53 results on '"Luche, H"'
Search Results
2. ROCK mediates dendritic cell migration within lymphatic capillaries under steady state and inflammation: 120
- Author
-
Nitschké, M., Aebischer, D., Lucic, M., Luche, H., Fehling, J., Biehlmaier, O., and Halin, C.
- Published
- 2012
3. P723: LOSS OF THE NK MOTIF AND ANKYRIN REPEAT DOMAIN 1 (KANK1) LEADS TO LYMPHOID COMPARTMENT DYSREGUATION IN MICE.
- Author
-
Almosailleakh, M., Narcisi, S., Roger Michel Côme, C., Luche, H., and Grønbæk, K.
- Published
- 2022
- Full Text
- View/download PDF
4. Incontinentia pigmenti underlies thymic dysplasia, autoantibodies to type I IFNs, and viral diseases.
- Author
-
Rosain J, Le Voyer T, Liu X, Gervais A, Polivka L, Cederholm A, Berteloot L, Parent AV, Pescatore A, Spinosa E, Minic S, Kiszewski AE, Tsumura M, Thibault C, Esnaola Azcoiti M, Martinovic J, Philippot Q, Khan T, Marchal A, Charmeteau-De Muylder B, Bizien L, Deswarte C, Hadjem L, Fauvarque MO, Dorgham K, Eriksson D, Falcone EL, Puel M, Ünal S, Geraldo A, Le Floc'h C, Li H, Rheault S, Muti C, Bobrie-Moyrand C, Welfringer-Morin A, Fuleihan RL, Lévy R, Roelens M, Gao L, Materna M, Pellegrini S, Piemonti L, Catherinot E, Goffard JC, Fekkar A, Sacko-Sow A, Soudée C, Boucherit S, Neehus AL, Has C, Hübner S, Blanchard-Rohner G, Amador-Borrero B, Utsumi T, Taniguchi M, Tani H, Izawa K, Yasumi T, Kanai S, Migaud M, Aubart M, Lambert N, Gorochov G, Picard C, Soudais C, L'Honneur AS, Rozenberg F, Milner JD, Zhang SY, Vabres P, Trpinac D, Marr N, Boddaert N, Desguerre I, Pasparakis M, Miller CN, Poziomczyk CS, Abel L, Okada S, Jouanguy E, Cheynier R, Zhang Q, Cobat A, Béziat V, Boisson B, Steffann J, Fusco F, Ursini MV, Hadj-Rabia S, Bodemer C, Bustamante J, Luche H, Puel A, Courtois G, Bastard P, Landegren N, Anderson MS, and Casanova JL
- Subjects
- Humans, Female, Child, Child, Preschool, Virus Diseases immunology, Infant, Adult, Adolescent, Young Adult, Interferon Type I immunology, Interferon Type I metabolism, Autoantibodies immunology, Thymus Gland immunology, Thymus Gland pathology, Incontinentia Pigmenti immunology, Incontinentia Pigmenti genetics, Incontinentia Pigmenti pathology, I-kappa B Kinase genetics, I-kappa B Kinase immunology
- Abstract
Human inborn errors of thymic T cell tolerance underlie the production of autoantibodies (auto-Abs) neutralizing type I IFNs, which predispose to severe viral diseases. We analyze 131 female patients with X-linked dominant incontinentia pigmenti (IP), heterozygous for loss-of-function (LOF) NEMO variants, from 99 kindreds in 10 countries. Forty-seven of these patients (36%) have auto-Abs neutralizing IFN-α and/or IFN-ω, a proportion 23 times higher than that for age-matched female controls. This proportion remains stable from the age of 6 years onward. On imaging, female patients with IP have a small, abnormally structured thymus. Auto-Abs against type I IFNs confer a predisposition to life-threatening viral diseases. By contrast, patients with IP lacking auto-Abs against type I IFNs are at no particular risk of viral disease. These results suggest that IP accelerates thymic involution, thereby underlying the production of auto-Abs neutralizing type I IFNs in at least a third of female patients with IP, predisposing them to life-threatening viral diseases., (© 2024 Rosain et al.)
- Published
- 2024
- Full Text
- View/download PDF
5. SMAC mimetic drives microglia phenotype and glioblastoma immune microenvironment.
- Author
-
Snacel-Fazy E, Soubéran A, Grange M, Joseph K, Colin C, Morando P, Luche H, Pagano A, Brustlein S, Debarbieux F, Toutain S, Siret C, van de Pavert SA, Rougon G, Figarella-Branger D, Ravi VM, Tabouret E, and Tchoghandjian A
- Subjects
- Animals, Humans, Mice, Brain Neoplasms immunology, Brain Neoplasms pathology, Apoptosis Regulatory Proteins metabolism, Mice, Inbred C57BL, Mitochondrial Proteins metabolism, Cell Line, Tumor, Tumor-Associated Macrophages immunology, Tumor-Associated Macrophages metabolism, Tumor-Associated Macrophages drug effects, Intracellular Signaling Peptides and Proteins metabolism, Mice, Transgenic, Glioblastoma immunology, Glioblastoma pathology, Tumor Microenvironment drug effects, Tumor Microenvironment immunology, Microglia drug effects, Microglia metabolism, Microglia immunology, Phenotype
- Abstract
Tumor-associated macrophages/microglia (TAMs) are highly plastic and heterogeneous immune cells that can be immune-supportive or tumor-supportive depending of the microenvironment. TAMs are the most abundant immune cells in glioblastoma (GB), and play a key role in immunosuppression. Therefore, TAMs reprogramming toward immune-supportive cells is a promising strategy to overcome immunosuppression. By leveraging scRNAseq human GB databases, we identified that Inhibitor of Apoptosis Proteins (IAP) were expressed by TAMs. To investigate their role in TAMs-related immunosuppression, we antagonized IAP using the central nervous system permeant SMAC mimetic GDC-0152 (SMg). On explants and cultured immune cells isolated from human GB samples, SMg modified TAMs activity. We showed that SMg treatment promoted microglia pro-apoptotic and anti-tumoral function via caspase-3 pro-inflammatory cleavage and the inhibition of tumoroids growth. Then we designed a relevant immunogenic mouse GB model to decipher the spatio-temporal densities, distribution, phenotypes and function of TAMs with or without SMg treatment. We used 3D imaging techniques, a transgenic mouse with fluorescent TAM subsets and mass cytometry. We confirmed that SMg promoted microglia activation, antigen-presenting function and tumor infiltration. In addition, we observed a remodeling of blood vessels, a decrease in anti-inflammatory macrophages and an increased level of monocytes and their mo-DC progeny. This remodeling of the TAM landscape is associated with an increase in CD8 T cell density and activation. Altogether, these results demonstrated that SMg drives the immunosuppressive basal microglia toward an active phenotype with pro-apoptotic and anti-tumoral function and modifies the GB immune landscape. This identifies IAP as targets of choice for a potential mechanism-based therapeutic strategy and SMg as a promising molecule for this application., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. CD5L as a promising biological therapeutic for treating sepsis.
- Author
-
Oliveira L, Silva MC, Gomes AP, Santos RF, Cardoso MS, Nóvoa A, Luche H, Cavadas B, Amorim I, Gärtner F, Malissen B, Mallo M, and Carmo AM
- Subjects
- Animals, Mice, Cecum surgery, Chemokine CXCL1 metabolism, Chemokine CXCL1 genetics, Disease Models, Animal, Ligation, Lipopolysaccharides, Neutrophil Infiltration drug effects, Phagocytosis, Pore Forming Cytotoxic Proteins metabolism, Recombinant Proteins therapeutic use, Recombinant Proteins administration & dosage, Shock, Septic immunology, Mice, Inbred C57BL, Mice, Knockout, Neutrophils immunology, Neutrophils metabolism, Sepsis immunology, Sepsis drug therapy, Apoptosis Regulatory Proteins therapeutic use, Receptors, Scavenger therapeutic use
- Abstract
Sepsis results from systemic, dysregulated inflammatory responses to infection, culminating in multiple organ failure. Here, we demonstrate the utility of CD5L for treating experimental sepsis caused by cecal ligation and puncture (CLP). We show that CD5L's important features include its ability to enhance neutrophil recruitment and activation by increasing circulating levels of CXCL1, and to promote neutrophil phagocytosis. CD5L-deficient mice exhibit impaired neutrophil recruitment and compromised bacterial control, rendering them susceptible to attenuated CLP. CD5L
-/- peritoneal cells from mice subjected to medium-grade CLP exhibit a heightened pro-inflammatory transcriptional profile, reflecting a loss of control of the immune response to the infection. Intravenous administration of recombinant CD5L (rCD5L) in immunocompetent C57BL/6 wild-type (WT) mice significantly ameliorates measures of disease in the setting of high-grade CLP-induced sepsis. Furthermore, rCD5L lowers endotoxin and damage-associated molecular pattern (DAMP) levels, and protects WT mice from LPS-induced endotoxic shock. These findings warrant the investigation of rCD5L as a possible treatment for sepsis in humans., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
7. The immunopathological landscape of human pre-TCRα deficiency: From rare to common variants.
- Author
-
Materna M, Delmonte OM, Bosticardo M, Momenilandi M, Conrey PE, Charmeteau-De Muylder B, Bravetti C, Bellworthy R, Cederholm A, Staels F, Ganoza CA, Darko S, Sayed S, Le Floc'h C, Ogishi M, Rinchai D, Guenoun A, Bolze A, Khan T, Gervais A, Krüger R, Völler M, Palterer B, Sadeghi-Shabestari M, Langlois de Septenville A, Schramm CA, Shah S, Tello-Cajiao JJ, Pala F, Amini K, Campos JS, Lima NS, Eriksson D, Lévy R, Seeleuthner Y, Jyonouchi S, Ata M, Al Ali F, Stittrich A, Deswarte C, Pereira A, Mégret J, Le Voyer T, Bastard P, Berteloot L, Dussiot M, Vladikine N, Cardenas PP, Jouanguy E, Alqahtani M, Hasan A, Thanaraj TA, Rosain J, Al Qureshah F, Sabato V, Alyanakian MA, Leruez-Ville M, Rozenberg F, Haddad E, Regueiro JR, Toribio ML, Kelsen JR, Salehi M, Nasiri S, Torabizadeh M, Rokni-Zadeh H, Changi-Ashtiani M, Vatandoost N, Moravej H, Akrami SM, Mazloomrezaei M, Cobat A, Meyts I, Toyofuku E, Nishimura M, Moriya K, Mizukami T, Imai K, Abel L, Malissen B, Al-Mulla F, Alkuraya FS, Parvaneh N, von Bernuth H, Beetz C, Davi F, Douek DC, Cheynier R, Langlais D, Landegren N, Marr N, Morio T, Shahrooei M, Schrijvers R, Henrickson SE, Luche H, Notarangelo LD, Casanova JL, and Béziat V
- Subjects
- Humans, Cell Differentiation, Homozygote, Loss of Function Mutation, Lymphocyte Count, Alleles, Infections immunology, Lymphoproliferative Disorders immunology, Pedigree, Male, Female, Middle Aged, Aged, Aged, 80 and over, Autoimmunity genetics, Intraepithelial Lymphocytes immunology, Receptors, Antigen, T-Cell, alpha-beta genetics, Membrane Glycoproteins genetics
- Abstract
We describe humans with rare biallelic loss-of-function PTCRA variants impairing pre-α T cell receptor (pre-TCRα) expression. Low circulating naive αβ T cell counts at birth persisted over time, with normal memory αβ and high γδ T cell counts. Their TCRα repertoire was biased, which suggests that noncanonical thymic differentiation pathways can rescue αβ T cell development. Only a minority of these individuals were sick, with infection, lymphoproliferation, and/or autoimmunity. We also report that 1 in 4000 individuals from the Middle East and South Asia are homozygous for a common hypomorphic PTCRA variant. They had normal circulating naive αβ T cell counts but high γδ T cell counts. Although residual pre-TCRα expression drove the differentiation of more αβ T cells, autoimmune conditions were more frequent in these patients compared with the general population.
