Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 969
Filtrar
1.
Nature ; 628(8008): 612-619, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38509366

RESUMEN

There is increasing interest in how immune cells in the meninges-the membranes that surround the brain and spinal cord-contribute to homeostasis and disease in the central nervous system1,2. The outer layer of the meninges, the dura mater, has recently been described to contain both innate and adaptive immune cells, and functions as a site for B cell development3-6. Here we identify organized lymphoid structures that protect fenestrated vasculature in the dura mater. The most elaborate of these dural-associated lymphoid tissues (DALT) surrounded the rostral-rhinal confluence of the sinuses and included lymphatic vessels. We termed this structure, which interfaces with the skull bone marrow and a comparable venous plexus at the skull base, the rostral-rhinal venolymphatic hub. Immune aggregates were present in DALT during homeostasis and expanded with age or after challenge with systemic or nasal antigens. DALT contain germinal centre B cells and support the generation of somatically mutated, antibody-producing cells in response to a nasal pathogen challenge. Inhibition of lymphocyte entry into the rostral-rhinal hub at the time of nasal viral challenge abrogated the generation of germinal centre B cells and class-switched plasma cells, as did perturbation of B-T cell interactions. These data demonstrate a lymphoid structure around vasculature in the dura mater that can sample antigens and rapidly support humoral immune responses after local pathogen challenge.


Asunto(s)
Duramadre , Inmunidad Humoral , Tejido Linfoide , Venas , Administración Intranasal , Antígenos/administración & dosificación , Antígenos/inmunología , Médula Ósea/inmunología , Sistema Nervioso Central/irrigación sanguínea , Sistema Nervioso Central/inmunología , Duramadre/irrigación sanguínea , Duramadre/inmunología , Centro Germinal/citología , Centro Germinal/inmunología , Vasos Linfáticos/inmunología , Tejido Linfoide/irrigación sanguínea , Tejido Linfoide/inmunología , Células Plasmáticas/inmunología , Cráneo/irrigación sanguínea , Linfocitos T/inmunología , Venas/fisiología , Humanos , Masculino , Femenino , Adulto , Persona de Mediana Edad , Animales , Ratones , Anciano de 80 o más Años
2.
Nature ; 627(8003): 399-406, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38448581

RESUMEN

Immune cells rely on transient physical interactions with other immune and non-immune populations to regulate their function1. To study these 'kiss-and-run' interactions directly in vivo, we previously developed LIPSTIC (labelling immune partnerships by SorTagging intercellular contacts)2, an approach that uses enzymatic transfer of a labelled substrate between the molecular partners CD40L and CD40 to label interacting cells. Reliance on this pathway limited the use of LIPSTIC to measuring interactions between CD4+ T helper cells and antigen-presenting cells, however. Here we report the development of a universal version of LIPSTIC (uLIPSTIC), which can record physical interactions both among immune cells and between immune and non-immune populations irrespective of the receptors and ligands involved. We show that uLIPSTIC can be used, among other things, to monitor the priming of CD8+ T cells by dendritic cells, reveal the steady-state cellular partners of regulatory T cells and identify germinal centre-resident T follicular helper cells on the basis of their ability to interact cognately with germinal centre B cells. By coupling uLIPSTIC with single-cell transcriptomics, we build a catalogue of the immune populations that physically interact with intestinal epithelial cells at the steady state and profile the evolution of the interactome of lymphocytic choriomeningitis virus-specific CD8+ T cells in multiple organs following systemic infection. Thus, uLIPSTIC provides a broadly useful technology for measuring and understanding cell-cell interactions across multiple biological systems.


Asunto(s)
Linfocitos B , Linfocitos T CD8-positivos , Comunicación Celular , Células Dendríticas , Células Epiteliales , Células T Auxiliares Foliculares , Linfocitos T Reguladores , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Comunicación Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Ligandos , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Células T Auxiliares Foliculares/citología , Células T Auxiliares Foliculares/inmunología , Linfocitos B/citología , Linfocitos B/inmunología , Centro Germinal/citología , Análisis de Expresión Génica de una Sola Célula , Células Epiteliales/citología , Células Epiteliales/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Especificidad de Órganos
3.
Nature ; 627(8003): 407-415, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38383779

