Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 34
Filtrer
Plus de filtres










Base de données
Gamme d'année
2.
Nat Commun ; 15(1): 4182, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38755157

RÉSUMÉ

Bone marrow plasma cells (BMPC) are the correlate of humoral immunity, consistently releasing antibodies into the bloodstream. It remains unclear if BMPC reflect different activation environments or maturation of their precursors. Here we define human BMPC heterogeneity and track the recruitment of antibody-secreting cells (ASC) from SARS-CoV-2 vaccine immune reactions to the bone marrow (BM). Trajectories based on single-cell transcriptomes and repertoires of peripheral and BM ASC reveal sequential colonisation of BMPC compartments. In activated B cells, IL-21 suppresses CD19 expression, indicating that CD19low-BMPC are derived from follicular, while CD19high-BMPC originate from extrafollicular immune reactions. In primary immune reactions, both CD19low- and CD19high-BMPC compartments are populated. In secondary immune reactions, most BMPC are recruited to CD19high-BMPC compartments, reflecting their origin from extrafollicular reactivations of memory B cells. A pattern also observable in vaccinated-convalescent individuals and upon diphtheria/tetanus/pertussis recall-vaccination. Thus, BMPC diversity reflects the evolution of a given humoral immune response.


Sujet(s)
Antigènes CD19 , Moelle osseuse , Interleukines , Plasmocytes , Humains , Plasmocytes/immunologie , Interleukines/immunologie , Interleukines/métabolisme , Moelle osseuse/immunologie , Antigènes CD19/immunologie , Antigènes CD19/métabolisme , Immunité humorale/immunologie , COVID-19/immunologie , COVID-19/virologie , SARS-CoV-2/immunologie , Cellules de la moelle osseuse/immunologie , Cellules de la moelle osseuse/cytologie , Analyse sur cellule unique , Adulte , Lymphocytes B/immunologie , Cellules productrices d'anticorps/immunologie , Femelle , Mâle , Vaccination , Adulte d'âge moyen , Vaccin diphtérie-tétanos-coqueluche/immunologie
3.
Nat Immunol ; 25(2): 256-267, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38172258

RÉSUMÉ

The pleiotropic alarmin interleukin-33 (IL-33) drives type 1, type 2 and regulatory T-cell responses via its receptor ST2. Subset-specific differences in ST2 expression intensity and dynamics suggest that transcriptional regulation is key in orchestrating the context-dependent activity of IL-33-ST2 signaling in T-cell immunity. Here, we identify a previously unrecognized alternative promoter in mice and humans that is located far upstream of the curated ST2-coding gene and drives ST2 expression in type 1 immunity. Mice lacking this promoter exhibit a selective loss of ST2 expression in type 1- but not type 2-biased T cells, resulting in impaired expansion of cytotoxic T cells (CTLs) and T-helper 1 cells upon viral infection. T-cell-intrinsic IL-33 signaling via type 1 promoter-driven ST2 is critical to generate a clonally diverse population of antiviral short-lived effector CTLs. Thus, lineage-specific alternative promoter usage directs alarmin responsiveness in T-cell subsets and offers opportunities for immune cell-specific targeting of the IL-33-ST2 axis in infections and inflammatory diseases.


Sujet(s)
Protéine-1 analogue au récepteur de l'interleukin-1 , Interleukine-33 , Animaux , Humains , Souris , Alarmines , Antiviraux , Protéine-1 analogue au récepteur de l'interleukin-1/génétique , Protéine-1 analogue au récepteur de l'interleukin-1/métabolisme , Interleukine-33/génétique , Sous-populations de lymphocytes T/métabolisme
4.
PLoS Biol ; 21(11): e3002015, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37983263