- Published
- 2024
- Full Text
- View/download PDF
8. A novel mass cytometry protocol optimized for immunophenotyping of low-frequency antigen-specific T cells.
- Author
-
Balz K, Grange M, Pegel U, Karamya ZA, Mello M, Zhou X, Berger T, Bloch K, Dunham D, Chinthrajah S, Nadeau K, Luche H, and Skevaki C
- Subjects
- Humans, Animals, Mice, Flow Cytometry methods, Immunophenotyping, Staining and Labeling, CD8-Positive T-Lymphocytes, Antigens, T-Lymphocytes
- Abstract
Understanding antigen-specific T-cell responses, for example, following virus infections or allergen exposure, is of high relevance for the development of vaccines and therapeutics. We aimed on optimizing immunophenotyping of T cells after antigen stimulation by improving staining procedures for flow and mass cytometry. Our method can be used for primary cells of both mouse and human origin for the detection of low-frequency T-cell response using a dual-barcoding system for individual samples and conditions. First, live-cell barcoding was performed using anti-CD45 antibodies prior to an in vitro T-cell stimulation assay. Second, to discriminate between stimulation conditions and prevent cell loss, sample barcoding was combined with a commercial barcoding solution. This dual-barcoding approach is cell sparing and, therefore, particularly relevant for samples with low cell numbers. To further reduce cell loss and to increase debarcoding efficiency of multiplexed samples, we combined our dual-barcoding approach with a new centrifugation-free washing system by laminar flow (Curiox™). Finally, to demonstrate the benefits of our established protocol, we assayed virus-specific T-cell response in SARS-CoV-2-vaccinated and SARS-CoV-2-infected patients and compared with healthy non-exposed individuals by a high-parameter CyTOF analysis. We could reveal a heterogeneity of phenotypes among responding CD4, CD8, and gd-T cells following antigen-specific stimulations. Our protocol allows to assay antigen-specific responses of minute populations of T cells to virus-derived peptides, allergens, or other antigens from the same donor sample, in order to investigate qualitative and quantitative differences., Competing Interests: For CS: Consultancy and research funding, Bencard Allergie and Thermo Fisher Scientific; Research Funding, Mead Johnson Nutrition MJN., (Copyright © 2024 Balz, Grange, Pegel, Karamya, Mello, Zhou, Berger, Bloch, Dunham, Chinthrajah, Nadeau, Luche and Skevaki.)
- Published
- 2024
- Full Text
- View/download PDF
9. Loss of the KN Motif and AnKyrin Repeat Domain 1 (KANK1) Leads to Lymphoid Compartment Dysregulation in Murine Model.
- Author
-
Almosailleakh M, Bentivegna S, Narcisi S, Benquet SJ, Gillberg L, Montaño-Almendras CP, Savickas S, Schoof EM, Wegener A, Luche H, Jensen HE, Côme C, and Grønbæk K
- Subjects
- Animals, Humans, Mice, Ankyrin Repeat genetics, Cytoskeletal Proteins metabolism, Disease Models, Animal, Adaptor Proteins, Signal Transducing metabolism, Neoplasms
- Abstract
The KN Motif and AnKyrin Repeat Domain 1 ( KANK1 ) is proposed as a tumour suppressor gene, as its expression is reduced or absent in several types of tumour tissue, and over-expressing the protein inhibited the proliferation of tumour cells in solid cancer models. We report a novel germline loss of heterozygosity mutation encompassing the KANK1 gene in a young patient diagnosed with myelodysplastic neoplasm (MDS) with no additional disease-related genomic aberrations. To study the potential role of KANK1 in haematopoiesis, we generated a new transgenic mouse model with a confirmed loss of KANK1 expression. KANK1 knockout mice did not develop any haematological abnormalities; however, the loss of its expression led to alteration in the colony forming and proliferative potential of bone marrow (BM) cells and a decrease in hematopoietic stem and progenitor cells (HSPCs) population frequency. A comprehensive marker expression analysis of lineage cell populations indicated a role for Kank1 in lymphoid cell development, and total protein analysis suggests the involvement of Kank1 in BM cells' cytoskeleton formation and mobility.
- Published
- 2023
- Full Text
- View/download PDF
10. Anti-HVEM mAb therapy improves antitumoral immunity both in vitro and in vivo, in a novel transgenic mouse model expressing human HVEM and BTLA molecules challenged with HVEM expressing tumors.
- Author
-
Demerlé C, Gorvel L, Mello M, Pastor S, Degos C, Zarubica A, Angelis F, Fiore F, Nunes JA, Malissen B, Greillier L, Guittard G, Luche H, Barlesi F, and Olive D
- Subjects
- Animals, Humans, Mice, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal therapeutic use, Mice, Inbred C57BL, Mice, Transgenic, Receptors, Immunologic metabolism, CD8-Positive T-Lymphocytes metabolism, Neoplasms, Receptors, Tumor Necrosis Factor, Member 14 immunology, Receptors, Tumor Necrosis Factor, Member 14 metabolism
- Abstract
Background: Tumor necrosis factor superfamily member 14 (TNFRSF14)/herpes virus entry mediator (HVEM) is the ligand for B and T lymphocyte attenuator (BTLA) and CD160-negative immune co-signaling molecules as well as viral proteins. Its expression is dysregulated with an overexpression in tumors and a connection with tumors of adverse prognosis., Methods: We developed C57BL/6 mouse models co-expressing human (hu)BTLA and huHVEM as well as antagonistic monoclonal antibodies (mAbs) that completely prevent the interactions of HVEM with its ligands., Results: Here, we show that the anti-HVEM18-10 mAb increases primary human αβ-T cells activity alone (CIS-activity) or in the presence of HVEM-expressing lung or colorectal cancer cells in vitro (TRANS-activity). Anti-HVEM18-10 synergizes with antiprogrammed death-ligand 1 (anti-PD-L1) mAb to activate T cells in the presence of PD-L1-positive tumors, but is sufficient to trigger T cell activation in the presence of PD-L1-negative cells. In order to better understand HVEM18-10 effects in vivo and especially disentangle its CIS and TRANS effects, we developed a knockin (KI) mouse model expressing human BTLA (huBTLA
+/+ ) and a KI mouse model expressing both huBTLA+/+ /huHVEM+/+ (double KI (DKI)). In vivo preclinical experiments performed in both mouse models showed that HVEM18-10 treatment was efficient to decrease human HVEM+ tumor growth. In the DKI model, anti-HVEM18-10 treatment induces a decrease of exhausted CD8+ T cells and regulatory T cells and an increase of effector memory CD4+ T cells within the tumor. Interestingly, mice which completely rejected tumors (±20%) did not develop tumors on rechallenge in both settings, therefore showing a marked T cell-memory phenotype effect., Conclusions: Altogether, our preclinical models validate anti-HVEM18-10 as a promising therapeutic antibody to use in clinics as a monotherapy or in combination with existing immunotherapies (antiprogrammed cell death protein 1/anti-PD-L1/anti-cytotoxic T-lymphocyte antigen-4 (CTLA-4))., Competing Interests: Competing interests: DO is a cofounder and shareholder of Imcheck Therapeutics, Alderaan Biotechnology, Emergence Therapeutics, and Stealth IO. HL is a co-founder and scientific advisor of JC discovery. FB reports payment or honoraria for lectures, presentations, speaker bureaus, manuscript writing, or educational events from AstraZeneca, Bayer, Bristol-Myers Squibb, Boehringer Ingelheim, Eli Lilly Oncology, F Hoffmann-La Roche, Novartis, Merck, MSD, Pierre Fabre, Pfizer, and Takeda, outside the submitted work. The other authors do not declare any conflict of interest., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2023
- Full Text
- View/download PDF
11. High-throughput mass cytometry staining for deep phenotyping of human natural killer cells.
- Author
-
Ben Amara A, Rouviere MS, Fattori S, Wlosik J, Gregori E, Boucherit N, Bernard PL, Nunès JA, Vey N, Luche H, Gorvel L, Olive D, and Chretien AS
- Subjects
- Humans, Flow Cytometry methods, Antibodies, Staining and Labeling, Leukocytes, Mononuclear, Killer Cells, Natural
- Abstract
This protocol details the step-by-step procedure for in-depth immune phenotyping of peripheral blood natural killer (NK) cells from clinical samples by mass cytometry. The protocol consists of three main steps: PBMC incubation with a mix of metal-conjugated antibodies for extracellular phenotyping followed by fixation, permeabilization and incubation with a mix of metal-conjugated antibodies for staining of intracellular proteins, and sample acquisition on a mass cytometer. High-dimensional analysis enables the visualization of NK cell subsets and their phenotypical characteristics. For complete details on the use and execution of this protocol, please refer to Chretien et al. (2021)., Competing Interests: Declaration of interests E.G. is currently employed by Fluidigm. D.O. is a cofounder and shareholder of Imcheck Therapeutics, Alderaan Biotechnology, Emergence Therapeutics, and Stealth IO., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
12. Single-cell transcriptomics uncovers an instructive T-cell receptor role in adult γδ T-cell lineage commitment.
- Author
-
Scaramuzzino S, Potier D, Ordioni R, Grenot P, Payet-Bornet D, Luche H, and Malissen B
- Subjects
- Animals, Cell Proliferation genetics, Cells, Cultured, Female, Male, Mice, Mice, Inbred C57BL, Receptors, Antigen, T-Cell, alpha-beta genetics, Signal Transduction genetics, CD4-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes metabolism, Cell Lineage genetics, Receptors, Antigen, T-Cell, gamma-delta genetics, Transcriptome genetics
- Abstract
After entering the adult thymus, bipotent T-cell progenitors give rise to αβ or γδ T cells. To determine whether the γδ T-cell receptor (TCR) has an instructive role in γδ T-cell lineage commitment or only "confirms" a pre-established γδ Τ-cell lineage state, we exploited mice lacking expression of LAT, an adaptor required for γδ TCR signaling. Although these mice showed a T-cell development block at the CD4
- CD8- double-negative third (DN3) stage, 0.3% of their DN3 cells expressed intermediate levels of γδ TCR (further referred to as γδint ) at their surface. Single-cell transcriptomics of LAT-deficient DN3 γδint cells demonstrated no sign of commitment to the γδ T-cell lineage, apart from γδ TCR expression. Although the lack of LAT is thought to tightly block DN3 cell development, we unexpectedly found that 25% of LAT-deficient DN3 γδint cells were actively proliferating and progressed up to the DN4 stage. However, even those cells failed to turn on the transcriptional program associated with the γδ T-cell lineage. Therefore, the γδ TCR-LAT signaling axis builds upon a γδ T-cell uncommitted lineage state to fully instruct adult γδ T-cell lineage specification., (© 2022 The Authors. Published under the terms of the CC BY 4.0 license.)- Published
- 2022
- Full Text
- View/download PDF
13. Conditional Deletion of Pdcd1 Identifies the Cell-Intrinsic Action of PD-1 on Functional CD8 T Cell Subsets for Antitumor Efficacy.
- Author
-
Raghavan S, Tovbis-Shifrin N, Kochel C, Sawant A, Mello M, Sathe M, Blumenschein W, Muise ES, Chackerian A, Pinheiro EM, Rosahl TW, Luche H, and de Waal Malefyt R
- Subjects
- Animals, CD8-Positive T-Lymphocytes drug effects, Female, Immune Checkpoint Inhibitors pharmacology, Lymphocytes, Tumor-Infiltrating drug effects, Mice, Mice, Inbred C57BL, Mice, Knockout, Programmed Cell Death 1 Receptor deficiency, CD8-Positive T-Lymphocytes immunology, Lymphocytes, Tumor-Infiltrating immunology, Neoplasms, Experimental immunology, Programmed Cell Death 1 Receptor immunology, T-Lymphocyte Subsets immunology
- Abstract
Programmed cell death-1 (PD-1) blockade has a profound effect on the ability of the immune system to eliminate tumors, but many questions remain about the cell types involved and the underlying mechanisms of immune activation. To shed some light on this, the cellular and molecular events following inhibition of PD-1 signaling was investigated in the MC-38 colon carcinoma model using constitutive (PD-1 KO) and conditional (PD1cKO) mice and in wild-type mice treated with PD-1 antibody. The impact on both tumor growth and the development of tumor immunity was assessed. In the PD-1cKO mice, a complete deletion of Pdcd1 in tumor-infiltrating T cells (TILs) after tamoxifen treatment led to the inhibition of tumor growth of both small and large tumors. Extensive immune phenotypic analysis of the TILs by flow and mass cytometry identified 20-different T cell subsets of which specifically 5-CD8 positive ones expanded in all three models after PD-1 blockade. All five subsets expressed granzyme B and interferon gamma (IFNγ). Gene expression analysis of the tumor further supported the phenotypic analysis in both PD-1cKO- and PD-1 Ab-treated mice and showed an upregulation of pathways related to CD4 and CD8 T-cell activation, enhanced signaling through costimulatory molecules and IFNγ, and non-T-cell processes. Altogether, using PD-1cKO mice, we define the intrinsic nature of PD-1 suppression of CD8 T-cell responses in tumor immunity., Competing Interests: Authors SR, NT-S, AS, MS, EP and AC were employed by Merck & Co., Inc., Kenilworth, NJ, USA. CK and RW were employed by Merck & Co., Inc., Kenilworth, NJ, USA at the time of the study and are currently employed by Synthekine. WB, EM, and TR are currently employed by Merck & Co., Inc., Kenilworth, NJ, USA., (Copyright © 2021 Raghavan, Tovbis-Shifrin, Kochel, Sawant, Mello, Sathe, Blumenschein, Muise, Chackerian, Pinheiro, Rosahl, Luche and de Waal Malefyt.)