RESUMEN

Neuromyelitis optica is a paradigmatic autoimmune disease of the central nervous system, in which the water-channel protein AQP4 is the target antigen1. The immunopathology in neuromyelitis optica is largely driven by autoantibodies to AQP42. However, the T cell response that is required for the generation of these anti-AQP4 antibodies is not well understood. Here we show that B cells endogenously express AQP4 in response to activation with anti-CD40 and IL-21 and are able to present their endogenous AQP4 to T cells with an AQP4-specific T cell receptor (TCR). A population of thymic B cells emulates a CD40-stimulated B cell transcriptome, including AQP4 (in mice and humans), and efficiently purges the thymic TCR repertoire of AQP4-reactive clones. Genetic ablation of Aqp4 in B cells rescues AQP4-specific TCRs despite sufficient expression of AQP4 in medullary thymic epithelial cells, and B-cell-conditional AQP4-deficient mice are fully competent to raise AQP4-specific antibodies in productive germinal-centre responses. Thus, the negative selection of AQP4-specific thymocytes is dependent on the expression and presentation of AQP4 by thymic B cells. As AQP4 is expressed in B cells in a CD40-dependent (but not AIRE-dependent) manner, we propose that thymic B cells might tolerize against a group of germinal-centre-associated antigens, including disease-relevant autoantigens such as AQP4.


Asunto(s)
Acuaporina 4 , Autoanticuerpos , Autoantígenos , Linfocitos B , Tolerancia Inmunológica , Neuromielitis Óptica , Animales , Humanos , Ratones , Proteína AIRE , Acuaporina 4/deficiencia , Acuaporina 4/genética , Acuaporina 4/inmunología , Acuaporina 4/metabolismo , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Antígenos CD40/inmunología , Centro Germinal/citología , Centro Germinal/inmunología , Neuromielitis Óptica/inmunología , Neuromielitis Óptica/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Timo/citología , Timo/inmunología , Células Epiteliales Tiroideas/inmunología , Células Epiteliales Tiroideas/metabolismo , Transcriptoma
4.
J Virol ; 97(12): e0100823, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-37962378

RESUMEN

IMPORTANCE: The human gammaherpesviruses Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus are etiologic agents of numerous B cell lymphomas. A hallmark of gammaherpesvirus infection is their ability to establish lifelong latency in B cells. However, the specific mechanisms that mediate chronic infection in B cells in vivo remain elusive. Cellular E3 ubiquitin ligases regulate numerous biological processes by catalyzing ubiquitylation and modifying protein location, function, or half-life. Many viruses hijack host ubiquitin ligases to evade antiviral host defense and promote viral fitness. Here, we used the murine gammaherpesvirus 68 in vivo system to demonstrate that the E3 ligase Cul4b is essential for this virus to establish latency in germinal center B cells. These findings highlight an essential role for this E3 ligase in promoting chronic gammaherpesvirus infection in vivo and suggest that targeted inhibition of E3 ligases may provide a novel and effective intervention strategy against gammaherpesvirus-associated diseases.


Asunto(s)
Linfocitos B , Gammaherpesvirinae , Infecciones por Herpesviridae , Infección Persistente , Animales , Ratones , Linfocitos B/enzimología , Linfocitos B/metabolismo , Linfocitos B/virología , Proteínas Cullin/metabolismo , Gammaherpesvirinae/fisiología , Centro Germinal/citología , Centro Germinal/virología , Infecciones por Herpesviridae/enzimología , Infecciones por Herpesviridae/virología , Infección Persistente/enzimología , Infección Persistente/virología , Ubiquitinas/metabolismo , Latencia del Virus
5.
Nature ; 617(7961): 592-598, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37011668

RESUMEN

The primary two-dose SARS-CoV-2 mRNA vaccine series are strongly immunogenic in humans, but the emergence of highly infectious variants necessitated additional doses and the development of vaccines aimed at the new variants1-4. SARS-CoV-2 booster immunizations in humans primarily recruit pre-existing memory B cells5-9. However, it remains unclear whether the additional doses induce germinal centre reactions whereby re-engaged B cells can further mature, and whether variant-derived vaccines can elicit responses to variant-specific epitopes. Here we show that boosting with an mRNA vaccine against the original monovalent SARS-CoV-2 mRNA vaccine or the bivalent B.1.351 and B.1.617.2 (Beta/Delta) mRNA vaccine induced robust spike-specific germinal centre B cell responses in humans. The germinal centre response persisted for at least eight weeks, leading to significantly more mutated antigen-specific bone marrow plasma cell and memory B cell compartments. Spike-binding monoclonal antibodies derived from memory B cells isolated from individuals boosted with either the original SARS-CoV-2 spike protein, bivalent Beta/Delta vaccine or a monovalent Omicron BA.1-based vaccine predominantly recognized the original SARS-CoV-2 spike protein. Nonetheless, using a more targeted sorting approach, we isolated monoclonal antibodies that recognized the BA.1 spike protein but not the original SARS-CoV-2 spike protein from individuals who received the mRNA-1273.529 booster; these antibodies were less mutated and recognized novel epitopes within the spike protein, suggesting that they originated from naive B cells. Thus, SARS-CoV-2 booster immunizations in humans induce robust germinal centre B cell responses and can generate de novo B cell responses targeting variant-specific epitopes.