RÉSUMÉ

Throughout life, hematopoietic stem cells (HSCs), residing in bone marrow (BM), continuously regenerate erythroid/megakaryocytic, myeloid, and lymphoid cell lineages. This steady-state hematopoiesis from HSC and multipotent progenitors (MPPs) in BM can be perturbed by stress. The molecular controls of how stress can impact hematopoietic output remain poorly understood. MicroRNAs (miRNAs) as posttranscriptional regulators of gene expression have been found to control various functions in hematopoiesis. We find that the miR-221/222 cluster, which is expressed in HSC and in MPPs differentiating from them, perturbs steady-state hematopoiesis in ways comparable to stress. We compare pool sizes and single-cell transcriptomes of HSC and MPPs in unperturbed or stress-perturbed, miR-221/222-proficient or miR-221/222-deficient states. MiR-221/222 deficiency in hematopoietic cells was induced in C57BL/6J mice by conditional vav-cre-mediated deletion of the floxed miR-221/222 gene cluster. Social stress as well as miR-221/222 deficiency, alone or in combination, reduced HSC pools 3-fold and increased MPPs 1.5-fold. It also enhanced granulopoisis in the spleen. Furthermore, combined stress and miR-221/222 deficiency increased the erythroid/myeloid/granulocytic precursor pools in BM. Differential expression analyses of single-cell RNAseq transcriptomes of unperturbed and stressed, proficient HSC and MPPs detected more than 80 genes, selectively up-regulated in stressed cells, among them immediate early genes (IEGs). The same differential single-cell transcriptome analyses of unperturbed, miR-221/222-proficient with deficient HSC and MPPs identified Fos, Jun, JunB, Klf6, Nr4a1, Ier2, Zfp36-all IEGs-as well as CD74 and Ly6a as potential miRNA targets. Three of them, Klf6, Nr4a1, and Zfp36, have previously been found to influence myelogranulopoiesis. Together with increased levels of Jun, Fos forms increased amounts of the heterodimeric activator protein-1 (AP-1), which is known to control the expression of the selectively up-regulated expression of the IEGs. The comparisons of single-cell mRNA-deep sequencing analyses of socially stressed with miR-221/222-deficient HSC identify 5 of the 7 Fos/AP-1-controlled IEGs, Ier2, Jun, Junb, Klf6, and Zfp36, as common activators of HSC from quiescence. Combined with stress, miR-221/222 deficiency enhanced the Fos/AP-1/IEG pathway, extended it to MPPs, and increased the number of granulocyte precursors in BM, inducing selective up-regulation of genes encoding heat shock proteins Hspa5 and Hspa8, tubulin-cytoskeleton-organizing proteins Tuba1b, Tubb 4b and 5, and chromatin remodeling proteins H3f3b, H2afx, H2afz, and Hmgb2. Up-regulated in HSC, MPP1, and/or MPP2, they appear as potential regulators of stress-induced, miR-221/222-dependent increased granulocyte differentiation. Finally, stress by serial transplantations of miR-221/222-deficient HSC selectively exhausted their lymphoid differentiation capacities, while retaining their ability to home to BM and to differentiate to granulocytes. Thus, miR-221/222 maintains HSC quiescence and multipotency by suppressing Fos/AP-1/IEG-mediated activation and by suppressing enhanced stress-like differentiation to granulocytes. Since miR-221/222 is also expressed in human HSC, controlled induction of miR-221/222 in HSC should improve BM transplantations.


Sujet(s)
microARN , Facteur de transcription AP-1 , Animaux , Humains , Souris , Différenciation cellulaire , Granulocytes , Cellules souches hématopoïétiques , Souris de lignée C57BL , microARN/génétique , microARN/métabolisme , Facteur de transcription AP-1/métabolisme
5.
Proc Natl Acad Sci U S A ; 120(29): e2207993120, 2023 07 18.
Article de Anglais | MEDLINE | ID: mdl-37428931

RÉSUMÉ

Osteoarthritis (OA) is a joint disease featuring cartilage breakdown and chronic pain. Although age and joint trauma are prominently associated with OA occurrence, the trigger and signaling pathways propagating their pathogenic aspects are ill defined. Following long-term catabolic activity and traumatic cartilage breakdown, debris accumulates and can trigger Toll-like receptors (TLRs). Here we show that TLR2 stimulation suppressed the expression of matrix proteins and induced an inflammatory phenotype in human chondrocytes. Further, TLR2 stimulation impaired chondrocyte mitochondrial function, resulting in severely reduced adenosine triphosphate (ATP) production. RNA-sequencing analysis revealed that TLR2 stimulation upregulated nitric oxide synthase 2 (NOS2) expression and downregulated mitochondria function-associated genes. NOS inhibition partially restored the expression of these genes, and rescued mitochondrial function and ATP production. Correspondingly, Nos2-/- mice were protected from age-related OA development. Taken together, the TLR2-NOS axis promotes human chondrocyte dysfunction and murine OA development, and targeted interventions may provide therapeutic and preventive approaches in OA.


