15 results on '"Chloé Beuraud"'
Search Results
2. 749 A novel translational mouse model for assessment of human STING-targeting therapies
- Author
-
Gaëlle Martin, Fabiane Sônego, Audrey Beringer, Chloé Beuraud, Yacine Cherifi, Patricia Isnard-Petit, Kader Thiam, and Angela Pappalardo
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
3. 908 Humanized model for assessment of therapies targeting either lymphoid or myeloid compartment: enhanced evaluation of clinical relevancy & translatability
- Author
-
Charles Dumontet, Gaëlle Martin, Chloé Beuraud, Yacine Cherifi, Alexandre Fraichard, Patricia Isnard-Petit, Kader Thiam, Angus Sinclair, Poonam Yakkundi, Florence Renart-Depontieu, Morgane Denis, Léa Magadoux, Elsa Kress, Ludovic Bourre, Dean Campbell, and Astrid Doerner
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
4. 14 Novel CD3 epsilon humanized N-terminal epitope model for assessment of efficacy of T-cell engagers
- Author
-
Gaëlle Martin, Fabiane Sônego, Audrey Beringer, Chloé Beuraud, Yacine Cherifi, Alexandre Fraichard, Patricia Isnard-Petit, and Kader Thiam
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
5. Abstract 3203: EVOcells Oncology: Tailored genetic engineering of iPSC-derived immune effector cells and combination with the right biologic therapeutics result in optimal killing of primary tumor cells from patients
- Author
-
Michael Esquerré, Audrey Holtzinger, Nadja Wagner, Monika Braun, Mélanie Pichery, Stefanie Pfaender, Stephanie Sontag, Kathrin Haake, Michela Mirenda, Michael Paillasse, Davide Grandolfo, Chloé Beuraud, Mandy Richter, Philip Hublitz, Julien Bousquet, Marion Fabre, Mylène Gador, Daniel Sommermeyer, Tanja Schneider, Oriane Bombarde, Camille Esquerré, Loic Ysebaert, Fabien Despas, Matthias Austen, Andreas Scheel, and Markus Dangl
- Subjects
Cancer Research ,Oncology - Abstract
Current autologous cell therapies, with blockbuster products on the market, have been leading for a decade to unprecedented clinical successes in patients with hematological malignancies. However, these patient-derived T-cell therapies are facing many challenges. The use of GMP iPSC lines to produce immune effector cells will reduce the complexity of the manufacturing process and will provide an unlimited source of starting material. The goal of the EVOcells Oncology platform is to offer a truly allogeneic cell therapy platform to treat a broad number of cancer patients with consistent quality and scalability of the final product. Besides, the versatility of our platform to produce different immune cell types combined to customized genetic engineering strategies will bring cell therapy to the level of personalized medicine. Our “off-the-shelf” cell therapy platform has already validated two pillars: iPSC-derived NK cells (iNK) and iPSC-derived Macrophages (iMACs). Through multiple genetic engineering strategies specific to each immune cell type, we are developing a comprehensive portfolio of cell therapy products to address specific tumor escape mechanism in liquid and solid tumors. Our initial effort aimed to develop these two innate immune cell types to propose efficacious cell therapies with an increased safety profile as they have low risk of graft-versus-host disease (GvHD) or CRS (Cytokine Release Syndrome). Thanks to the expression of a broad pattern of activatory receptors, iNK cells form Immunological Synapses with tumor cells leading in turn to efficient killing with and without addition of a CAR construct. Besides, we demonstrated the possibility to combine “naked” iNK cells with marketed therapeutic monoclonal antibodies (mAb) to further improve their efficacy. At the end of the differentiation process, iMACs are showing a M0 like phenotype with high plasticity allowing the in vitro differentiation of the cells towards either a M1 or a M2 polarization in response to the appropriate stimulations. iMACs produce key macrophages cytokines and are able to kill tumor cells via ADCP (Antibody-Dependent-Cell-Phagocytosis) mechanism when combined to a therapeutic mAb. Thanks to our collaboration with clinicians at the IUCT-Oncopole (Toulouse Cancer Hospital), we were able to identify appropriate cancer indications and further demonstrate in a translational fashion that both iNK and iMACs are able to kill primary resistant tumor cells which were isolated from patient’ samples. Taken together, these results are showing the versatility and the breadth of our EVOcells Oncology platform to produce a true arsenal of cell therapies and its potential for future clinical development. Citation Format: Michael Esquerré, Audrey Holtzinger, Nadja Wagner, Monika Braun, Mélanie Pichery, Stefanie Pfaender, Stephanie Sontag, Kathrin Haake, Michela Mirenda, Michael Paillasse, Davide Grandolfo, Chloé Beuraud, Mandy Richter, Philip Hublitz, Julien Bousquet, Marion Fabre, Mylène Gador, Daniel Sommermeyer, Tanja Schneider, Oriane Bombarde, Camille Esquerré, Loic Ysebaert, Fabien Despas, Matthias Austen, Andreas Scheel, Markus Dangl. EVOcells Oncology: Tailored genetic engineering of iPSC-derived immune effector cells and combination with the right biologic therapeutics result in optimal killing of primary tumor cells from patients [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3203.
