355 results on '"Dutertre, Charles-Antoine"'
Search Results
102. Monocyte-derived dendritic cells from HLA-B27+ axial spondyloarthritis (SpA) patients display altered functional capacity and deregulated gene expression
- Author
-
Talpin, Alice, primary, Costantino, Félicie, additional, Bonilla, Nelly, additional, Leboime, Ariane, additional, Letourneur, Franck, additional, Jacques, Sébastien, additional, Dumont, Florent, additional, Amraoui, Sonia, additional, Dutertre, Charles-Antoine, additional, Garchon, Henri-Jean, additional, Breban, Maxime, additional, and Chiocchia, Gilles, additional
- Published
- 2014
- Full Text
- View/download PDF
103. Apoptotic cell capture by DCs induces unexpectedly robust autologous CD4+T-cell responses
- Author
-
Valente, Michael, primary, Baey, Camille, additional, Louche, Pauline, additional, Dutertre, Charles-Antoine, additional, Vimeux, Lene, additional, Marañón, Concepción, additional, Hosmalin, Anne, additional, and Feuillet, Vincent, additional
- Published
- 2014
- Full Text
- View/download PDF
104. TLR3–Responsive, XCR1+, CD141(BDCA-3)+/CD8α+-Equivalent Dendritic Cells Uncovered in Healthy and Simian Immunodeficiency Virus–Infected Rhesus Macaques
- Author
-
Dutertre, Charles-Antoine, primary, Jourdain, Jean-Pierre, additional, Rancez, Magali, additional, Amraoui, Sonia, additional, Fossum, Even, additional, Bogen, Bjarne, additional, Sanchez, Cindy, additional, Couëdel-Courteille, Anne, additional, Richard, Yolande, additional, Dalod, Marc, additional, Feuillet, Vincent, additional, Cheynier, Rémi, additional, and Hosmalin, Anne, additional
- Published
- 2014
- Full Text
- View/download PDF
105. Deciphering the Stromal and Hematopoietic Cell Network of the Adventitia from Non-Aneurysmal and Aneurysmal Human Aorta
- Author
-
Dutertre, Charles-Antoine, primary, Clement, Marc, additional, Morvan, Marion, additional, Schäkel, Knut, additional, Castier, Yves, additional, Alsac, Jean-Marc, additional, Michel, Jean-Baptiste, additional, and Nicoletti, Antonino, additional
- Published
- 2014
- Full Text
- View/download PDF
106. Activation of the Receptor NKG2D Leads to Production of Th17 Cytokines in CD4+ T Cells of Patients With Crohn's Disease
- Author
-
Pariente, Benjamin, primary, Mocan, Iulia, additional, Camus, Matthieu, additional, Dutertre, Charles–Antoine, additional, Ettersperger, Julien, additional, Cattan, Pierre, additional, Gornet, Jean–Marc, additional, Dulphy, Nicolas, additional, Charron, Dominique, additional, Lémann, Marc, additional, Toubert, Antoine, additional, and Allez, Matthieu, additional
- Published
- 2011
- Full Text
- View/download PDF
107. ID: 127: Inhibition of inflammasome signaling in the bat immune system
- Author
-
Irving, Aaron Trent, Ahn, Matae, Dutertre, Charles-Antoine, and Wang, Linfa
- Published
- 2015
- Full Text
- View/download PDF
108. The XC chemokine receptor 1 is a conserved selective marker of mammalian cells homologous to mouse CD8α+ dendritic cells
- Author
-
Crozat, Karine, primary, Guiton, Rachel, additional, Contreras, Vanessa, additional, Feuillet, Vincent, additional, Dutertre, Charles-Antoine, additional, Ventre, Erwan, additional, Vu Manh, Thien-Phong, additional, Baranek, Thomas, additional, Storset, Anne K., additional, Marvel, Jacqueline, additional, Boudinot, Pierre, additional, Hosmalin, Anne, additional, Schwartz-Cornil, Isabelle, additional, and Dalod, Marc, additional
- Published
- 2010
- Full Text
- View/download PDF
109. M1793 TH17 Cells in Crohn's Disease Express an Innate Immune Receptor: the Natural Killer Activating Receptor 2d
- Author
-
Pariente, Benjamin, primary, Mocan, Iulia, additional, Camus, Matthieu, additional, Dutertre, Charles-Antoine, additional, Cattan, Pierre, additional, Dulphy, Nicolas, additional, Gornet, Jean-Marc, additional, Lemann, Marc, additional, Toubert, Antoine, additional, and Allez, Matthieu, additional
- Published
- 2010
- Full Text
- View/download PDF
110. IL-23 and IL-12p70 production by monocytes and dendritic cells in primary HIV-1 infection
- Author
-
Louis, Stéphanie, primary, Dutertre, Charles-Antoine, additional, Vimeux, Lene, additional, Fery, Ludovic, additional, Henno, Lucy, additional, Diocou, Seckou, additional, Kahi, Sandrine, additional, Deveau, Christiane, additional, Meyer, Laurence, additional, Goujard, Cécile, additional, and Hosmalin, Anne, additional
- Published
- 2010
- Full Text
- View/download PDF
111. Les anticorps : mieux les connaître pour mieux s’en servir
- Author
-
Abès, Riad, primary, Dutertre, Charles-Antoine, additional, and Teillaud, Jean-Luc, additional
- Published
- 2009
- Full Text
- View/download PDF
112. R603: A New Low Dose Efficient Anti-CD20 Immunotherapy for CLL Patients ?
- Author
-
Le Garff-Tavernier, Magali, primary, Decocq, Julie, primary, de Romeuf, Christophe, primary, Teillaud, Jean-Luc, primary, Dutertre, Charles-Antoine, primary, Debre, Patrice, primary, Merle-Beral, Helene, primary, and Vieillard, Vincent, primary
- Published
- 2008
- Full Text
- View/download PDF
113. Immune control of tumors: host immune response and antibody-based immunotherapy
- Author
-
Abès, Riad, primary, Héritier, Sébastien, additional, Dutertre, Charles-Antoine, additional, and Teillaud, Jean-Luc, additional
- Published
- 2008
- Full Text
- View/download PDF
114. A novel subset of NK cells expressing high levels of inhibitory FcγRIIB modulating antibody-dependent function
- Author
-
Dutertre, Charles-Antoine, primary, Bonnin-Gélizé, Emmanuelle, additional, Pulford, Karen, additional, Bourel, Dominique, additional, Fridman, Wolf-Herman, additional, and Teillaud, Jean-Luc, additional
- Published
- 2008
- Full Text
- View/download PDF
115. Can NK Cells Play a Role in Anti-CD20 Immunotherapy for CLL Patients?.
- Author
-
Le Garff-Tavernier, Magali, primary, De Romeuf, Christophe, additional, Teillaud, Jean-Luc, additional, Decocq, Julie, additional, Dutertre, Charles-Antoine, additional, Debre, Patrice, additional, Merle-Beral, Helene, additional, and Vieillard, Vincent, additional
- Published
- 2007
- Full Text
- View/download PDF
116. Le deuxième acte des anticorps monoclonaux : de nouvelles molécules pour de nouveaux défis
- Author
-
Dutertre, Charles-Antoine, primary and Teillaud, Jean-Luc, additional
- Published
- 2006
- Full Text
- View/download PDF
117. Monocyte-derived dendritic cells from HLA-B27+ axial spondyloarthritis (SpA) patients display altered functional capacity and deregulated gene expression.
- Author
-
Talpin, Alice, Costantino, Félicie, Bonilla, Nelly, Leboime, Ariane, Letourneur, Franck, Jacques, Sébastien, Dumont, Florent, Amraoui, Sonia, Dutertre, Charles-Antoine, Garchon, Henri-Jean, Breban, Maxime, and Chiocchia, Gilles
- Published
- 2014
- Full Text
- View/download PDF
118. Apoptotic cell capture by DCs induces unexpectedly robust autologous CD4+ T-cell responses.
- Author
-
Valente, Michael, Baey, Camille, Louche, Pauline, Dutertre, Charles ‐ Antoine, Vimeux, Lene, Marañón, Concepción, Hosmalin, Anne, and Feuillet, Vincent
- Abstract
Apoptotic cells represent an important source of self-antigens and their engulfment by dendritic cells (DCs) is usually considered to be related to tolerance induction. We report here an unexpectedly high level of human CD4
+ T-cell proliferation induced by autologous DCs loaded with autologous apoptotic cells, due to the activation of more than 10% of naive CD4+ T cells. This proliferation is not due to an increase in the costimulatory capacity of DCs, but is dependent on apoptotic cell-associated material processed through an endo-lysosomal pathway and presented on DC MHC class II molecules. Autologous CD4+ T cells stimulated with apoptotic cell-loaded DCs exhibit suppressive capacities. However, in the presence of bacterial lipopolysaccharide, apoptotic cell-loaded DCs induce the generation of IL-17-producing cells. Thus, apoptotic cell engulfment by DCs may lead to increased autologous responses, initially generating CD4+ T cells with suppressive capacities able to differentiate into Th17 cells in the presence of a bacterial danger signal such as LPS. [ABSTRACT FROM AUTHOR]- Published
- 2014
- Full Text
- View/download PDF
119. TLR3-Responsive, XCR1+, CD141(BDCA-3)+/CD8α+- Equivalent Dendritic Cells Uncovered in Healthy and Simian Immunodeficiency Virus-Infected Rhesus Macaques.