Asunto(s)
Linfocitos B , Vacunas contra la COVID-19 , COVID-19 , Centro Germinal , Inmunización Secundaria , Humanos , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19/inmunología , COVID-19/prevención & control , COVID-19/virología , Vacunas contra la COVID-19/administración & dosificación , Vacunas contra la COVID-19/inmunología , SARS-CoV-2/genética , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Linfocitos B/citología , Linfocitos B/inmunología , Centro Germinal/citología , Centro Germinal/inmunología , Células Plasmáticas/citología , Células Plasmáticas/inmunología , Células B de Memoria/citología , Células B de Memoria/inmunología , Epítopos de Linfocito B/genética , Epítopos de Linfocito B/inmunología
6.
Cell ; 186(1): 131-146.e13, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-36565697

RESUMEN

Germinal centers (GCs) form in secondary lymphoid organs in response to infection and immunization and are the source of affinity-matured B cells. The duration of GC reactions spans a wide range, and long-lasting GCs (LLGCs) are potentially a source of highly mutated B cells. We show that rather than consisting of continuously evolving B cell clones, LLGCs elicited by influenza virus or SARS-CoV-2 infection in mice are sustained by progressive replacement of founder clones by naive-derived invader B cells that do not detectably bind viral antigens. Rare founder clones that resist replacement for long periods are enriched in clones with heavily mutated immunoglobulins, including some with very high affinity for antigen, that can be recalled by boosting. Our findings reveal underappreciated aspects of the biology of LLGCs generated by respiratory virus infection and identify clonal replacement as a potential constraint on the development of highly mutated antibodies within these structures.


Asunto(s)
Linfocitos B , Centro Germinal , Infecciones por Virus ARN , Animales , Ratones , Linfocitos B/citología , Linfocitos B/inmunología , Células Clonales , COVID-19 , Centro Germinal/citología , Centro Germinal/inmunología , SARS-CoV-2 , Gripe Humana , Infecciones por Virus ARN/inmunología , Infecciones por Virus ARN/patología , Infecciones por Virus ARN/virología
7.
Nature ; 613(7945): 735-742, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36473496

RESUMEN

Feedback inhibition of humoral immunity by antibodies was first documented in 19091. Subsequent studies showed that, depending on the context, antibodies can enhance or inhibit immune responses2,3. However, little is known about how pre-existing antibodies influence the development of memory B cells. Here we examined the memory B cell response in individuals who received two high-affinity anti-SARS-CoV-2 monoclonal antibodies and subsequently two doses of an mRNA vaccine4-8. We found that the recipients of the monoclonal antibodies produced antigen-binding and neutralizing titres that were only fractionally lower compared than in control individuals. However, the memory B cells of the individuals who received the monoclonal antibodies differed from those of control individuals in that they predominantly expressed low-affinity IgM antibodies that carried small numbers of somatic mutations and showed altered receptor binding domain (RBD) target specificity, consistent with epitope masking. Moreover, only 1 out of 77 anti-RBD memory antibodies tested neutralized the virus. The mechanism underlying these findings was examined in experiments in mice that showed that germinal centres formed in the presence of the same antibodies were dominated by low-affinity B cells. Our results indicate that pre-existing high-affinity antibodies bias germinal centre and memory B cell selection through two distinct mechanisms: (1) by lowering the activation threshold for B cells, thereby permitting abundant lower-affinity clones to participate in the immune response; and (2) through direct masking of their cognate epitopes. This may in part explain the shifting target profile of memory antibodies elicited by booster vaccinations9.