- Published
- 2023
- Full Text
- View/download PDF
6. 749 A novel translational mouse model for assessment of human STING-targeting therapies
- Author
-
Chloé Beuraud, Patricia Isnard-Petit, Yacine Cherifi, Fabiane Sônego, Kader Thiam, Audrey Beringer, Angela Pappalardo, and Gaëlle Martin
- Subjects
Pharmacology ,Cancer Research ,business.industry ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Bioinformatics ,eye diseases ,Sting ,Oncology ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business ,RC254-282 - Abstract
BackgroundAlthough Immune checkpoint inhibitors (ICI)-targeting therapies have revolutionized the treatment of cancer, several tumors do not respond to those therapies. Preclinical and clinical evidences suggest that STING is a promising target to improve the immunogenicity of tumors, turning them responsive to ICI, and enhancing anti-tumor response. DMXAA failed to show efficacy in clinical trials, despite its encouraging anti-tumor response in preclinical phase, highlighting the need of accurate translational preclinical models. On top of the specificity barrier reported for STING-targeting agents, the heterogeneity of STING variants and their variability in the response to STING-targeting therapies, brings another level of complexity in preclinical evaluation of anti-STING therapies. Here, we report the generation of STING humanized (hSTING) mouse models enabling the in vivo assessment of STING-targeting agents.MethodsHuman STING variants show a high heterogenicity and population stratification. Different variants, and isoforms, respond differently to STING agonists. We developed mouse models expressing the main human STING variants and isoforms found in the population to recapitulate this complexity. Human STING was inserted by knock-in at the endogenous locus to enable a physiological expression pattern of STING while invalidating the mouse gene. Herein, we will focus on the human STING full length H323 model.ResultsT and B lymphocytes, NK, DCs and monocytes frequence in the spleen, bone marrow and blood were found to be similar in hSTING and WT mice, suggesting that the humanization of STING did not alter the immune cell distribution in these compartments. These cells express human STING, while no expression of mouse STING was observed. Splenocytes isolated from hSTING and WT mice produced IL-6 and IFN-γ upon activation with 2'3'-cGAMP, a cyclic dinucleotide with activity towards both mouse and human STING. Similarly, a mouse and human STING agonist induced the activation of DCs in both hSTING and WT mice, as observed by the increased expression of CD80/CD86 on DCs ex vivo and in vivo. Moreover, systemic production of IL-6, IFN-γ and TNF-α in response to this STING agonist was observed and suggest that human STING is functional in hSTING mice. As expected, hSTING mice did not respond to activation with DMXAA in vivo, whereas this agonist induced the systemic production of cytokines and activation of DCs in WT mice.ConclusionsThe novel hSTING model described here supports the assessment of human STING-targeting agents in immuno-oncology and inflammation. Intercrosses of this model with ICI humanized models could support the assessment of combination therapies
- Published
- 2021
7. 908 Humanized model for assessment of therapies targeting either lymphoid or myeloid compartment: enhanced evaluation of clinical relevancy & translatability
- Author
-
Gaëlle Martin, Patricia Isnard-Petit, Astrid Doerner, Poonam Yakkundi, Alexandre Fraichard, Morgane Denis, Elsa Kress, Yacine Cherifi, Charles Dumontet, Florence Renart-Depontieu, Dean Campbell, Chloé Beuraud, Ludovic Bourre, Angus M. Sinclair, Kader Thiam, and Léa Magadoux
- Subjects
Pharmacology ,Cancer Research ,Myeloid ,business.industry ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.anatomical_structure ,Oncology ,medicine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Compartment (pharmacokinetics) ,business ,RC254-282 - Abstract
BackgroundThe breakthrough of immunotherapies has unleashed new hope and new success for cancer therapy. However, the choice of a preclinical model is one of the main challenges as they are important for evaluation of translatability to help support testing in clinical studies, including potential efficacy and tolerability of immunotherapies during preclinical development.The development of mouse models featuring a human immune system (HIS) provides new paths for the investigation of the efficacy of immunotherapies in preclinical models engrafted with human tumors. Although these models provided a breakthrough in the assessment of immune targeting agents, they also come with a few significant caveats. These include: a lack of a mature human myeloid compartment in the mouse, and a short life span of the model when this compartment is promoted at non-physiological levels via the over-expression of human cytokines. Here, we report a novel mouse model (BRGSF-HIS), featuring functional human lymphoid and myeloid compartments. The human immune response of this model was assessed through examination of the immune cells composition in the tumor microenvironment (TME), and its ability to respond to biologics known to trigger cytokine release syndrome (CRS).MethodsBRGSF (balb/C Rag2-/-, IL2Rg-/-, SIRPaNOD and Flt3-/-) is a highly immunodeficient mouse, with reduced murine myeloid cells, which allows long term CD34+ HSC-engraftment and development of human lymphoid and myeloid compartments (human CD141+ and CD1c+ DC subsets, CD123+ pDC, CD14+ monocytes), in blood, spleen and bone marrow. The engraftment is stable for over twelve months with no side effects. The effect of exogenous human Flt3 ligand (Flt3L) on the composition of TME in A549 model, and an anti-CD3 antibody (OKT3)-induced CRS, were assessed.ResultsExogenous human Flt3L significantly and transiently increased the proportion of human myeloid cells. This can be recalled by continuous dosing of Flt3L. Assessment of tumor immunobiology in A549 model, showed increased tumor-infiltrating T-cells (mainly CD8+ T-cells) and myeloid cells, while tumor-infiltrating NK cells were decreased. The presence of myeloid cells provides a new opportunity for assessment of myeloid targeted therapies, as proven using pDC-depleting antibodies. OKT3 administration resulted in CRS symptoms including a temperature drop, body weight loss and a change in serum cytokine levels. Symptoms were mitigated upon administration of tocilizumab, suggesting the contribution of the myeloid compartment in the response observed.ConclusionsThese data demonstrate that BRGSF-HIS mice support development of functional human myeloid cells and that this mouse model enables preclinical evaluation of cancer immunotherapy in vivo.