- Author
-
Dutertre, Charles-Antoine, Jourdain, Jean-Pierre, Rancez, Magali, Amraoui, Sonia, Fossum, Even, Bogen, Bjarne, Sanchez, Cindy, Couédel-Courteille, Anne, Richard, Yolande, Dalod, Marc, Feuillet, Vincent, Cheynier, Rémi, and Hosmalin, Anne
- Subjects
- *
DENDRITIC cells , *SIMIAN immunodeficiency virus , *RHESUS monkeys , *CELL adhesion molecules , *SPLEEN , *BLOOD - Abstract
In mice, CD8α+ myeloid dendritic cells (mDC) optimally cross-present Ags to CD8+ T cells and respond strongly to TLR3 ligands. Although equivalent DC have been identified by comparative genomic analysis and functional studies in humans as XCR1+CD141 (BDCA-3)+Clec9A+cell adhesion molecule 1+ mDC, and in sheep as CD26+ mDC, these cells remained elusive in nonhuman primates. To remedy this situation, we delineated precisely DC and monocyte populations by 12-color flow cytometry and transcriptomic analyses in healthy rhesus macaques. We identified a new mDC population, with strong phenotypic and transcriptional homology to human CD141+ and murine CD8α+ mDC, including XCR1 membrane expression as a conserved specific marker. In contrast, high CD11c expression was not characteristic of mDC in macaques, but of CD16+ monocytes. Like their human and murine homologs, simian XCR1+ mDC had much stronger responses to TLR3 stimulation than other myeloid cells. The importance of this new mDC population was tested in SIVmac251 infection, the most relevant animal model for pathogenic HIV-1 infection and vaccination. This population increased sharply and transiently during acute infection, but was reduced in blood and spleen during advanced disease. The identification of XCR1+ mDC in rhesus macaques opens new avenues for future preclinical vaccinal studies and highlights XCR1 as a prime candidate for targeted vaccine delivery. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
120. Activating and inhibitory Fc&ggr; receptors in immunotherapy:being the actor or being the target.
- Author
-
Abès, Riad, Dutertre, Charles-Antoine, Agnelli, Lauren, and Teillaud, Jean-Luc
- Subjects
IMMUNOTHERAPY ,ANTIBODY-dependent cell cytotoxicity ,BISPECIFIC antibodies ,MONOCLONAL antibodies ,IMMUNOREGULATION ,IMMUNOGLOBULIN G - Abstract
Membrane FC&ggr; receptors (FC&ggr;Rs) can act either as potent activators of effector cell functions or as inhibitors of receptor-mediated cell activation following engagement by IgG antibodies bound to their target molecules. The remarkable ability of activating FC&ggr;Rs to trigger antibody-dependent cellular cytotoxicity, cytokine release and phagocytosis/endocytosis followed by antigen presentation has stimulated the development of a number of therapeutic monoclonal antibodies whose Fc regions have been engineered to optimize their effector functions, mostly their killing activities. Conversely, the demonstration that inhibitory FC&ggr;Rs can block or downmodulate effector functions has led to the concept that targeting these receptors is of interest in a number of pathologies. The use of bispecific antibodies leading to the crosslinking of FC&ggr;RIIB with activating receptors could induce immunomodulation in autoimmune or allergic diseases. Alternatively, the use of cytotoxic/antagonist anti-FC&ggr;RIIB antibodies could kill FC&ggr;RIIB-positive tumor cells or prevent the downmodulation of activating receptors. Thus, antibodies engineered to preferentially target activating or inhibitory FC&ggr;Rs are currently being designed for therapeutic use. INSET: Key issues. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
121. Monocyte-derived dendritic cells from HLA-B27+axial spondyloarthritis (SpA) patients display altered functional capacity and deregulated gene expression
- Author
-
Talpin, Alice, Costantino, Félicie, Bonilla, Nelly, Leboime, Ariane, Letourneur, Franck, Jacques, Sébastien, Dumont, Florent, Amraoui, Sonia, Dutertre, Charles-Antoine, Garchon, Henri-Jean, Breban, Maxime, and Chiocchia, Gilles
- Abstract
This study aimed to compare the functional capacity and gene expression profile of monocyte-derived dendritic cells (MD-DCs) in HLA-B27+axial spondyloarthritis (SpA) patients and healthy controls. MD-DCs were differentiated with interleukin 4 (IL-4) and granulocyte-macrophage colony-stimulating factor (GM-CSF) for seven days, starting from purified CD14+monocytes and stimulated with lipopolysaccharide (LPS) for six and twenty four hours. Their capacity to stimulate allogeneic CD4+T cells from unrelated healthy donor was tested. Transcriptomic study was performed with Affymetrix HuGene 1.0 ST microarrays. Gene expression levels were compared between patients and controls using a multivariate design under a linear model (LIMMA). Real-time quantitative PCR (qRT-PCR) was performed for validation of the most striking gene expression differences. The stimulatory capacity of allogeneic CD4+T cells by MD-DCs from SpA patients was decreased. Transcriptomic analysis revealed 81 genes differentially expressed in MD-DCs between SpA patients and controls (P<0.01 and fold-change <0.66 or >1.5). Four selected genes were validated by qRT-PCR: ADAMTS15, CITED2, F13A1and SELL. Expression levels of ADAMTS15and CITED2,encoding a metallopeptidase and a transcription factor, respectively, were inversely correlated with each other (R = 0.75, P= 0.0003). Furthermore, in silicoanalysis identified several genes of the Wnt signaling pathway having expression co-regulated with CITED2. This study revealed altered function and gene expression pattern in MD-DCs from HLA-B27+axial SpA. Co-expression study showed an inverse correlation between ADAMTS15and CITED2. Moreover, the Wnt signaling pathway appeared as deregulated in SpA MD-DCs, a finding which may be connected to Th17-driven inflammatory responses.
- Published
- 2014
- Full Text
- View/download PDF
122. Pivotal role of M-DC8+monocytes from viremic HIV-infected patients in TNFα overproduction in response to microbial products
- Author
-
Dutertre, Charles-Antoine, Amraoui, Sonia, DeRosa, Annalisa, Jourdain, Jean-Pierre, Vimeux, Lene, Goguet, Matthieu, Degrelle, Séverine, Feuillet, Vincent, Liovat, Anne-Sophie, Müller-Trutwin, Michaela, Decroix, Nipa, Deveau, Christiane, Meyer, Laurence, Goujard, Cécile, Loulergue, Pierre, Launay, Odile, Richard, Yolande, and Hosmalin, Anne
- Abstract
HIV infects activated CD4+T cells and induces their depletion. Progressive HIV infection leading to AIDS is fueled by chronic immune hyperactivation, mediated by inflammatory cytokines like TNFα. This has been related to intestinal epithelial damage and microbial LPS translocation into the circulation. Using 11-color flow cytometry, cell sorting, and cell culture, we investigated the numbers and TNFα production of fully defined circulating dendritic cell and monocyte populations during HIV-1 infection. In 15 viremic, untreated patients, compared with 8 treated, virologically suppressed patients or to 13 healthy blood donors, circulating CD141 (BDCA-3)+and CD1c (BDCA-1)+dendritic cell counts were reduced. Conversely, CD14+CD16++monocyte counts were increased, particularly those expressing M-DC8, while classical CD14++CD16−M-DC8−monocyte numbers were unchanged. Blood mononuclear cells from viremic patients produced more TNFα in response to LPS than those from virologically suppressed patients. M-DC8+monocytes were mostly responsible for this overproduction. Moreover, M-DC8+monocytes differentiated in vitro from classical monocytes using M-CSF and GM-CSF, which is increased in viremic patient's plasma. This M-DC8+monocyte population, which is involved in the pathogenesis of chronic inflammatory diseases like Crohn disease, might thus be considered as a major actor in the immune hyperactivation fueling HIV infection progression.
- Published
- 2012
- Full Text
- View/download PDF
123. NF-κB and TET2 promote macrophage reprogramming in hypoxia that overrides the immunosuppressive effects of the tumor microenvironment.
- Author
-
de la Calle-Fabregat, Carlos, Calafell-Segura, Josep, Gardet, Margaux, Dunsmore, Garett, Mulder, Kevin, Ciudad, Laura, Silvin, Aymeric, Moreno-Càceres, Joaquim, Corbí, Ángel L., Muñoz-Pinedo, Cristina, Michels, Judith, Gouy, Sébastien, Dutertre, Charles-Antoine, Rodríguez-Ubreva, Javier, Ginhoux, Florent, and Ballestar, Esteban
- Subjects
- *
DNA demethylation , *IMMUNE response , *OVARIAN tumors , *TUMOR microenvironment , *MACROPHAGES - Abstract
Macrophages orchestrate tissue homeostasis and immunity. In the tumor microenvironment (TME), macrophage presence is largely associated with poor prognosis because of their reprogramming into immunosuppressive cells. We investigated the effects of hypoxia, a TME-associated feature, on the functional, epigenetic, and transcriptional reprogramming of macrophages and found that hypoxia boosts their immunogenicity. Hypoxic inflammatory macrophages are characterized by a cluster of proinflammatory genes undergoing ten-eleven translocation-mediated DNA demethylation and overexpression. These genes are regulated by NF-κB, while HIF1a dominates the transcriptional reprogramming, demonstrated through ChIP-seq and pharmacological inhibition. In bladder and ovarian carcinomas, hypoxic inflammatory macrophages are enriched in immune-infiltrated tumors, correlating with better patient prognoses. Coculture assays and cell-cell communication analyses support that hypoxic-activated macrophages enhance T cell-mediated responses. The NF-κB-associated hypomethylation signature is displayed by a subset of hypoxic inflammatory macrophages, isolated from ovarian tumors. Our results challenge paradigms regarding the effects of hypoxia on macrophages and highlight actionable target cells to modulate anticancer immune responses. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