- Published
- 2021
- Full Text
- View/download PDF
14. High-dimensional mass cytometry analysis of NK cell alterations in AML identifies a subgroup with adverse clinical outcome.
- Author
-
Chretien AS, Devillier R, Granjeaud S, Cordier C, Demerle C, Salem N, Wlosik J, Orlanducci F, Gorvel L, Fattori S, Hospital MA, Pakradouni J, Gregori E, Paul M, Rochigneux P, Pagliardini T, Morey M, Fauriat C, Dulphy N, Toubert A, Luche H, Malissen M, Blaise D, Nunès JA, Vey N, and Olive D
- Subjects
- Antigens, CD immunology, Humans, Immunophenotyping, Lymphocyte Activation immunology, Remission Induction, Treatment Outcome, Flow Cytometry methods, Killer Cells, Natural immunology, Leukemia, Myeloid, Acute immunology, Leukemia, Myeloid, Acute pathology
- Abstract
Natural killer (NK) cells are major antileukemic immune effectors. Leukemic blasts have a negative impact on NK cell function and promote the emergence of phenotypically and functionally impaired NK cells. In the current work, we highlight an accumulation of CD56
- CD16+ unconventional NK cells in acute myeloid leukemia (AML), an aberrant subset initially described as being elevated in patients chronically infected with HIV-1. Deep phenotyping of NK cells was performed using peripheral blood from patients with newly diagnosed AML ( n = 48, HEMATOBIO cohort, NCT02320656) and healthy subjects ( n = 18) by mass cytometry. We showed evidence of a moderate to drastic accumulation of CD56- CD16+ unconventional NK cells in 27% of patients. These NK cells displayed decreased expression of NKG2A as well as the triggering receptors NKp30 and NKp46, in line with previous observations in HIV-infected patients. High-dimensional characterization of these NK cells highlighted a decreased expression of three additional major triggering receptors required for NK cell activation, NKG2D, DNAM-1, and CD96. A high proportion of CD56- CD16+ NK cells at diagnosis was associated with an adverse clinical outcome and decreased overall survival (HR = 0.13; P = 0.0002) and event-free survival (HR = 0.33; P = 0.018) and retained statistical significance in multivariate analysis. Pseudotime analysis of the NK cell compartment highlighted a disruption of the maturation process, with a bifurcation from conventional NK cells toward CD56- CD16+ NK cells. Overall, our data suggest that the accumulation of CD56- CD16+ NK cells may be the consequence of immune escape from innate immunity during AML progression., Competing Interests: The authors declare no competing interest., (Copyright © 2021 the Author(s). Published by PNAS.)- Published
- 2021
- Full Text
- View/download PDF
15. LymphoAtlas: a dynamic and integrated phosphoproteomic resource of TCR signaling in primary T cells reveals ITSN2 as a regulator of effector functions.
- Author
-
Locard-Paulet M, Voisinne G, Froment C, Goncalves Menoita M, Ounoughene Y, Girard L, Gregoire C, Mori D, Martinez M, Luche H, Garin J, Malissen M, Burlet-Schiltz O, Malissen B, Gonzalez de Peredo A, and Roncagalli R
- Subjects
- Animals, Antibodies pharmacology, CD4-Positive T-Lymphocytes immunology, Chromatography, Liquid, Computational Biology, Gene Expression Regulation drug effects, Gene Expression Regulation genetics, Gene Expression Regulation immunology, Lymphocyte Activation drug effects, Lymphocyte Activation immunology, Mice, Mice, Inbred C57BL, Phosphorylation, Protein Biosynthesis drug effects, Protein Biosynthesis genetics, Protein Biosynthesis immunology, Signal Transduction immunology, Tandem Mass Spectrometry, Time Factors, Adaptor Proteins, Vesicular Transport metabolism, CD4-Positive T-Lymphocytes drug effects, Phosphoproteins metabolism, Protein Kinases metabolism, Proteomics, Receptors, Antigen, T-Cell metabolism, Signal Transduction genetics
- Abstract
T-cell receptor (TCR) ligation-mediated protein phosphorylation regulates the activation, cellular responses, and fates of T cells. Here, we used time-resolved high-resolution phosphoproteomics to identify, quantify, and characterize the phosphorylation dynamics of thousands of phosphorylation sites in primary T cells during the first 10 min after TCR stimulation. Bioinformatic analysis of the data revealed a coherent orchestration of biological processes underlying T-cell activation. In particular, functional modules associated with cytoskeletal remodeling, transcription, translation, and metabolic processes were mobilized within seconds after TCR engagement. Among proteins whose phosphorylation was regulated by TCR stimulation, we demonstrated, using a fast-track gene inactivation approach in primary lymphocytes, that the ITSN2 adaptor protein regulated T-cell effector functions. This resource, called LymphoAtlas, represents an integrated pipeline to further decipher the organization of the signaling network encoding T-cell activation. LymphoAtlas is accessible to the community at: https://bmm-lab.github.io/LymphoAtlas., (© 2020 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2020
- Full Text
- View/download PDF
16. Quantitative interactomics in primary T cells unveils TCR signal diversification extent and dynamics.
- Author
-
Voisinne G, Kersse K, Chaoui K, Lu L, Chaix J, Zhang L, Goncalves Menoita M, Girard L, Ounoughene Y, Wang H, Burlet-Schiltz O, Luche H, Fiore F, Malissen M, Gonzalez de Peredo A, Liang Y, Roncagalli R, and Malissen B
- Subjects
- Animals, CD4-Positive T-Lymphocytes metabolism, Chromatography, Affinity methods, Mass Spectrometry methods, Mice, Mice, Transgenic, Primary Cell Culture, Protein Interaction Mapping methods, Receptors, Antigen, T-Cell immunology, Signal Transduction genetics, CD4-Positive T-Lymphocytes immunology, Protein Interaction Maps immunology, Receptors, Antigen, T-Cell metabolism, Signal Transduction immunology
- Abstract
The activation of T cells by the T cell antigen receptor (TCR) results in the formation of signaling protein complexes (signalosomes), the composition of which has not been analyzed at a systems level. Here, we isolated primary CD4
+ T cells from 15 gene-targeted mice, each expressing one tagged form of a canonical protein of the TCR-signaling pathway. Using affinity purification coupled with mass spectrometry, we analyzed the composition and dynamics of the signalosomes assembling around each of the tagged proteins over 600 s of TCR engagement. We showed that the TCR signal-transduction network comprises at least 277 unique proteins involved in 366 high-confidence interactions, and that TCR signals diversify extensively at the level of the plasma membrane. Integrating the cellular abundance of the interacting proteins and their interaction stoichiometry provided a quantitative and contextual view of each documented interaction, permitting anticipation of whether ablation of a single interacting protein can impinge on the whole TCR signal-transduction network.- Published
- 2019
- Full Text
- View/download PDF
17. 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
18. Diversity of innate immune cell subsets across spatial and temporal scales in an EAE mouse model.
- Author
-
Caravagna C, Jaouën A, Desplat-Jégo S, Fenrich KK, Bergot E, Luche H, Grenot P, Rougon G, Malissen M, and Debarbieux F
- Subjects
- Animals, Dendritic Cells pathology, Encephalomyelitis, Autoimmune, Experimental genetics, Encephalomyelitis, Autoimmune, Experimental pathology, Leukocytes pathology, Mice, Mice, Transgenic, Microglia pathology, Multiple Sclerosis genetics, Multiple Sclerosis immunology, Multiple Sclerosis pathology, Spinal Cord pathology, Dendritic Cells immunology, Encephalomyelitis, Autoimmune, Experimental immunology, Immunity, Innate, Leukocytes immunology, Microglia immunology, Spinal Cord immunology
- Abstract
In both multiple sclerosis and its model experimental autoimmune encephalomyelitis (EAE), the extent of resident microglia activation and infiltration of monocyte-derived cells to the CNS is positively correlated to tissue damage. To address the phenotype characterization of different cell subsets, their spatio-temporal distributions and contributions to disease development we induced EAE in Thy1-CFP//LysM-EGFP//CD11c-EYFP reporter mice. We combined high content flow cytometry, immunofluorescence and two-photon imaging in live mice and identified a stepwise program of inflammatory cells accumulation. First on day 10 after induction, EGFP
+ neutrophils and monocytes invade the spinal cord parenchyma through the meninges rather than by extravasion. This event occurs just before axonal losses in the white matter. Once in the parenchyma, monocytes mature into EGFP+ /EYFP+ monocyte-derived dendritic cells (moDCs) whose density is maximal on day 17 when the axonal degradation and clinical signs stabilize. Meanwhile, microglia is progressively activated in the grey matter and subsequently recruited to plaques to phagocyte axon debris. LysM-EGFP//CD11c-EYFP mice appear as a powerful tool to differentiate moDCs from macrophages and to study the dynamics of immune cell maturation and phenotypic evolution in EAE.- Published
- 2018
- Full Text
- View/download PDF
19. In Utero Administration of Drugs Targeting Microglia Improves the Neurodevelopmental Outcome Following Cytomegalovirus Infection of the Rat Fetal Brain.
- Author
-
Cloarec R, Bauer S, Teissier N, Schaller F, Luche H, Courtens S, Salmi M, Pauly V, Bois E, Pallesi-Pocachard E, Buhler E, Michel FJ, Gressens P, Malissen M, Stamminger T, Streblow DN, Bruneau N, and Szepetowski P
- Abstract
Congenital cytomegalovirus (CMV) infections represent one leading cause of neurodevelopmental disorders. Recently, we reported on a rat model of CMV infection of the developing brain in utero , characterized by early and prominent infection and alteration of microglia-the brain-resident mononuclear phagocytes. Besides their canonical function against pathogens, microglia are also pivotal to brain development. Here we show that CMV infection of the rat fetal brain recapitulated key postnatal phenotypes of human congenital CMV including increased mortality, sensorimotor impairment reminiscent of cerebral palsy, hearing defects, and epileptic seizures. The possible influence of early microglia alteration on those phenotypes was then questioned by pharmacological targeting of microglia during pregnancy. One single administration of clodronate liposomes in the embryonic brains at the time of CMV injection to deplete microglia, and maternal feeding with doxycyxline throughout pregnancy to modify microglia in the litters' brains, were both associated with dramatic improvements of survival, body weight gain, sensorimotor development and with decreased risk of epileptic seizures. Improvement of microglia activation status did not persist postnatally after doxycycline discontinuation; also, active brain infection remained unchanged by doxycycline. Altogether our data indicate that early microglia alteration, rather than brain CMV load per se , is instrumental in influencing survival and the neurological outcomes of CMV-infected rats, and suggest that microglia might participate in the neurological outcome of congenital CMV in humans. Furthermore this study represents a first proof-of-principle for the design of microglia-targeted preventive strategies in the context of congenital CMV infection of the brain.