- Published
- 2021
8. 15 A novel CD28 humanized mouse model for efficacy assessment of CD28-targeting therapies
- Author
-
Fabiane Sônego, Alexandre Fraichard, Kader Thiam, Chloé Beuraud, Patricia Isnard-Petit, Audrey Beringer, Yacine Cherifi, and Gaëlle Martin
- Subjects
medicine.medical_treatment ,T cell ,CD3 ,CD28 ,Biology ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,In vitro ,Immune system ,Cytokine ,medicine.anatomical_structure ,Humanized mouse ,medicine ,Cancer research ,biology.protein ,CD8 - Abstract
Background Immuno-intervention through targeting of activating and inhibitory immune checkpoints (ICP), has shown promising results in the clinic over the last years. To facilitate these researches, mouse models expressing humanized ICP instead of their mouse counterparts were developed. Herein, we describe a novel CD28 humanized mouse model (hCD28 model), designed to test compounds targeting human CD28 (hCD28). Methods Human and mouse CD28 (mCD28) have different signalling responses, with hCD28 being known for inducing higher levels of pro-inflammatory cytokines upon stimulation with ligands/superagonists. This can be explained by the expression of CD28i, a hCD28 amplifier isoform which is not found in mouse. Additionally, evidences suggested that the different signalling between human and mCD28 relies on one amino acid change in the intracellular domain (ICD).1 Because the hCD28 model was developed to assess hCD28-targeting therapeutics, we decided to keep the expression of both canonical and CD28i isoforms to avoid undermining the biological effects of the testing antibodies. Although keeping the human ICD could favour the evaluation of cytokine production and therefore the safety of the test therapeutics, we decided to keep the mouse ICD to enable a proper interaction of CD28 with its signalling partners, allowing a physiological stimulation of CD28 in efficacy studies. Results hCD28 mice express hCD28 on T cells and the frequency of CD3 T cells is comparable in both WT and hCD28 mice. Stimulation of hCD28 mice-isolated T cells with hCD28 ligands and agonist antibodies resulted in T cell proliferation and cytokine production, suggesting that hCD28 is functional in mouse cells. MC38 uptake rate and kinetic of growth were comparable in WT and hCD28 mice, suggesting no major defect in the immune response in the hCD28 mice. Importantly, splenocytes and tumor draining lymph nodes cells isolated from tumor-bearing hCD28 mice showed higher production of IL-2 and IFN-gamma upon in vitro re-challenged with MC38 when compared to WT cells. Since the frequency of CD3 cells (Treg, CD4+ and CD8+) is comparable to WT mice, this could be explained by the expression of the amplifier CD28i isoform, which is absent in WT mice. Conclusions The hCD28 model described here supports the efficacy assessment of hCD28-targeting biologics, enabling PK/PD studies as hCD28 expression levels and pattern are physiological. However, after careful consideration of the CD28 biology, we decided to keep the mouse ICD, although it triggers lower pro-inflammatory cytokine production than CD28 human ICD. As such, this model is not suitable for toxicology/safety studies. Reference Porciello N, Grazioli P, Campese AF, et al. A non-conserved amino acid variant regulates differential signalling between human and mouse CD28. Nat Commun 2018; 9:1–16.