124. Single-cell immunology: Past, present, and future.
- Author
-
Ginhoux, Florent, Yalin, Adam, Dutertre, Charles Antoine, and Amit, Ido
- Subjects
- *
IMMUNOLOGY , *CLINICAL immunology , *IMMUNE system , *GENOMICS - Abstract
The immune system is a complex, dynamic, and plastic ecosystem composed of multiple cell types that constantly sense and interact with their local microenvironment to protect from infection and maintain homeostasis. For over a century, great efforts and ingenuity have been applied to the characterization of immune cells and their microenvironments, but traditional marker-based and bulk technologies left key questions unanswered. In the past decade, the advent of single-cell genomic approaches has revolutionized our knowledge of the cellular and molecular makeup of the immune system. In this perspective, we outline the past, present, and future applications of single-cell genomics in immunology and discuss how the integration of multiomics at the single-cell level will pave the way for future advances in immunology research and clinical translation. In this perspective, Ginhoux, Yalin, Dutertre, and Amit outline the past, present, and future applications of single-cell genomics in immunology and discuss how the integration of multiomics at the single-cell level will pave the way for future advances in immunology research and clinical translation. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
125. HIV-1 Reservoirs Form in Urethral Tissue Macrophages of Patients Under Antiretroviral Therapy
- Author
-
Sennepin, Alexis, Real, Fernando, Dutertre, Charles Antoine, Schmitt, Alain, Ganor, Yonatan, Rémi Cheynier, Eugenin, Eliseo, Revol, Marc, Cristofari, Sara, Hosmalin, Anne, and Bomsel, Morgane
126. Mapping the human DC lineage through the integration of high-dimensional techniques
- Author
-
See, Peter, Dutertre, Charles-Antoine, Chen, Jinmiao, Günther, Patrick, McGovern, Naomi, Irac, Sergio Erdal, Gunawan, Merry, Beyer, Marc, Händler, Kristian, Duan, Kaibo, Sumatoh, Hermi Rizal Bin, Ruffin, Nicolas, Jouve, Mabel, Gea-Mallorquí, Ester, Hennekam, Raoul CM, Lim, Tony, Yip, Chan Chung, Wen, Ming, Malleret, Benoit, Low, Ivy, Shadan, Nurhidaya Binte, Fen, Charlene Foong Shu, Tay, Alicia, Lum, Josephine, Zolezzi, Francesca, Larbi, Anis, Poidinger, Michael, Chan, Jerry KY, Chen, Qingfeng, Rénia, Laurent, Haniffa, Muzlifah, Benaroch, Philippe, Schlitzer, Andreas, Schultze, Joachim L, Newell, Evan W, and Ginhoux, Florent
- Subjects
Blood Cells ,Sequence Analysis, RNA ,Humans ,Cell Differentiation ,Cell Lineage ,Dendritic Cells ,Cell Separation ,Single-Cell Analysis ,3. Good health ,Unsupervised Machine Learning
127. Prevalence and functional profile of SARS-CoV-2 T cells in asymptomatic Kenyan adults.
- Author
-
Samandari, Taraz, Ongalo, Joshua B., McCarthy, Kimberly D., Biegon, Richard K., Madiega, Philister A., Mithika, Anne, Orinda, Joseph, Mboya, Grace M., Mwaura, Patrick, Anzala, Omu, Onyango, Clayton, Oluoch, Fredrick O., Osoro, Eric, Dutertre, Charles-Antoine, Tan, Nicole, Hang, Shou Kit, Hariharaputran, Smrithi, Lye, David C., Herman-Roloff, Amy, and Le Bert, Nina
- Subjects
- *
T cells , *KENYANS , *SARS-CoV-2 , *CYTOSKELETAL proteins , *COVID-19 - Abstract
BACKGROUND. SARS-CoV-2 infection in Africa has been characterized by a less severe disease profile than what has been observed elsewhere, but the profile of SARS-CoV-2-specific adaptive immunity in these mainly asymptomatic patients has not, to our knowledge, been analyzed. METHODS. We collected blood samples from residents of rural Kenya (n = 80), who had not experienced any respiratory symptoms or had contact with individuals with COVID-19 and had not received COVID-19 vaccines. We analyzed spike-specific antibodies and T cells specific for SARS-CoV-2 structural (membrane, nucleocapsid, and spike) and accessory (ORF3a, ORF7, ORF8) proteins. Pre-pandemic blood samples collected in Nairobi (n = 13) and blood samples from mild-to-moderately symptomatic COVID-19 convalescent patients (n = 36) living in the urban environment of Singapore were also studied. RESULTS. Among asymptomatic Africans, we detected anti-spike antibodies in 41.0% of the samples and T cell responses against 2 or more SARS-CoV-2 proteins in 82.5% of samples examined. Such a pattern was absent in the pre-pandemic samples. Furthermore, distinct from cellular immunity in European and Asian COVID-19 convalescents, we observed strong T cell immunogenicity against viral accessory proteins (ORF3a, ORF8) but not structural proteins, as well as a higher IL-10/IFN-γ cytokine ratio profile. CONCLUSIONS. The high incidence of T cell responses against different SARS-CoV-2 proteins in seronegative participants suggests that serosurveys underestimate SARS-CoV-2 prevalence in settings where asymptomatic infections prevail. The functional and antigen-specific profile of SARS-CoV-2-specific T cells in African individuals suggests that environmental factors can play a role in the development of protective antiviral immunity. FUNDING. US Centers for Disease Control and Prevention, Division of Global Health Protection; the Singapore Ministry of Health's National Medical Research Council (COVID19RF3-0060, COVID19RF-001, COVID19RF-008, MOH-StaR17Nov-0001). [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
128. Activation of the Receptor NKG2D Leads to Production of Th17 Cytokines in CD4+ T Cells of Patients With Crohn's Disease.
- Author
-
Pariente, Benjamin, Mocan, Iulia, Camus, Matthieu, Dutertre, Charles–Antoine, Ettersperger, Julien, Cattan, Pierre, Gornet, Jean–Marc, Dulphy, Nicolas, Charron, Dominique, Lémann, Marc, Toubert, Antoine, and Allez, Matthieu
- Subjects
CYTOKINES ,CROHN'S disease ,INFLAMMATION ,MUCOUS membranes ,IMMUNE response ,INFLAMMATORY bowel diseases ,INTERFERONS - Abstract
Background & Aims: The natural killer group 2 member D (NKG2D) is a stimulatory receptor expressed on a subset of mucosal and peripheral CD4
+ T cells in patients with Crohn''s disease (CD) and other inflammatory diseases. Ligand activation of NKG2D in patients induces CD4+ T cells to release T-helper (Th) 1 cytokines and become cytotoxic. We investigated the Th17 cytokines produced by T cells that express NKG2D in blood and intestinal mucosa samples from patients with CD. Methods: We isolated CD4+ T cells from peripheral blood and lamina propria samples of patients with CD or ulcerative colitis (UC) and healthy individuals (controls). We analyzed the phenotype and functions of the CD4+ NKG2D+ T cells and the cytokines they produce in response to NKG2D stimulation. Results: In patients with CD, CD4+ T cells that express NKG2D produced high levels of interleukin (IL)-17 and IL-22 and expressed high levels of CCR6, the IL-23 receptor, CD161, and RORC (a transcription factor that regulates expression of Th17 cytokines). CD4+ T cells that produced IL-17 expressed high levels of NKG2D and CD161. Costimulation of NKG2D and the T-cell receptor (TCR) significantly increased production of IL-17 and tumor necrosis factor α by CD4+ T cells, compared with activation of only the TCR. CD4+ NKG2D+ T cells also responded to Th17 polarization. Conclusions: NKG2D is a functional marker of CD4+ T cells that produce IL-17 in patients with CD, via costimulation of the TCR and NKG2D. Reagents developed to block NKG2D might reduce gastrointestinal inflammation in patients with CD. [Copyright &y& Elsevier]- Published
- 2011
- Full Text
- View/download PDF
129. Systemic vaccination induces CD8+ T cells and remodels the tumor microenvironment.
- Author
-
Baharom, Faezzah, Ramirez-Valdez, Ramiro A., Khalilnezhad, Ahad, Khalilnezhad, Shabnam, Dillon, Marlon, Hermans, Dalton, Fussell, Sloane, Tobin, Kennedy K.S., Dutertre, Charles-Antoine, Lynn, Geoffrey M., Müller, Sören, Ginhoux, Florent, Ishizuka, Andrew S., and Seder, Robert A.