- Published
- 2018
- Full Text
- View/download PDF
20. Atp6ap2 ablation in adult mice impairs viability through multiple organ deficiencies.
- Author
-
Wendling O, Champy MF, Jaubert S, Pavlovic G, Dubos A, Lindner L, Jacobs H, Mark M, Combe R, Da Cruz IG, Luche H, Mudgett JS, Rosahl T, Sorg T, Malissen M, Reilly PT, and Hérault Y
- Subjects
- Animals, Gene Knockout Techniques, Mice, Survival Analysis, Multiple Organ Failure mortality, Multiple Organ Failure pathology, Proton-Translocating ATPases deficiency, Receptors, Cell Surface deficiency
- Abstract
ATP6AP2 codes for the (pro)renin receptor and is an essential component of vacuolar H+ ATPase. Activating (pro)renin for conversion of Angiotensinogen to Angiotensin makes ATP6AP2 attractive for drug intervention. Tissue-specific ATP6AP2 inactivation in mouse suggested a strong impact on various organs. Consistent with this, we found that embryonic ablation of Atp6ap2 resulted in both male hemizygous lethality and female haploinsufficiency. Next, we examined the phenotype of an induced inactivation in the adult animal, most akin to detect potential effect of functional interference of ATP6AP2 through drug therapy. Induced ablation of Atp6ap2, even without equal efficiency in all tissues (aorta, brain and kidney), resulted in rapid lethality marked by weight loss, changes in nutritional as well as blood parameters, leukocyte depletion, and bone marrow hypoplasia. Upon Atp6ap2 ablation, the colon demonstrated a rapid disruption of crypt morphology, aberrant proliferation, cell-death activation, as well as generation of microadenomas. Consequently, disruption of ATP6AP2 is extremely poorly tolerated in the adult, and severely affects various organ systems demonstrating that ATP6AP2 is an essential gene implicated in basic cellular mechanisms and necessary for multiple organ function. Accordingly, any potential drug targeting of this gene product must be strictly assessed for safety.
- Published
- 2017
- Full Text
- View/download PDF
21. Natural Killer Defective Maturation Is Associated with Adverse Clinical Outcome in Patients with Acute Myeloid Leukemia.
- Author
-
Chretien AS, Fauriat C, Orlanducci F, Galseran C, Rey J, Bouvier Borg G, Gautherot E, Granjeaud S, Hamel-Broza JF, Demerle C, Ifrah N, Lacombe C, Cornillet-Lefebvre P, Delaunay J, Toubert A, Gregori E, Luche H, Malissen M, Arnoulet C, Nunes JA, Vey N, and Olive D
- Abstract
Accumulating evidence highlights natural killer (NK) cell parameters as potential prognostic factors in cancer patients, which provides a strong rationale for developing therapeutic strategies aiming at restoring NK cell. However, reaching this point warrants better characterization of tumor-induced NK cell alterations. Our group recently reported heterogeneous NK maturation in acute myeloid leukemia (AML) patients. However, the clinical significance of such observations remained to be assessed on a larger cohort of patients. NK maturation based on expression of CD56, CD57, and KIR was assessed by flow cytometry in newly diagnosed AML patients ( N = 87 patients from GOELAMS-LAM-IR-2006 multicenter trial). Clinical outcome was evaluated with regard to NK maturation profiles. Unsupervised integrated analysis of NK maturation markers confirmed the existence of three distinct groups of patients [hypomaturation (24.1%), intermediate maturation (66.7%), and hypermaturation (9.2%)]. In univariate analysis, significant differences in overall survival (OS) ( P = 0.0006) and relapse-free survival (RFS) ( P < 0.0001) were observed among these different groups. Patients with hypomaturation profile had reduced OS, with 3-year OS rates of 12.5 vs 57.1 and 57.4% for patients with intermediate and hypermaturation, respectively. Consistently, patients with hypomaturation profile had reduced RFS, with 3-year RFS rates of 0 vs 52.6 and 73.3% for patients with intermediate and hypermaturation, respectively. In multivariate Cox regression models, NK hypomaturation remained significantly associated with reduced OS and RFS, independent of other factors [hazard ratio (HR) = 4.15, P = 0.004 and HR = 8.23, P = 0.003, respectively]. NK maturation defects were further explored by mass cytometry and revealed that NK hypomaturation profile is associated with a reduced frequency of memory-like NK cells. In conclusion, besides classical alterations of NK triggering and inhibitory receptors expression in AML, we confirm that the homeostasis of NK maturation can be modified in the context of AML, notably with a deep maturation blockade in almost 10% patients.
- Published
- 2017
- Full Text
- View/download PDF
22. Unsupervised High-Dimensional Analysis Aligns Dendritic Cells across Tissues and Species.
- Author
-
Guilliams M, Dutertre CA, Scott CL, McGovern N, Sichien D, Chakarov S, Van Gassen S, Chen J, Poidinger M, De Prijck S, Tavernier SJ, Low I, Irac SE, Mattar CN, Sumatoh HR, Low GHL, Chung TJK, Chan DKH, Tan KK, Hon TLK, Fossum E, Bogen B, Choolani M, Chan JKY, Larbi A, Luche H, Henri S, Saeys Y, Newell EW, Lambrecht BN, Malissen B, and Ginhoux F
- Subjects
- Animals, Cell Differentiation physiology, Flow Cytometry, Humans, Inflammation pathology, Macaca, Mice, Mice, Inbred C57BL, Dendritic Cells physiology
- Abstract
Dendritic cells (DCs) are professional antigen-presenting cells that hold great therapeutic potential. Multiple DC subsets have been described, and it remains challenging to align them across tissues and species to analyze their function in the absence of macrophage contamination. Here, we provide and validate a universal toolbox for the automated identification of DCs through unsupervised analysis of conventional flow cytometry and mass cytometry data obtained from multiple mouse, macaque, and human tissues. The use of a minimal set of lineage-imprinted markers was sufficient to subdivide DCs into conventional type 1 (cDC1s), conventional type 2 (cDC2s), and plasmacytoid DCs (pDCs) across tissues and species. This way, a large number of additional markers can still be used to further characterize the heterogeneity of DCs across tissues and during inflammation. This framework represents the way forward to a universal, high-throughput, and standardized analysis of DC populations from mutant mice and human patients., (Copyright © 2016 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
23. Broad and Largely Concordant Molecular Changes Characterize Tolerogenic and Immunogenic Dendritic Cell Maturation in Thymus and Periphery.
- Author
-
Ardouin L, Luche H, Chelbi R, Carpentier S, Shawket A, Montanana Sanchis F, Santa Maria C, Grenot P, Alexandre Y, Grégoire C, Fries A, Vu Manh TP, Tamoutounour S, Crozat K, Tomasello E, Jorquera A, Fossum E, Bogen B, Azukizawa H, Bajenoff M, Henri S, Dalod M, and Malissen B
- Subjects
- Animals, Antigen Presentation, Cell Differentiation, Cells, Cultured, Interferon Regulatory Factors genetics, Lymphocyte Activation, Mice, Mice, Inbred C57BL, Mice, Knockout, Receptors, Chemokine metabolism, Toll-Like Receptors immunology, Transcriptome, Virus Replication, Central Tolerance, Dendritic Cells immunology, Peripheral Tolerance, T-Lymphocytes, Regulatory immunology, Thymus Gland immunology
- Abstract
Dendritic cells (DCs) are instrumental in the initiation of T cell responses, but how thymic and peripheral tolerogenic DCs differ globally from Toll-like receptor (TLR)-induced immunogenic DCs remains unclear. Here, we show that thymic XCR1(+) DCs undergo a high rate of maturation, accompanied by profound gene-expression changes that are essential for central tolerance and also happen in germ-free mice. Those changes largely overlap those occurring during tolerogenic and, more unexpectedly, TLR-induced maturation of peripheral XCR1(+) DCs, arguing against the commonly held view that tolerogenic DCs undergo incomplete maturation. Interferon-stimulated gene (ISG) expression was among the few discriminators of immunogenic and tolerogenic XCR1(+) DCs. Tolerogenic XCR1(+) thymic DCs were, however, unique in expressing ISGs known to restrain virus replication. Therefore, a broad functional convergence characterizes tolerogenic and immunogenic XCR1(+) DC maturation in the thymus and periphery, maximizing antigen presentation and signal delivery to developing and to conventional and regulatory mature T cells., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
24. Cytomegalovirus Infection of the Rat Developing Brain In Utero Prominently Targets Immune Cells and Promotes Early Microglial Activation.
- Author
-
Cloarec R, Bauer S, Luche H, Buhler E, Pallesi-Pocachard E, Salmi M, Courtens S, Massacrier A, Grenot P, Teissier N, Watrin F, Schaller F, Adle-Biassette H, Gressens P, Malissen M, Stamminger T, Streblow DN, Bruneau N, and Szepetowski P
- Subjects
- Animals, Cell Lineage, Cytomegalovirus Infections immunology, Flow Cytometry, Macrophage Activation, Microglia immunology, Rats, Reverse Transcriptase Polymerase Chain Reaction, Brain embryology, Cytomegalovirus Infections pathology, Microglia pathology, Muromegalovirus pathogenicity
- Abstract
Background: Congenital cytomegalovirus infections are a leading cause of neurodevelopmental disorders in human and represent a major health care and socio-economical burden. In contrast with this medical importance, the pathophysiological events remain poorly known. Murine models of brain cytomegalovirus infection, mostly neonatal, have brought recent insights into the possible pathogenesis, with convergent evidence for the alteration and possible involvement of brain immune cells., Objectives and Methods: In order to confirm and expand those findings, particularly concerning the early developmental stages following infection of the fetal brain, we have created a model of in utero cytomegalovirus infection in the developing rat brain. Rat cytomegalovirus was injected intraventricularly at embryonic day 15 (E15) and the brains analyzed at various stages until the first postnatal day, using a combination of gene expression analysis, immunohistochemistry and multicolor flow cytometry experiments., Results: Rat cytomegalovirus infection was increasingly seen in various brain areas including the choroid plexi and the ventricular and subventricular areas and was prominently detected in CD45low/int, CD11b+ microglial cells, in CD45high, CD11b+ cells of the myeloid lineage including macrophages, and in CD45+, CD11b- lymphocytes and non-B non-T cells. In parallel, rat cytomegalovirus infection of the developing rat brain rapidly triggered a cascade of pathophysiological events comprising: chemokines upregulation, including CCL2-4, 7 and 12; infiltration by peripheral cells including B-cells and monocytes at E17 and P1, and T-cells at P1; and microglia activation at E17 and P1., Conclusion: In line with previous findings in neonatal murine models and in human specimen, our study further suggests that neuroimmune alterations might play critical roles in the early stages following cytomegalovirus infection of the brain in utero. Further studies are now needed to determine which role, whether favorable or detrimental, those putative double-edge swords events actually play.
- Published
- 2016
- Full Text
- View/download PDF
25. Phenotypic dynamics of microglial and monocyte-derived cells in glioblastoma-bearing mice.
- Author
-
Ricard C, Tchoghandjian A, Luche H, Grenot P, Figarella-Branger D, Rougon G, Malissen M, and Debarbieux F
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Brain Neoplasms metabolism, Brain Neoplasms pathology, Cell Line, Tumor, Dendritic Cells cytology, Dendritic Cells metabolism, Female, Flow Cytometry, Glioblastoma metabolism, Glioblastoma pathology, Humans, Luminescent Proteins genetics, Luminescent Proteins metabolism, Male, Mice, Mice, Transgenic, Microglia metabolism, Microglia pathology, Microscopy, Fluorescence, Multiphoton, Middle Aged, Monocytes metabolism, Monocytes pathology, Neoplasm Transplantation, Phenotype, Young Adult, Brain Neoplasms diagnostic imaging, Dendritic Cells pathology, Glioblastoma diagnostic imaging, Microglia cytology, Monocytes cytology, Multimodal Imaging methods
- Abstract
Inflammatory cells, an integral component of tumor evolution, are present in Glioblastomas multiforme (GBM). To address the cellular basis and dynamics of the inflammatory microenvironment in GBM, we established an orthotopic syngenic model by grafting GL261-DsRed cells in immunocompetent transgenic LysM-EGFP//CD11c-EYFP reporter mice. We combined dynamic spectral two-photon imaging with multiparametric cytometry and multicolor immunostaining to characterize spatio-temporal distribution, morphology and activity of microglia and blood-derived infiltrating myeloid cells in live mice. Early stages of tumor development were dominated by microglial EYFP(+) cells invading the tumor, followed by massive recruitment of circulating LysM-EGFP(+) cells. Fluorescent invading cells were conventional XCR1(+) and monocyte-derived dendritic cells distributed in subpopulations of different maturation stages, located in different areas relative to the tumor core. The lethal stage of the disease was characterized by the progressive accumulation of EGFP(+)/EYFP(+) monocyte-derived dendritic cells. This local phenotypic regulation of monocyte subtypes marked a transition in the immune response.