- Published
- 2020
9. Abstract 1811: A novel OX40 humanized mouse model for efficacy assessment of OX40-targeting therapies
- Author
-
Gaëlle Martin, Kader Thiam, Yacine Cherifi, Audrey Beringer, Chloé Beuraud, Patricia Isnard Petit, Fabiane Sônego, and Alexandre Fraichard
- Subjects
Cancer Research ,education.field_of_study ,medicine.medical_treatment ,Biology ,Immune checkpoint ,OX40 ligand ,medicine.anatomical_structure ,Cytokine ,Immune system ,Oncology ,Mechanism of action ,Humanized mouse ,Cancer research ,medicine ,Cytotoxic T cell ,Bone marrow ,medicine.symptom ,education - Abstract
Immunotherapies through targeting of activating and inhibitory immune checkpoints (ICP) has revolutionized the treatment of cancers. To facilitate the translation of basic research into clinical application of these immunotherapies, we developed a pipeline of immunocompetent mouse models expressing humanized ICP instead of their mouse counterparts, thus compounds can be tested in the absence of endogenous cross-reacting mouse targets. The humanization strategy is target-dependent but ensures that the biology of the target, its physiological regulation and interacting partners are preserved. Herein, we describe a novel OX40 humanized mouse model (hOX40 model), designed to assess the efficacy and mechanism of action of compounds targeting human OX40 (hOX40). The expression of the fully human OX40 protein (human extracellular, transmembrane and intracellular domains) did not alter the distribution of immune cells in spleen, blood and bone marrow. hOX40 was expressed on CD4 and CD8 T cells upon activation and the kinetic of expression mirrored the expression of mouse OX40 in WT cells activated under the same conditions. Treg cells isolated from hOX40 mice expressed hOX40 and this expression was upregulated upon activation, also recapitulating the expression of mouse OX40 in WT cells, and suggesting that the regulation of OX40 expression was kept in the hOX40 mice. hOX40 was also found to be functional on CD4 T cells, as evidenced by the increased proliferation and cytokine production upon stimulation with human OX40 ligand. Importantly, hOX40 mice bearing MC38 tumors showed a tumor growth inhibition upon treatment with an anti-human OX40 therapy with agonist activity. This model should also enable the investigation of mechanism of action as well as tumor immunological memory, as a similar design of the hOX40 model was used for the assessment of a bispecific therapy targeting OX40 and CTLA-4 (Kvarnhammar et.al, 2019). The hOX40 mouse model is currently being crossed with other models expressing humanized immune checkpoint to enable the assessment of bispecifics and combo therapies. Citation Format: Fabiane Sônego, Gaëlle Martin, Audrey Beringer, Chloé Beuraud, Yacine Cherifi, Alexandre Fraichard, Patricia Isnard Petit, Kader Thiam. A novel OX40 humanized mouse model for efficacy assessment of OX40-targeting therapies [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1811.