- Subjects
- *
TUMOR microenvironment , *T cells , *TYPE I interferons , *MYELOID cells , *CD8 antigen , *PEPTIDE receptors , *CELL physiology - Abstract
Therapeutic cancer vaccines are designed to increase tumor-specific T cell immunity. However, suppressive mechanisms within the tumor microenvironment (TME) may limit T cell function. Here, we assessed how the route of vaccination alters intratumoral myeloid cells. Using a self-assembling nanoparticle vaccine that links tumor antigen peptides to a Toll-like receptor 7/8 agonist (SNP-7/8a), we treated tumor-bearing mice subcutaneously (SNP-SC) or intravenously (SNP-IV). Both routes generated antigen-specific CD8+ T cells that infiltrated tumors. However, only SNP-IV mediated tumor regression, dependent on systemic type I interferon at the time of boost. Single-cell RNA-sequencing revealed that intratumoral monocytes expressing an immunoregulatory gene signature (Chil3 , Anxa2 , Wfdc17) were reduced after SNP-IV boost. In humans, the Chil3 + monocyte gene signature is enriched in CD16– monocytes and associated with worse outcomes. Our results show that the generation of tumor-specific CD8+ T cells combined with remodeling of the TME is a promising approach for tumor immunotherapy. [Display omitted] • Tumor-specific T cells are necessary but not sufficient for therapeutic efficacy • IV vaccination promotes tumor regression by remodeling the TME • Systemic IFN-I following IV vaccination alters intratumoral Chil3 + monocytes • Enrichment of human homologs of Chil3 + monocytes is associated with worse outcomes Intravenous administration of a cancer vaccine promotes tumor regression via antigen-specific CD8+ T cells and type I interferon-dependent modulation of the tumor microenvironment. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
130. A TLR3 Ligand Reestablishes Chemotherapeutic Responses in the Context of FPR1 Deficiency
- Author
-
Francesca Castoldi, Maria Chiara Maiuri, Julie Le Naour, Julien Taieb, Allan Sauvat, Suzette Delaloge, Charles-Antoine Dutertre, Jessica Zucman-Rossi, Juliette Paillet, Erika Vacchelli, Liwei Zhao, Giulia Cerrato, Guido Kroemer, Claire Mulot, Isabelle Martins, Oliver Kepp, Federico Pietrocola, Pierre Laurent-Puig, Aymeric Silvin, Florent Ginhoux, Fabrice Andre, Peng Liu, Laurence Zitvogel, Sandy Adjemian, Cornelia Richter, Zoltan Szallasi, Peter Vandenabeele, Zsofia Sztupinszki, Jonathan Pol, Gautier Stoll, Khady Mangane, Centre de Recherche des Cordeliers (CRC (UMR_S_1138 / U1138)), École Pratique des Hautes Études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Université Paris Cité (UPCité), Institut Gustave Roussy (IGR), Institut Universitaire de France (IUF), Ministère de l'Education nationale, de l’Enseignement supérieur et de la Recherche (M.E.N.E.S.R.), Universidade de São Paulo = University of São Paulo (USP), Boston Children's Hospital, Harvard Medical School [Boston] (HMS), Cancer Research and Personalized Medicine - CARPEM [Paris], Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Hôpital Cochin [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Necker - Enfants Malades [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Immunologie des tumeurs et immunothérapie (UMR 1015), Université Paris-Sud - Paris 11 (UP11)-Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM), Agency for science, technology and research [Singapore] (A*STAR), Singapore Immunology Network (SIgN), Biomedical Sciences Institute (BMSI), School of Medicine [Shanghai Jiaotong University], Shanghai Jiaotong University, Métabolisme, Cancer et Immunité (CRC - UMR_S 1138), Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Université Paris Cité (UPCité)-Centre de Recherche des Cordeliers (CRC (UMR_S_1138 / U1138)), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Université Paris Cité (UPCité)-École Pratique des Hautes Études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), University of Oslo (UiO), Direction de la recherche [Gustave Roussy], Département de médecine oncologique [Gustave Roussy], Pathologie mammaire, Institut Gustave Roussy (IGR)-Institut Gustave Roussy (IGR), Immunologie anti-tumorale et immunothérapie des cancers (ITIC), Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Saclay, ANR-16-RHUS-0008,LUMIERE,LUMIERE(2016), European Project: 825410,ONCOBIOME, Le Naour, Julie, Liu, Peng, Zhao, Liwei, Adjemian, Sandy, Sztupinszki, Zsofia, Taieb, Julien, Mulot, Claire, Silvin, Aymeric, Dutertre, Charles-Antoine, Ginhoux, Florent, Sauvat, Allan, Cerrato, Giulia, Castoldi, Francesca, Martins, Isabelle, Stoll, Gautier, Paillet, Juliette, Mangane, Khady, Richter, Cornelia, Kepp, Oliver, Maiuri, Maria Chiara, Pietrocola, Federico, Vandenabeele, Peter, André, Fabrice, Delaloge, Suzette, Szallasi, Zoltan, Laurent-Puig, Pierre, Zucman-Rossi, Jessica, Zitvogel, Laurence, Pol, Jonathan G, Vacchelli, Erika, Kroemer, Guido, École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Université Paris Cité (UPC), Universidade de São Paulo (USP), Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Université Paris Cité (UPC)-Centre de Recherche des Cordeliers (CRC (UMR_S_1138 / U1138)), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Université Paris Cité (UPC)-École pratique des hautes études (EPHE), and Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPC)
- Subjects
0301 basic medicine ,Anthracycline ,medicine.medical_treatment ,[SDV]Life Sciences [q-bio] ,Mice, Transgenic ,Ligands ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Medicine ,Animals ,Humans ,ComputingMilieux_MISCELLANEOUS ,business.industry ,Pattern recognition receptor ,Wild type ,Immunotherapy ,Receptors, Formyl Peptide ,Toll-Like Receptor 3 ,Disease Models, Animal ,030104 developmental biology ,Cell Transformation, Neoplastic ,Poly I-C ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,TLR3 ,Cancer research ,business ,Colorectal Neoplasms ,Annexin A1 - Abstract
For anthracycline-based chemotherapy to be immunogenic, dying cancer cells must release annexin A1 (ANXA1) that subsequently interacts with the pattern recognition receptor, formyl peptide receptor 1 (FPR1), on the surface of dendritic cells (DC). Approximately 30% of individuals bear loss-of-function alleles of FPR1, calling for strategies to ameliorate their anticancer immune response. Here, we show that immunotherapy with a ligand of Toll-like receptor-3, polyinosinic:polycytidylic acid (pIC), restores the deficient response to chemotherapy of tumors lacking ANXA1 developing in immunocompetent mice or those of normal cancers growing in FPR1-deficient mice. This effect was accompanied by improved DC- and T-lymphocyte–mediated anticancer immunity. Of note, carcinogen-induced breast cancers precociously developed in FPR1-deficient mice as compared with wild-type controls. A similar tendency for earlier cancer development was found in patients carrying the loss-of-function allele of FPR1. These findings have potential implications for the clinical management of FPR1-deficient patients. Significance: The loss-of-function variant rs867228 in FPR1, harbored by approximately 30% of the world population, is associated with the precocious manifestation of breast, colorectal, esophageal, and head and neck carcinomas. pIC restores deficient chemotherapeutic responses in mice lacking Fpr1, suggesting a personalized strategy for compensating for the FPR1 defect. This article is highlighted in the In This Issue feature, p. 211
- Published
- 2021
- Full Text
- View/download PDF
131. Non-terminally exhausted tumor-resident memory HBV-specific T cell responses correlate with relapse-free survival in hepatocellular carcinoma.
- Author
-
Cheng, Yang, Gunasegaran, Bavani, Singh, Harsimran D., Dutertre, Charles-Antoine, Loh, Chiew Yee, Lim, Jia Qi, Crawford, Jeremy Chase, Lee, Hong Kai, Zhang, Xiaomeng, Lee, Bernett, Becht, Etienne, Lim, Wan Jun, Yeong, Joe, Chan, Chung Yip, Chung, Alexander, Goh, Brian K.P., Chow, Pierce K.H., Chan, Jerry K.Y., Ginhoux, Florent, and Tai, David
- Subjects
- *
T cells , *CHRONIC hepatitis B , *HEPATOCELLULAR carcinoma , *IMMUNE checkpoint proteins , *HEPATITIS B virus , *CELL populations , *AUTOBIOGRAPHICAL memory , *BLOOD cells - Abstract
Hepatocellular carcinoma (HCC) often develops following chronic hepatitis B virus (HBV) infection and responds poorly to immune checkpoint blockade. Here, we examined the antigen specificities of HCC-infiltrating T cells and their relevance to tumor control. Using highly multiplexed peptide-MHC tetramer staining of unexpanded cells from blood, liver, and tumor tissues from 46 HCC patients, we detected 91 different antigen-specific CD8+ T cell populations targeting HBV, neoantigen, tumor-associated, and disease-unrelated antigens. Parallel high-dimensional analysis delineated five distinct antigen-specific tissue-resident memory T (Trm) cell populations. Intratumoral and intrahepatic HBV-specific T cells were enriched for two Trm cell subsets that were PD-1lo TOX lo, despite being clonally expanded. High frequencies of intratumoral terminally exhausted T cells were uncommon. Patients with tumor-infiltrating HBV-specific CD8+ Trm cells exhibited longer-term relapse-free survival. Thus, non-terminally exhausted HBV-specific CD8+ Trm cells show hallmarks of active involvement and effective antitumor response, implying that these cells could be harnessed for therapeutic purposes. [Display omitted] • Broad analysis of CD8+ T cell antigen specificity in HBV-HCC patient blood, liver, tumor • Tumor-infiltrating HBV-specific T cells correlate with superior relapse-free survival • Five Trm subsets are enriched in liver- and tumor-infiltrating HBV-specific T cells • HBV-specific Trm cells have expanded TCR clones and lack hallmarks of terminal exhaustion Hepatocellular carcinoma (HCC) often develops following chronic hepatitis B virus (HBV) infection and responds poorly to immune checkpoint blockade. Cheng et al. examine the antigen specificities of HCC-infiltrating T cells and reveal that HBV-specific Trm cells are phenotypically distinct, clonally expanded, and are not terminally exhausted, suggesting that these cells may be harnessed for therapeutic purposes. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
132. Microbial exposure during early human development primes fetal immune cells.
- Author
-
Mishra, Archita, Lai, Ghee Chuan, Yao, Leong Jing, Aung, Thet Tun, Shental, Noam, Rotter-Maskowitz, Aviva, Shepherdson, Edwin, Singh, Gurmit Singh Naranjan, Pai, Rhea, Shanti, Adhika, Wong, Regina Men Men, Lee, Andrea, Khyriem, Costerwell, Dutertre, Charles Antoine, Chakarov, Svetoslav, Srinivasan, K.G., Shadan, Nurhidaya Binte, Zhang, Xiao-Meng, Khalilnezhad, Shabnam, and Cottier, Fabien
- Subjects
- *
FETAL development , *T cells , *FETAL tissues , *CELL analysis , *FETUS , *HUMAN embryology , *LYMPH nodes , *PHENOTYPES - Abstract
The human fetal immune system begins to develop early during gestation; however, factors responsible for fetal immune-priming remain elusive. We explored potential exposure to microbial agents in utero and their contribution toward activation of memory T cells in fetal tissues. We profiled microbes across fetal organs using 16S rRNA gene sequencing and detected low but consistent microbial signal in fetal gut, skin, placenta, and lungs in the 2nd trimester of gestation. We identified several live bacterial strains including Staphylococcus and Lactobacillus in fetal tissues, which induced in vitro activation of memory T cells in fetal mesenteric lymph node, supporting the role of microbial exposure in fetal immune-priming. Finally, using SEM and RNA-ISH, we visualized discrete localization of bacteria-like structures and eubacterial-RNA within 14th weeks fetal gut lumen. These findings indicate selective presence of live microbes in fetal organs during the 2nd trimester of gestation and have broader implications toward the establishment of immune competency and priming before birth. [Display omitted] • Human fetuses in 2nd trimester show T cell diversity with effector-memory phenotype • Fetal organs show diverse bacterial genera that can be cultured and propagated • Bacterial structures with mucin-like threads are visualized in 14-weeks EGA fetal gut • Fetal bacteria induce syngeneic memory T cell activation in fetal mLN T cells Analysis of human fetal tissues and the placenta in the 2nd trimester of gestation identifies live bacterial strains that are able to induce the activation of memory T cells in the fetal mesenteric lymph node, thus providing insights into early life immunity. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
133. Guidelines for preparation and flow cytometry analysis of human nonlymphoid tissue DC.
- Author
-
Dudziak D, Heger L, Agace WW, Bakker J, de Gruijl TD, Dress RJ, Dutertre CA, Fenton TM, Fransen MF, Ginhoux F, Heyman O, Horev Y, Hornsteiner F, Kandiah V, Kles P, Lubin R, Mizraji G, Prokopi A, Saar O, Sopper S, Stoitzner P, Strandt H, Sykora MM, Toffoli EC, Tripp CH, van Pul K, van de Ven R, Wilensky A, Yona S, and Zelle-Rieser C
- Abstract
This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy, and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs, and various nonlymphoid tissues. Within this article, detailed protocols are presented that allow for the generation of single-cell suspensions from human nonlymphoid tissues including lung, skin, gingiva, intestine as well as from tumors and tumor-draining lymph nodes with a subsequent analysis of dendritic cells by flow cytometry. Further, prepared single-cell suspensions can be subjected to other applications including cellular enrichment procedures, RNA sequencing, functional assays, etc. While all protocols were written by experienced scientists who routinely use them in their work, this article was also peer-reviewed by leading experts and approved by all co-authors, making it an essential resource for basic and clinical DC immunologists., (© 2024 The Author(s). European Journal of Immunology published by Wiley‐VCH GmbH.)