Asunto(s)
Anticuerpos Antivirales , Linfocitos B , Vacunas contra la COVID-19 , COVID-19 , Retroalimentación Fisiológica , Memoria Inmunológica , Vacunación , Vacunas de ARNm , Animales , Ratones , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19/inmunología , COVID-19/prevención & control , COVID-19/terapia , COVID-19/virología , SARS-CoV-2/inmunología , Vacunas de ARNm/inmunología , Vacunas contra la COVID-19/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Epítopos de Linfocito B/química , Epítopos de Linfocito B/inmunología , Inmunoglobulina M/inmunología , Centro Germinal/citología , Centro Germinal/inmunología , Inmunización Secundaria , Hipermutación Somática de Inmunoglobulina
8.
Nature ; 609(7929): 998-1004, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36131022

RESUMEN

Germinal centres are the engines of antibody evolution. Here, using human immunodeficiency virus (HIV) Env protein immunogen priming in rhesus monkeys followed by a long period without further immunization, we demonstrate germinal centre B (BGC) cells that last for at least 6 months. A 186-fold increase in BGC cells was present by week 10 compared with conventional immunization. Single-cell transcriptional profiling showed that both light- and dark-zone germinal centre states were sustained. Antibody somatic hypermutation of BGC cells continued to accumulate throughout the 29-week priming period, with evidence of selective pressure. Env-binding BGC cells were still 49-fold above baseline at 29 weeks, which suggests that they could remain active for even longer periods of time. High titres of HIV-neutralizing antibodies were generated after a single booster immunization. Fully glycosylated HIV trimer protein is a complex antigen, posing considerable immunodominance challenges for B cells1,2. Memory B cells generated under these long priming conditions had higher levels of antibody somatic hypermutation, and both memory B cells and antibodies were more likely to recognize non-immunodominant epitopes. Numerous BGC cell lineage phylogenies spanning more than the 6-month germinal centre period were identified, demonstrating continuous germinal centre activity and selection for at least 191 days with no further antigen exposure. A long-prime, slow-delivery (12 days) immunization approach holds promise for difficult vaccine targets and suggests that patience can have great value for tuning of germinal centres to maximize antibody responses.


Asunto(s)
Afinidad de Anticuerpos , Linfocitos B , Movimiento Celular , Células Clonales , Centro Germinal , Anticuerpos Anti-VIH , Inmunización , Animales , Anticuerpos Neutralizantes/genética , Anticuerpos Neutralizantes/inmunología , Afinidad de Anticuerpos/genética , Afinidad de Anticuerpos/inmunología , Linfocitos B/citología , Linfocitos B/inmunología , Células Clonales/citología , Células Clonales/inmunología , Epítopos de Linfocito B/inmunología , Perfilación de la Expresión Génica , Centro Germinal/citología , Centro Germinal/inmunología , Anticuerpos Anti-VIH/genética , Anticuerpos Anti-VIH/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Humanos , Inmunización Secundaria , Macaca mulatta/inmunología , Macaca mulatta/virología , Células B de Memoria/citología , Células B de Memoria/inmunología , Análisis de la Célula Individual , Hipermutación Somática de Inmunoglobulina/genética , Hipermutación Somática de Inmunoglobulina/inmunología , Factores de Tiempo , Productos del Gen env del Virus de la Inmunodeficiencia Humana/administración & dosificación , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología
9.
Nature ; 608(7924): 724-732, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35948631

RESUMEN

The lymphocyte genome is prone to many threats, including programmed mutation during differentiation1, antigen-driven proliferation and residency in diverse microenvironments. Here, after developing protocols for expansion of single-cell lymphocyte cultures, we sequenced whole genomes from 717 normal naive and memory B and T cells and haematopoietic stem cells. All lymphocyte subsets carried more point mutations and structural variants than haematopoietic stem cells, with higher burdens in memory cells than in naive cells, and with T cells accumulating mutations at a higher rate throughout life. Off-target effects of immunological diversification accounted for approximately half of the additional differentiation-associated mutations in lymphocytes. Memory B cells acquired, on average, 18 off-target mutations genome-wide for every on-target IGHV mutation during the germinal centre reaction. Structural variation was 16-fold higher in lymphocytes than in stem cells, with around 15% of deletions being attributable to off-target recombinase-activating gene activity. DNA damage from ultraviolet light exposure and other sporadic mutational processes generated hundreds to thousands of mutations in some memory cells. The mutation burden and signatures of normal B cells were broadly similar to those seen in many B-cell cancers, suggesting that malignant transformation of lymphocytes arises from the same mutational processes that are active across normal ontogeny. The mutational landscape of normal lymphocytes chronicles the off-target effects of programmed genome engineering during immunological diversification and the consequences of differentiation, proliferation and residency in diverse microenvironments.