Sujet(s)
Cartilage articulaire , Arthrose , Humains , Souris , Animaux , Chondrocytes/métabolisme , Récepteur de type Toll-2/génétique , Récepteur de type Toll-2/métabolisme , Arthrose/métabolisme , Récepteurs de type Toll/métabolisme , Cartilage articulaire/métabolisme , Cellules cultivées
6.
Nat Commun ; 14(1): 791, 2023 02 11.
Article de Anglais | MEDLINE | ID: mdl-36774347

RÉSUMÉ

Prolonged lung pathology has been associated with COVID-19, yet the cellular and molecular mechanisms behind this chronic inflammatory disease are poorly understood. In this study, we combine advanced imaging and spatial transcriptomics to shed light on the local immune response in severe COVID-19. We show that activated adventitial niches are crucial microenvironments contributing to the orchestration of prolonged lung immunopathology. Up-regulation of the chemokines CCL21 and CCL18 associates to endothelial-to-mesenchymal transition and tissue fibrosis within these niches. CCL21 over-expression additionally links to the local accumulation of T cells expressing the cognate receptor CCR7. These T cells are imprinted with an exhausted phenotype and form lymphoid aggregates that can organize in ectopic lymphoid structures. Our work proposes immune-stromal interaction mechanisms promoting a self-sustained and non-resolving local immune response that extends beyond active viral infection and perpetuates tissue remodeling.


Sujet(s)
COVID-19 , Chimiokine CCL21 , Chimiokines CC , Humains , COVID-19/immunologie , Fibrose , Poumon , Lymphocytes T/immunologie
7.
Front Bioeng Biotechnol ; 10: 1046127, 2022.
Article de Anglais | MEDLINE | ID: mdl-36479429

RÉSUMÉ

The isolation of chondrocytes from human articular cartilage for single-cell RNA sequencing requires extensive and prolonged tissue digestion at 37 C. Modulations of the transcriptional activity likely take place during this period such that the transcriptomes of isolated human chondrocytes no longer match their original status in vivo. Here, we optimized the human chondrocyte isolation procedure to maximally preserve the in vivo transcriptome. Cartilage tissues were transferred into a hypoxia chamber (4% O2) immediately after being removed from OA patients and minced finely. Collagenase II at concentrations of 0.02%, 0.1%, 0.25%, 0.5%, 1%, and 2% was applied for 0.5, 1, 2, 4, and 18 h to digest the minced tissue. Actinomycin D (ActD) was added to test its capacity in stabilizing the transcriptome. Cell yield, viability, cell size, and transcriptome were determined using counter chamber, flow cytometry, and RNA sequencing (RNA-seq). Collagenase II at 2% concentration released small chondrocytes from cartilage matrix during the first digestion hour and started to release large cells thereafter, reaching a complete release at 4 h. During 4-h digestions, collagenase II at 2% and 1% but not at lower concentrations yielded maximal release also of the large chondrocyte population. RNA-seq analysis revealed that a 4-h digestion period with 1% or 2% collagenase II plus Actinomycin D optimally preserved the transcriptome. Thus, this study provides an isolation protocol for single chondrocytes from human articular cartilage optimized for transcriptome preservation and RNA-seq analysis.

8.
Nature ; 611(7937): 794-800, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-36323785

RÉSUMÉ

Protective immunity relies on the interplay of innate and adaptive immune cells with complementary and redundant functions. Innate lymphoid cells (ILCs) have recently emerged as tissue-resident, innate mirror images of the T cell system, with which they share lineage-specifying transcription factors and effector machinery1. Located at barrier surfaces, ILCs are among the first responders against invading pathogens and thus could potentially determine the outcome of the immune response2. However, so far it has not been possible to dissect the unique contributions of ILCs to protective immunity owing to limitations in specific targeting of ILC subsets. Thus, all of the available data have been generated either in mice lacking the adaptive immune system or with tools that also affect other immune cell subsets. In addition, it has been proposed that ILCs might be dispensable for a proper immune response because other immune cells could compensate for their absence3-7. Here we report the generation of a mouse model based on the neuromedin U receptor 1 (Nmur1) promoter as a driver for simultaneous expression of Cre recombinase and green fluorescent protein, which enables gene targeting in group 2 ILCs (ILC2s) without affecting other innate and adaptive immune cells. Using Cre-mediated gene deletion of Id2 and Gata3 in Nmur1-expressing cells, we generated mice with a selective and specific deficiency in ILC2s. ILC2-deficient mice have decreased eosinophil counts at steady state and are unable to recruit eosinophils to the airways in models of allergic asthma. Further, ILC2-deficient mice do not mount an appropriate immune and epithelial type 2 response, resulting in a profound defect in worm expulsion and a non-protective type 3 immune response. In total, our data establish non-redundant functions for ILC2s in the presence of adaptive immune cells at steady state and during disease and argue for a multilayered organization of the immune system on the basis of a spatiotemporal division of labour.