- Published
- 2024
- Full Text
- View/download PDF
134. TREM2-Expressing Multinucleated Giant Macrophages Are a Biomarker of Good Prognosis in Head and Neck Squamous Cell Carcinoma.
- Author
-
Gessain G, Anzali AA, Lerousseau M, Mulder K, Bied M, Auperin A, Stockholm D, Signolle N, Sassi F, Marques Da Costa ME, Marchais A, Sayadi A, Weidner D, Uderhardt S, Blampey Q, Nakkireddy SR, Broutin S, Dutertre CA, Busson P, Walter T, Marhic A, Moya-Plana A, Guerlain J, Breuskin I, Casiraghi O, Gorphe P, Classe M, Scoazec JY, Blériot C, and Ginhoux F
- Subjects
- Humans, Prognosis, Giant Cells pathology, Giant Cells metabolism, Membrane Glycoproteins metabolism, Biomarkers, Tumor metabolism, Receptors, Immunologic metabolism, Receptors, Immunologic genetics, Macrophages metabolism, Squamous Cell Carcinoma of Head and Neck metabolism, Squamous Cell Carcinoma of Head and Neck pathology, Squamous Cell Carcinoma of Head and Neck genetics, Head and Neck Neoplasms metabolism, Head and Neck Neoplasms pathology, Head and Neck Neoplasms diagnosis, Head and Neck Neoplasms genetics
- Abstract
Significance: Novel individual biomarkers are needed to guide therapeutic decisions for patients with head and neck cancer. We report for the first time, granulomas of TREM2-expressing multinucleated giant macrophages in keratin-rich tumor niches, as a biomarker of favorable prognosis and developed a deep-learning model to automate its quantification on routinely stained pathological slides., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
135. Sopa: a technology-invariant pipeline for analyses of image-based spatial omics.
- Author
-
Blampey Q, Mulder K, Gardet M, Christodoulidis S, Dutertre CA, André F, Ginhoux F, and Cournède PH
- Subjects
- Humans, Image Processing, Computer-Assisted methods, Single-Cell Analysis methods, Computational Biology methods, Transcriptome, Animals, Software
- Abstract
Spatial omics data allow in-depth analysis of tissue architectures, opening new opportunities for biological discovery. In particular, imaging techniques offer single-cell resolutions, providing essential insights into cellular organizations and dynamics. Yet, the complexity of such data presents analytical challenges and demands substantial computing resources. Moreover, the proliferation of diverse spatial omics technologies, such as Xenium, MERSCOPE, CosMX in spatial-transcriptomics, and MACSima and PhenoCycler in multiplex imaging, hinders the generality of existing tools. We introduce Sopa ( https://github.com/gustaveroussy/sopa ), a technology-invariant, memory-efficient pipeline with a unified visualizer for all image-based spatial omics. Built upon the universal SpatialData framework, Sopa optimizes tasks like segmentation, transcript/channel aggregation, annotation, and geometric/spatial analysis. Its output includes user-friendly web reports and visualizer files, as well as comprehensive data files for in-depth analysis. Overall, Sopa represents a significant step toward unifying spatial data analysis, enabling a more comprehensive understanding of cellular interactions and tissue organization in biological systems., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
136. A new step in understanding mouse cDC ontogeny.
- Author
-
Dutertre CA
- Subjects
- Animals, Mice
- Published
- 2024
- Full Text
- View/download PDF
137. BCL2 Inhibition Reveals a Dendritic Cell-Specific Immune Checkpoint That Controls Tumor Immunosurveillance.
- Author
-
Zhao L, Liu P, Mao M, Zhang S, Bigenwald C, Dutertre CA, Lehmann CHK, Pan H, Paulhan N, Amon L, Buqué A, Yamazaki T, Galluzzi L, Kloeckner B, Silvin A, Pan Y, Chen H, Tian AL, Ly P, Dudziak D, Zitvogel L, Kepp O, and Kroemer G
- Subjects
- Humans, Animals, Mice, Dendritic Cells, Programmed Cell Death 1 Receptor, Monitoring, Immunologic, Mice, Knockout, Proto-Oncogene Proteins c-bcl-2 genetics, Neoplasms drug therapy, Neoplasms genetics, Antineoplastic Agents therapeutic use
- Abstract
We developed a phenotypic screening platform for the functional exploration of dendritic cells (DC). Here, we report a genome-wide CRISPR screen that revealed BCL2 as an endogenous inhibitor of DC function. Knockout of BCL2 enhanced DC antigen presentation and activation as well as the capacity of DCs to control tumors and to synergize with PD-1 blockade. The pharmacologic BCL2 inhibitors venetoclax and navitoclax phenocopied these effects and caused a cDC1-dependent regression of orthotopic lung cancers and fibrosarcomas. Thus, solid tumors failed to respond to BCL2 inhibition in mice constitutively devoid of cDC1, and this was reversed by the infusion of DCs. Moreover, cDC1 depletion reduced the therapeutic efficacy of BCL2 inhibitors alone or in combination with PD-1 blockade and treatment with venetoclax caused cDC1 activation, both in mice and in patients. In conclusion, genetic and pharmacologic BCL2 inhibition unveils a DC-specific immune checkpoint that restrains tumor immunosurveillance., Significance: BCL2 inhibition improves the capacity of DCs to stimulate anticancer immunity and restrain cancer growth in an immunocompetent context but not in mice lacking cDC1 or mature T cells. This study indicates that BCL2 blockade can be used to sensitize solid cancers to PD-1/PD-L1-targeting immunotherapy. This article is featured in Selected Articles from This Issue, p. 2293., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
138. iPS-cell-derived microglia promote brain organoid maturation via cholesterol transfer.
- Author
-
Park DS, Kozaki T, Tiwari SK, Moreira M, Khalilnezhad A, Torta F, Olivié N, Thiam CH, Liani O, Silvin A, Phoo WW, Gao L, Triebl A, Tham WK, Gonçalves L, Kong WT, Raman S, Zhang XM, Dunsmore G, Dutertre CA, Lee S, Ong JM, Balachander A, Khalilnezhad S, Lum J, Duan K, Lim ZM, Tan L, Low I, Utami KH, Yeo XY, Di Tommaso S, Dupuy JW, Varga B, Karadottir RT, Madathummal MC, Bonne I, Malleret B, Binte ZY, Wei Da N, Tan Y, Wong WJ, Zhang J, Chen J, Sobota RM, Howland SW, Ng LG, Saltel F, Castel D, Grill J, Minard V, Albani S, Chan JKY, Thion MS, Jung SY, Wenk MR, Pouladi MA, Pasqualini C, Angeli V, Cexus ONF, and Ginhoux F
- Subjects
- Animals, Humans, Mice, Cell Differentiation, Axons, Cell Proliferation, Esters metabolism, Lipid Droplets metabolism, Brain cytology, Brain metabolism, Induced Pluripotent Stem Cells cytology, Microglia cytology, Microglia metabolism, Neurogenesis, Organoids cytology, Organoids metabolism, Cholesterol metabolism, Neural Stem Cells cytology, Neural Stem Cells metabolism
- Abstract
Microglia are specialized brain-resident macrophages that arise from primitive macrophages colonizing the embryonic brain
1 . Microglia contribute to multiple aspects of brain development, but their precise roles in the early human brain remain poorly understood owing to limited access to relevant tissues2-6 . The generation of brain organoids from human induced pluripotent stem cells recapitulates some key features of human embryonic brain development7-10 . However, current approaches do not incorporate microglia or address their role in organoid maturation11-21 . Here we generated microglia-sufficient brain organoids by coculturing brain organoids with primitive-like macrophages generated from the same human induced pluripotent stem cells (iMac)22 . In organoid cocultures, iMac differentiated into cells with microglia-like phenotypes and functions (iMicro) and modulated neuronal progenitor cell (NPC) differentiation, limiting NPC proliferation and promoting axonogenesis. Mechanistically, iMicro contained high levels of PLIN2+ lipid droplets that exported cholesterol and its esters, which were taken up by NPCs in the organoids. We also detected PLIN2+ lipid droplet-loaded microglia in mouse and human embryonic brains. Overall, our approach substantially advances current human brain organoid approaches by incorporating microglial cells, as illustrated by the discovery of a key pathway of lipid-mediated crosstalk between microglia and NPCs that leads to improved neurogenesis., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