Asunto(s)
Linfocitos , Mutación , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/patología , Diferenciación Celular , Proliferación Celular , Microambiente Celular , Daño del ADN/genética , Daño del ADN/efectos de la radiación , Centro Germinal/citología , Centro Germinal/inmunología , Humanos , Memoria Inmunológica/genética , Linfocitos/citología , Linfocitos/inmunología , Linfocitos/metabolismo , Linfocitos/patología , Neoplasias/genética , Neoplasias/patología
10.
Biochem Biophys Res Commun ; 618: 86-92, 2022 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-35716600

RESUMEN

The transcription factor Bach2 serves as a crucial regulator of the germinal center (GC) reaction, which is required for production of high-affinity antibodies and establishment of long-lived B cell memory. However, the stage at which Bach2 controls the GC programs and the precise mechanism underlying these processes remain poorly understood. In this study, we show that genetic ablation of Bach2 in GC B cells of mice impairs their survival and maintenance, and memory B cell formation. These defects can be rescued by enforced expression of anti-apoptotic gene Bcl2. As expected, Bach2-deficient GC B cells are defective in antibody affinity maturation, but have normal somatic hyper mutation and class switch recombination of immunoglobulin genes. Mechanistically, Bach2 controls the GC programs by directly repressing pro-apoptotic gene Bim and a set of genes involved in cell stress response and metabolic processes. Thus, our work reveals the precise roles of Bach2 in the GC biology, and demonstrates that Bach2 acts as a crucial survival regulator of GC B cells, providing a key mechanism underlying GC B maintenance and B cell memory formation.


Asunto(s)
Linfocitos B , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Centro Germinal , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Supervivencia Celular/fisiología , Centro Germinal/citología , Centro Germinal/inmunología , Memoria Inmunológica , Ratones , Factores de Transcripción/metabolismo
12.
Nature ; 604(7904): 141-145, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35168246

RESUMEN

Germinal centres (GC) are lymphoid structures in which B cells acquire affinity-enhancing somatic hypermutations (SHM), with surviving clones differentiating into memory B cells (MBCs) and long-lived bone marrow plasma cells1-5 (BMPCs). SARS-CoV-2 mRNA vaccination induces a persistent GC response that lasts for at least six months in humans6-8. The fate of responding GC B cells as well as the functional consequences of such persistence remain unknown. Here, we detected SARS-CoV-2 spike protein-specific MBCs in 42 individuals who had received two doses of the SARS-CoV-2 mRNA vaccine BNT162b2 six month earlier. Spike-specific IgG-secreting BMPCs were detected in 9 out of 11 participants. Using a combined approach of sequencing the B cell receptors of responding blood plasmablasts and MBCs, lymph node GC B cells and plasma cells and BMPCs from eight individuals and expression of the corresponding monoclonal antibodies, we tracked the evolution of 1,540 spike-specific B cell clones. On average, early blood spike-specific plasmablasts exhibited the lowest SHM frequencies. By contrast, SHM frequencies of spike-specific GC B cells increased by 3.5-fold within six months after vaccination. Spike-specific MBCs and BMPCs accumulated high levels of SHM, which corresponded with enhanced anti-spike antibody avidity in blood and enhanced affinity as well as neutralization capacity of BMPC-derived monoclonal antibodies. We report how the notable persistence of the GC reaction induced by SARS-CoV-2 mRNA vaccination in humans culminates in affinity-matured long-term antibody responses that potently neutralize the virus.


Asunto(s)
Linfocitos B , Vacuna BNT162 , Centro Germinal , Vacunación , Anticuerpos Monoclonales , Anticuerpos Antivirales , Linfocitos B/citología , Linfocitos B/inmunología , Vacuna BNT162/administración & dosificación , Vacuna BNT162/inmunología , COVID-19/inmunología , COVID-19/prevención & control , COVID-19/virología , Centro Germinal/citología , Centro Germinal/inmunología , Humanos , ARN Mensajero/genética , SARS-CoV-2/genética , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología
13.
J Immunol ; 208(5): 1057-1065, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35149531

RESUMEN

T follicular regulatory (Tfr) cells are a subset of CD4+ T cells that express CXCR5 and migrate into germinal centers (GCs). They regulate GC reactions by communicating with T follicular helper (Tfh) and B cells. TNF inhibitors are used in inflammatory diseases; however, the generation of autoantibodies or anti-drug Abs sometimes causes problems. Because TNFR2 signaling is important for suppressive functions of regulatory T cells, we investigated the role of TNFR2 on human Tfr cells. Tfr cells stimulated with MR2-1 (an anti-TNFR2 agonistic Ab) were analyzed for cell proliferation, Foxp3 expression, and surface molecules. Tfh/B cell proliferation, IgM production, and differentiation in cocultures with MR2-1-stimulated Tfr cells were examined. Tfr cells express a high level of TNFR2. MR2-1 stimulation altered the gene expression profile of Tfr cells. Cell proliferation and Foxp3 expression of Tfr cells were enhanced by MR2-1. MR2-1-stimulated Tfr cells expressed ICOS and Programmed cell death protein 1 and significantly suppressed Tfh/B cell proliferation, IgM production, and B cell differentiation. TNFR2-stimulated Tfr cells retained the migration function according to the CXCL13 gradient. In conclusion, we showed that TNFR2-stiumulated Tfr cells can regulate Tfh and B cells. Aberrant antibody production during TNF inhibitor treatment might be, at least in part, associated with TNFR2 signaling inhibition in Tfr cells. In addition, expansion and maturation of Tfr cells via TNFR2 stimulation in vitro may be useful for a cell-based therapy in inflammatory and autoimmune diseases to control GC reactions.