Sujet(s)
Système immunitaire , Immunité innée , Lymphocytes , Animaux , Souris , Asthme/génétique , Asthme/immunologie , Asthme/anatomopathologie , Modèles animaux de maladie humaine , Granulocytes éosinophiles/anatomopathologie , Immunité innée/immunologie , Lymphocytes/classification , Lymphocytes/immunologie , Protéines à fluorescence verte , Système immunitaire/cytologie , Système immunitaire/immunologie , Système immunitaire/anatomopathologie
10.
Nat Commun ; 13(1): 3059, 2022 06 01.
Article de Anglais | MEDLINE | ID: mdl-35650194

RÉSUMÉ

Bone growth requires a specialised, highly angiogenic blood vessel subtype, so-called type H vessels, which pave the way for osteoblasts surrounding these vessels. At the end of adolescence, type H vessels differentiate into quiescent type L endothelium lacking the capacity to promote bone growth. Until now, the signals that switch off type H vessel identity and thus limit adolescent bone growth have remained ill defined. Here we show that mechanical forces, associated with increased body weight at the end of adolescence, trigger the mechanoreceptor PIEZO1 and thereby mediate enhanced production of the kinase FAM20C in osteoblasts. FAM20C, the major kinase of the secreted phosphoproteome, phosphorylates dentin matrix protein 1, previously identified as a key factor in bone mineralization. Thereupon, dentin matrix protein 1 is secreted from osteoblasts in a burst-like manner. Extracellular dentin matrix protein 1 inhibits vascular endothelial growth factor signalling by preventing phosphorylation of vascular endothelial growth factor receptor 2. Hence, secreted dentin matrix protein 1 transforms type H vessels into type L to limit bone growth activity and enhance bone mineralization. The discovered mechanism may suggest new options for the treatment of diseases characterised by aberrant activity of bone and vessels such as osteoarthritis, osteoporosis and osteosarcoma.


Sujet(s)
Calcification physiologique , Néovascularisation physiologique , Contrainte mécanique , Adolescent , Développement osseux , Trame osseuse , Protéines de la matrice extracellulaire , Humains , Canaux ioniques , Morphogenèse , Phosphoprotéines , Facteur de croissance endothéliale vasculaire de type A , Récepteur-2 au facteur croissance endothéliale vasculaire
11.
Mucosal Immunol ; 15(3): 480-490, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-35169232

RÉSUMÉ

Immunosuppressive Interleukin (IL)-10 production by pro-inflammatory CD4+ T cells is a central self-regulatory function to limit aberrant inflammation. Still, the molecular mediators controlling IL-10 expression in human CD4+ T cells are largely undefined. Here, we identify a Notch/STAT3 signaling-module as a universal molecular switch to induce IL-10 expression across human naïve and major effector CD4+ T cell subsets. IL-10 induction was transient, jointly controlled by the transcription factors Blimp-1/c-Maf and accompanied by upregulation of several co-inhibitory receptors, including LAG-3, CD49b, PD-1, TIM-3 and TIGIT. Consistent with a protective role of IL-10 in inflammatory bowel diseases (IBD), effector CD4+ T cells from Crohn's disease patients were defective in Notch/STAT3-induced IL-10 production and skewed towards an inflammatory Th1/17 cell phenotype. Collectively, our data identify a Notch/STAT3-Blimp-1/c-Maf axis as a common anti-inflammatory pathway in human CD4+ T cells, which is defective in IBD and thus may represent an attractive therapeutic target.