- Published
- 2016
- Full Text
- View/download PDF
26. Promiscuous Foxp3-cre activity reveals a differential requirement for CD28 in Foxp3⁺ and Foxp3⁻ T cells.
- Author
-
Franckaert D, Dooley J, Roos E, Floess S, Huehn J, Luche H, Fehling HJ, Liston A, Linterman MA, and Schlenner SM
- Subjects
- Animals, Autoimmunity genetics, CD28 Antigens genetics, Cell Differentiation genetics, Cell Lineage genetics, Cell Survival genetics, Clonal Selection, Antigen-Mediated genetics, Forkhead Transcription Factors genetics, Gene Expression Regulation, Developmental, Homeostasis, Mice, Mice, Transgenic, Signal Transduction genetics, CD28 Antigens metabolism, Forkhead Transcription Factors metabolism, T-Lymphocyte Subsets physiology, T-Lymphocytes, Regulatory physiology
- Abstract
Costimulatory signals by CD28 are critical for thymic regulatory T-cell (Treg) development. To determine the functional relevance of CD28 for peripheral Treg post thymic selection, we crossed the widely used Forkhead box protein 3 (Foxp3)-CreYFP mice to mice bearing a conditional Cd28 allele. Treg-specific CD28 deficiency provoked a severe autoimmune syndrome as a result of a strong disadvantage in competitive fitness and proliferation of CD28-deficient Tregs. By contrast, Treg survival and lineage integrity were not affected by the lack of CD28. This data demonstrate that, even after the initial induction requirement, Treg maintain a higher dependency on CD28 signalling than conventional T cells for homeostasis. In addition, we found the Foxp3-CreYFP allele to be a hypomorph, with reduced Foxp3 protein levels. Furthermore, we report here the stochastic activity of the Foxp3-CreYFP allele in non-Tregs, sufficient to recombine some conditional alleles (including Cd28) but not others (including R26-RFP). This hypomorphism and 'leaky' expression of the Foxp3-CreYFP allele should be considered when analysing the conditionally mutated Treg.
- Published
- 2015
- Full Text
- View/download PDF
27. Evaluating the efficiency of isotope transmission for improved panel design and a comparison of the detection sensitivities of mass cytometer instruments.
- Author
-
Tricot S, Meyrand M, Sammicheli C, Elhmouzi-Younes J, Corneau A, Bertholet S, Malissen M, Le Grand R, Nuti S, Luche H, and Cosma A
- Subjects
- Algorithms, Animals, Antibodies immunology, Cells, Cultured, Flow Cytometry instrumentation, Fluorescent Dyes, Isotopes metabolism, Mice, Mice, Inbred C57BL, Spleen cytology, Flow Cytometry methods, Lanthanoid Series Elements metabolism, Mass Spectrometry methods, Staining and Labeling methods
- Abstract
The recent introduction of mass cytometry, a technique coupling a cell introduction system generating a stream of single cells with mass spectrometry, has greatly increased the number of parameters that can be measured per single cell. As with all new technology there is a need for dissemination of standardization and quality control procedures. Here, we characterize variations in sensitivity observed across the mass range of a mass cytometer, using different lanthanide tags. We observed a five-fold difference in lanthanide detection over the mass range and demonstrated that each instrument has its own sensitivity pattern. Therefore, the selection of lanthanide combinations is a key step in the establishment of a staining panel for mass cytometry-based experiments, particularly for multicenter studies. We propose the sensitivity pattern as the basis for panel design, instrument standardization and future implementation of normalization algorithms., (© 2015 International Society for Advancement of Cytometry.)
- Published
- 2015
- Full Text
- View/download PDF
28. Conditional ablation of NKp46+ cells using a novel Ncr1(greenCre) mouse strain: NK cells are essential for protection against pulmonary B16 metastases.
- Author
-
Merzoug LB, Marie S, Satoh-Takayama N, Lesjean S, Albanesi M, Luche H, Fehling HJ, Di Santo JP, and Vosshenrich CA
- Subjects
- Animals, Antigens, Ly biosynthesis, B-Lymphocytes immunology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Green Fluorescent Proteins genetics, Interleukin Receptor Common gamma Subunit genetics, Lung Neoplasms immunology, Melanoma, Experimental immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, Natural Cytotoxicity Triggering Receptor 1 biosynthesis, Antigens, Ly genetics, Killer Cells, Natural immunology, Lung Neoplasms secondary, Melanoma, Experimental pathology, Natural Cytotoxicity Triggering Receptor 1 genetics
- Abstract
To study gene functions specifically in NKp46+ cells we developed novel Cre mice allowing for conditional gene targeting in cells expressing Ncr1 (encoding NKp46). We generated transgenic Ncr1(greenCre) mice carrying an EGFPcre fusion under the control of a proximal Ncr1 promoter that faithfully directed EGFPcre expression to NKp46+ cells from lymphoid and nonlymphoid tissues. This approach allowed for direct detection of Cre-expressing NKp46+ cells via their GFP signature by flow cytometry and histology. Cre was functional as evidenced by the NKp46+ cell-specific expression of RFP in Ncr1(greenCre) Rosa-dtRFP reporter mice. We generated Ncr1(greenCre) Il2rg(fl/fl) mice that lack NKp46+ cells in an otherwise intact hematopoietic environment. Il2rg encodes the common gamma chain (γc ), which is an essential receptor subunit for cytokines (IL-2, -4, -7, -9, -15, and -21) that stimulate lymphocyte development and function. In Ncr1(greenCre) Il2rg(fl/fl) mice, NK cells are severely reduced and the few remaining NKp46+ cells escaping γc deletion failed to express GFP. Using this new NK-cell-deficient model, we demonstrate that the homeostasis of NKp46+ cells from all tissues (including the recently described intraepithelial ILC1 subset) requires Il2rg. Finally, Ncr1(greenCre) Il2rg(fl/fl) mice are unable to reject B16 lung metastases demonstrating the essential role of NKp46+ cells in antimelanoma immune responses., (© 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2014
- Full Text
- View/download PDF
29. Improved method to retain cytosolic reporter protein fluorescence while staining for nuclear proteins.
- Author
-
Heinen AP, Wanke F, Moos S, Attig S, Luche H, Pal PP, Budisa N, Fehling HJ, Waisman A, and Kurschus FC
- Subjects
- Animals, Cytoplasm metabolism, Fixatives, Fluorescent Dyes, Forkhead Transcription Factors, Mice, Mice, Inbred C57BL, Mice, Transgenic, Mycobacterium tuberculosis immunology, Nuclear Receptor Subfamily 1, Group F, Member 3, Staining and Labeling, T-Box Domain Proteins, T-Lymphocytes cytology, Tissue Fixation methods, Cytokines analysis, Flow Cytometry methods, Nuclear Proteins analysis, Transcription Factors analysis
- Abstract
Staining of transcription factors (TFs) together with retention of fluorescent reporter proteins is hindered by loss of fluorescence using current available methods. In this study, it is shown that current TF staining protocols do not destroy fluorescent proteins (FPs) but rather that fixation is not sufficient to retain FPs in the cytosol of the permeabilized cells. In this article, a simple and reliable protocol is elaborated, which allows efficient TF and cytokine staining while retaining FPs inside fixed cells., (© 2014 International Society for Advancement of Cytometry.)
- Published
- 2014
- Full Text
- View/download PDF
30. Temporal lineage tracing of Aire-expressing cells reveals a requirement for Aire in their maturation program.
- Author
-
Nishikawa Y, Nishijima H, Matsumoto M, Morimoto J, Hirota F, Takahashi S, Luche H, Fehling HJ, Mouri Y, and Matsumoto M
- Subjects
- Animals, Apoptosis genetics, Apoptosis immunology, Autoantigens immunology, Autoantigens metabolism, B7-1 Antigen immunology, B7-1 Antigen metabolism, Cell Differentiation genetics, Cell Lineage genetics, Cells, Cultured, Cross-Priming genetics, Cross-Priming immunology, Epithelial Cells metabolism, Flow Cytometry, Immunohistochemistry, Kinetics, Luminescent Proteins genetics, Luminescent Proteins metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Thymocytes cytology, Thymocytes immunology, Thymocytes metabolism, Thymus Gland cytology, Time Factors, Transcription Factors genetics, Transcription Factors metabolism, AIRE Protein, Cell Differentiation immunology, Cell Lineage immunology, Epithelial Cells immunology, Transcription Factors immunology
- Abstract
Understanding the cellular dynamics of Aire-expressing lineage(s) among medullary thymic epithelial cells (AEL-mTECs) is essential for gaining insight into the roles of Aire in establishment of self-tolerance. In this study, we monitored the maturation program of AEL-mTECs by temporal lineage tracing, in which bacterial artificial chromosome transgenic mice expressing tamoxifen-inducible Cre recombinase under control of the Aire regulatory element were crossed with reporter strains. We estimated that the half-life of AEL-mTECs subsequent to Aire expression was ∼7-8 d, which was much longer than that reported previously, owing to the existence of a post-Aire stage. We found that loss of Aire did not alter the overall lifespan of AEL-mTECs, inconsistent with the previous notion that Aire expression in medullary thymic epithelial cells (mTECs) might result in their apoptosis for efficient cross-presentation of self-antigens expressed by AEL-mTECs. In contrast, Aire was required for the full maturation program of AEL-mTECs, as exemplified by the lack of physiological downregulation of CD80 during the post-Aire stage in Aire-deficient mice, thus accounting for the abnormally increased CD80(high) mTECs seen in such mice. Of interest, increased CD80(high) mTECs in Aire-deficient mice were not mTEC autonomous and were dependent on cross-talk with thymocytes. These results further support the roles of Aire in the differentiation program of AEL-mTECs.
- Published
- 2014
- Full Text
- View/download PDF
31. Origins and functional specialization of macrophages and of conventional and monocyte-derived dendritic cells in mouse skin.
- Author
-
Tamoutounour S, Guilliams M, Montanana Sanchis F, Liu H, Terhorst D, Malosse C, Pollet E, Ardouin L, Luche H, Sanchez C, Dalod M, Malissen B, and Henri S
- Subjects
- Animals, Antigens, Differentiation analysis, CD11b Antigen analysis, Cell Lineage, Chemotaxis, Leukocyte, Chromatography, Gel, Dendritic Cells immunology, Dermatitis, Contact immunology, Dermatitis, Contact pathology, Dermis cytology, Gene Expression Regulation, Developmental, Immunophenotyping methods, Langerhans Cells cytology, Langerhans Cells immunology, Lymphocyte Cooperation, Macrophages physiology, Mice, Microbiota immunology, Monocytes cytology, Principal Component Analysis, Radiation Chimera, Receptors, CCR2 analysis, Receptors, IgG analysis, Skin immunology, Skin microbiology, Specific Pathogen-Free Organisms, Staining and Labeling methods, Transcriptome, Dendritic Cells cytology, Macrophages cytology, Skin cytology
- Abstract
In the skin, the lack of markers permitting the unambiguous identification of macrophages and of conventional and monocyte-derived dendritic cells (DCs) complicates understanding of their contribution to skin integrity and to immune responses. By combining CD64 and CCR2 staining, we successfully identified each of these cell types and studied their origin, transcriptomic signatures, and migratory and T cell stimulatory properties. We also analyzed the impact of microbiota on their development and their contribution to skin inflammation during contact hypersensitivity. Dermal macrophages had a unique scavenging role and were unable to migrate and activate T cells. Conventional dermal DCs excelled both at migrating and activating T cells. In the steady-state dermis, monocyte-derived DCs are continuously generated by extravasated Ly-6C(hi) monocytes. Their T cell stimulatory capacity combined with their poor migratory ability made them particularly suited to activate skin-tropic T cells. Therefore, a high degree of functional specialization occurs among the mononuclear phagocytes of the skin., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