Asunto(s)
Linfocitos B/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Células T Auxiliares Foliculares/inmunología , Linfocitos T Reguladores/inmunología , Enfermedades Autoinmunes/terapia , Linfocitos B/citología , Antígeno B7-H1/metabolismo , Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Proliferación Celular , Quimiocina CXCL13/metabolismo , Factores de Transcripción Forkhead/biosíntesis , Perfilación de la Expresión Génica , Centro Germinal/citología , Humanos , Inmunoglobulina M/biosíntesis , Proteína Coestimuladora de Linfocitos T Inducibles/biosíntesis , Activación de Linfocitos/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Receptores CXCR5/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/antagonistas & inhibidores , Transducción de Señal/inmunología , Factores de Necrosis Tumoral/metabolismo
14.
Cell Rep ; 38(8): 110399, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35139367

RESUMEN

Follicular helper T (Tfh) cells promote, whereas follicular regulatory T (Tfr) cells restrain, germinal center (GC) reactions. However, the precise roles of these cells in the complex GC reaction remain poorly understood. Here, we perturb Tfh or Tfr cells after SARS-CoV-2 spike protein vaccination in mice. We find that Tfh cells promote the frequency and somatic hypermutation (SHM) of Spike-specific GC B cells and regulate clonal diversity. Tfr cells similarly control SHM and clonal diversity in the GC but do so by limiting clonal competition. In addition, deletion of Tfh or Tfr cells during primary vaccination results in changes in SHM after vaccine boosting. Aged mice, which have altered Tfh and Tfr cells, have lower GC responses, presenting a bimodal distribution of SHM. Together, these data demonstrate that GC responses to SARS-CoV-2 spike protein vaccines require a fine balance of positive and negative follicular T cell help to optimize humoral immunity.


Asunto(s)
COVID-19/prevención & control , Centro Germinal/inmunología , Glicoproteína de la Espiga del Coronavirus/administración & dosificación , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Envejecimiento , Animales , Anticuerpos Antivirales/sangre , Linfocitos B/inmunología , Linfocitos B/metabolismo , COVID-19/virología , Centro Germinal/citología , Centro Germinal/metabolismo , Inmunidad Humoral , Ratones , Ratones Endogámicos C57BL , SARS-CoV-2/inmunología , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/inmunología , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Vacunación , Vacunas de Subunidad/inmunología
15.
Front Immunol ; 12: 744573, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34925321

RESUMEN

B cell affinity maturation occurs in the germinal center (GC). Light-zone (LZ) GC B cells (BGC-cells) interact with follicular dendritic cells (FDCs) and compete for the limited, sequential help from T follicular helper cells needed to escape from apoptosis and complete their differentiation. The highest-affinity LZ BGC-cells enter the cell cycle and differentiate into PCs, following a dramatic epigenetic reorganization that induces transcriptome changes in general and the expression of the PRDM1 gene in particular. Human PC precursors are characterized by the loss of IL-4/STAT6 signaling and the absence of CD23 expression. Here, we studied the fate of human LZ BGC-cells as a function of their CD23 expression. We first showed that CD23 expression was restricted to the GC LZ, where it was primarily expressed by FDCs; less than 10% of tonsil LZ BGC-cells were positive. Sorted LZ BGC-cells left in culture and stimulated upregulated CD23 expression but were unable to differentiate into PCs - in contrast to cells that did not upregulate CD23 expression. An in-depth analysis (including single-cell gene expression) showed that stimulated CD23-negative LZ BGC-cells differentiated into plasmablasts and time course of gene expression changes delineates the transcriptional program that sustains PC differentiation. In particular, we identified a B cell proliferation signature supported by a transient MYC gene expression. Overall, the CD23 marker might be of value in answering questions about the differentiation of normal BGC-cells and allowed us to propose an instructive LZ BGC-cells maturation and fate model.


Asunto(s)
Linfocitos B/inmunología , Diferenciación Celular/inmunología , Centro Germinal/inmunología , Activación de Linfocitos/inmunología , Células Plasmáticas/inmunología , Linfocitos B/citología , Linfocitos B/metabolismo , Centro Germinal/citología , Humanos , Células Plasmáticas/citología , Receptores de IgE/metabolismo , Transcripción Genética
16.
Nat Commun ; 12(1): 6687, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34795279

RESUMEN

Pathogenic autoantibodies contribute to tissue damage and clinical decline in autoimmune disease. Follicular T cells are central regulators of germinal centers, although their contribution to autoantibody-mediated disease remains unclear. Here we perform single cell RNA and T cell receptor (TCR) sequencing of follicular T cells in a mouse model of autoantibody-mediated disease, allowing for analyses of paired transcriptomes and unbiased TCRαß repertoires at single cell resolution. A minority of clonotypes are preferentially shared amongst autoimmune follicular T cells and clonotypic expansion is associated with differential gene signatures in autoimmune disease. Antigen prediction using algorithmic and machine learning approaches indicates convergence towards shared specificities between non-autoimmune and autoimmune follicular T cells. However, differential autoimmune transcriptional signatures are preserved even amongst follicular T cells with shared predicted specificities. These results demonstrate that follicular T cells are phenotypically distinct in B cell-driven autoimmune disease, providing potential therapeutic targets to modulate autoantibody development.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Linfocitos B/inmunología , Perfilación de la Expresión Génica/métodos , Centro Germinal/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/metabolismo , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Células Clonales/inmunología , Células Clonales/metabolismo , Centro Germinal/citología , Centro Germinal/metabolismo , Ratones Endogámicos C57BL , Microscopía Confocal , RNA-Seq/métodos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de la Célula Individual/métodos , Linfocitos T Colaboradores-Inductores/metabolismo
17.
Front Immunol ; 12: 731100, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34603308

RESUMEN

Coronavirus disease 2019 (COVID-19), caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a serious infectious disease that has led to a global pandemic with high morbidity and mortality. High-affinity neutralizing antibody is important for controlling infection, which is closely regulated by follicular helper T (Tfh) cells. Tfh cells play a central role in promoting germinal center reactions and driving cognate B cell differentiation for antibody secretion. Available studies indicate a close relationship between virus-specific Tfh cell-mediated immunity and SARS-CoV-2 infection progression. Although several lines of evidence have suggested that Tfh cells contribute to the control of SARS-CoV-2 infection by eliciting neutralizing antibody productions, further studies are needed to elucidate Tfh-mediated effector mechanisms in anti-SARS-CoV-2 immunity. Here, we summarize the functional features and roles of virus-specific Tfh cells in the immunopathogenesis of SARS-CoV-2 infection and in COVID-19 vaccines, and highlight the potential of targeting Tfh cells as therapeutic strategy against SARS-CoV-2 infection.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19/inmunología , SARS-CoV-2/inmunología , Células T Auxiliares Foliculares/inmunología , Formación de Anticuerpos/inmunología , Linfocitos B/inmunología , COVID-19/patología , Vacunas contra la COVID-19/inmunología , Diferenciación Celular/inmunología , Centro Germinal/citología , Centro Germinal/inmunología , Humanos , Activación de Linfocitos/inmunología , Células T Auxiliares Foliculares/citología
18.
Front Immunol ; 12: 734282, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34616402

RESUMEN

Germinal center (GC) reactions are vital to the correct functioning of the adaptive immune system, through formation of high affinity, class switched antibodies. GCs are transient anatomical structures in secondary lymphoid organs where specific B cells, after recognition of antigen and with T cell help, undergo class switching. Subsequently, B cells cycle between zones of proliferation and somatic hypermutation and zones where renewed antigen acquisition and T cell help allows for selection of high affinity B cells (affinity maturation). Eventually GC B cells first differentiate into long-lived memory B cells (MBC) and finally into plasma cells (PC) that partially migrate to the bone marrow to encapsulate into long-lived survival niches. The regulation of GC reactions is a highly dynamically coordinated process that occurs between various cells and molecules that change in their signals. Here, we present a system-level perspective of T cell-mediated GC B cell differentiation, presenting and discussing the experimental and computational efforts on the regulation of the GCs. We aim to integrate Systems Biology with B cell biology, to advance elucidation of the regulation of high-affinity, class switched antibody formation, thus to shed light on the delicate functioning of the adaptive immune system. Specifically, we: i) review experimental findings of internal and external factors driving various GC dynamics, such as GC initiation, maturation and GCBC fate determination; ii) draw comparisons between experimental observations and mathematical modeling investigations; and iii) discuss and reflect on current strategies of modeling efforts, to elucidate B cell behavior during the GC tract. Finally, perspectives are specifically given on to the areas where a Systems Biology approach may be useful to predict novel GCBC-T cell interaction dynamics.


Asunto(s)
Linfocitos B/inmunología , Comunicación Celular , Diferenciación Celular , Centro Germinal/inmunología , Activación de Linfocitos , Modelos Inmunológicos , Biología de Sistemas , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Linfocitos B/metabolismo , Proliferación Celular , Centro Germinal/citología , Centro Germinal/metabolismo , Humanos , Fenotipo , Linfocitos T Colaboradores-Inductores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Int J Mol Sci ; 22(20)2021 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-34681663

RESUMEN

The role of extracellular vesicles (EVs) proteome in diffuse large B-cell lymphoma (DLBCL) pathology, subclassification, and patient screening is unexplored. We analyzed by state-of-the-art mass spectrometry the whole cell and secreted extracellular vesicles (EVs) proteomes of different molecular subtypes of DLBCL, germinal center B cell (GCB subtype), and activated B cell (ABC subtype). After quality control assessment, we compared whole-cell and secreted EVs proteomes of the two cell-of-origin (COO) categories, GCB and ABC subtypes, resulting in 288/1115 significantly differential expressed proteins from the whole-cell proteome and 228/608 proteins from EVs (adjust p-value < 0.05/p-value < 0.05). In our preclinical model system, we demonstrated that the EV proteome and the whole-cell proteome possess the capacity to separate cell lines into ABC and GCB subtypes. KEGG functional analysis and GO enrichment analysis for cellular component, molecular function, and biological process of differential expressed proteins (DEP) between ABC and GCB EVs showed a significant enrichment of pathways involved in immune response function. Other enriched functional categories for DEPs constitute cellular signaling and intracellular trafficking such as B-cell receptor (BCR), Fc_gamma R-mediated phagocytosis, ErbB signaling, and endocytosis. Our results suggest EVs can be explored as a tool for patient diagnosis, follow-up, and disease monitoring. Finally, this study proposes novel drug targets based on highly expressed proteins, for which antitumor drugs are available suggesting potential combinatorial therapies for aggressive forms of DLBCL. Data are available via ProteomeXchange with identifier PXD028267.


Asunto(s)
Vesículas Extracelulares/metabolismo , Linfoma de Células B Grandes Difuso/patología , Proteoma/análisis , Proteómica/métodos , Linfocitos B/metabolismo , Línea Celular Tumoral , Centro Germinal/citología , Centro Germinal/metabolismo , Humanos , Linfoma de Células B Grandes Difuso/metabolismo , Espectrometría de Masas
20.
J Exp Med ; 218(12)2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34665220

RESUMEN

T follicular helper cell (TFH)-dependent antibody responses are critical for long-term immunity. Antibody responses are diminished in early life, limiting long-term protective immunity and allowing prolonged or recurrent infection, which may be important for viral lung infections that are highly prevalent in infancy. In a murine model using respiratory syncytial virus (RSV), we show that TFH and the high-affinity antibody production they promote are vital for preventing disease on RSV reinfection. Following a secondary RSV infection, TFH-deficient mice had significantly exacerbated disease characterized by delayed viral clearance, increased weight loss, and immunopathology. TFH generation in early life was compromised by heightened IL-2 and STAT5 signaling in differentiating naive T cells. Neutralization of IL-2 during early-life RSV infection resulted in a TFH-dependent increase in antibody-mediated immunity and was sufficient to limit disease severity upon reinfection. These data demonstrate the importance of TFH in protection against recurrent RSV infection and highlight a mechanism by which this is suppressed in early life.


Asunto(s)
Interleucina-2/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Células T Auxiliares Foliculares/inmunología , Células T Auxiliares Foliculares/virología , Factores de Edad , Animales , Anticuerpos Antivirales , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Subgrupos de Linfocitos B/virología , Femenino , Centro Germinal/citología , Centro Germinal/inmunología , Centro Germinal/virología , Inmunidad Humoral , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-2/fisiología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Mutantes , Embarazo , Proteínas Proto-Oncogénicas c-bcl-6/genética , Proteínas Proto-Oncogénicas c-bcl-6/inmunología , Reinfección/inmunología , Reinfección/virología , Infecciones por Virus Sincitial Respiratorio/metabolismo , Factor de Transcripción STAT5/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...