Sujet(s)
Maladie de Crohn , Maladies inflammatoires intestinales , Animaux , Maladie de Crohn/métabolisme , Humains , Maladies inflammatoires intestinales/métabolisme , Interleukine-10/métabolisme , Souris , Souris knockout , Protéines proto-oncogènes c-maf/génétique , Protéines proto-oncogènes c-maf/métabolisme , Facteur de transcription STAT-3/métabolisme , Lymphocytes auxiliaires Th1/métabolisme
12.
Arthritis Rheumatol ; 74(6): 934-947, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-34962360

RÉSUMÉ

OBJECTIVE: Patients with autoimmune inflammatory rheumatic diseases receiving rituximab (RTX) therapy are at higher risk of poor COVID-19 outcomes and show substantially impaired humoral immune response to anti-SARS-CoV-2 vaccine. However, the complex relationship between antigen-specific B cells and T cells and the level of B cell repopulation necessary to achieve anti-vaccine responses remain largely unknown. METHODS: Antibody responses to SARS-CoV-2 vaccines and induction of antigen-specific B and CD4/CD8 T cell subsets were studied in 19 patients with rheumatoid arthritis (RA) or antineutrophil cytoplasmic antibody-associated vasculitis receiving RTX, 12 patients with RA receiving other therapies, and 30 healthy controls after SARS-CoV-2 vaccination with either messenger RNA or vector-based vaccines. RESULTS: A minimum of 10 B cells per microliter (0.4% of lymphocytes) in the peripheral circulation appeared to be required for RTX-treated patients to mount seroconversion to anti-S1 IgG upon SARS-CoV-2 vaccination. RTX-treated patients who lacked IgG seroconversion showed reduced receptor-binding domain-positive B cells (P = 0.0005), a lower frequency of Tfh-like cells (P = 0.0481), as well as fewer activated CD4 (P = 0.0036) and CD8 T cells (P = 0.0308) compared to RTX-treated patients who achieved IgG seroconversion. Functionally relevant B cell depletion resulted in impaired interferon-γ secretion by spike-specific CD4 T cells (P = 0.0112, r = 0.5342). In contrast, antigen-specific CD8 T cells were reduced in both RA patients and RTX-treated patients, independently of IgG formation. CONCLUSION: In RTX-treated patients, a minimum of 10 B cells per microliter in the peripheral circulation is a candidate biomarker for a high likelihood of an appropriate cellular and humoral response after SARS-CoV-2 vaccination. Mechanistically, the data emphasize the crucial role of costimulatory B cell functions for the proper induction of CD4 responses propagating vaccine-specific B cell and plasma cell differentiation.


Sujet(s)
Polyarthrite rhumatoïde , COVID-19 , Anticorps antiviraux , Polyarthrite rhumatoïde/traitement médicamenteux , COVID-19/prévention et contrôle , Vaccins contre la COVID-19/usage thérapeutique , Numération cellulaire , Humains , Immunité humorale , Immunoglobuline G , Rituximab/usage thérapeutique , SARS-CoV-2 , Vaccination/méthodes
13.
Nature ; 600(7888): 295-301, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34695836

RÉSUMÉ

SARS-CoV-2 is a single-stranded RNA virus that causes COVID-19. Given its acute and often self-limiting course, it is likely that components of the innate immune system play a central part in controlling virus replication and determining clinical outcome. Natural killer (NK) cells are innate lymphocytes with notable activity against a broad range of viruses, including RNA viruses1,2. NK cell function may be altered during COVID-19 despite increased representation of NK cells with an activated and adaptive phenotype3,4. Here we show that a decline in viral load in COVID-19 correlates with NK cell status and that NK cells can control SARS-CoV-2 replication by recognizing infected target cells. In severe COVID-19, NK cells show defects in virus control, cytokine production and cell-mediated cytotoxicity despite high expression of cytotoxic effector molecules. Single-cell RNA sequencing of NK cells over the time course of the COVID-19 disease spectrum reveals a distinct gene expression signature. Transcriptional networks of interferon-driven NK cell activation are superimposed by a dominant transforming growth factor-ß (TGFß) response signature, with reduced expression of genes related to cell-cell adhesion, granule exocytosis and cell-mediated cytotoxicity. In severe COVID-19, serum levels of TGFß peak during the first two weeks of infection, and serum obtained from these patients severely inhibits NK cell function in a TGFß-dependent manner. Our data reveal that an untimely production of TGFß is a hallmark of severe COVID-19 and may inhibit NK cell function and early control of the virus.