- Published
- 2021
- Full Text
- View/download PDF
10. CCR10 + ILC2s with ILC1-like properties exhibit a protective function in severe allergic asthma
- Author
-
Vincent Lombardi, Rachel Golub, Sylvie Chollet-Martin, C. Neukirch, Chloé Beuraud, Emmanuel Nony, Sonia Luce, Sabi Airouche, Thibaut Perchet, Véronique Baron-Bodo, Philippe Moingeon, Michel Aubier, Laurent Mascarell, Emmanuel Naline, S. Horiot, Philippe Devillier, Stallergenes SA (Stallergenes), Stallergenes, Hôpital Foch [Suresnes], Université Paris-Saclay, AP-HP - Hôpital Bichat - Claude Bernard [Paris], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Université Paris Diderot - Paris 7 (UPD7), Institut National de la Santé et de la Recherche Médicale (INSERM), Lymphopoïèse (Lymphopoïèse (UMR_1223 / U1223 / U-Pasteur_4)), Institut Pasteur [Paris]-Université Paris Diderot - Paris 7 (UPD7)-Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris-Sud - Paris 11 (UP11), and Institut Pasteur [Paris] (IP)-Université Paris Diderot - Paris 7 (UPD7)-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
0301 basic medicine ,medicine.diagnostic_test ,business.industry ,Immunology ,medicine.disease ,Pathophysiology ,3. Good health ,Flow cytometry ,Allergic inflammation ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030228 respiratory system ,Amphiregulin ,Downregulation and upregulation ,Immunology and Allergy ,Medicine ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,CCL27 ,CCR10 ,business ,Asthma - Abstract
International audience; We previously showed that patients with severe allergic asthma have high numbers of circulating ILC2s expressing CCR10.METHOD:Herein, CCR10+ ILC2s were further analyzed in the blood of healthy individuals or patients with allergic and non-allergic asthma. Characteristics of human CCR10+ and CCR10- ILC2s were assessed by flow cytometry as well as single-cell multiplex RT-qPCR. The role of CCR10+ ILC2s in asthma pathophysiology was studied in allergen-treated mice.RESULTS:When compared to healthy controls, CCR10+ ILC2s are enriched in the blood of both allergic and non-allergic severe asthmatic patients, and these cells are recruited to the lungs. Plasma concentrations of the CCR10 ligand CCL27 are significantly increased in severe asthmatics when compared to non-asthmatic patients. CCR10+ ILC2s secrete little TH 2 cytokines, but exhibit ILC1-like properties, including a capacity to produce IFN-γ. Also, single-cell analysis reveals that the CCR10+ ILC2 subset is enriched in cells expressing amphiregulin. CCR10+ ILC2 depletion, as well as blocking of IFN-γ activity, exacerbates airway hyperreactivity in allergen-challenged mice, providing evidence for a protective role of these cells in allergic inflammation.CONCLUSIONS:Frequencies of circulating CCR10+ ILC2s and CCL27 plasma concentrations represent candidate markers of asthma severity. The characterization of CCR10+ ILC2s in human samples and in mouse asthma models suggests that these cells downregulate allergic inflammation through IFN-γ production.
