicale Institution (IFR150)-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Fédératif de Recherche Bio-médicale Institution (IFR150), Service Diabétologie [CHU Toulouse], Pôle Cardiovasculaire et Métabolique [CHU Toulouse], and Centre Hospitalier Universitaire de Toulouse (CHU Toulouse)-Centre Hospitalier Universitaire de Toulouse (CHU Toulouse)
- Subjects
medicine.medical_specialty ,Adipose tissue macrophages ,medicine.medical_treatment ,Blotting, Western ,Immunology ,Biology ,Proinflammatory cytokine ,Mice ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Immunology and Allergy ,Macrophage ,10. No inequality ,Cells, Cultured ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Estradiol ,Reverse Transcriptase Polymerase Chain Reaction ,Estrogen Receptor alpha ,Flow Cytometry ,Cell biology ,Mice, Inbred C57BL ,Toll-Like Receptor 4 ,Endocrinology ,Cytokine ,Thioglycolates ,Macrophages, Peritoneal ,TLR4 ,Cytokines ,Female ,Inflammation Mediators ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Estrogen receptor alpha ,030217 neurology & neurosurgery ,Ex vivo ,Signal Transduction - Abstract
17β-estradiol (E2) has been shown to promote the expression of inflammatory mediators by LPS-activated tissue resident macrophages through estrogen receptor α (ERα) signaling. However, it remained to be determined whether E2 similarly influences macrophages effector functions under inflammatory conditions in vivo, and whether this action of E2 resulted from a direct effect on macrophages. We show in this study that chronic E2 administration to ovariectomized mice significantly increased both cytokine (IL-1β, IL-6, and TNF-α) and inducible NO synthase mRNA abundance in thioglycolate (TGC)-elicited macrophages. The proinflammatory action of E2 was also evidenced at the level of released IL-1β and IL-6 by ex vivo LPS-activated macrophages. E2 concomitantly inhibited PI3K activity as well as Akt phosphorylation in TGC-elicited macrophages, suggesting that E2 promoted TLR-dependent macrophage activation by alleviating this suppressive signaling pathway. Indeed, this effect was abolished in the presence of the inhibitor wortmannin, demonstrating a key functional link between inhibition of PI3K activity and the E2 action on macrophage functions. Endogenous estrogens levels circulating in ovary-intact mice were sufficient to promote the above described actions. Finally, thanks to a CreLox strategy, targeted disruption of ERα gene in macrophages totally abolished the effect of E2 on the expression of inflammatory mediators by both resident and TGC-elicited peritoneal macrophages. In conclusion, we demonstrate that estrogens, through the activation of ERα in macrophages in vivo, enhance their ability to produce inflammatory mediators and cytokines upon subsequent TLR activation.
- Published
- 2010
- Full Text
- View/download PDF
45. Selective immunosuppression by administration of major histocompatibility complex (MHC) class II-binding peptides. I. Evidence for in vivo MHC blockade preventing T cell activation
- Author
-
A Sette, Luciano Adorini, A Dragomir, Jean-Charles Guéry, and J Leighton
- Subjects
T cell ,T-Lymphocytes ,Immunology ,Antigen presentation ,Molecular Sequence Data ,Antigen-Presenting Cells ,chemical and pharmacologic phenomena ,Biology ,Major histocompatibility complex ,Lymphocyte Activation ,Binding, Competitive ,Antigen ,medicine ,Immune Tolerance ,Immunology and Allergy ,Amino Acid Sequence ,Antigen-presenting cell ,MHC class II ,Hybridomas ,Antigen processing ,Histocompatibility Antigens Class II ,hemic and immune systems ,Articles ,MHC restriction ,Molecular biology ,medicine.anatomical_structure ,biology.protein ,Muramidase ,Cell Division - Abstract
Draining lymph node cells (LNC) from mice immunized with hen egg white lysozyme (HEL) display at their surface antigen-MHC complexes able to stimulate, in the absence of any further antigen addition, HEL peptide-specific, class II-restricted T cell hybridomas. Chloroquine addition to these LNC cultures fails to inhibit antigen presentation, indicating that antigenic complexes of class II molecules and HEL peptides are formed in vivo. MHC class II restriction of antigen presentation by LNC from HEL-primed mice was verified by the use of anti-class II monoclonal antibodies. Coinjection of HEL and the I-Ak-binding peptide HEL 112-129 in mice of H-2k haplotype inhibits the ability of LNC to stimulate I-Ak-restricted, HEL 46-61-specific T cell hybridomas. Similar results are obtained in mice coinjected with the HEL peptides 46-61 and 112-129. Inhibition of T hybridoma activation can also be observed using as antigen-presenting cells irradiated, T cell-depleted LNC from mice coinjected with HEL 46-61 and HEL 112-129, ruling out the possible role of either specific or nonspecific suppressor T cells. Inhibition of T cell proliferation is associated with MHC-specific inhibition of antigen presentation and with occupancy by the competitor of class II binding sites, as measured by activation of peptide-specific T cell hybridomas. These results demonstrate that administration of MHC class II binding peptide competitors selectively inhibits antigen presentation to class II-restricted T cells, indicating competitive blockade of class II molecules in vivo.