32. Self-antigen-driven activation induces instability of regulatory T cells during an inflammatory autoimmune response.
- Author
-
Bailey-Bucktrout SL, Martinez-Llordella M, Zhou X, Anthony B, Rosenthal W, Luche H, Fehling HJ, and Bluestone JA
- Subjects
- Animals, CD4-Positive T-Lymphocytes immunology, Cell Lineage, Central Nervous System immunology, DNA Methylation, Down-Regulation immunology, Forkhead Transcription Factors biosynthesis, Forkhead Transcription Factors genetics, Genes, Reporter, Lymph Nodes immunology, Lymphocyte Activation, Mice, Mice, Inbred C57BL, Myelin-Oligodendrocyte Glycoprotein immunology, Peptide Fragments immunology, Receptors, Interleukin-2 physiology, Recombinant Fusion Proteins immunology, Regulatory Sequences, Nucleic Acid, Specific Pathogen-Free Organisms, Autoantigens immunology, Encephalomyelitis, Autoimmune, Experimental immunology, Forkhead Transcription Factors physiology, Gene Expression Regulation immunology, T-Lymphocytes, Regulatory immunology
- Abstract
Stable Foxp3 expression is crucial for regulatory T (Treg) cell function. We observed that antigen-driven activation and inflammation in the CNS promoted Foxp3 instability selectively in the autoreactive Treg cells that expressed high amounts of Foxp3 before experimental autoimmune encephalitis induction. Treg cells with a demethylated Treg-cell-specific demethylated region in the Foxp3 locus downregulated Foxp3 transcription in the inflamed CNS during the induction phase of the response. Stable Foxp3 expression returned at the population level with the resolution of inflammation or was rescued by IL-2-anti-IL-2 complex treatment during the antigen priming phase. Thus, a subset of fully committed self-antigen-specific Treg cells lost Foxp3 expression during an inflammatory autoimmune response and might be involved in inadequate control of autoimmunity. These results have important implications for Treg cell therapies and give insights into the dynamics of the Treg cell network during autoreactive CD4(+) T cell effector responses in vivo., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
33. Identification of a new stromal cell type involved in the regulation of inflamed B cell follicles.
- Author
-
Mionnet C, Mondor I, Jorquera A, Loosveld M, Maurizio J, Arcangeli ML, Ruddle NH, Nowak J, Aurrand-Lions M, Luche H, and Bajénoff M
- Subjects
- Animals, Chemokine CXCL13 metabolism, Dendritic Cells pathology, Fibroblasts pathology, Lymph Nodes pathology, Lymphocyte Depletion, Lymphocytes pathology, Mice, Receptors, CXCR5 deficiency, Receptors, CXCR5 metabolism, Receptors, Complement 3d metabolism, Stromal Cells metabolism, Stromal Cells pathology, T-Lymphocytes, B-Lymphocytes pathology, Inflammation immunology, Inflammation pathology
- Abstract
Lymph node (LN) stromal cells provide survival signals and adhesive substrata to lymphocytes. During an immune response, B cell follicles enlarge, questioning how LN stromal cells manage these cellular demands. Herein, we used a murine fate mapping system to describe a new stromal cell type that resides in the T cell zone of resting LNs. We demonstrated that upon inflammation, B cell follicles progressively trespassed into the adjacent T cell zone and surrounded and converted these stromal cells into CXCL13 secreting cells that in return delineated the new boundaries of the growing follicle. Acute B cell ablation in inflamed LNs abolished CXCL13 secretion in these cells, while LT-β deficiency in B cells drastically affected this conversion. Altogether, we reveal the existence of a dormant stromal cell subset that can be functionally awakened by B cells to delineate the transient boundaries of their expanding territories upon inflammation., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2013
- Full Text
- View/download PDF
34. TRAF6 is essential for maintenance of regulatory T cells that suppress Th2 type autoimmunity.
- Author
-
Muto G, Kotani H, Kondo T, Morita R, Tsuruta S, Kobayashi T, Luche H, Fehling HJ, Walsh M, Choi Y, and Yoshimura A
- Subjects
- Animals, Cell Proliferation, Female, Forkhead Transcription Factors metabolism, Integrases metabolism, Interleukin-2 metabolism, Lymphocyte Activation immunology, Mice, Mice, Knockout, Phenotype, STAT5 Transcription Factor metabolism, TNF Receptor-Associated Factor 6 deficiency, Autoimmunity immunology, T-Lymphocytes, Regulatory immunology, TNF Receptor-Associated Factor 6 metabolism, Th2 Cells immunology
- Abstract
Regulatory T cells (Tregs) maintain immune homeostasis by limiting inflammatory responses. TRAF6 plays a key role in the regulation of innate and adaptive immunity by mediating signals from various receptors including the T-cell receptor (TCR). T cell-specific deletion of TRAF6 has been shown to induce multiorgan inflammatory disease, but the role of TRAF6 in Tregs remains to be investigated. Here, we generated Treg-specific TRAF6-deficient mice using Foxp3-Cre and TRAF6-flox mice. Treg-specific TRAF6-deficient (cKO) mice developed allergic skin diseases, arthritis, lymphadenopathy and hyper IgE phenotypes. Although TRAF6-deficient Tregs possess similar in vitro suppression activity compared to wild-type Tregs, TRAF6-deficient Tregs did not suppress colitis in lymphopenic mice very efficiently due to reduced number of Foxp3-positive cells. In addition, the fraction of TRAF6-deficient Tregs was reduced compared with wild-type Tregs in female cKO mice without inflammation. Moreover, adoptive transfer of Foxp3 (+) Tregs into Rag2(-/-) mice revealed that TRAF6-deficient Tregs converted into Foxp3(-) cells more rapidly than WT Tregs under lymphopenic conditions. Fate-mapping analysis also revealed that conversion of Tregs from Foxp3(+) to Foxp3(-) (exFoxp3 cells) was accelerated in TRAF6-deficient Tregs. These data indicate that TRAF6 in Tregs plays important roles in the maintenance of Foxp3 in Tregs and in the suppression of pathogenic Th2 type conversion of Tregs.
- Published
- 2013
- Full Text
- View/download PDF
35. Multicolor fate mapping of Langerhans cell homeostasis.
- Author
-
Ghigo C, Mondor I, Jorquera A, Nowak J, Wienert S, Zahner SP, Clausen BE, Luche H, Malissen B, Klauschen F, and Bajénoff M
- Subjects
- Animals, Color, Imaging, Three-Dimensional, Inflammation pathology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Cell Lineage, Cytological Techniques methods, Homeostasis, Langerhans Cells pathology
- Abstract
Langerhans cells (LCs) constitute a network of immune sentinels in the skin epidermis that is seeded during embryogenesis. Whereas the development of LCs has been extensively studied, much less is known about the homeostatic renewal of adult LCs in "nonmanipulated" animals. Here, we present a new multicolor fluorescent fate mapping system and quantification approach to investigate adult LC homeostasis. This novel approach enables us to propose and provide evidence for a model in which the adult epidermal LC network is not formed by mature coequal LCs endowed with proliferative capabilities, but rather constituted by adjacent proliferative units composed of "dividing" LCs and their terminally differentiated daughter cells. Altogether, our results demonstrate the general utility of our novel fate-mapping system to follow cell population dynamics in vivo and to establish an alternative model for LC homeostasis.
- Published
- 2013
- Full Text
- View/download PDF
36. In vivo fate mapping identifies pre-TCRα expression as an intra- and extrathymic, but not prethymic, marker of T lymphopoiesis.
- Author
-
Luche H, Nageswara Rao T, Kumar S, Tasdogan A, Beckel F, Blum C, Martins VC, Rodewald HR, and Fehling HJ
- Subjects
- Animals, Bone Marrow immunology, Cell Differentiation genetics, Gene Expression Regulation genetics, Membrane Glycoproteins genetics, Mice, Mice, Knockout, Receptors, Antigen, T-Cell, alpha-beta genetics, Receptors, Antigen, T-Cell, gamma-delta genetics, Receptors, Antigen, T-Cell, gamma-delta immunology, Thymocytes cytology, Thymus Gland cytology, Cell Differentiation immunology, Gene Expression Regulation immunology, Lymphopoiesis physiology, Membrane Glycoproteins immunology, Receptors, Antigen, T-Cell, alpha-beta immunology, Thymocytes immunology, Thymus Gland immunology
- Abstract
Expression of the pre-T cell receptor α (pTα) gene has been exploited in previous studies as a molecular marker to identify tiny cell populations in bone marrow (BM) and blood that were suggested to contain physiologically relevant thymus settling progenitors (TSPs). But to what extent these cells genuinely contribute to thymopoiesis has remained obscure. We have generated a novel pTα(iCre) knockin mouse line and performed lineage-tracing experiments to precisely quantitate the contribution of pTα-expressing progenitors to distinct differentiation pathways and to the genealogy of mature hematopoietic cells under physiological in vivo conditions. Using these mice in combination with fluorescent reporter strains, we observe highly consistent labeling patterns that identify pTα expression as a faithful molecular marker of T lineage commitment. Specifically, the fate of pTα-expressing progenitors was found to include all αβ and most γδ T cells but, in contrast to previous assumptions, to exclude B, NK, and thymic dendritic cells. Although we could detect small numbers of T cell progenitors with a history of pTα expression in BM and blood, our data clearly exclude these populations as physiologically important precursors of thymopoiesis and indicate that they instead belong to a pathway of T cell maturation previously defined as extrathymic.
- Published
- 2013
- Full Text
- View/download PDF
37. Differential requirement for ROCK in dendritic cell migration within lymphatic capillaries in steady-state and inflammation.
- Author
-
Nitschké M, Aebischer D, Abadier M, Haener S, Lucic M, Vigl B, Luche H, Fehling HJ, Biehlmaier O, Lyck R, and Halin C
- Subjects
- Animals, Cell Adhesion Molecules genetics, Cell Adhesion Molecules metabolism, Crosses, Genetic, Dendritic Cells drug effects, Dendritic Cells immunology, Dendritic Cells pathology, Dermatitis, Contact drug therapy, Dermatitis, Contact immunology, Dermatitis, Contact pathology, Endothelium, Lymphatic drug effects, Endothelium, Lymphatic metabolism, Endothelium, Lymphatic pathology, Intercellular Adhesion Molecule-1 metabolism, Luminescent Proteins genetics, Luminescent Proteins metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Microscopy, Fluorescence, Microscopy, Video, Protein Kinase Inhibitors pharmacology, Radiation Chimera, Recombinant Fusion Proteins metabolism, Skin drug effects, Skin immunology, Skin metabolism, Skin pathology, Up-Regulation drug effects, rho-Associated Kinases antagonists & inhibitors, rho-Associated Kinases genetics, Cell Movement drug effects, Dendritic Cells metabolism, Dermatitis, Contact metabolism, Endothelium, Lymphatic immunology, rho-Associated Kinases metabolism
- Abstract
Dendritic cell (DC) migration via lymphatic vessels to draining lymph nodes (dLNs) is crucial for the initiation of adaptive immunity. We imaged this process by intravital microscopy (IVM) in the ear skin of transgenic mice bearing red-fluorescent vasculature and yellow-fluorescent DCs. DCs within lymphatic capillaries were rarely transported by flow, but actively migrated within lymphatics and were significantly faster than in the interstitium. Pharmacologic blockade of the Rho-associated protein kinase (ROCK), which mediates nuclear contraction and de-adhesion from integrin ligands, significantly reduced DC migration from skin to dLNs in steady-state. IVM revealed that ROCK blockade strongly reduced the velocity of interstitial DC migration, but only marginally affected intralymphatic DC migration. By contrast, during tissue inflammation, ROCK blockade profoundly decreased both interstitial and intralymphatic DC migration. Inhibition of intralymphatic migration was paralleled by a strong up-regulation of ICAM-1 in lymphatic endothelium, suggesting that during inflammation ROCK mediates de-adhesion of DC-expressed integrins from lymphatic-expressed ICAM-1. Flow chamber assays confirmed an involvement of lymphatic-expressed ICAM-1 and DC-expressed ROCK in DC crawling on lymphatic endothelium. Overall, our findings further define the role of ROCK in DC migration to dLNs and reveal a differential requirement for ROCK in intralymphatic DC crawling during steady-state and inflammation.
- Published
- 2012
- Full Text
- View/download PDF
38. Plasticity of Foxp3(+) T cells reflects promiscuous Foxp3 expression in conventional T cells but not reprogramming of regulatory T cells.