Sujet(s)
COVID-19/immunologie , Cellules tueuses naturelles/immunologie , SARS-CoV-2/immunologie , Facteur de croissance transformant bêta/immunologie , Atlas comme sujet , Régulation de l'expression des gènes/immunologie , Humains , Immunité innée , Grippe humaine/immunologie , Cellules tueuses naturelles/anatomopathologie , RNA-Seq , Analyse sur cellule unique , Facteurs temps , Facteur de croissance transformant bêta/sang , Charge virale/immunologie , Réplication virale/immunologie
14.
Am J Respir Crit Care Med ; 204(12): 1403-1417, 2021 12 15.
Article de Anglais | MEDLINE | ID: mdl-34534436

RÉSUMÉ

Rationale: Pulmonary sarcoidosis is generally presumed to be a T-helper cell type 1- and macrophage-driven disease. However, mouse models have recently revealed that chronically inflamed lung tissue can also comprise T follicular helper (Tfh)-like cells and represents a site of active T-cell/B-cell cooperation. Objectives: To assess the role of pulmonary Tfh- and germinal center-like lymphocytes in sarcoidosis. Methods: BAL fluid, lung tissue, and peripheral blood samples from patients with sarcoidosis were analyzed by flow cytometry, immunohistology, RNA sequencing, and in vitro T-cell/B-cell cooperation assays for phenotypic and functional characterization of germinal center-like reactions in inflamed tissue. Measurements and Main Results: We identified a novel population of Tfh-like cells characterized by high expression of the B helper molecules CD40L and IL-21 in BAL of patients with sarcoidosis. Transcriptome analysis further confirmed a phenotype that was both Tfh-like and tissue resident. BAL T cells provided potent help for B cells to differentiate into antibody-producing cells. In lung tissue, we observed large peribronchial infiltrates with T and B cells in close contact, and many IgA+ plasmablasts. Most clusters were nonectopic; that is, they did not contain follicular dendritic cells. Patients with sarcoidosis also showed elevated levels of PD-1high CXCR5- CD40Lhigh ICOShigh Tfh-like cells, but not classical CXCR5+ Tfh cells, in the blood. Conclusions: Active T-cell/B-cell cooperation and local production of potentially pathogenic antibodies in the inflamed lung represents a novel pathomechanism in sarcoidosis and should be considered from both diagnostic and therapeutic perspectives.


Sujet(s)
Lymphocytes B/immunologie , Centre germinatif/immunologie , Poumon/immunologie , Sarcoïdose pulmonaire/immunologie , Lymphocytes T auxiliaires folliculaires/immunologie , Adulte , Sujet âgé , Marqueurs biologiques/sang , Liquide de lavage bronchoalvéolaire/cytologie , Liquide de lavage bronchoalvéolaire/immunologie , Études cas-témoins , Femelle , Cytométrie en flux , Humains , Poumon/anatomopathologie , Mâle , Adulte d'âge moyen , Sarcoïdose pulmonaire/sang , Sarcoïdose pulmonaire/anatomopathologie
15.
Sci Immunol ; 6(60)2021 06 15.
Article de Anglais | MEDLINE | ID: mdl-34131023

RÉSUMÉ

Patients with kidney failure are at increased risk for SARS-CoV-2 infection making effective vaccinations a critical need. It is not known how well mRNA vaccines induce B and plasma cell responses in dialysis patients (DP) or kidney transplant recipients (KTR) compared to healthy controls (HC). We studied humoral and B cell responses of 35 HC, 44 DP and 40 KTR. Markedly impaired anti-BNT162b2 responses were identified among KTR and DP compared to HC. In DP, the response was delayed (3-4 weeks after boost) and reduced with anti-S1 IgG and IgA positivity in 70.5% and 68.2%, respectively. In contrast, KTR did not develop IgG responses except one patient who had a prior unrecognized infection and developed anti-S1 IgG. The majority of antigen-specific B cells (RBD+) were identified in the plasmablast or post-switch memory B cell compartments in HC, whereas RBD+ B cells were enriched among pre-switch and naïve B cells from DP and KTR. The frequency and absolute number of antigen-specific circulating plasmablasts in the cohort correlated with the Ig response, a characteristic not reported for other vaccinations. In conclusion, these data indicated that immunosuppression resulted in impaired protective immunity after mRNA vaccination, including Ig induction with corresponding generation of plasmablasts and memory B cells. Thus, there is an urgent need to improve vaccination protocols in patients after kidney transplantation or on chronic dialysis.