- Published
- 1992
46. Natural killer cells recruited into lymph nodes inhibit alloreactive T-cell activation through perforin-mediated killing of donor allogeneic dendritic cells
- Author
-
Jean-Charles Guéry, Sophie Laffont, Cyril Seillet, John R. Ortaldo, Jérôme D. Coudert, Centre de Physiopathologie Toulouse Purpan (CPTP), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Laboratory of experimental immunology, Frederick National Laboratory for Cancer Research (FNLCR), financement Etablissement Français des Greffes, collaboration avec John Ortaldo (NCIF, MD, and USA)
- Subjects
CD4-Positive T-Lymphocytes ,Cytotoxicity, Immunologic ,Graft Rejection ,transplant rejection: responses to alloantigens ,medicine.medical_treatment ,T cell ,Immunology ,Priming (immunology) ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Biochemistry ,DC biology ,03 medical and health sciences ,Interleukin 21 ,Mice ,0302 clinical medicine ,NK-92 ,Cell Movement ,medicine ,Animals ,Interleukin-7 receptor ,030304 developmental biology ,Innate immunity ,0303 health sciences ,biology ,Perforin ,NK cell biology ,Cell Biology ,Hematology ,Dendritic Cells ,Skin Transplantation ,3. Good health ,Killer Cells, Natural ,medicine.anatomical_structure ,biology.protein ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Lymph Nodes ,CD8 ,030215 immunology ,Allotransplantation - Abstract
11 pages; International audience; Natural killer (NK)-cell alloreactivity is exploited in bone marrow transplantation to improve clinical outcome. Likewise, in solid organ transplantation, it has been recently shown that recipient NK cells may limit alloreactive T-cell responses through their capacity to prevent the persistence of graft-derived allogeneic dendritic cells (DCs). In a model of CD4(+) T cell-mediated allogeneic skin graft rejection, we show that the absence of host NK-cell alloreactivity was characterized by enhanced expansion of alloreactive effector T lymphocytes, including Th2 cells, and massive eosinophilic infiltrates in the rejected tissues. In CD8(+) T cell-deficient C57BL/6 (H-2(b)) recipients injected with allogeneic BALB/c (H-2(d)) DCs, we demonstrated that NK cells expressing the H-2D(d)-specific Ly49D activating receptor were implicated in the regulation of alloreactive CD4(+) T-cell responses. Moreover, we showed that Ly49D(+) CD127(-) NK cells were recruited within DC draining lymph nodes and rapidly eliminated allogeneic H-2(d) DCs through the perforin pathway. In normal mice, we further demonstrated that NK cells by quickly eliminating allogeneic DCs strongly inhibited alloreactive CD8(+) T-cell responses. Thus, NK cells act as early regulators of alloreactive T-cell priming in allotransplantation through their capacity to kill allogeneic DCs in draining lymph nodes.