139. Dendritic cell type 3 arises from Ly6C + monocyte-dendritic cell progenitors.
- Author
-
Liu Z, Wang H, Li Z, Dress RJ, Zhu Y, Zhang S, De Feo D, Kong WT, Cai P, Shin A, Piot C, Yu J, Gu Y, Zhang M, Gao C, Chen L, Wang H, Vétillard M, Guermonprez P, Kwok I, Ng LG, Chakarov S, Schlitzer A, Becher B, Dutertre CA, Su B, and Ginhoux F
- Subjects
- Mice, Humans, Animals, Phenotype, Cells, Cultured, Dendritic Cells, Cell Differentiation, Monocytes, Stem Cells
- Abstract
Conventional dendritic cells (cDCs) are professional antigen-presenting cells that control the adaptive immune response. Their subsets and developmental origins have been intensively investigated but are still not fully understood as their phenotypes, especially in the DC2 lineage and the recently described human DC3s, overlap with monocytes. Here, using LEGENDScreen to profile DC vs. monocyte lineages, we found sustained expression of FLT3 and CD45RB through the whole DC lineage, allowing DCs and their precursors to be distinguished from monocytes. Using fate mapping models, single-cell RNA sequencing and adoptive transfer, we identified a lineage of murine CD16/32
+ CD172a+ DC3, distinct from DC2, arising from Ly6C+ monocyte-DC progenitors (MDPs) through Lyz2+ Ly6C+ CD11c- pro-DC3s, whereas DC2s develop from common DC progenitors (CDPs) through CD7+ Ly6C+ CD11c+ pre-DC2s. Corresponding DC subsets, developmental stages, and lineages exist in humans. These findings reveal DC3 as a DC lineage phenotypically related to but developmentally different from monocytes and DC2s., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
140. A microbiota-modulated checkpoint directs immunosuppressive intestinal T cells into cancers.
- Author
-
Fidelle M, Rauber C, Alves Costa Silva C, Tian AL, Lahmar I, de La Varende AM, Zhao L, Thelemaque C, Lebhar I, Messaoudene M, Pizzato E, Birebent R, Mbogning Fonkou MD, Zoppi S, Reni A, Dalban C, Leduc M, Ferrere G, Durand S, Ly P, Silvin A, Mulder K, Dutertre CA, Ginhoux F, Yonekura S, Roberti MP, Tidjani-Alou M, Terrisse S, Chen J, Kepp O, Schippers A, Wagner N, Suárez-Gosálvez J, Kobold S, Fahrner JE, Richard C, Bosq J, Lordello L, Vitali G, Galleron N, Quinquis B, Le Chatelier E, Blanchard L, Girard JP, Jarry A, Gervois N, Godefroy E, Labarrière N, Koschny R, Daillère R, Besse B, Truntzer C, Ghiringhelli F, Coatnoan N, Mhanna V, Klatzmann D, Drubay D, Albiges L, Thomas AM, Segata N, Danlos FX, Marabelle A, Routy B, Derosa L, Kroemer G, and Zitvogel L
- Subjects
- Animals, Humans, Mice, Bacteria immunology, Cell Movement, Fecal Microbiota Transplantation, Interleukin-17 metabolism, Th17 Cells immunology, Gastrointestinal Tract immunology, Gastrointestinal Tract microbiology, Anti-Bacterial Agents adverse effects, Cell Adhesion Molecules metabolism, Drug Resistance, Neoplasm, Gastrointestinal Microbiome immunology, Immune Checkpoint Inhibitors therapeutic use, Immune Tolerance drug effects, Immunologic Surveillance, Integrins metabolism, Mucoproteins metabolism, Neoplasms immunology, Neoplasms therapy
- Abstract
Antibiotics (ABX) compromise the efficacy of programmed cell death protein 1 (PD-1) blockade in cancer patients, but the mechanisms underlying their immunosuppressive effects remain unknown. By inducing the down-regulation of mucosal addressin cell adhesion molecule 1 (MAdCAM-1) in the ileum, post-ABX gut recolonization by Enterocloster species drove the emigration of enterotropic α4β7
+ CD4+ regulatory T 17 cells into the tumor. These deleterious ABX effects were mimicked by oral gavage of Enterocloster species, by genetic deficiency, or by antibody-mediated neutralization of MAdCAM-1 and its receptor, α4β7 integrin. By contrast, fecal microbiota transplantation or interleukin-17A neutralization prevented ABX-induced immunosuppression. In independent lung, kidney, and bladder cancer patient cohorts, low serum levels of soluble MAdCAM-1 had a negative prognostic impact. Thus, the MAdCAM-1-α4β7 axis constitutes an actionable gut immune checkpoint in cancer immunosurveillance.- Published
- 2023
- Full Text
- View/download PDF
141. Dual ontogeny of disease-associated microglia and disease inflammatory macrophages in aging and neurodegeneration.
- Author
-
Silvin A, Uderhardt S, Piot C, Da Mesquita S, Yang K, Geirsdottir L, Mulder K, Eyal D, Liu Z, Bridlance C, Thion MS, Zhang XM, Kong WT, Deloger M, Fontes V, Weiner A, Ee R, Dress R, Hang JW, Balachander A, Chakarov S, Malleret B, Dunsmore G, Cexus O, Chen J, Garel S, Dutertre CA, Amit I, Kipnis J, and Ginhoux F
- Subjects
- Aging, Animals, Brain pathology, Humans, Macrophages pathology, Membrane Glycoproteins, Mice, Receptors, Immunologic, Alzheimer Disease genetics, Microglia pathology
- Abstract
Brain macrophage populations include parenchymal microglia, border-associated macrophages, and recruited monocyte-derived cells; together, they control brain development and homeostasis but are also implicated in aging pathogenesis and neurodegeneration. The phenotypes, localization, and functions of each population in different contexts have yet to be resolved. We generated a murine brain myeloid scRNA-seq integration to systematically delineate brain macrophage populations. We show that the previously identified disease-associated microglia (DAM) population detected in murine Alzheimer's disease models actually comprises two ontogenetically and functionally distinct cell lineages: embryonically derived triggering receptor expressed on myeloid cells 2 (TREM2)-dependent DAM expressing a neuroprotective signature and monocyte-derived TREM2-expressing disease inflammatory macrophages (DIMs) accumulating in the brain during aging. These two distinct populations appear to also be conserved in the human brain. Herein, we generate an ontogeny-resolved model of brain myeloid cell heterogeneity in development, homeostasis, and disease and identify cellular targets for the treatment of neurodegeneration., Competing Interests: Declaration of interests A.S. and F.G. are inventors on a patent filed, owned, and managed by A∗ccelerate technologies Pte Ltd, A(∗)STAR, Singapore, on technology related to the work presented in this manuscript., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
142. Tissue-resident FOLR2 + macrophages associate with CD8 + T cell infiltration in human breast cancer.
- Author
-
Nalio Ramos R, Missolo-Koussou Y, Gerber-Ferder Y, Bromley CP, Bugatti M, Núñez NG, Tosello Boari J, Richer W, Menger L, Denizeau J, Sedlik C, Caudana P, Kotsias F, Niborski LL, Viel S, Bohec M, Lameiras S, Baulande S, Lesage L, Nicolas A, Meseure D, Vincent-Salomon A, Reyal F, Dutertre CA, Ginhoux F, Vimeux L, Donnadieu E, Buttard B, Galon J, Zelenay S, Vermi W, Guermonprez P, Piaggio E, and Helft J
- Subjects
- Breast immunology, CD8-Positive T-Lymphocytes, Female, Folate Receptor 2, Humans, Lymphocytes, Tumor-Infiltrating, Prognosis, Breast Neoplasms epidemiology, Breast Neoplasms immunology, Macrophages
- Abstract
Macrophage infiltration is a hallmark of solid cancers, and overall macrophage infiltration correlates with lower patient survival and resistance to therapy. Tumor-associated macrophages, however, are phenotypically and functionally heterogeneous. Specific subsets of tumor-associated macrophage might be endowed with distinct roles on cancer progression and antitumor immunity. Here, we identify a discrete population of FOLR2
+ tissue-resident macrophages in healthy mammary gland and breast cancer primary tumors. FOLR2+ macrophages localize in perivascular areas in the tumor stroma, where they interact with CD8+ T cells. FOLR2+ macrophages efficiently prime effector CD8+ T cells ex vivo. The density of FOLR2+ macrophages in tumors positively correlates with better patient survival. This study highlights specific roles for tumor-associated macrophage subsets and paves the way for subset-targeted therapeutic interventions in macrophages-based cancer therapies., Competing Interests: Declaration of interests The authors J.H., R.N.R., E.P., and P.G. and their institutions have filed a patent related to this work., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
143. Twin study reveals non-heritable immune perturbations in multiple sclerosis.
- Author
-
Ingelfinger F, Gerdes LA, Kavaka V, Krishnarajah S, Friebel E, Galli E, Zwicky P, Furrer R, Peukert C, Dutertre CA, Eglseer KM, Ginhoux F, Flierl-Hecht A, Kümpfel T, De Feo D, Schreiner B, Mundt S, Kerschensteiner M, Hohlfeld R, Beltrán E, and Becher B
- Subjects
- Genetic Predisposition to Disease genetics, Humans, Interleukin-2 genetics, OX40 Ligand, Twins, Dizygotic genetics, Twins, Monozygotic genetics, Multiple Sclerosis
- Abstract
Multiple sclerosis (MS) is a chronic inflammatory disorder of the central nervous system underpinned by partially understood genetic risk factors and environmental triggers and their undefined interactions
1,2 . Here we investigated the peripheral immune signatures of 61 monozygotic twin pairs discordant for MS to dissect the influence of genetic predisposition and environmental factors. Using complementary multimodal high-throughput and high-dimensional single-cell technologies in conjunction with data-driven computational tools, we identified an inflammatory shift in a monocyte cluster of twins with MS, coupled with the emergence of a population of IL-2 hyper-responsive transitional naive helper T cells as MS-related immune alterations. By integrating data on the immune profiles of healthy monozygotic and dizygotic twin pairs, we estimated the variance in CD25 expression by helper T cells displaying a naive phenotype to be largely driven by genetic and shared early environmental influences. Nonetheless, the expanding helper T cells of twins with MS, which were also elevated in non-twin patients with MS, emerged independent of the individual genetic makeup. These cells expressed central nervous system-homing receptors, exhibited a dysregulated CD25-IL-2 axis, and their proliferative capacity positively correlated with MS severity. Together, our matched-pair analysis of the extended twin approach allowed us to discern genetically and environmentally determined features of an MS-associated immune signature., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