Sujet(s)
Anticorps antiviraux/sang , Vaccins contre la COVID-19/immunologie , COVID-19/prévention et contrôle , Sujet immunodéprimé , Transplantation rénale , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Anticorps antiviraux/immunologie , Vaccin BNT162 , COVID-19/immunologie , Femelle , Humains , Immunité humorale/effets des médicaments et des substances chimiques , Immunité humorale/immunologie , Mâle , Adulte d'âge moyen , Dialyse rénale , SARS-CoV-2 , Receveurs de transplantation
16.
Nat Commun ; 12(1): 1961, 2021 03 30.
Article de Anglais | MEDLINE | ID: mdl-33785765

RÉSUMÉ

The pathogenesis of severe COVID-19 reflects an inefficient immune reaction to SARS-CoV-2. Here we analyze, at the single cell level, plasmablasts egressed into the blood to study the dynamics of adaptive immune response in COVID-19 patients requiring intensive care. Before seroconversion in response to SARS-CoV-2 spike protein, peripheral plasmablasts display a type 1 interferon-induced gene expression signature; however, following seroconversion, plasmablasts lose this signature, express instead gene signatures induced by IL-21 and TGF-ß, and produce mostly IgG1 and IgA1. In the sustained immune reaction from COVID-19 patients, plasmablasts shift to the expression of IgA2, thereby reflecting an instruction by TGF-ß. Despite their continued presence in the blood, plasmablasts are not found in the lungs of deceased COVID-19 patients, nor does patient IgA2 binds to the dominant antigens of SARS-CoV-2. Our results thus suggest that, in severe COVID-19, SARS-CoV-2 triggers a chronic immune reaction that is instructed by TGF-ß, and is distracted from itself.


Sujet(s)
Anticorps antiviraux/immunologie , COVID-19/immunologie , SARS-CoV-2/immunologie , Facteur de croissance transformant bêta/immunologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , COVID-19/virologie , Femelle , Humains , Immunoglobuline A/immunologie , Immunoglobuline G/immunologie , Interleukines/immunologie , Mâle , Adulte d'âge moyen , Plasmocytes/immunologie , SARS-CoV-2/génétique , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/immunologie
17.
Allergy ; 76(4): 1109-1122, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-32895937

RÉSUMÉ

BACKGROUND: Vitamin A regulates the adaptive immune response and a modulatory impact on type I allergy is discussed. The cellular mechanisms are largely unknown. OBJECTIVE: To determine the vitamin A-responding specific lymphocyte reaction in vivo. METHODS: Antigen-specific B and T lymphocytes were analyzed in an adoptive transfer airway inflammation mouse model in response to 9-cis retinoic acid (9cRA) and after lymphocyte-specific genetic targeting of the receptor RARα. Flow cytometry, quantitative PCR, next-generation sequencing, and specific Ig-ELISA were used to characterize the cells functionally. RESULTS: Systemic 9cRA profoundly enhanced the specific IgA-secreting B-cell frequencies in the lung tissue and serum IgA while reducing serum IgE concentrations. RARα overexpression in antigen-specific B cells promoted differentiation into plasmablasts at the expense of germinal center B cells. In antigen-specific T cells, RARα strongly promoted the differentiation of T follicular helper cells followed by an enhanced germinal center response. CONCLUSIONS: 9cRA signaling via RARα impacts the allergen-specific immunoglobulin response directly by the differentiation of B cells and indirectly by promoting T follicular helper cells.


Sujet(s)
Hypersensibilité , Rétinol , Animaux , Différenciation cellulaire , Centre germinatif , Souris , Lymphocytes T auxiliaires folliculaires , Lymphocytes T auxiliaires
18.
Eur J Immunol ; 51(4): 915-929, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33296081

RÉSUMÉ

T lymphocytes accumulate in inflamed tissues of patients with chronic inflammatory diseases (CIDs) and express pro-inflammatory cytokines upon re-stimulation in vitro. Further, a significant genetic linkage to MHC genes suggests that T lymphocytes play an important role in the pathogenesis of CIDs including juvenile idiopathic arthritis (JIA). However, the functions of T lymphocytes in established disease remain elusive. Here we dissect the transcriptional and the clonal heterogeneity of synovial T lymphocytes in JIA patients by single-cell RNA sequencing combined with T cell receptor profiling on the same cells. We identify clonally expanded subpopulations of T lymphocytes expressing genes reflecting recent activation by antigen in situ. A PD-1+ TOX+ EOMES+ population of CD4+ T lymphocytes expressed immune regulatory genes and chemoattractant genes for myeloid cells. A PD-1+ TOX+ BHLHE40+ population of CD4+ , and a mirror population of CD8+ T lymphocytes expressed genes driving inflammation, and genes supporting B lymphocyte activation in situ. This analysis points out that multiple types of T lymphocytes have to be targeted for therapeutic regeneration of tolerance in arthritis.