- Published
- 2008
- Full Text
- View/download PDF
47. A spontaneous hybridoma producing autoanti-idiotypic antibodies that recognizea Vx−associated idiotope in mercury-induced autoimmunity
- Author
-
Philippe Druet and Jean-Charles Guéry
- Subjects
Idiotype ,Renal glomerulus ,medicine.drug_class ,Blotting, Western ,Kidney Glomerulus ,Immunology ,medicine.disease_cause ,Monoclonal antibody ,Autoimmune Diseases ,Autoimmunity ,Immunoglobulin kappa-Chains ,Glomerulonephritis ,Western blot ,Rats, Inbred BN ,medicine ,Animals ,Immunology and Allergy ,Autoantibodies ,Hybridomas ,biology ,medicine.diagnostic_test ,Antibodies, Monoclonal ,Idiotopes ,Molecular biology ,Antibodies, Anti-Idiotypic ,Rats ,Mercuric Chloride ,Monoclonal ,biology.protein ,Antibody - Abstract
Anti-idiotypic (Id) antibodies have been suggested to play a role in the self regulation process observed in Brown-Norway rats developing mercury-induced autoimmunity. However, the presence of such antibodies has not yet been directly demonstrated. For that purpose, spleen cells from a mercury-injected rat were fused and the resulting hybridomas tested for their anti-Id activity against monoclonal anti-glomerular basement membrane (GBM) antibodies produced in this model. A monoclonal antibody (mAb) was obtained that specifically reacted in an enzyme-linked immunosorbent assay with an anti-GBM mAb and to a much lesser extent with another one produced in the same fusion. In Western blot experiments this autoanti-Id mAb reacted under reducing conditions with the kappa L chains but not with the H chains of the two anti-GBM mAb. It did not react with the kappa L chains of eight other rat mAb. This mAb is therefore an autoanti-Id mAb that recognizes a V kappa-associated Id expressed on two anti-GBM mAb. These results demonstrate that anti-GBM antibodies and their corresponding autoanti-Id antibodies are simultaneously produced during this disease. Whether or not these autoanti-Id antibodies have a regulatory and/or a pathogenic role in this disease remains to be established.
- Published
- 1990
- Full Text
- View/download PDF
48. Specificity and cross-reactive idiotypes of anti-glomerular basement membrane autoantibodies in HgCl2-induced autoimmune glomerulonephritis
- Author
-
François Hirsch, Chantal Mandet, Jean-Charles Guéry, E. Druet, Denis Glotz, Philippe Druet, and E. de Heer
- Subjects
medicine.drug_class ,Kidney Glomerulus ,Immunology ,Fluorescent Antibody Technique ,Cross Reactions ,Immunofluorescence ,Monoclonal antibody ,Autoantigens ,Basement Membrane ,Autoimmune Diseases ,Glomerulonephritis ,Immunoglobulin Idiotypes ,Antibody Specificity ,Rats, Inbred BN ,medicine ,Animals ,Immunology and Allergy ,Autoantibodies ,Basement membrane ,biology ,medicine.diagnostic_test ,Glomerular basement membrane ,Autoantibody ,Antibodies, Monoclonal ,medicine.disease ,Molecular biology ,Rats ,medicine.anatomical_structure ,Polyclonal antibodies ,Mercuric Chloride ,biology.protein ,Laminin ,Antibody - Abstract
Mercury-induced autoimmune glomerulonephritis in the Brown-Norway (BN) rat is characterized by the successive appearance of linear and granular glomerular IgG deposits. Anti-laminin autoantibodies represent the major part of the anti-glomerular basement membrane (GBM) antibodies produced in this model. Fusions were performed in this model and four anti-GBM monoclonal antibodies (mAb) were obtained. Three of them were laminin specific. Using rabbit anti-idiotype antibodies, cross-reactive idiotypes (CRId) were characterized on anti-laminin antibodies. They were expressed on the three anti-laminin mAb, on kidney-eluted and circulating anti-laminin antibodies. CRId-bearing immunoglobulins were detected transiently in the circulation and paralleled the anti-laminin antibody activity. By immunofluorescence studies on kidney cryostat sections two different CRId were defined. One was localized close to the antigen-combining site since it was not revealed on kidney-bound antibodies, in contrast with the second CRId. This latter CRId was also found deposited in a typical linear pattern in the early phase of the disease and in a granular pattern in the late phase, demonstrating that these CRId are components of immune deposits. Taken together, these results suggest that in this model of T-dependent polyclonal B cell activation, restricted sets of V genes encode for at least a part of the anti-GBM autoantibodies.
- Published
- 1990
- Full Text
- View/download PDF
49. Calcium channel blocker prevents T helper type 2 cell-mediated airway inflammation
- Author
-
Bruno Gomes, Catherine Leclerc, Pierre Paulet, Bernard Mariamé, Marilena Djata Cabral, Jean-Charles Guéry, Alexandra Gallard, Marc Moreau, Philippe Druet, Magali Savignac, Lucette Pelletier, Centre de Physiopathologie Toulouse Purpan (CPTP), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre de biologie du développement (CBD), Centre de Biologie Intégrative (CBI), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Centre National de la Recherche Scientifique (CNRS)-Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Centre National de la Recherche Scientifique (CNRS)-Centre National de la Recherche Scientifique (CNRS), Centre National de la Recherche Scientifique (CNRS)-Université Toulouse III - Paul Sabatier (UT3), and Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Centre de Biologie Intégrative (CBI)
- Subjects
MESH: Inflammation ,CD4-Positive T-Lymphocytes ,Intracellular Fluid ,MESH: Asthma ,medicine.medical_treatment ,MESH: Calcium Channel Blockers ,Critical Care and Intensive Care Medicine ,Lymphocyte Activation ,Severity of Illness Index ,Mice ,0302 clinical medicine ,MESH: Animals ,MESH: Administration, Intranasal ,Lung ,0303 health sciences ,Mice, Inbred BALB C ,biology ,MESH: Dendritic Cells ,MESH: Intracellular Fluid ,MESH: Enzyme-Linked Immunosorbent Assay ,MESH: CD4-Positive T-Lymphocytes ,respiratory system ,Calcium Channel Blockers ,3. Good health ,medicine.anatomical_structure ,Cytokine ,MESH: Calcium ,Female ,medicine.symptom ,Bronchoalveolar Lavage Fluid ,MESH: Injections, Intraperitoneal ,Injections, Intraperitoneal ,medicine.drug ,Pulmonary and Respiratory Medicine ,MESH: Mice, Transgenic ,Ovalbumin ,Nicardipine ,MESH: Mice, Inbred BALB C ,Inflammation ,Enzyme-Linked Immunosorbent Assay ,Mice, Transgenic ,MESH: Nicardipine ,03 medical and health sciences ,Th2 Cells ,MESH: Th2 Cells ,Intensive care ,MESH: Severity of Illness Index ,MESH: Cell Proliferation ,medicine ,Animals ,MESH: Lung ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,MESH: Lymphocyte Activation ,MESH: Mice ,B cell ,Interleukin 4 ,Administration, Intranasal ,030304 developmental biology ,Cell Proliferation ,MESH: Ovalbumin ,business.industry ,MESH: Bronchoalveolar Lavage Fluid ,T lymphocyte ,Dendritic Cells ,Asthma ,respiratory tract diseases ,Disease Models, Animal ,Immunology ,biology.protein ,Calcium ,MESH: Disease Models, Animal ,business ,MESH: Female ,030215 immunology - Abstract
RATIONALE: Ca(2+) signaling controls the production of T helper (Th) type 2 cytokines known to be deleterious in asthma. Recently, we showed that Ca(2+) signaling was dihydropyridine (DHP)-sensitive in Th2 lymphocytes and that the DHP derivate, nicardipine, used in the treatment of cardiovascular pathologies, prevents Th2-dependent B cell polyclonal activation. OBJECTIVES: We tested the effect of nicardipine in experimental allergic asthma. METHODS: BALB/c mice immunized with ovalbumin (OVA) in alum and challenged with intranasal OVA were treated with nicardipine once the Th2 response, or even airway inflammation, was induced. We also tested the effect of nicardipine in asthma induced by transferring OVA-specific Th2 cells in BALB/c mice exposed to intranasal OVA. We checked the impact of nicardipine on T-cell responses and airway inflammation. MEASUREMENTS AND MAIN RESULTS: Nicardipine inhibited in vitro Ca(2+) response in Th2 cells. In vivo, it impeded the development of Th2-mediated airway inflammation and reduced the capacity of lymphocytes from lung-draining lymph nodes to secrete Th2, but not Th1, cytokines. Nicardipine did not affect antigen presentation to CD4(+) T lymphocytes, nor the initial localization of Th2 cells into the lungs of mice exposed to intranasal OVA; however, it reduced the production of type 2 cytokines and the amplification of the Th2 response in mice with asthma. Conversely, nicardipine had no effect on Th1-mediated airway inflammation. CONCLUSIONS: Nicardipine improves experimental asthma by impairing Th2-dependent inflammation. This study could provide a rationale for developing drugs selectively targeting DHP receptors of Th2 lymphocytes, potentially beneficial in the treatment of asthma.