144. A single-cell and spatially resolved atlas of human breast cancers.
- Author
-
Wu SZ, Al-Eryani G, Roden DL, Junankar S, Harvey K, Andersson A, Thennavan A, Wang C, Torpy JR, Bartonicek N, Wang T, Larsson L, Kaczorowski D, Weisenfeld NI, Uytingco CR, Chew JG, Bent ZW, Chan CL, Gnanasambandapillai V, Dutertre CA, Gluch L, Hui MN, Beith J, Parker A, Robbins E, Segara D, Cooper C, Mak C, Chan B, Warrier S, Ginhoux F, Millar E, Powell JE, Williams SR, Liu XS, O'Toole S, Lim E, Lundeberg J, Perou CM, and Swarbrick A
- Subjects
- B-Lymphocytes immunology, B7-H1 Antigen genetics, Biomarkers, Tumor genetics, Breast Neoplasms immunology, CD8-Positive T-Lymphocytes immunology, Endothelial Cells metabolism, Female, Gene Expression Regulation, Neoplastic genetics, Humans, Macrophages cytology, Macrophages immunology, Membrane Proteins genetics, Myeloid Cells immunology, Myeloid Cells metabolism, Sequence Analysis, RNA, Tumor Microenvironment, Tumor Suppressor Proteins genetics, Breast Neoplasms genetics, Breast Neoplasms pathology, Single-Cell Analysis, Transcriptome genetics
- Abstract
Breast cancers are complex cellular ecosystems where heterotypic interactions play central roles in disease progression and response to therapy. However, our knowledge of their cellular composition and organization is limited. Here we present a single-cell and spatially resolved transcriptomics analysis of human breast cancers. We developed a single-cell method of intrinsic subtype classification (SCSubtype) to reveal recurrent neoplastic cell heterogeneity. Immunophenotyping using cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) provides high-resolution immune profiles, including new PD-L1/PD-L2
+ macrophage populations associated with clinical outcome. Mesenchymal cells displayed diverse functions and cell-surface protein expression through differentiation within three major lineages. Stromal-immune niches were spatially organized in tumors, offering insights into antitumor immune regulation. Using single-cell signatures, we deconvoluted large breast cancer cohorts to stratify them into nine clusters, termed 'ecotypes', with unique cellular compositions and clinical outcomes. This study provides a comprehensive transcriptional atlas of the cellular architecture of breast cancer., (© 2021. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2021
- Full Text
- View/download PDF
145. Cross-tissue single-cell landscape of human monocytes and macrophages in health and disease.
- Author
-
Mulder K, Patel AA, Kong WT, Piot C, Halitzki E, Dunsmore G, Khalilnezhad S, Irac SE, Dubuisson A, Chevrier M, Zhang XM, Tam JKC, Lim TKH, Wong RMM, Pai R, Khalil AIS, Chow PKH, Wu SZ, Al-Eryani G, Roden D, Swarbrick A, Chan JKY, Albani S, Derosa L, Zitvogel L, Sharma A, Chen J, Silvin A, Bertoletti A, Blériot C, Dutertre CA, and Ginhoux F
- Subjects
- Arthritis, Rheumatoid immunology, COVID-19 immunology, Gene Expression genetics, Gene Expression Profiling, Humans, Interferon-gamma immunology, L-Amino Acid Oxidase metabolism, Liver Cirrhosis immunology, Macrophages immunology, Neoplasms immunology, RNA, Small Cytoplasmic genetics, Single-Cell Analysis, T-Lymphocytes, Regulatory immunology, Transcriptome immunology, Dendritic Cells immunology, Gene Expression immunology, Monocytes immunology, Transcriptome genetics, Tumor-Associated Macrophages immunology
- Abstract
Mononuclear phagocytes (MNPs) encompass dendritic cells, monocytes, and macrophages (MoMac), which exhibit antimicrobial, homeostatic, and immunoregulatory functions. We integrated 178,651 MNPs from 13 tissues across 41 datasets to generate a MNP single-cell RNA compendium (MNP-VERSE), a publicly available tool to map MNPs and define conserved gene signatures of MNP populations. Next, we generated a MoMac-focused compendium that revealed an array of specialized cell subsets widely distributed across multiple tissues. Specific pathological forms were expanded in cancer and inflammation. All neoplastic tissues contained conserved tumor-associated macrophage populations. In particular, we focused on IL4I1
+ CD274(PD-L1)+ IDO1+ macrophages, which accumulated in the tumor periphery in a T cell-dependent manner via interferon-γ (IFN-γ) and CD40/CD40L-induced maturation from IFN-primed monocytes. IL4I1_Macs exhibited immunosuppressive characteristics through tryptophan degradation and promoted the entry of regulatory T cell into tumors. This integrated analysis provides a robust online-available platform for uniform annotation and dissection of specific macrophage functions in healthy and pathological states., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
146. Atopic dermatitis microbiomes stratify into ecologic dermotypes enabling microbial virulence and disease severity.
- Author
-
Tay ASL, Li C, Nandi T, Chng KR, Andiappan AK, Mettu VS, de Cevins C, Ravikrishnan A, Dutertre CA, Wong XFCC, Ng AHQ, Matta SA, Ginhoux F, Rötzschke O, Chew FT, Tang MBY, Yew YW, Nagarajan N, and Common JEA
- Subjects
- Adolescent, Adult, Bacteria genetics, Bacteria pathogenicity, Biomarkers blood, Cytokines blood, Dermatitis, Atopic blood, Dermatitis, Atopic immunology, Dermatitis, Atopic metabolism, Female, Humans, Male, Middle Aged, Phenotype, Severity of Illness Index, Skin chemistry, Skin metabolism, Skin Tests, Virulence genetics, Water metabolism, Young Adult, Dermatitis, Atopic microbiology, Microbiota, Skin microbiology
- Abstract
Background: Atopic dermatitis (AD) is a common skin disease affecting up to 20% of the global population, with significant clinical heterogeneity and limited information about molecular subtypes and actionable biomarkers. Although alterations in the skin microbiome have been described in subjects with AD during progression to flare state, the prognostic value of baseline microbiome configurations has not been explored., Objective: Our aim was to identify microbial signatures on AD skin that are predictive of disease fate., Methods: Nonlesional skin of patients with AD and healthy control subjects were sampled at 2 time points separated by at least 4 weeks. Using whole metagenome analysis of skin microbiomes of patients with AD and control subjects (n = 49 and 189 samples), we identified distinct microbiome configurations (dermotypes A and B). Blood was collected for immunophenotyping, and skin surface samples were analyzed for correlations with natural moisturizing factors and antimicrobial peptides., Results: Dermotypes were robust and validated across 2 additional cohorts (63 individuals), with strong enrichment of subjects with AD in dermotype B. Dermotype B was characterized by reduced microbial richness, depletion of Cutibacterium acnes, Dermacoccus and Methylobacterium species, individual-specific outlier abundance of Staphylococcus species (eg, S epidermidis, S capitis, S aureus), and enrichment in metabolic pathways (eg, branched chain amino acids and arginine biosynthesis) and virulence genes (eg, β-toxin, δ-toxin) that defined a pathogenic ecology. Skin surface and circulating host biomarkers exhibited a distinct microbial-associated signature that was further reflected in more severe itching, frequent flares, and increased disease severity in patients harboring the dermotype B microbiome., Conclusion: We report distinct clusters of microbial profiles that delineate the role of microbiome configurations in AD heterogeneity, highlight a mechanism for ongoing inflammation, and provide prognostic utility toward microbiome-based disease stratification., (Copyright © 2020 American Academy of Allergy, Asthma & Immunology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
147. [Identification of circulating inflammatory dendritic cells by a multi-omic approach].
- Author
-
Dutertre CA and Ginhoux F
- Subjects
- Algorithms, Cell Separation methods, Dendritic Cells metabolism, Gene Expression Profiling, Humans, Inflammation genetics, Inflammation metabolism, Inflammation pathology, Lipopolysaccharide Receptors metabolism, Lupus Erythematosus, Systemic genetics, Lupus Erythematosus, Systemic metabolism, Lupus Erythematosus, Systemic pathology, Proteomics methods, Dendritic Cells pathology, Genomics methods, Inflammation immunology, Lupus Erythematosus, Systemic immunology
- Published
- 2020
- Full Text
- View/download PDF
148. Guidelines for the use of flow cytometry and cell sorting in immunological studies (second edition).
- Author
-
Cossarizza A, Chang HD, Radbruch A, Acs A, Adam D, Adam-Klages S, Agace WW, Aghaeepour N, Akdis M, Allez M, Almeida LN, Alvisi G, Anderson G, Andrä I, Annunziato F, Anselmo A, Bacher P, Baldari CT, Bari S, Barnaba V, Barros-Martins J, Battistini L, Bauer W, Baumgart S, Baumgarth N, Baumjohann D, Baying B, Bebawy M, Becher B, Beisker W, Benes V, Beyaert R, Blanco A, Boardman DA, Bogdan C, Borger JG, Borsellino G, Boulais PE, Bradford JA, Brenner D, Brinkman RR, Brooks AES, Busch DH, Büscher M, Bushnell TP, Calzetti F, Cameron G, Cammarata I, Cao X, Cardell SL, Casola S, Cassatella MA, Cavani A, Celada A, Chatenoud L, Chattopadhyay PK, Chow S, Christakou E, Čičin-Šain L, Clerici M, Colombo FS, Cook L, Cooke A, Cooper AM, Corbett AJ, Cosma A, Cosmi L, Coulie PG, Cumano A, Cvetkovic L, Dang VD, Dang-Heine C, Davey MS, Davies D, De Biasi S, Del Zotto G, Dela Cruz GV, Delacher M, Della Bella S, Dellabona P, Deniz G, Dessing M, Di Santo JP, Diefenbach A, Dieli F, Dolf A, Dörner T, Dress RJ, Dudziak D, Dustin M, Dutertre CA, Ebner F, Eckle SBG, Edinger M, Eede P, Ehrhardt GRA, Eich M, Engel P, Engelhardt B, Erdei A, Esser C, Everts B, Evrard M, Falk CS, Fehniger TA, Felipo-Benavent M, Ferry H, Feuerer M, Filby A, Filkor K, Fillatreau S, Follo M, Förster I, Foster J, Foulds GA, Frehse B, Frenette PS, Frischbutter S, Fritzsche W, Galbraith DW, Gangaev A, Garbi N, Gaudilliere B, Gazzinelli RT, Geginat J, Gerner W, Gherardin NA, Ghoreschi K, Gibellini L, Ginhoux F, Goda K, Godfrey DI, Goettlinger C, González-Navajas JM, Goodyear CS, Gori A, Grogan JL, Grummitt D, Grützkau A, Haftmann C, Hahn J, Hammad H, Hämmerling G, Hansmann L, Hansson G, Harpur CM, Hartmann S, Hauser A, Hauser AE, Haviland DL, Hedley D, Hernández DC, Herrera G, Herrmann M, Hess C, Höfer T, Hoffmann P, Hogquist K, Holland T, Höllt T, Holmdahl R, Hombrink P, Houston JP, Hoyer BF, Huang B, Huang FP, Huber JE, Huehn J, Hundemer M, Hunter CA, Hwang WYK, Iannone A, Ingelfinger F, Ivison SM, Jäck HM, Jani PK, Jávega B, Jonjic S, Kaiser T, Kalina T, Kamradt T, Kaufmann SHE, Keller B, Ketelaars SLC, Khalilnezhad A, Khan S, Kisielow J, Klenerman P, Knopf J, Koay HF, Kobow K, Kolls JK, Kong WT, Kopf M, Korn T, Kriegsmann K, Kristyanto H, Kroneis T, Krueger A, Kühne J, Kukat C, Kunkel D, Kunze-Schumacher H, Kurosaki T, Kurts C, Kvistborg P, Kwok I, Landry J, Lantz O, Lanuti P, LaRosa F, Lehuen A, LeibundGut-Landmann S, Leipold MD, Leung LYT, Levings MK, Lino AC, Liotta F, Litwin V, Liu Y, Ljunggren HG, Lohoff M, Lombardi G, Lopez L, López-Botet M, Lovett-Racke AE, Lubberts E, Luche H, Ludewig B, Lugli E, Lunemann S, Maecker HT, Maggi L, Maguire O, Mair F, Mair KH, Mantovani A, Manz RA, Marshall AJ, Martínez-Romero A, Martrus G, Marventano I, Maslinski W, Matarese G, Mattioli AV, Maueröder C, Mazzoni A, McCluskey J, McGrath M, McGuire HM, McInnes IB, Mei HE, Melchers F, Melzer S, Mielenz D, Miller SD, Mills KHG, Minderman H, Mjösberg J, Moore J, Moran B, Moretta L, Mosmann TR, Müller S, Multhoff G, Muñoz LE, Münz C, Nakayama T, Nasi M, Neumann K, Ng LG, Niedobitek A, Nourshargh S, Núñez G, O'Connor JE, Ochel A, Oja A, Ordonez D, Orfao A, Orlowski-Oliver E, Ouyang W, Oxenius A, Palankar R, Panse I, Pattanapanyasat K, Paulsen M, Pavlinic D, Penter L, Peterson P, Peth C, Petriz J, Piancone F, Pickl WF, Piconese S, Pinti M, Pockley AG, Podolska MJ, Poon Z, Pracht K, Prinz I, Pucillo CEM, Quataert SA, Quatrini L, Quinn KM, Radbruch H, Radstake TRDJ, Rahmig S, Rahn HP, Rajwa B, Ravichandran G, Raz Y, Rebhahn JA, Recktenwald D, Reimer D, Reis e Sousa C, Remmerswaal EBM, Richter L, Rico LG, Riddell A, Rieger AM, Robinson JP, Romagnani C, Rubartelli A, Ruland J, Saalmüller A, Saeys Y, Saito T, Sakaguchi S, Sala-de-Oyanguren F, Samstag Y, Sanderson S, Sandrock I, Santoni A, Sanz RB, Saresella M, Sautes-Fridman C, Sawitzki B, Schadt L, Scheffold A, Scherer HU, Schiemann M, Schildberg FA, Schimisky E, Schlitzer A, Schlosser J, Schmid S, Schmitt S, Schober K, Schraivogel D, Schuh W, Schüler T, Schulte R, Schulz AR, Schulz SR, Scottá C, Scott-Algara D, Sester DP, Shankey TV, Silva-Santos B, Simon AK, Sitnik KM, Sozzani S, Speiser DE, Spidlen J, Stahlberg A, Stall AM, Stanley N, Stark R, Stehle C, Steinmetz T, Stockinger H, Takahama Y, Takeda K, Tan L, Tárnok A, Tiegs G, Toldi G, Tornack J, Traggiai E, Trebak M, Tree TIM, Trotter J, Trowsdale J, Tsoumakidou M, Ulrich H, Urbanczyk S, van de Veen W, van den Broek M, van der Pol E, Van Gassen S, Van Isterdael G, van Lier RAW, Veldhoen M, Vento-Asturias S, Vieira P, Voehringer D, Volk HD, von Borstel A, von Volkmann K, Waisman A, Walker RV, Wallace PK, Wang SA, Wang XM, Ward MD, Ward-Hartstonge KA, Warnatz K, Warnes G, Warth S, Waskow C, Watson JV, Watzl C, Wegener L, Weisenburger T, Wiedemann A, Wienands J, Wilharm A, Wilkinson RJ, Willimsky G, Wing JB, Winkelmann R, Winkler TH, Wirz OF, Wong A, Wurst P, Yang JHM, Yang J, Yazdanbakhsh M, Yu L, Yue A, Zhang H, Zhao Y, Ziegler SM, Zielinski C, Zimmermann J, and Zychlinsky A
- Subjects
- Consensus, Humans, Phenotype, Allergy and Immunology standards, Cell Separation methods, Cell Separation standards, Flow Cytometry methods, Flow Cytometry standards
- Abstract
These guidelines are a consensus work of a considerable number of members of the immunology and flow cytometry community. They provide the theory and key practical aspects of flow cytometry enabling immunologists to avoid the common errors that often undermine immunological data. Notably, there are comprehensive sections of all major immune cell types with helpful Tables detailing phenotypes in murine and human cells. The latest flow cytometry techniques and applications are also described, featuring examples of the data that can be generated and, importantly, how the data can be analysed. Furthermore, there are sections detailing tips, tricks and pitfalls to avoid, all written and peer-reviewed by leading experts in the field, making this an essential research companion., (© 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2019
- Full Text
- View/download PDF
149. Apoptotic cell capture by DCs induces unexpectedly robust autologous CD4+ T-cell responses.
- Author
-
Valente M, Baey C, Louche P, Dutertre CA, Vimeux L, Marañón C, Hosmalin A, and Feuillet V
- Subjects
- Antigen Presentation, Autoantigens immunology, Cells, Cultured, Histocompatibility Antigens Class II immunology, Humans, Interleukin-17 immunology, Lipopolysaccharides immunology, Lymphocyte Activation, Th17 Cells immunology, Apoptosis immunology, CD4-Positive T-Lymphocytes immunology, Dendritic Cells immunology
- Abstract
Apoptotic cells represent an important source of self-antigens and their engulfment by dendritic cells (DCs) is usually considered to be related to tolerance induction. We report here an unexpectedly high level of human CD4(+) T-cell proliferation induced by autologous DCs loaded with autologous apoptotic cells, due to the activation of more than 10% of naive CD4(+) T cells. This proliferation is not due to an increase in the costimulatory capacity of DCs, but is dependent on apoptotic cell-associated material processed through an endo-lysosomal pathway and presented on DC MHC class II molecules. Autologous CD4(+) T cells stimulated with apoptotic cell-loaded DCs exhibit suppressive capacities. However, in the presence of bacterial lipopolysaccharide, apoptotic cell-loaded DCs induce the generation of IL-17-producing cells. Thus, apoptotic cell engulfment by DCs may lead to increased autologous responses, initially generating CD4(+) T cells with suppressive capacities able to differentiate into Th17 cells in the presence of a bacterial danger signal such as LPS., (© 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2014
- Full Text
- View/download PDF
150. [Antibodies: better knowledge for a better use].
- Author
-
Abès R, Dutertre CA, and Teillaud JL
- Subjects
- Animals, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal immunology, Antibody Specificity, Antigens, Neoplasm immunology, Antigens, Surface immunology, Complementarity Determining Regions chemistry, Complementarity Determining Regions immunology, Cytokines antagonists & inhibitors, Cytokines immunology, Drug Delivery Systems, Epitopes immunology, Humans, Immunoconjugates therapeutic use, Immunoglobulin Fab Fragments chemistry, Immunoglobulin Fab Fragments immunology, Immunoglobulin Fc Fragments chemistry, Immunoglobulin Fc Fragments immunology, Mice, Neoplasms immunology, Neoplasms therapy, Protein Engineering, Risk Assessment, Antibodies, Monoclonal therapeutic use, Antigen-Antibody Reactions immunology
- Abstract
The therapeutic use of monoclonal antibodies is growing exponentially. Our knowledge on antibody structure, in particular that of IgG1, largely used in the clinic, has progressed remarkably. However, some formidable challenges still remain to be confronted, among which the increase of a yet-limited antibody efficacy, the lowering of the frequency of serious clinical adverse events, and the establishment of pre-clinical models that can be reliably extrapolated to humans represent major goals. The selection of relevant target antigens with regard to the pathology to be treated and to the expected effects of the antibody used is also a critical parameter. Facing these challenges, the amazing molecular plasticity of antibodies, as well as new antibody engineering approaches based on the most recent insights on the structure and biology of antibodies and their targets represent areas of research that will make monoclonal antibodies remarkable drugs for human health in a near future.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.