- Published
- 2019
- Full Text
- View/download PDF
11. Circulating innate lymphoid cells are differentially regulated in allergic and nonallergic subjects
- Author
-
Sonia Luce, Catherine Neukirch, Chloé Beuraud, Vincent Lombardi, S. Horiot, Véronique Baron-Bodo, Peter S. Linsley, Hélène Moussu, Philippe Moingeon, Sylvie Chollet-Martin, Lise Morizur, Michel Aubier, and Erik Wambre
- Subjects
0301 basic medicine ,Immunology ,Gene Expression ,Bioinformatics ,Immunophenotyping ,03 medical and health sciences ,0302 clinical medicine ,Antigens, CD ,Humans ,Immunology and Allergy ,Medicine ,Cell Lineage ,Lymphocytes ,Conjunctivitis, Allergic ,business.industry ,Innate lymphoid cell ,Flow Cytometry ,Rhinitis, Allergic ,Asthma ,Immunity, Innate ,030104 developmental biology ,030228 respiratory system ,Desensitization, Immunologic ,Case-Control Studies ,Leukocytes, Mononuclear ,Cytokines ,business - Published
- 2016
- Full Text
- View/download PDF
12. CCR10
- Author
-
Chloé, Beuraud, Vincent, Lombardi, Sonia, Luce, Stéphane, Horiot, Emmanuel, Naline, Catherine, Neukirch, Sabi, Airouche, Thibaut, Perchet, Rachel, Golub, Philippe, Devillier, Sylvie, Chollet-Martin, Véronique, Baron-Bodo, Emmanuel, Nony, Michel, Aubier, Laurent, Mascarell, and Philippe, Moingeon
- Subjects
Receptors, CCR10 ,Allergens ,Severity of Illness Index ,Asthma ,Immunity, Innate ,Lymphocyte Subsets ,Disease Models, Animal ,Interferon-gamma ,Mice ,Animals ,Cytokines ,Humans ,Disease Susceptibility ,Lymphocyte Count ,Biomarkers - Abstract
We previously showed that patients with severe allergic asthma have high numbers of circulating ILC2s expressing CCR10.Herein, CCR10When compared to healthy controls, CCR10Frequencies of circulating CCR10
- Published
- 2018
13. Protein kinase C θ controls type 2 innate lymphoid cell and TH2 responses to house dust mite allergen
- Author
-
Marc Le Bert, Nathalie Rouxel, Quentin Marquant, Pauline Chenuet, Fabrice Trovero, Chloé Beuraud, Dieudonnée Togbe, Bernhard Ryffel, Louis Fauconnier, Tiffany Marchiol, Laurent Mascarell, Fahima Madouri, Margaux Gallerand, Vincent Lombardi, Valérie F. J. Quesniaux, François Erard, Aurélie Ledru, Lionel Apetoh, Immunologie et Neurogénétique Expérimentales et Moléculaires ( INEM ), Université d'Orléans ( UO ) -Centre National de la Recherche Scientifique ( CNRS ), Stallergenes Greer (France, Antony), Artimmune, privé, Eco-Anthropologie et Ethnobiologie ( EAE ), Centre National de la Recherche Scientifique ( CNRS ) -Université Paris Diderot - Paris 7 ( UPD7 ) -Muséum National d'Histoire Naturelle ( MNHN ), Equipe CD4DNA (LNC - U1231), Lipides - Nutrition - Cancer [Dijon - U1231] ( LNC ), Université de Bourgogne ( UB ) -AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Université de Bourgogne ( UB ) -AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Institut National de la Santé et de la Recherche Médicale ( INSERM ), Institute of Immunology and Infectious Disease and Molecular Medicine, Immunologie et Neurogénétique Expérimentales et Moléculaires (INEM), Université d'Orléans (UO)-Centre National de la Recherche Scientifique (CNRS), Eco-Anthropologie et Ethnobiologie (EAE), Muséum national d'Histoire naturelle (MNHN)-Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS), Lipides - Nutrition - Cancer [Dijon - U1231] (LNC), and Université de Bourgogne (UB)-Institut National de la Santé et de la Recherche Médicale (INSERM)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Université de Bourgogne (UB)-Institut National de la Santé et de la Recherche Médicale (INSERM)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement
- Subjects