- Author
-
Miyao T, Floess S, Setoguchi R, Luche H, Fehling HJ, Waldmann H, Huehn J, and Hori S
- Subjects
- Animals, CD2 Antigens genetics, CD2 Antigens metabolism, CD4-Positive T-Lymphocytes cytology, Cell Differentiation, Cell Lineage genetics, Cell Lineage immunology, DNA Methylation, Epigenesis, Genetic, Forkhead Transcription Factors genetics, Gene Expression, Humans, Immunologic Memory, In Vitro Techniques, Inflammation immunology, Inflammation metabolism, Inflammation pathology, Interleukin-2 Receptor alpha Subunit metabolism, Lymphocyte Activation, Lymphopenia immunology, Lymphopenia metabolism, Lymphopenia pathology, Mice, Mice, Knockout, Mice, Transgenic, T-Lymphocyte Subsets cytology, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, T-Lymphocytes, Regulatory cytology, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, Forkhead Transcription Factors metabolism, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory metabolism
- Abstract
The emerging notion of environment-induced reprogramming of Foxp3(+) regulatory T (Treg) cells into helper T (Th) cells remains controversial. By genetic fate mapping or adoptive transfers, we have identified a minor population of nonregulatory Foxp3(+) T cells exhibiting promiscuous and transient Foxp3 expression, which gave rise to Foxp3(-) ("exFoxp3") Th cells and selectively accumulated in inflammatory cytokine milieus or in lymphopenic environments including those in early ontogeny. In contrast, Treg cells did not undergo reprogramming under those conditions irrespective of their thymic or peripheral origins. Moreover, although a few Treg cells transiently lose Foxp3 expression, such "latent" Treg cells retained their memory and robustly re-expressed Foxp3 and suppressive function upon activation. This study establishes that Treg cells constitute a stable cell lineage, whose committed state in a changing environment is ensured by DNA demethylation of the Foxp3 locus irrespectively of ongoing Foxp3 expression., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
39. CD64 expression distinguishes monocyte-derived and conventional dendritic cells and reveals their distinct role during intramuscular immunization.
- Author
-
Langlet C, Tamoutounour S, Henri S, Luche H, Ardouin L, Grégoire C, Malissen B, and Guilliams M
- Subjects
- Adjuvants, Immunologic, Alum Compounds, Animals, Cell Differentiation, Immunization, Injections, Intramuscular, Interferon-gamma biosynthesis, Mice, Receptors, IgG metabolism, T-Lymphocytes immunology, Vaccines administration & dosage, Vaccines immunology, Dendritic Cells immunology, Monocytes immunology, Muscle, Skeletal immunology, Receptors, IgG biosynthesis
- Abstract
Although most vaccines are administered i.m., little is known about the dendritic cells (DCs) that are present within skeletal muscles. In this article, we show that expression of CD64, the high-affinity IgG receptor FcγRI, distinguishes conventional DCs from monocyte-derived DCs (Mo-DCs). By using such a discriminatory marker, we defined the distinct DC subsets that reside in skeletal muscles and identified their migratory counterparts in draining lymph nodes (LNs). We further used this capability to analyze the functional specialization that exists among muscle DCs. After i.m. administration of Ag adsorbed to alum, we showed that alum-injected muscles contained large numbers of conventional DCs that belong to the CD8α(+)- and CD11b(+)-type DCs. Both conventional DC types were capable of capturing Ag and of migrating to draining LNs, where they efficiently activated naive T cells. In alum-injected muscles, Mo-DCs were as numerous as conventional DCs, but only a small fraction migrated to draining LNs. Therefore, alum by itself poorly induces Mo-DCs to migrate to draining LNs. We showed that addition of small amounts of LPS to alum enhanced Mo-DC migration. Considering that migratory Mo-DCs had, on a per cell basis, a higher capacity to induce IFN-γ-producing T cells than conventional DCs, the addition of LPS to alum enhanced the overall immunogenicity of Ags presented by muscle-derived DCs. Therefore, a full understanding of the role of adjuvants during i.m. vaccination needs to take into account the heterogeneous migratory and functional behavior of muscle DCs and Mo-DCs revealed in this study.
- Published
- 2012
- Full Text
- View/download PDF
40. Differential processing of self-antigens by subsets of thymic stromal cells.
- Author
-
Guerder S, Viret C, Luche H, Ardouin L, and Malissen B
- Subjects
- CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Epithelial Cells immunology, Humans, Receptors, Antigen, T-Cell immunology, Antigen Presentation immunology, Antigen-Presenting Cells immunology, Autoantigens immunology, Thymus Gland immunology
- Abstract
The stromal network of the thymus provides a unique environment that supports the development of mature CD4(+) and CD8(+) T cells expressing a very diverse repertoire of T cell receptors (TCR) with limited reactivity to self-antigens. Thymic cortical epithelial cells (cTECs) are specialized antigen-presenting cells (APCs) that promote the positive selection of developing thymocytes while medullary thymic epithelial cells (mTECs) and thymic dendritic cells (tDCs) induce central tolerance to self-antigens. Recent studies showed that cTECs express a unique set of proteases involved in the generation of self-peptides presented by major-histocompatibility encoded molecules (pMHC) and consequently may express a unique set of pMHC complexes. Conversely, the stromal cells of the medulla developed several mechanisms to mirror as closely as possible the constellation of self-peptides derived from peripheral tissues. Here, we discuss how these different features allow for the development of a highly diverse but poorly self-reactive repertoire of functional T cells., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
41. Fate mapping analysis of lymphoid cells expressing the NKp46 cell surface receptor.
- Author
-
Narni-Mancinelli E, Chaix J, Fenis A, Kerdiles YM, Yessaad N, Reynders A, Gregoire C, Luche H, Ugolini S, Tomasello E, Walzer T, and Vivier E
- Subjects
- Animals, Antigens, Ly genetics, Cell Differentiation, Cell Lineage, Intestines cytology, Liver cytology, Lymphoid Tissue cytology, Mice, Mice, Transgenic, Natural Cytotoxicity Triggering Receptor 1 genetics, Antigens, Ly metabolism, Lymphoid Tissue metabolism, Natural Cytotoxicity Triggering Receptor 1 metabolism, T-Lymphocytes metabolism
- Abstract
NKp46 is a cell surface receptor expressed on natural killer (NK) cells, on a minute subset of T cells, and on a population of innate lymphoid cells that produce IL-22 and express the transcription factor retinoid-related orphan receptor (ROR)-γt, referred to as NK cell receptor (NKR)(+)ROR-γt(+) cells. Here we describe Nkp46(iCre) knock-in mice in which the gene encoding the improved Cre (iCre) recombinase was inserted into the Nkp46 locus. This mouse was used to noninvasively trace cells expressing NKp46 in vivo. Fate mapping experiments demonstrated the stable expression of NKp46 on NK cells and allowed a reappraisal of the sequential steps of NK cell maturation. NKp46 genetic tracing also showed that gut NKR(+)ROR-γt(+) and NK cells represent two distinct lineages. In addition, the genetic heterogeneity of liver NK cells was evidenced. Finally, Nkp46(iCre) mice also represent a unique mouse model of conditional mutagenesis specifically in NKp46(+) cells, paving the way for further developments in the biology of NKp46(+) NK, T, and NKR(+)ROR-γt(+) cells.
- Published
- 2011
- Full Text
- View/download PDF
42. Cutting edge: expression of XCR1 defines mouse lymphoid-tissue resident and migratory dendritic cells of the CD8α+ type.
- Author
-
Crozat K, Tamoutounour S, Vu Manh TP, Fossum E, Luche H, Ardouin L, Guilliams M, Azukizawa H, Bogen B, Malissen B, Henri S, and Dalod M
- Subjects
- Animals, Antigens, CD biosynthesis, Cell Movement genetics, DNA Fingerprinting, Dendritic Cells classification, Dendritic Cells cytology, Genetic Markers immunology, Humans, Integrin alpha Chains biosynthesis, Lymphoid Tissue cytology, Lymphoid Tissue metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Receptors, Chemokine genetics, Transcription, Genetic immunology, CD8 Antigens biosynthesis, Cell Movement immunology, Dendritic Cells immunology, Lymphoid Tissue immunology, Receptors, Chemokine biosynthesis, Receptors, G-Protein-Coupled biosynthesis
- Abstract
Subsets of dendritic cells (DCs) have been described according to their functions and anatomical locations. Conventional DC subsets are defined by reciprocal expression of CD11b and CD8α in lymphoid tissues (LT), and of CD11b and CD103 in non-LT (NLT). Spleen CD8α(+) and dermal CD103(+) DCs share a high efficiency for Ag cross-presentation and a developmental dependency on specific transcription factors. However, it is not known whether all NLT-derived CD103(+) DCs and LT-resident CD8α(+) DCs are similar despite their different anatomical locations. XCR1 was previously described as exclusively expressed on mouse spleen CD8α(+) DCs and human blood BDCA3(+) DCs. In this article, we showed that LT-resident CD8α(+) DCs and NLT-derived CD103(+) DCs specifically express XCR1 and are characterized by a unique transcriptional fingerprint, irrespective of their tissue of origin. Therefore, CD8α(+) DCs and CD103(+) DCs belong to a common DC subset which is unequivocally identified by XCR1 expression throughout the body.
- Published
- 2011
- Full Text
- View/download PDF
43. The earliest intrathymic precursors of CD8α(+) thymic dendritic cells correspond to myeloid-type double-negative 1c cells.
- Author
-
Luche H, Ardouin L, Teo P, See P, Henri S, Merad M, Ginhoux F, and Malissen B
- Subjects
- Animals, Antigens, Surface genetics, Antigens, Surface immunology, Antigens, Surface metabolism, CD8 Antigens metabolism, Cell Differentiation immunology, Dendritic Cells metabolism, Female, Flow Cytometry, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Hyaluronan Receptors immunology, Hyaluronan Receptors metabolism, Interferon Regulatory Factors immunology, Interferon Regulatory Factors metabolism, Interleukin-2 Receptor alpha Subunit immunology, Interleukin-2 Receptor alpha Subunit metabolism, Lectins, C-Type genetics, Lectins, C-Type immunology, Lectins, C-Type metabolism, Male, Mannose-Binding Lectins genetics, Mannose-Binding Lectins immunology, Mannose-Binding Lectins metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Models, Immunological, Myeloid Cells metabolism, Spleen cytology, Spleen immunology, Spleen metabolism, T-Lymphocytes immunology, T-Lymphocytes metabolism, Thymus Gland cytology, Thymus Gland metabolism, Time Factors, CD8 Antigens immunology, Dendritic Cells immunology, Myeloid Cells immunology, Thymus Gland immunology
- Abstract
The dendritic cells (DCs) present in lymphoid and non-lymphoid organs are generated from progenitors with myeloid-restricted potential. However, in the thymus a major subset of DCs expressing CD8α and langerin (CD207) appears to stand apart from all other DCs in that it is thought to derive from progenitors with lymphoid potential. Using mice expressing a fluorescent reporter and a diphtheria toxin receptor under the control of the cd207 gene, we demonstrated that CD207(+) CD8α(+) thymic DCs do not share a common origin with T cells but originate from intrathymic precursors that express markers that are normally present on all (CD11c(+) and MHCII molecules) or on some (CD207, CD135, CD8α, CX3CR1) DC subsets. Those intrathymic myeloid-type precursors correspond to CD44(+) CD25(-) double-negative 1c (DN1c) cells and are continuously renewed from bone marrow-derived canonical DC precursors. In conclusion, our results demonstrate that the earliest intrathymic precursors of CD8α(+) thymic DCs correspond to myeloid-type DN1c cells and support the view that under physiological conditions myeloid-restricted progenitors generate the whole constellation of DCs present in the body including the thymus., (Copyright © 2011 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2011
- Full Text
- View/download PDF
44. Specific erythroid-lineage defect in mice conditionally deficient for Mediator subunit Med1.
- Author
-
Stumpf M, Yue X, Schmitz S, Luche H, Reddy JK, and Borggrefe T
- Subjects
- Animals, Cell Line, Enhancer Elements, Genetic, Gene Expression Regulation, Hematopoietic Stem Cells cytology, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Lymphopoiesis physiology, Mediator Complex Subunit 1 genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Transcription Factors genetics, Transcription Factors metabolism, beta-Globins genetics, beta-Globins metabolism, Cell Lineage, Hematopoietic Stem Cells physiology, Mediator Complex Subunit 1 metabolism
- Abstract
The Mediator complex forms the bridge between transcriptional activators and the RNA polymerase II. Med1 (also known as PBP or TRAP220) is a key component of Mediator that interacts with nuclear hormone receptors and GATA transcription factors. Here, we show dynamic recruitment of GATA-1, TFIIB, Mediator, and RNA polymerase II to the β-globin locus in induced mouse erythroid leukemia cells and in an erythropoietin-inducible hematopoietic progenitor cell line. Using Med1 conditional knockout mice, we demonstrate a specific block in erythroid development but not in myeloid or lymphoid development, highlighted by the complete absence of β-globin gene expression. Thus, Mediator subunit Med1 plays a pivotal role in erythroid development and in β-globin gene activation.