- Published
- 2007
- Full Text
- View/download PDF
50. Estrogen enhances susceptibility to experimental autoimmune myasthenia gravis by promoting type 1-polarized immune responses
- Author
-
Laurent Delpy, Corinne Bruand, Lucile Garidou, Jean-Charles Guéry, Abdelhadi Saoudi, Victorine Douin-Echinard, Physiologie Moléculaire de la Réponse Immune et des Lymphoproliférations (PMRIL), Université de Limoges (UNILIM)-Génomique, Environnement, Immunité, Santé, Thérapeutique (GEIST FR CNRS 3503)-Centre National de la Recherche Scientifique (CNRS), Institut de médecine moléculaire de Rangueil (I2MR), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-IFR150-Institut National de la Santé et de la Recherche Médicale (INSERM), Departement /u563 : Immunologie et Maladies infectieuses, Centre de Physiopathologie Toulouse Purpan (CPTP), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Toulouse III - Paul Sabatier (UT3), and Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
MESH: Interleukin-12 ,MESH: Spleen ,medicine.disease_cause ,Autoimmunity ,Pathogenesis ,Mice ,0302 clinical medicine ,Immunology and Allergy ,Receptors, Cholinergic ,MESH: Animals ,Cells, Cultured ,0303 health sciences ,Estradiol ,MESH: Antigen-Presenting Cells ,Interleukin-12 ,3. Good health ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Female ,Disease Susceptibility ,MESH: Estradiol ,MESH: Cells, Cultured ,medicine.medical_specialty ,medicine.drug_class ,Immunology ,MESH: Disease Susceptibility ,Antigen-Presenting Cells ,MESH: Myasthenia Gravis, Autoimmune, Experimental ,03 medical and health sciences ,Immune system ,MESH: Mice, Inbred C57BL ,Internal medicine ,MESH: Cell Proliferation ,medicine ,Animals ,MESH: Mice ,030304 developmental biology ,Cell Proliferation ,Autoimmune disease ,MESH: Receptors, Cholinergic ,business.industry ,Autoantibody ,Th1 Cells ,medicine.disease ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,Mice, Inbred C57BL ,Endocrinology ,MESH: Th1 Cells ,Estrogen ,business ,MESH: Female ,030217 neurology & neurosurgery ,Spleen ,Hormone - Abstract
Myasthenia gravis (MG) is an organ-specific autoimmune disease caused in most cases by autoantibodies against the nicotinic acetylcholine receptor (AChR). It is now well documented that many autoimmune diseases, including MG, are more prevalent in women than in men, and that fluctuations in disease severity occur during pregnancy. These observations raise the question of the potential role of sex hormones, such as estrogens, as mediators of sex differences in autoimmunity. In the present study, we have analyzed the effect of 17β-estradiol (E2) on the pathogenesis of experimental autoimmune myasthenia gravis (EAMG), an animal model of MG. We show that treatment with E2 before Ag priming is necessary and sufficient to promote AChR-specific Th1 cell expansion in vivo. This time-limited exposure to E2 enhances the production of anti-AChR IgG2ab (specific for b allotype; e.g., B6) and IgG2b, but not IgG1, and significantly increases the severity of EAMG in mice. Interestingly, the E2-mediated augmentation in AChR-specific Th1 response correlates with an enhanced production of IL-12 by splenic APCs through the recruitment of CD8α+ dendritic cells. These data provide the first evidence that estrogen enhances EAMG, and sheds some light on the role of sex hormones in immune responses and susceptibility to autoimmune disease in women.
- Published
- 2005
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.