0301 basic medicine ,Adoptive cell transfer ,medicine.medical_treatment ,Immunology ,interferon regulatory factor 4 ,innate lymphoid cells ,Biology ,House dust mite ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Immunology and Allergy ,Protein kinase C ,Innate lymphoid cell ,FOXP3 ,protein kinase Cθ ,Dendritic cell ,[SDV.SP]Life Sciences [q-bio]/Pharmaceutical sciences ,Eosinophil ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,Cytokine ,[ SDV.SP ] Life Sciences [q-bio]/Pharmaceutical sciences ,nuclear factor of activated T cells ,eosinophils ,Cell activation ,allergic asthma ,030215 immunology - Abstract
Background Protein kinase C (PKC) θ, a serine/threonine kinase, is involved in T H 2 cell activation and proliferation. Type 2 innate lymphoid cells (ILC2s) resemble T H 2 cells and produce the T H 2 cytokines IL-5 and IL-13 but lack antigen-specific receptors. The mechanism by which PKC-θ drives innate immune cells to instruct T H 2 responses in patients with allergic lung inflammation remains unknown. Objectives We hypothesized that PKC-θ contributes to ILC2 activation and might be necessary for ILC2s to instruct the T H 2 response. Methods PRKCQ gene expression was assessed in innate lymphoid cell subsets purified from human PBMCs and mouse lung ILC2s. ILC2 activation and eosinophil recruitment, T H 2-related cytokine and chemokine production, lung histopathology, interferon regulatory factor 4 (IRF4) mRNA expression, and nuclear factor of activated T cells (NFAT1) protein expression were determined. Adoptive transfer of ILC2s from wild-type mice was performed in wild-type and PKC-θ–deficient (PKC-θ −/− ) mice. Results Here we report that PKC-θ is expressed in both human and mouse ILC2s. Mice lacking PKC-θ had reduced ILC2 numbers, T H 2 cell numbers and activation, airway hyperresponsiveness, and expression of the transcription factors IRF4 and NFAT1. Importantly, adoptive transfer of ILC2s restored eosinophil influx and IL-4, IL-5 and IL-13 production in lung tissue, as well as T H 2 cell activation. The pharmacologic PKC-θ inhibitor (Compound 20) administered during allergen challenge reduced ILC2 numbers and activation, as well as airway inflammation and IRF4 and NFAT1 expression. Conclusions Therefore our findings identify PKC-θ as a critical factor for ILC2 activation that contributes to T H 2 cell differentiation, which is associated with IRF4 and NFAT1 expression in allergic lung inflammation.
- Published
- 2017
- Full Text
- View/download PDF
14. Sialylated Fetuin-A as a candidate predictive biomarker for successful grass pollen allergen immunotherapy
- Author
-
Vincent Lombardi, Thierry Batard, Laurent Mascarell, Emmanuel Nony, Chloé Beuraud, K. Jain, Philippe Moingeon, Sylvie Chollet-Martin, Christine Kellenberger, Christian Beauvallet, Philippe Devillier, Noémie Caillot, Sabi Airouche, Henri Chabre, Sonia Luce, S. Horiot, Sandrine Mariano, Véronique Baron-Bodo, Julien Bouley, Stallergenes Greer, Génétique Animale et Biologie Intégrative (GABI), Institut National de la Recherche Agronomique (INRA)-AgroParisTech, Hôpital Foch [Suresnes], UPRES EA220, Inflammation, Chimiokines et Immunopathologie [Châtenay-Malabry], Université Paris-Sud - Paris 11 (UP11)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Saclay, Architecture et fonction des macromolécules biologiques (AFMB), Centre National de la Recherche Scientifique (CNRS)-Aix Marseille Université (AMU)-Institut National de la Recherche Agronomique (INRA), CIFRE fellowship from ANRT (Association Nationale de la Recherche et de la Technologie), and Institut National de la Recherche Agronomique (INRA)-Aix Marseille Université (AMU)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Lipopolysaccharides ,0301 basic medicine ,Allergy ,Ovalbumin ,alpha-2-HS-Glycoprotein ,medicine.medical_treatment ,[SDV]Life Sciences [q-bio] ,Immunology ,Biology ,Poaceae ,medicine.disease_cause ,Allergic sensitization ,03 medical and health sciences ,Allergen ,Double-Blind Method ,medicine ,otorhinolaryngologic diseases ,Animals ,Humans ,Immunology and Allergy ,Gene Silencing ,Receptor ,Allergen immunotherapy ,Mice, Inbred BALB C ,Sublingual Immunotherapy ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,Rhinitis, Allergic, Seasonal ,Dendritic Cells ,Immunotherapy ,Dendritic cell ,Allergens ,medicine.disease ,Fetuin ,3. Good health ,Fetuin-A ,[SDV.GEN.GA]Life Sciences [q-bio]/Genetics/Animal genetics ,030104 developmental biology ,biology.protein ,Pollen ,biomarker ,serum ,Biomarkers - Abstract
International audience; Background: Eligibility to immunotherapy is based on the determination of IgE reactivity to a specific allergen by means of skin prick or in vitro testing. Biomarkers predicting the likelihood of clinical improvement during immunotherapy would significantly improve patient selection. Methods: Proteins were differentially assessed by using 2-dimensional differential gel electrophoresis and label-free mass spectrometry in pretreatment sera obtained from clinical responders and nonresponders within a cohort of 82 patients with grass pollen allergy receiving sublingual immunotherapy or placebo. Functional studies of Fetuin-A (FetA) were conducted by using gene silencing in a mouse asthma model, human dendritic cell in vitro stimulation assays, and surface plasmon resonance. Results: Analysis by using quantitative proteomics of pretreatment sera from patients with grass pollen allergy reveals that high levels of O-glycosylated sialylated FetA isoforms are found in patients exhibiting a strong decrease in rhinoconjunctivitis symptoms after sublingual immunotherapy. Although FetA is involved in numerous inflammatory conditions, its potential role in allergy is unknown. In vivo silencing of the FETUA gene in BALB/c mice results in a dramatic upregulation of airway hyperresponsiveness, lung resistance, and T(H)2 responses after allergic sensitization to ovalbumin. Both sialylated and nonsialytated FetA bind to LPS, but only the former synergizes with LPS and grass pollen or mite allergens to enhance the Toll-like receptor 4-mediated proallergic properties of human dendritic cells. Conclusions: As a reflection of the patient's inflammatory status, pretreatment levels of sialylated FetA in the blood are indicative of the likelihood of clinical responses during grass pollen immunotherapy.
- Published
- 2017
- Full Text
- View/download PDF
15. Protein kinase Cθ controls type 2 innate lymphoid cell and T
- Author
-
Fahima, Madouri, Pauline, Chenuet, Chloé, Beuraud, Louis, Fauconnier, Tiffany, Marchiol, Nathalie, Rouxel, Aurélie, Ledru, Margaux, Gallerand, Vincent, Lombardi, Laurent, Mascarell, Quentin, Marquant, Lionel, Apetoh, François, Erard, Marc, Le Bert, Fabrice, Trovero, Valérie F J, Quesniaux, Bernhard, Ryffel, and Dieudonnée, Togbe
- Subjects
Male ,Mice, Knockout ,NFATC Transcription Factors ,Cell Differentiation ,Dipeptides ,Allergens ,Asthma ,Immunity, Innate ,Isoenzymes ,Mice, Inbred C57BL ,Leukocyte Count ,Protein Kinase C-theta ,Interferon Regulatory Factors ,Animals ,Cytokines ,Humans ,Female ,Antigens, Dermatophagoides ,Lymphocytes ,Bronchoalveolar Lavage Fluid ,Lung ,Protein Kinase Inhibitors ,Protein Kinase C - Abstract
Protein kinase C (PKC) θ, a serine/threonine kinase, is involved in TWe hypothesized that PKC-θ contributes to ILC2 activation and might be necessary for ILC2s to instruct the TPRKCQ gene expression was assessed in innate lymphoid cell subsets purified from human PBMCs and mouse lung ILC2s. ILC2 activation and eosinophil recruitment, THere we report that PKC-θ is expressed in both human and mouse ILC2s. Mice lacking PKC-θ had reduced ILC2 numbers, TTherefore our findings identify PKC-θ as a critical factor for ILC2 activation that contributes to T
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.