- Published
- 2010
- Full Text
- View/download PDF
45. In vivo imaging of partially reversible th17 cell-induced neuronal dysfunction in the course of encephalomyelitis.
- Author
-
Siffrin V, Radbruch H, Glumm R, Niesner R, Paterka M, Herz J, Leuenberger T, Lehmann SM, Luenstedt S, Rinnenthal JL, Laube G, Luche H, Lehnardt S, Fehling HJ, Griesbeck O, and Zipp F
- Subjects
- Animals, Apoptosis, Axons physiology, Calcium metabolism, Cell Communication, Cell Movement, Cells, Cultured, Mice, Mice, Inbred C57BL, Receptors, N-Methyl-D-Aspartate physiology, Synapses physiology, Encephalomyelitis, Autoimmune, Experimental immunology, Interleukin-17 physiology, Neurons physiology, T-Lymphocytes, Helper-Inducer physiology
- Abstract
Neuronal damage in autoimmune neuroinflammation is the correlate for long-term disability in multiple sclerosis (MS) patients. Here, we investigated the role of immune cells in neuronal damage processes in animal models of MS by monitoring experimental autoimmune encephalomyelitis (EAE) by using two-photon microscopy of living anaesthetized mice. In the brainstem, we detected sustained interaction between immune and neuronal cells, particularly during disease peak. Direct interaction of myelin oligodendrocyte glycoprotein (MOG)-specific Th17 and neuronal cells in demyelinating lesions was associated with extensive axonal damage. By combining confocal, electron, and intravital microscopy, we showed that these contacts remarkably resembled immune synapses or kinapses, albeit with the absence of potential T cell receptor engagement. Th17 cells induced severe, localized, and partially reversible fluctuation in neuronal intracellular Ca(2+) concentration as an early sign of neuronal damage. These results highlight the central role of the Th17 cell effector phenotype for neuronal dysfunction in chronic neuroinflammation., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
46. Temporal specification of blood progenitors from mouse embryonic stem cells and induced pluripotent stem cells.
- Author
-
Irion S, Clarke RL, Luche H, Kim I, Morrison SJ, Fehling HJ, and Keller GM
- Subjects
- Activins administration & dosage, Animals, Bone Morphogenetic Protein 4 administration & dosage, Cell Differentiation drug effects, Cell Differentiation physiology, Cell Line, Embryonic Stem Cells drug effects, Embryonic Stem Cells metabolism, Gene Expression, HMGB Proteins genetics, HMGB Proteins metabolism, Hematopoiesis drug effects, Hematopoiesis genetics, Hematopoietic Stem Cells drug effects, Hematopoietic Stem Cells metabolism, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Lymphopoiesis drug effects, Lymphopoiesis genetics, Lymphopoiesis physiology, Membrane Glycoproteins metabolism, Mice, Models, Biological, Platelet Membrane Glycoprotein IIb metabolism, Pluripotent Stem Cells drug effects, Pluripotent Stem Cells metabolism, Receptors, Complement metabolism, Recombinant Proteins administration & dosage, SOXF Transcription Factors genetics, SOXF Transcription Factors metabolism, Time Factors, Transcription Factors genetics, Transcription Factors metabolism, Vascular Endothelial Growth Factor A administration & dosage, Vascular Endothelial Growth Factor Receptor-2 metabolism, Embryonic Stem Cells cytology, Hematopoiesis physiology, Hematopoietic Stem Cells cytology, Pluripotent Stem Cells cytology
- Abstract
The efficient and reproducible generation of differentiated progenitors from pluripotent stem cells requires the recapitulation of appropriate developmental stages and pathways. Here, we have used the combination of activin A, BMP4 and VEGF under serum-free conditions to induce hematopoietic differentiation from both embryonic and induced pluripotent stem cells, with the aim of modeling the primary sites of embryonic hematopoiesis. We identified two distinct Flk1-positive hematopoietic populations that can be isolated based on temporal patterns of emergence. The earliest arising population displays characteristics of yolk sac hematopoiesis, whereas a late developing Flk1-positive population appears to reflect the para-aortic splanchnopleura hematopoietic program, as it has reduced primitive erythroid capacity and substantially enhanced myeloid and lymphoid potential compared with the earlier wave. These differences between the two populations are accompanied by differences in the expression of Sox17 and Hoxb4, as well as in the cell surface markers AA4.1 and CD41. Together, these findings support the interpretation that the two populations are representative of the early sites of mammalian hematopoiesis.
- Published
- 2010
- Full Text
- View/download PDF
47. Preferential localization of IgG memory B cells adjacent to contracted germinal centers.
- Author
-
Aiba Y, Kometani K, Hamadate M, Moriyama S, Sakaue-Sawano A, Tomura M, Luche H, Fehling HJ, Casellas R, Kanagawa O, Miyawaki A, and Kurosaki T
- Subjects
- ADP-ribosyl Cyclase 1 immunology, ADP-ribosyl Cyclase 1 metabolism, Animals, B-Lymphocytes cytology, B-Lymphocytes metabolism, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, Chickens, Flow Cytometry, Germinal Center metabolism, Immunization, Immunoglobulin G genetics, Immunoglobulin G metabolism, Immunohistochemistry, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Nitrophenols chemistry, Phenylacetates chemistry, Spleen cytology, Spleen immunology, Spleen metabolism, gamma-Globulins chemistry, gamma-Globulins immunology, B-Lymphocytes immunology, Germinal Center immunology, Immunoglobulin G immunology, Immunologic Memory immunology
- Abstract
It has long been presumed that after leaving the germinal centers (GCs), memory B cells colonize the marginal zone or join the recirculating pool. Here we demonstrate the preferential localization of nitrophenol-chicken gamma-globulin-induced CD38(+)IgG1(+) memory B cells adjacent to contracted GCs in the spleen. The memory B cells in this region proliferated after secondary immunization, a response that was abolished by depletion of CD4(+) T cells. We also found that these IgG1(+) memory B cells could present antigen on their surface, and that this activity was required for their activation. These results implicate this peri-GC region as an important site for survival and reactivation of memory B cells.
- Published
- 2010
- Full Text
- View/download PDF
48. Asynchronous RAG-1 expression during B lymphopoiesis.
- Author
-
Welner RS, Esplin BL, Garrett KP, Pelayo R, Luche H, Fehling HJ, and Kincade PW
- Subjects
- Animals, Antigens, Ly biosynthesis, Antigens, Ly genetics, B-Lymphocytes metabolism, Cell Differentiation genetics, Cell Differentiation immunology, Cell Lineage genetics, Cell Lineage immunology, Cells, Cultured, Gene Knock-In Techniques, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells immunology, Hematopoietic Stem Cells metabolism, Homeodomain Proteins physiology, Lymphopoiesis genetics, Membrane Proteins biosynthesis, Membrane Proteins genetics, Mice, Mice, Inbred C57BL, Mice, Transgenic, Proto-Oncogene Proteins c-kit biosynthesis, Proto-Oncogene Proteins c-kit genetics, Recombination, Genetic, B-Lymphocytes cytology, B-Lymphocytes immunology, Homeodomain Proteins biosynthesis, Homeodomain Proteins genetics, Lymphopoiesis immunology
- Abstract
Changes in cell surface markers and patterns of gene expression are commonly used to construct sequences of events in hematopoiesis. However, the order may not be as rigid as once thought and it is unclear which changes represent the best milestones of differentiation. We developed a fate-mapping model where cells with a history of RAG-1 expression are permanently marked by red fluorescence. This approach is valuable for appreciating lymphoid-lineage relationships without need for irradiation and transplantation. Hematopoietic stem cells (HSC) as well as myeloid and dendritic cell progenitors were unlabeled. Also as expected, most previously identified RAG-1(+) early lymphoid progenitors in bone marrow and all lymphoid-affiliated cells were marked. Of particular interest, there was heterogeneity among canonical common lymphoid progenitors (CLP) in bone marrow. Labeled CLP expressed slightly higher levels of IL-7Ralpha, displayed somewhat less c-Kit, and generated CD19(+) lymphocytes faster than the unlabeled CLP. Furthermore, CLP with a history of RAG-1 expression were much less likely to generate dendritic and NK cells. The RAG-1-marked CLP were lineage stable even when exposed to LPS, while unlabeled CLP were redirected to become dendritic cells in response to this TLR4 ligand. These findings indicate that essential events in B lymphopoiesis are not tightly synchronized. Some progenitors with increased probability of becoming lymphocytes express RAG-1 while still part of the lineage marker-negative Sca-1(+)c-Kit(high) (LSK) fraction. Other progenitors first activate this locus after c-Kit levels have diminished and cell surface IL-7 receptors are detectable.
- Published
- 2009
- Full Text
- View/download PDF
49. Intestinal lamina propria dendritic cell subsets have different origin and functions.
- Author
-
Varol C, Vallon-Eberhard A, Elinav E, Aychek T, Shapira Y, Luche H, Fehling HJ, Hardt WD, Shakhar G, and Jung S
- Subjects
- Animals, Antigens, CD immunology, CD11b Antigen immunology, Cell Differentiation, Cell Line, Tumor, Colitis immunology, Colitis pathology, Disease Models, Animal, Granulocyte-Macrophage Colony-Stimulating Factor immunology, Homeostasis, Integrin alpha Chains immunology, Mice, Mice, Inbred C57BL, Phenotype, Receptors, Interleukin-8A immunology, fms-Like Tyrosine Kinase 3 immunology, Cell Lineage, Dendritic Cells cytology, Dendritic Cells immunology, Intestinal Mucosa cytology, Intestinal Mucosa immunology
- Abstract
The intestinal immune system discriminates between tolerance toward the commensal microflora and robust responses to pathogens. Maintenance of this critical balance is attributed to mucosal dendritic cells (DCs) residing in organized lymphoid tissue and dispersed in the subepithelial lamina propria. In situ parameters of lamina propria DCs (lpDCs) remain poorly understood. Here, we combined conditional cell ablation and precursor-mediated in vivo reconstitution to establish that lpDC subsets have distinct origins and functions. CD103(+) CX(3)CR1(-) lpDCs arose from macrophage-DC precursors (MDPs) via DC-committed intermediates (pre-cDCs) through a Flt3L growth-factor-mediated pathway. CD11b(+) CD14(+) CX(3)CR1(+) lpDCs were derived from grafted Ly6C(hi) but not Ly6C(lo) monocytes under the control of GM-CSF. Mice reconstituted exclusively with CX(3)CR1(+) lpDCs when challenged in an innate colitis model developed severe intestinal inflammation that was driven by graft-derived TNF-alpha-secreting CX(3)CR1(+) lpDCs. Our results highlight the critical importance of the lpDC subset balance for robust gut homeostasis.
- Published
- 2009
- Full Text
- View/download PDF
50. The patch-like pattern of OR37 receptors is formed by turning off gene expression in non-appropriate areas.
- Author
-
Strotmann J, Bader A, Luche H, Fehling HJ, and Breer H
- Subjects
- Animals, Bacterial Proteins genetics, Integrases genetics, Integrases metabolism, Luminescent Proteins genetics, Mice, Mice, Transgenic, RNA, Messenger metabolism, Receptors, Odorant genetics, Gene Expression Regulation, Olfactory Bulb metabolism, Receptors, Odorant metabolism, Vomeronasal Organ metabolism
- Abstract
Olfactory sensory neurons (OSNs) expressing the same odorant receptor (OR) gene are generally widely dispersed throughout the olfactory epithelium (OE). In contrast, OSNs expressing any member from the special OR37 subfamily are concentrated in a small patch in the centre of the OE. To evaluate whether transcription of OR37 genes is only possible in the patch region, or if they can generally be chosen also in non-appropriate areas, a transgenic approach was employed that permanently labelled all cells which ever transcribed a representative OR37 gene. It was found that - in addition to cells inside the patch - numerous cells outside were labelled, indicating that they had transcribed the OR37 gene, but then turned it off while choosing another OR gene. Permanent expression of the OR37 gene was exclusively maintained in the patch. The results suggest that mechanisms acting downstream of an initial OR gene choice restrict OR37 expression to the patch.
- 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.