Sujet(s)
Antigènes/immunologie , Arthrite juvénile/immunologie , Facteurs de transcription à motif basique hélice-boucle-hélice/immunologie , Protéines HMG/immunologie , Protéines à homéodomaine/immunologie , Récepteur-1 de mort cellulaire programmée/immunologie , Protéines à domaine boîte-T/immunologie , Lymphocytes T/immunologie , Arthrite juvénile/génétique , Arthrite juvénile/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Cellules cultivées , Analyse de profil d'expression de gènes/méthodes , Protéines HMG/métabolisme , Protéines à homéodomaine/métabolisme , Humains , Récepteur-1 de mort cellulaire programmée/métabolisme , RNA-Seq/méthodes , Récepteurs aux antigènes des cellules T/génétique , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/métabolisme , Analyse sur cellule unique/méthodes , Protéines à domaine boîte-T/métabolisme , Lymphocytes T/métabolisme , Transcriptome/génétique , Transcriptome/immunologie
19.
Immunity ; 53(5): 1015-1032.e8, 2020 11 17.
Article de Anglais | MEDLINE | ID: mdl-33207209

RÉSUMÉ

Solitary intestinal lymphoid tissues such as cryptopatches (CPs) and isolated lymphoid follicles (ILFs) constitute steady-state activation hubs containing group 3 innate lymphoid cells (ILC3) that continuously produce interleukin (IL)-22. The outer surface of CPs and ILFs is demarcated by a poorly characterized population of CD11c+ cells. Using genome-wide single-cell transcriptional profiling of intestinal mononuclear phagocytes and multidimensional flow cytometry, we found that CP- and ILF-associated CD11c+ cells were a transcriptionally distinct subset of intestinal cDCs, which we term CIA-DCs. CIA-DCs required programming by CP- and ILF-resident CCR6+ ILC3 via lymphotoxin-ß receptor signaling in cDCs. CIA-DCs differentially expressed genes associated with immunoregulation and were the major cellular source of IL-22 binding protein (IL-22BP) at steady state. Mice lacking CIA-DC-derived IL-22BP exhibited diminished expression of epithelial lipid transporters, reduced lipid resorption, and changes in body fat homeostasis. Our findings provide insight into the design principles of an immunoregulatory checkpoint controlling nutrient absorption.


Sujet(s)
Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Immunité innée , Sous-populations de lymphocytes/immunologie , Sous-populations de lymphocytes/métabolisme , Plaques de Peyer/cytologie , Plaques de Peyer/immunologie , Récepteurs aux interleukines/biosynthèse , Animaux , Marqueurs biologiques , Expression des gènes , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes , Immunophénotypage , Muqueuse intestinale/immunologie , Muqueuse intestinale/métabolisme , Métabolisme lipidique , Souris , Souris transgéniques , Petit ARN cytoplasmique/génétique , Récepteurs aux interleukines/génétique , Transduction du signal
20.
N Engl J Med ; 383(12): 1149-1155, 2020 09 17.
Article de Anglais | MEDLINE | ID: mdl-32937047

RÉSUMÉ

Daratumumab, a human monoclonal antibody that targets CD38, depletes plasma cells and is approved for the treatment of multiple myeloma. Long-lived plasma cells are implicated in the pathogenesis of systemic lupus erythematosus because they secrete autoantibodies, but they are unresponsive to standard immunosuppression. We describe the use of daratumumab that induced substantial clinical responses in two patients with life-threatening lupus, with the clinical responses sustained by maintenance therapy with belimumab, an antibody to B-cell activating factor. Significant depletion of long-lived plasma cells, reduction of interferon type I activity, and down-regulation of T-cell transcripts associated with chronic inflammation were documented. (Supported by the Deutsche Forschungsgemeinschaft and others.).


Sujet(s)
Antigènes CD38/antagonistes et inhibiteurs , Anticorps monoclonaux/usage thérapeutique , Immunosuppresseurs/usage thérapeutique , Lupus érythémateux disséminé/traitement médicamenteux , Glycoprotéines membranaires/antagonistes et inhibiteurs , Plasmocytes/effets des médicaments et des substances chimiques , Antigènes CD38/métabolisme , Adulte , Anticorps monoclonaux humanisés/usage thérapeutique , Créatinine/sang , Créatinine/urine , Régulation négative , Femelle , Humains , Interféron de type I/antagonistes et inhibiteurs , Chimiothérapie de maintenance , Glycoprotéines membranaires/métabolisme , Adulte d'âge moyen , Protéinurie , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE