Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Immunol ; 36: 339-357, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29356584

RESUMEN

Maintenance of immunological self-tolerance requires lymphocytes carrying self-reactive antigen receptors to be selectively prevented from mounting destructive or inflammatory effector responses. Classically, self-tolerance is viewed in terms of the removal, editing, or silencing of B and T cells that have formed self-reactive antigen receptors during their early development. However, B cells activated by foreign antigen can enter germinal centers (GCs), where they further modify their antigen receptor by somatic hypermutation (SHM) of their immunoglobulin genes. The inevitable emergence of activated, self-reactive GC B cells presents a unique challenge to the maintenance of self-tolerance that must be rapidly countered to avoid autoantibody production. Here we discuss current knowledge of the mechanisms that enforce B cell self-tolerance, with particular focus on the control of self-reactive GC B cells. We also consider how self-reactive GC B cells can escape self-tolerance to initiate autoantibody production or instead be redeemed via SHM and used in productive antibody responses.


Asunto(s)
Autoinmunidad , Linfocitos B/inmunología , Centro Germinal/inmunología , Animales , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Linfocitos B/metabolismo , Centro Germinal/metabolismo , Humanos , Tolerancia Inmunológica , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
2.
Cell ; 186(6): 1144-1161.e18, 2023 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-36868219

RESUMEN

Germinal centers (GCs) that form within lymphoid follicles during antibody responses are sites of massive cell death. Tingible body macrophages (TBMs) are tasked with apoptotic cell clearance to prevent secondary necrosis and autoimmune activation by intracellular self antigens. We show by multiple redundant and complementary methods that TBMs derive from a lymph node-resident, CD169-lineage, CSF1R-blockade-resistant precursor that is prepositioned in the follicle. Non-migratory TBMs use cytoplasmic processes to chase and capture migrating dead cell fragments using a "lazy" search strategy. Follicular macrophages activated by the presence of nearby apoptotic cells can mature into TBMs in the absence of GCs. Single-cell transcriptomics identified a TBM cell cluster in immunized lymph nodes which upregulated genes involved in apoptotic cell clearance. Thus, apoptotic B cells in early GCs trigger activation and maturation of follicular macrophages into classical TBMs to clear apoptotic debris and prevent antibody-mediated autoimmune diseases.


Asunto(s)
Centro Germinal , Ganglios Linfáticos , Macrófagos , Apoptosis , Linfocitos B , Ganglios Linfáticos/citología , Macrófagos/citología , Macrófagos/metabolismo
3.
Cell ; 184(5): 1330-1347.e13, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33636130

RESUMEN

Osteoclasts are large multinucleated bone-resorbing cells formed by the fusion of monocyte/macrophage-derived precursors that are thought to undergo apoptosis once resorption is complete. Here, by intravital imaging, we reveal that RANKL-stimulated osteoclasts have an alternative cell fate in which they fission into daughter cells called osteomorphs. Inhibiting RANKL blocked this cellular recycling and resulted in osteomorph accumulation. Single-cell RNA sequencing showed that osteomorphs are transcriptionally distinct from osteoclasts and macrophages and express a number of non-canonical osteoclast genes that are associated with structural and functional bone phenotypes when deleted in mice. Furthermore, genetic variation in human orthologs of osteomorph genes causes monogenic skeletal disorders and associates with bone mineral density, a polygenetic skeletal trait. Thus, osteoclasts recycle via osteomorphs, a cell type involved in the regulation of bone resorption that may be targeted for the treatment of skeletal diseases.


Asunto(s)
Resorción Ósea/patología , Osteoclastos/patología , Ligando RANK/metabolismo , Animales , Apoptosis , Resorción Ósea/metabolismo , Fusión Celular , Células Cultivadas , Humanos , Macrófagos/citología , Ratones , Osteocondrodisplasias/tratamiento farmacológico , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patología , Osteoclastos/metabolismo , Transducción de Señal
4.
Nat Immunol ; 24(3): 531-544, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36658240

RESUMEN

Immunoglobulin A (IgA) secretion by plasma cells, terminally differentiated B cells residing in the intestinal lamina propria, assures microbiome homeostasis and protects the host against enteric infections. Exposure to diet-derived and commensal-derived signals provides immune cells with organizing cues that instruct their effector function and dynamically shape intestinal immune responses at the mucosal barrier. Recent data have described metabolic and microbial inputs controlling T cell and innate lymphoid cell activation in the gut; however, whether IgA-secreting lamina propria plasma cells are tuned by local stimuli is completely unknown. Although antibody secretion is considered to be imprinted during B cell differentiation and therefore largely unaffected by environmental changes, a rapid modulation of IgA levels in response to intestinal fluctuations might be beneficial to the host. In the present study, we showed that dietary cholesterol absorption and commensal recognition by duodenal intestinal epithelial cells lead to the production of oxysterols, evolutionarily conserved lipids with immunomodulatory functions. Using conditional cholesterol 25-hydroxylase deleter mouse line we demonstrated that 7α,25-dihydroxycholesterol from epithelial cells is critical to restrain IgA secretion against commensal- and pathogen-derived antigens in the gut. Intestinal plasma cells sense oxysterols via the chemoattractant receptor GPR183 and couple their tissue positioning with IgA secretion. Our findings revealed a new mechanism linking dietary cholesterol and humoral immune responses centered around plasma cell localization for efficient mucosal protection.


Asunto(s)
Inmunidad Innata , Células Plasmáticas , Animales , Ratones , Colesterol en la Dieta , Células Epiteliales , Inmunoglobulina A , Mucosa Intestinal , Receptores Acoplados a Proteínas G , Intestinos
5.
Cell ; 180(5): 878-894.e19, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32059783

RESUMEN

Pathogenic autoantibodies arise in many autoimmune diseases, but it is not understood how the cells making them evade immune checkpoints. Here, single-cell multi-omics analysis demonstrates a shared mechanism with lymphoid malignancy in the formation of public rheumatoid factor autoantibodies responsible for mixed cryoglobulinemic vasculitis. By combining single-cell DNA and RNA sequencing with serum antibody peptide sequencing and antibody synthesis, rare circulating B lymphocytes making pathogenic autoantibodies were found to comprise clonal trees accumulating mutations. Lymphoma driver mutations in genes regulating B cell proliferation and V(D)J mutation (CARD11, TNFAIP3, CCND3, ID3, BTG2, and KLHL6) were present in rogue B cells producing the pathogenic autoantibody. Antibody V(D)J mutations conferred pathogenicity by causing the antigen-bound autoantibodies to undergo phase transition to insoluble aggregates at lower temperatures. These results reveal a pre-neoplastic stage in human lymphomagenesis and a cascade of somatic mutations leading to an iconic pathogenic autoantibody.


Asunto(s)
Autoanticuerpos/genética , Enfermedades Autoinmunes/genética , Linfocitos B/inmunología , Linfoma/genética , Animales , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Linfocitos B/patología , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Portadoras/genética , Evolución Clonal/genética , Evolución Clonal/inmunología , Ciclina D3/genética , Guanilato Ciclasa/genética , Humanos , Proteínas Inmediatas-Precoces/genética , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/inmunología , Proteínas Inhibidoras de la Diferenciación/genética , Linfoma/inmunología , Linfoma/patología , Ratones , Mutación/genética , Mutación/inmunología , Proteínas de Neoplasias/genética , Análisis de Secuencia de ADN/métodos , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteínas Supresoras de Tumor/genética , Recombinación V(D)J/genética
6.
Immunity ; 56(3): 562-575.e6, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36842431

RESUMEN

Dietary components and metabolites have a profound impact on immunity and inflammation. Here, we investigated how sensing of cholesterol metabolite oxysterols by γδ T cells impacts their tissue residency and function. We show that dermal IL-17-producing γδ T (Tγδ17) cells essential for skin-barrier homeostasis require oxysterols sensing through G protein receptor 183 (GPR183) for their development and inflammatory responses. Single-cell transcriptomics and murine reporter strains revealed that GPR183 on developing γδ thymocytes is needed for their maturation by sensing medullary thymic epithelial-cell-derived oxysterols. In the skin, basal keratinocytes expressing the oxysterol enzyme cholesterol 25-hydroxylase (CH25H) maintain dermal Tγδ17 cells. Diet-driven increases in oxysterols exacerbate Tγδ17-cell-mediated psoriatic inflammation, dependent on GPR183 on γδ T cells. Hence, cholesterol-derived oxysterols control spatially distinct but biologically linked processes of thymic education and peripheral function of dermal T cells, implicating diet as a focal parameter of dermal Tγδ17 cells.


Asunto(s)
Colesterol en la Dieta , Oxiesteroles , Humanos , Animales , Ratones , Oxiesteroles/metabolismo , Piel/metabolismo , Inflamación , Proteínas de Unión al GTP/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
7.
Nat Immunol ; 20(10): 1299-1310, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31534238

RESUMEN

Resisting and tolerating microbes are alternative strategies to survive infection, but little is known about the evolutionary mechanisms controlling this balance. Here genomic analyses of anatomically modern humans, extinct Denisovan hominins and mice revealed a TNFAIP3 allelic series with alterations in the encoded immune response inhibitor A20. Each TNFAIP3 allele encoded substitutions at non-catalytic residues of the ubiquitin protease OTU domain that diminished IκB kinase-dependent phosphorylation and activation of A20. Two TNFAIP3 alleles encoding A20 proteins with partial phosphorylation deficits seemed to be beneficial by increasing immunity without causing spontaneous inflammatory disease: A20 T108A;I207L, originating in Denisovans and introgressed in modern humans throughout Oceania, and A20 I325N, from an N-ethyl-N-nitrosourea (ENU)-mutagenized mouse strain. By contrast, a rare human TNFAIP3 allele encoding an A20 protein with 95% loss of phosphorylation, C243Y, caused spontaneous inflammatory disease in humans and mice. Analysis of the partial-phosphorylation A20 I325N allele in mice revealed diminished tolerance of bacterial lipopolysaccharide and poxvirus inoculation as tradeoffs for enhanced immunity.


Asunto(s)
Infecciones por Poxviridae/inmunología , Poxviridae/fisiología , Dominios Proteicos/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Alelos , Animales , Extinción Biológica , Humanos , Inmunidad , Inflamación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación Missense/genética , Fosforilación
8.
Immunity ; 55(8): 1414-1430.e5, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35896116

RESUMEN

Germinal centers (GCs), transient structures within B cell follicles and central to affinity maturation, require the coordinated behavior of T and B cells. IL-21, a pleiotropic T cell-derived cytokine, is key to GC biology through incompletely understood mechanisms. By genetically restricting production and receipt of IL-21 in vivo, we reveal how its independent actions on T and B cells combine to regulate the GC. IL-21 established the magnitude of the GC B cell response by promoting CD4+ T cell expansion and differentiation in a dose-dependent manner and with paracrine activity. Within GC, IL-21 specifically promoted B cell centroblast identity and, when bioavailability was high, plasma cell differentiation. Critically, these actions may occur irrespective of cognate T-B interactions, making IL-21 a general promoter of growth as distinct to a mediator of affinity-driven selection via synaptic delivery. This promiscuous activity of IL-21 explains the consequences of IL-21 deficiency on antibody-based immunity.


Asunto(s)
Sinapsis Inmunológicas , Linfocitos T Colaboradores-Inductores , Diferenciación Celular , Centro Germinal , Interleucinas
9.
Immunity ; 55(12): 2386-2404.e8, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36446385

RESUMEN

The association between cancer and autoimmune disease is unexplained, exemplified by T cell large granular lymphocytic leukemia (T-LGL) where gain-of-function (GOF) somatic STAT3 mutations correlate with co-existing autoimmunity. To investigate whether these mutations are the cause or consequence of CD8+ T cell clonal expansions and autoimmunity, we analyzed patients and mice with germline STAT3 GOF mutations. STAT3 GOF mutations drove the accumulation of effector CD8+ T cell clones highly expressing NKG2D, the receptor for stress-induced MHC-class-I-related molecules. This subset also expressed genes for granzymes, perforin, interferon-γ, and Ccl5/Rantes and required NKG2D and the IL-15/IL-2 receptor IL2RB for maximal accumulation. Leukocyte-restricted STAT3 GOF was sufficient and CD8+ T cells were essential for lethal pathology in mice. These results demonstrate that STAT3 GOF mutations cause effector CD8+ T cell oligoclonal accumulation and that these rogue cells contribute to autoimmune pathology, supporting the hypothesis that somatic mutations in leukemia/lymphoma driver genes contribute to autoimmune disease.


Asunto(s)
Enfermedades Autoinmunes , Leucemia Linfocítica Granular Grande , Animales , Ratones , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Linfocitos T CD8-positivos , Mutación con Ganancia de Función , Leucemia Linfocítica Granular Grande/genética , Leucemia Linfocítica Granular Grande/patología , Mutación , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
11.
Immunity ; 54(8): 1652-1664, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34380063

RESUMEN

Germinal center (GC) B cells are the source of the high-affinity, class-switched antibodies required for protective immunity. The unique biology of GC B cells involves iterative rounds of antibody gene somatic hypermutation coupled to multiple selection and differentiation pathways. Recent advances in areas such as single cell and gene editing technologies have shed new light upon these complex and dynamic processes. We review these findings here and integrate them into the current understanding of GC B cell replication and death, the retention of high-affinity and class-switched B cells in the GC, and differentiation into plasma and memory cell effectors. We also discuss how the biology of GC responses relates to vaccine effectiveness and outline current and future challenges in the field.


Asunto(s)
Linfocitos B/inmunología , Diferenciación Celular/inmunología , Centro Germinal/citología , Centro Germinal/inmunología , Cambio de Clase de Inmunoglobulina/inmunología , Afinidad de Anticuerpos/inmunología , Proliferación Celular , Humanos , Memoria Inmunológica/inmunología , Hipermutación Somática de Inmunoglobulina/inmunología , Vacunación
12.
Immunity ; 54(5): 988-1001.e5, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33857421

RESUMEN

Positive selection of high-affinity B cells within germinal centers (GCs) drives affinity maturation of antibody responses. Here, we examined the mechanism underlying the parallel transition from immunoglobulin M (IgM) to IgG. Early GCs contained mostly unswitched IgM+ B cells; IgG+ B cells subsequently increased in frequency, dominating GC responses 14-21 days after antigen challenge. Somatic hypermutation and generation of high-affinity clones occurred with equal efficiency among IgM+ and IgG+ GC B cells, and inactivation of Ig class-switch recombination did not prevent depletion of IgM+ GC B cells. Instead, high-affinity IgG+ GC B cells outcompeted high-affinity IgM+ GC B cells via a selective advantage associated with IgG antigen receptor structure but independent of the extended cytoplasmic tail. Thus, two parallel forms of GC B-cell-positive selection, based on antigen receptor variable and constant regions, respectively, operate in tandem to ensure high-affinity IgG antibodies predominate in mature serum antibody responses.


Asunto(s)
Linfocitos B/inmunología , Centro Germinal/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Animales , Formación de Anticuerpos/inmunología , Antígenos/inmunología , Femenino , Cambio de Clase de Inmunoglobulina/inmunología , Región Variable de Inmunoglobulina/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovinos/inmunología , Hipermutación Somática de Inmunoglobulina/inmunología
13.
Immunity ; 54(12): 2908-2921.e6, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34788600

RESUMEN

Viral mutations are an emerging concern in reducing SARS-CoV-2 vaccination efficacy. Second-generation vaccines will need to elicit neutralizing antibodies against sites that are evolutionarily conserved across the sarbecovirus subgenus. Here, we immunized mice containing a human antibody repertoire with diverse sarbecovirus receptor-binding domains (RBDs) to identify antibodies targeting conserved sites of vulnerability. Antibodies with broad reactivity against diverse clade B RBDs targeting the conserved class 4 epitope, with recurring IGHV/IGKV pairs, were readily elicited but were non-neutralizing. However, rare class 4 antibodies binding this conserved RBD supersite showed potent neutralization of SARS-CoV-2 and all variants of concern. Structural analysis revealed that the neutralizing ability of cross-reactive antibodies was reserved only for those with an elongated CDRH3 that extends the antiparallel beta-sheet RBD core and orients the antibody light chain to obstruct ACE2-RBD interactions. These results identify a structurally defined pathway for vaccine strategies eliciting escape-resistant SARS-CoV-2 neutralizing antibodies.


Asunto(s)
Betacoronavirus/fisiología , Vacunas contra la COVID-19/inmunología , Infecciones por Coronavirus/inmunología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Animales , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/metabolismo , Secuencia Conservada/genética , Evolución Molecular , Humanos , Inmunización , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Unión Proteica , Dominios Proteicos/genética , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Desarrollo de Vacunas
14.
Cell ; 162(4): 926-6.e1, 2015 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-26276638

RESUMEN

Dynamic interactions between B and T cells underpin the development of adaptive humoral immune responses to infections and vaccines. Recent advances in the molecular and spatiotemporal control of these interactions during primary responses have contributed greatly to elucidating the molecular pathogenesis of numerous immunodeficiency and autoimmune diseases. The next challenge is to determine how and where memory B and T cells interact during secondary responses to facilitate the rapid and robust response that characterizes anamnestic immunity.


Asunto(s)
Linfocitos B/metabolismo , Comunicación Celular , Linfocitos T/metabolismo , Animales , Formación de Anticuerpos , Linfocitos B/citología , Humanos , Memoria Inmunológica , Linfocitos T/citología , Linfocitos T Reguladores/citología
15.
Nature ; 608(7924): 757-765, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35948641

RESUMEN

The notion that mobile units of nucleic acid known as transposable elements can operate as genomic controlling elements was put forward over six decades ago1,2. However, it was not until the advancement of genomic sequencing technologies that the abundance and repertoire of transposable elements were revealed, and they are now known to constitute up to two-thirds of mammalian genomes3,4. The presence of DNA regulatory regions including promoters, enhancers and transcription-factor-binding sites within transposable elements5-8 has led to the hypothesis that transposable elements have been co-opted to regulate mammalian gene expression and cell phenotype8-14. Mammalian transposable elements include recent acquisitions and ancient transposable elements that have been maintained in the genome over evolutionary time. The presence of ancient conserved transposable elements correlates positively with the likelihood of a regulatory function, but functional validation remains an essential step to identify transposable element insertions that have a positive effect on fitness. Here we show that CRISPR-Cas9-mediated deletion of a transposable element-namely the LINE-1 retrotransposon Lx9c11-in mice results in an exaggerated and lethal immune response to virus infection. Lx9c11 is critical for the neogenesis of a non-coding RNA (Lx9c11-RegoS) that regulates genes of the Schlafen family, reduces the hyperinflammatory phenotype and rescues lethality in virus-infected Lx9c11-/- mice. These findings provide evidence that a transposable element can control the immune system to favour host survival during virus infection.


Asunto(s)
Elementos Transponibles de ADN , Interacciones Microbiota-Huesped , Inmunidad , Retroelementos , Virosis , Animales , Sistemas CRISPR-Cas/genética , Elementos Transponibles de ADN/genética , Elementos Transponibles de ADN/inmunología , Evolución Molecular , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Inmunidad/genética , Ratones , ARN no Traducido/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Retroelementos/genética , Retroelementos/inmunología , Virosis/genética , Virosis/inmunología
16.
Cell ; 150(1): 194-206, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22770220

RESUMEN

The differentiation of follicular dendritic cells (FDC) is essential to the remarkable microanatomic plasticity of lymphoid follicles. Here we show that FDC arise from ubiquitous perivascular precursors (preFDC) expressing platelet-derived growth factor receptor ß (PDGFRß). PDGFRß-Cre-driven reporter gene recombination resulted in FDC labeling, whereas conditional ablation of PDGFRß(+)-derived cells abolished FDC, indicating that FDC originate from PDGFRß(+) cells. Lymphotoxin-α-overexpressing prion protein (PrP)(+) kidneys developed PrP(+) FDC after transplantation into PrP(-) mice, confirming that preFDC exist outside lymphoid organs. Adipose tissue-derived PDGFRß(+) stromal-vascular cells responded to FDC maturation factors and, when transplanted into lymphotoxin ß receptor (LTßR)(-) kidney capsules, differentiated into Mfge8(+)CD21/35(+)FcγRIIß(+)PrP(+) FDC capable of trapping immune complexes and recruiting B cells. Spleens of lymphocyte-deficient mice contained perivascular PDGFRß(+) FDC precursors whose expansion required both lymphoid tissue inducer (LTi) cells and lymphotoxin. The ubiquity of preFDC and their strategic location at blood vessels may explain the de novo generation of organized lymphoid tissue at sites of lymphocytic inflammation.


Asunto(s)
Vasos Sanguíneos/citología , Células Dendríticas Foliculares/citología , Bazo/citología , Células Madre/citología , Animales , Linfocitos B/inmunología , Células Dendríticas Foliculares/inmunología , Células Dendríticas Foliculares/metabolismo , Centro Germinal/citología , Centro Germinal/inmunología , Centro Germinal/metabolismo , Inflamación/patología , Células Asesinas Naturales/inmunología , Ratones , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Organismos Libres de Patógenos Específicos , Bazo/metabolismo
17.
Immunity ; 47(6): 1142-1153.e4, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29262350

RESUMEN

Memory B cells (MBCs) and plasma cells (PCs) constitute the two cellular outputs of germinal center (GC) responses that together facilitate long-term humoral immunity. Although expression of the transcription factor BLIMP-1 identifies cells undergoing PC differentiation, no such marker exists for cells committed to the MBC lineage. Here, we report that the chemokine receptor CCR6 uniquely marks MBC precursors in both mouse and human GCs. CCR6+ GC B cells were highly enriched within the GC light zone (LZ), were the most quiescent of all GC B cells, exhibited a cell-surface phenotype and gene expression signature indicative of an MBC transition, and possessed the augmented response characteristics of MBCs. MBC precursors within the GC LZ predominantly possessed a low affinity for antigen but also included cells from within the high-affinity pool. These data indicate a fundamental dichotomy between the processes that drive MBC and PC differentiation during GC responses.


Asunto(s)
Centro Germinal/inmunología , Inmunidad Humoral , Células Plasmáticas/inmunología , Células Precursoras de Linfocitos B/inmunología , Receptores CCR6/inmunología , Animales , Antígeno B7-2/genética , Antígeno B7-2/inmunología , Diferenciación Celular , Linaje de la Célula/inmunología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Centro Germinal/citología , Humanos , Memoria Inmunológica , Inmunofenotipificación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Células Plasmáticas/citología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/inmunología , Células Precursoras de Linfocitos B/citología , Receptores CCR6/genética , Receptores CXCR4/genética , Receptores CXCR4/inmunología , Transducción de Señal
18.
Nat Immunol ; 14(5): 446-53, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23502855

RESUMEN

Spleen-resident dendritic cell (DC) populations occupy sentinel positions for the capture and presentation of blood-borne antigens. Here we found a difference in expression of the chemotactic receptor EBI2 (GPR183) on splenic DC subsets and that EBI2 regulated the positioning and homeostasis of DCs in the spleen. EBI2 and its main ligand, 7α,25-OHC, were required for the generation of the splenic CD4(+) DC subset and the localization of DCs in bridging channels. Absence of EBI2 from DCs resulted in defects in both the activation of CD4(+) T cells and the induction of antibody responses. Regulated expression of EBI2 on DC populations is therefore critical for the generation and correct positioning of splenic DCs and the initiation of immune responses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Quimiotaxis/inmunología , Células Dendríticas/inmunología , Activación de Linfocitos , Receptores Acoplados a Proteínas G/metabolismo , Bazo/inmunología , Animales , Formación de Anticuerpos/genética , Presentación de Antígeno/genética , Antígenos CD4/metabolismo , Movimiento Celular/genética , Movimiento Celular/inmunología , Células Cultivadas , Células Dendríticas/patología , Homeostasis/genética , Homeostasis/inmunología , Activación de Linfocitos/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptores Acoplados a Proteínas G/genética , Bazo/patología
19.
Immunity ; 42(4): 704-18, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25840682

RESUMEN

B helper follicular T (Tfh) cells are critical for long-term humoral immunity. However, it remains unclear how these cells are recruited and contribute to secondary immune responses. Here we show that primary Tfh cells segregate into follicular mantle (FM) and germinal center (GC) subpopulations that display distinct gene expression signatures. Restriction of the primary Tfh cell subpopulation in the GC was mediated by downregulation of chemotactic receptor EBI2. Following collapse of the GC, memory T cells persisted in the outer follicle where they scanned CD169(+) subcapsular sinus macrophages. Reactivation and intrafollicular expansion of these follicular memory T cells in the subcapsular region was followed by their extrafollicular dissemination via the lymphatic flow. These data suggest that Tfh cells integrate their antigen-experience history to focus T cell help within the GC during primary responses but act rapidly to provide systemic T cell help after re-exposure to the antigen.


Asunto(s)
Linfocitos B/citología , Linaje de la Célula/inmunología , Centro Germinal/citología , Inmunidad Humoral , Linfocitos T Colaboradores-Inductores/citología , Animales , Linfocitos B/inmunología , Diferenciación Celular , Linaje de la Célula/genética , Movimiento Celular/inmunología , Proliferación Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Centro Germinal/inmunología , Memoria Inmunológica , Ratones , Ratones Noqueados , Cultivo Primario de Células , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/inmunología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/genética , Lectina 1 Similar a Ig de Unión al Ácido Siálico/inmunología , Transducción de Señal , Linfocitos T Colaboradores-Inductores/inmunología
20.
Immunity ; 42(5): 890-902, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25979420

RESUMEN

The mechanistic links between genetic variation and autoantibody production in autoimmune disease remain obscure. Autoimmune lymphoproliferative syndrome (ALPS) is caused by inactivating mutations in FAS or FASL, with autoantibodies thought to arise through failure of FAS-mediated removal of self-reactive germinal center (GC) B cells. Here we show that FAS is in fact not required for this process. Instead, FAS inactivation led to accumulation of a population of unconventional GC B cells that underwent somatic hypermutation, survived despite losing antigen reactivity, and differentiated into a large population of plasma cells that included autoantibody-secreting clones. IgE(+) plasma cell numbers, in particular, increased after FAS inactivation and a major cohort of ALPS-affected patients were found to have hyper-IgE. We propose that these previously unidentified cells, designated "rogue GC B cells," are a major driver of autoantibody production and provide a mechanistic explanation for the linked production of IgE and autoantibodies in autoimmune disease.


Asunto(s)
Autoanticuerpos/inmunología , Linfocitos B/citología , Centro Germinal/citología , Centro Germinal/inmunología , Inmunoglobulina E/inmunología , Receptor fas/inmunología , Animales , Autoanticuerpos/biosíntesis , Linfocitos B/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoglobulina E/biosíntesis , Ratones , Reacción en Cadena de la Polimerasa , Receptor fas/deficiencia , Receptor fas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA