Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Sci Immunol ; 4(41)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31672862

RESUMEN

Measles is a disease caused by the highly infectious measles virus (MeV) that results in both viremia and lymphopenia. Lymphocyte counts recover shortly after the disappearance of measles-associated rash, but immunosuppression can persist for months to years after infection, resulting in increased incidence of secondary infections. Animal models and in vitro studies have proposed various immunological factors underlying this prolonged immune impairment, but the precise mechanisms operating in humans are unknown. Using B cell receptor (BCR) sequencing of human peripheral blood lymphocytes before and after MeV infection, we identified two immunological consequences from measles underlying immunosuppression: (i) incomplete reconstitution of the naïve B cell pool leading to immunological immaturity and (ii) compromised immune memory to previously encountered pathogens due to depletion of previously expanded B memory clones. Using a surrogate model of measles in ferrets, we investigated the clinical consequences of morbillivirus infection and demonstrated a depletion of vaccine-acquired immunity to influenza virus, leading to a compromised immune recall response and increased disease severity after secondary influenza virus challenge. Our results show that MeV infection causes changes in naïve and memory B lymphocyte diversity that persist after the resolution of clinical disease and thus contribute to compromised immunity to previous infections or vaccinations. This work highlights the importance of MeV vaccination not only for the control of measles but also for the maintenance of herd immunity to other pathogens, which can be compromised after MeV infection.


Asunto(s)
Linfocitos B/inmunología , Sarampión/inmunología , Receptores de Antígenos de Linfocitos B/genética , Adolescente , Animales , Linfocitos B/virología , Niño , Preescolar , Estudios de Cohortes , Hurones , Humanos , Terapia de Inmunosupresión , Masculino , Sarampión/virología , Virus del Sarampión/inmunología , Receptores de Antígenos de Linfocitos B/inmunología
2.
Nature ; 548(7669): 597-601, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28847005

RESUMEN

In two previously described donors, the extracellular domain of LAIR1, a collagen-binding inhibitory receptor encoded on chromosome 19 (ref. 1), was inserted between the V and DJ segments of an antibody. This insertion generated, through somatic mutations, broadly reactive antibodies against RIFINs, a type of variant antigen expressed on the surface of Plasmodium falciparum-infected erythrocytes. To investigate how frequently such antibodies are produced in response to malaria infection, we screened plasma from two large cohorts of individuals living in malaria-endemic regions. Here we report that 5-10% of malaria-exposed individuals, but none of the European blood donors tested, have high levels of LAIR1-containing antibodies that dominate the response to infected erythrocytes without conferring enhanced protection against febrile malaria. By analysing the antibody-producing B cell clones at the protein, cDNA and gDNA levels, we characterized additional LAIR1 insertions between the V and DJ segments and discovered a second insertion modality whereby the LAIR1 exon encoding the extracellular domain and flanking intronic sequences are inserted into the switch region. By exon shuffling, this mechanism leads to the production of bispecific antibodies in which the LAIR1 domain is precisely positioned at the elbow between the VH and CH1 domains. Additionally, in one donor the genomic DNA encoding the VH and CH1 domains was deleted, leading to the production of a camel-like LAIR1-containing antibody. Sequencing of the switch regions of memory B cells from European blood donors revealed frequent templated inserts originating from transcribed genes that, in rare cases, comprised exons with orientations and frames compatible with expression. These results reveal different modalities of LAIR1 insertion that lead to public and dominant antibodies against infected erythrocytes and suggest that insertion of templated DNA represents an additional mechanism of antibody diversification that can be selected in the immune response against pathogens and exploited for B cell engineering.


Asunto(s)
Anticuerpos Antiprotozoarios/química , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Donantes de Sangre , Malaria/inmunología , Mutagénesis Insercional , Plasmodium falciparum/inmunología , Receptores Inmunológicos/genética , Anticuerpos Antiprotozoarios/genética , Antígenos de Protozoos/metabolismo , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Eritrocitos/metabolismo , Eritrocitos/parasitología , Europa (Continente) , Femenino , Genes de las Cadenas Pesadas de las Inmunoglobulinas/genética , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Región de Cambio de la Inmunoglobulina/genética , Memoria Inmunológica , Intrones/genética , Malaria/epidemiología , Malaria/parasitología , Masculino , Plasmodium falciparum/metabolismo , Dominios Proteicos , Receptores Inmunológicos/química , Receptores Inmunológicos/inmunología , Moldes Genéticos , Exones VDJ/genética
3.
Blood ; 127(18): 2193-202, 2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-26907631

RESUMEN

Fas is a transmembrane receptor involved in the maintenance of tolerance and immune homeostasis. In murine models, it has been shown to be essential for deletion of autoreactive B cells in the germinal center. The role of Fas in human B-cell selection and in development of autoimmunity in patients carrying FAS mutations is unclear. We analyzed patients with either a somatic FAS mutation or a germline FAS mutation and somatic loss-of-heterozygosity, which allows comparing the fate of B cells with impaired vs normal Fas signaling within the same individual. Class-switched memory B cells showed: accumulation of FAS-mutated B cells; failure to enrich single V, D, J genes and single V-D, D-J gene combinations of the B-cell receptor variable region; increased frequency of variable regions with higher content of positively charged amino acids; and longer CDR3 and maintenance of polyreactive specificities. Importantly, Fas-deficient switched memory B cells showed increased rates of somatic hypermutation. Our data uncover a defect in B-cell selection in patients with FAS mutations, which has implications for the understanding of the pathogenesis of autoimmunity and lymphomagenesis of autoimmune lymphoproliferative syndrome.


Asunto(s)
Síndrome Linfoproliferativo Autoinmune/inmunología , Subgrupos de Linfocitos B/inmunología , Selección Clonal Mediada por Antígenos , Mutación , Receptor fas/fisiología , Apoptosis , Autoinmunidad , Línea Celular Transformada , Transformación Celular Neoplásica , Niño , Codón sin Sentido , Femenino , Mutación del Sistema de Lectura , Mutación de Línea Germinal , Heterocigoto , Humanos , Memoria Inmunológica , Pérdida de Heterocigocidad , Masculino , Análisis de Secuencia de ADN , Hipermutación Somática de Inmunoglobulina , Recombinación V(D)J , Receptor fas/deficiencia , Receptor fas/genética
4.
Nature ; 529(7584): 105-109, 2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26700814

RESUMEN

Plasmodium falciparum antigens expressed on the surface of infected erythrocytes are important targets of naturally acquired immunity against malaria, but their high number and variability provide the pathogen with a powerful means of escape from host antibodies. Although broadly reactive antibodies against these antigens could be useful as therapeutics and in vaccine design, their identification has proven elusive. Here we report the isolation of human monoclonal antibodies that recognize erythrocytes infected by different P. falciparum isolates and opsonize these cells by binding to members of the RIFIN family. These antibodies acquired broad reactivity through a novel mechanism of insertion of a large DNA fragment between the V and DJ segments. The insert, which is both necessary and sufficient for binding to RIFINs, encodes the entire 98 amino acid collagen-binding domain of LAIR1, an immunoglobulin superfamily inhibitory receptor encoded on chromosome 19. In each of the two donors studied, the antibodies are produced by a single expanded B-cell clone and carry distinct somatic mutations in the LAIR1 domain that abolish binding to collagen and increase binding to infected erythrocytes. These findings illustrate, with a biologically relevant example, a novel mechanism of antibody diversification by interchromosomal DNA transposition and demonstrate the existence of conserved epitopes that may be suitable candidates for the development of a malaria vaccine.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Variación Antigénica/inmunología , Antígenos de Protozoos/inmunología , Malaria/inmunología , Mutagénesis Insercional/genética , Plasmodium falciparum/inmunología , Receptores Inmunológicos/inmunología , Secuencia de Aminoácidos , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/uso terapéutico , Linfocitos B/citología , Linfocitos B/inmunología , Células Clonales/citología , Células Clonales/inmunología , Colágeno/inmunología , Colágeno/metabolismo , Secuencia Conservada/inmunología , Elementos Transponibles de ADN/genética , Elementos Transponibles de ADN/inmunología , Epítopos de Linfocito B/química , Epítopos de Linfocito B/inmunología , Eritrocitos/inmunología , Eritrocitos/metabolismo , Eritrocitos/parasitología , Humanos , Kenia , Malaria/parasitología , Vacunas contra la Malaria/química , Vacunas contra la Malaria/inmunología , Proteínas de la Membrana/química , Proteínas de la Membrana/inmunología , Datos de Secuencia Molecular , Estructura Terciaria de Proteína/genética , Proteínas Protozoarias/química , Proteínas Protozoarias/inmunología , Receptores Inmunológicos/química , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo
5.
J Virol ; 89(9): 4748-59, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25673724

RESUMEN

UNLABELLED: The B cell-activating factor (BAFF) is critical for B cell development and humoral immunity in mice and humans. While the role of BAFF in B cells has been widely described, its role in innate immunity remains unknown. Using BAFF receptor (BAFFR)-deficient mice, we characterized BAFFR-related innate and adaptive immune functions following infection with vesicular stomatitis virus (VSV) and lymphocytic choriomeningitis virus (LCMV). We identified a critical role for BAFFR signaling in the generation and maintenance of the CD169(+) macrophage compartment. Consequently, Baffr(-) (/) (-) mice exhibited limited induction of innate type I interferon production after viral infection. Lack of BAFFR signaling reduced virus amplification and presentation following viral infection, resulting in highly reduced antiviral adaptive immune responses. As a consequence, BAFFR-deficient mice showed exacerbated and fatal disease after viral infection. Mechanistically, transient lack of B cells in Baffr(-) (/) (-) animals resulted in limited lymphotoxin expression, which is critical for maintenance of CD169(+) cells. In conclusion, BAFFR signaling affects both innate and adaptive immune activation during viral infections. IMPORTANCE: Viruses cause acute and chronic infections in humans resulting in millions of deaths every year. Innate immunity is critical for the outcome of a viral infection. Innate type I interferon production can limit viral replication, while adaptive immune priming by innate immune cells induces pathogen-specific immunity with long-term protection. Here, we show that BAFFR deficiency not only perturbed B cells, but also resulted in limited CD169(+) macrophages. These macrophages are critical in amplifying viral particles to trigger type I interferon production and initiate adaptive immune priming. Consequently, BAFFR deficiency resulted in reduced enforced viral replication, limited type I interferon production, and reduced adaptive immunity compared to BAFFR-competent controls. As a result, BAFFR-deficient mice were predisposed to fatal viral infections. Thus, BAFFR expression is critical for innate immune activation and antiviral immunity.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Macrófagos/química , Macrófagos/inmunología , Receptores de Interleucina-4/deficiencia , Infecciones por Rhabdoviridae/inmunología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/análisis , Inmunidad Adaptativa , Animales , Inmunidad Innata , Interferón Tipo I/metabolismo , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones Noqueados , Transducción de Señal , Vesiculovirus/inmunología
6.
J Allergy Clin Immunol ; 134(2): 420-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24679343

RESUMEN

BACKGROUND: Five different G protein-coupled sphingosine-1-phosphate (S1P) receptors (S1P1-S1P5) regulate a variety of physiologic and pathophysiologic processes, including lymphocyte circulation, multiple sclerosis (MS), and cancer. Although B-lymphocyte circulation plays an important role in these processes and is essential for normal immune responses, little is known about S1P receptors in human B cells. OBJECTIVE: To explore their function and signaling, we studied B-cell lines and primary B cells from control subjects, patients with leukemia, patients with S1P receptor inhibitor-treated MS, and patients with primary immunodeficiencies. METHODS: S1P receptor expression was analyzed by using multicolor immunofluorescence microscopy and quantitative PCR. Transwell assays were used to study cell migration. S1P receptor internalization was visualized by means of time-lapse imaging with fluorescent S1P receptor fusion proteins expressed by using lentiviral gene transfer. B-lymphocyte subsets were characterized by means of flow cytometry and immunofluorescence microscopy. RESULTS: Showing that different B-cell populations express different combinations of S1P receptors, we found that S1P1 promotes migration, whereas S1P4 modulates and S1P2 inhibits S1P1 signals. Expression of CD69 in activated B lymphocytes and B cells from patients with chronic lymphocytic leukemia inhibited S1P-induced migration. Studying B-cell lines, normal B lymphocytes, and B cells from patients with primary immunodeficiencies, we identified Bruton tyrosine kinase, ß-arrestin 2, LPS-responsive beige-like anchor protein, dedicator of cytokinesis 8, and Wiskott-Aldrich syndrome protein as critical signaling components downstream of S1P1. CONCLUSION: Thus S1P receptor signaling regulates human B-cell circulation and might be a factor contributing to the pathology of MS, chronic lymphocytic leukemia, and primary immunodeficiencies.


Asunto(s)
Subgrupos de Linfocitos B/metabolismo , Inmunodeficiencia Variable Común/metabolismo , Leucemia Linfocítica Crónica de Células B/metabolismo , Esclerosis Múltiple/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Agammaglobulinemia Tirosina Quinasa , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/genética , Antígenos de Diferenciación de Linfocitos T/inmunología , Antígenos de Diferenciación de Linfocitos T/metabolismo , Arrestinas/genética , Arrestinas/inmunología , Arrestinas/metabolismo , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/patología , Línea Celular , Movimiento Celular , Inmunodeficiencia Variable Común/genética , Inmunodeficiencia Variable Común/inmunología , Inmunodeficiencia Variable Común/patología , Regulación de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/inmunología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/inmunología , Leucemia Linfocítica Crónica de Células B/patología , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Cultivo Primario de Células , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Isoformas de Proteínas/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/inmunología , Proteínas Tirosina Quinasas/metabolismo , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/inmunología , Transducción de Señal , Imagen de Lapso de Tiempo , Proteína del Síndrome de Wiskott-Aldrich/genética , Proteína del Síndrome de Wiskott-Aldrich/inmunología , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Arrestina beta 2 , beta-Arrestinas
7.
J Immunol ; 192(3): 1044-54, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24379121

RESUMEN

The peripheral B cell compartment is maintained by homeostatic proliferation and through replenishment by bone marrow precursors. Because hematopoietic stem cells cycle at a slow rate, replenishment must involve replication of precursor B cells. To study proliferation of early human B cell progenitors, we established a feeder cell-free in vitro system allowing the development of B cells from CD34(+) hematopoietic stem cells up to the stage of immature IgM(+) B cells. We found that pro-B and pre-B cells generated in vitro can proliferate autonomously and persist up to 7 wk in culture in the absence of signals induced by exogenously added cytokines. Nevertheless, addition of IL-7 enhanced pre-B cell expansion and inhibited maturation into IgM(+) B cells. The B cell precursor subsets replicating in vitro were highly similar to the bone marrow B cell precursors cycling in vivo. The autonomous proliferation of B cell precursor subsets in vitro and their long-term persistence implies that proliferation during pro-B and pre-B cell stages plays an important role in the homeostasis of the peripheral B cell compartment. Our in vitro culture can be used to study defects in B cell development or in reconstitution of the B cell pool after depletion and chemotherapy.


Asunto(s)
Linfocitos B/citología , Técnicas de Cultivo de Célula/métodos , Células Madre Hematopoyéticas/citología , Adulto , Animales , Médula Ósea , División Celular , Linaje de la Célula , Células Cultivadas , Técnicas de Cocultivo , Proteínas de Unión al ADN/deficiencia , Sangre Fetal/citología , Supervivencia de Injerto , Células Madre Hematopoyéticas/efectos de los fármacos , Xenoinjertos , Homeostasis , Humanos , Inmunoglobulina M/biosíntesis , Inmunofenotipificación , Interleucina-7/farmacología , Linfopoyesis/efectos de los fármacos , Ratones , Quimera por Radiación , Receptores de Interleucina-2/deficiencia , Factores de Tiempo , Adulto Joven
9.
J Allergy Clin Immunol ; 131(4): 959-71, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23465663

RESUMEN

B cells develop from hematopoietic precursor cells in an ordered maturation and selection process. Extensive studies with many different mouse mutants provided fundamental insights into this process. However, the characterization of genetic defects causing primary immunodeficiencies was essential in understanding human B-cell biology. Defects in pre-B-cell receptor components or in downstream signaling proteins, such as Bruton tyrosine kinase and B-cell linker protein, arrest development at the pre-B-cell stage. Defects in survival-regulating proteins, such as B-cell activator of the TNF-α family receptor (BAFF-R) or caspase recruitment domain-containing protein 11 (CARD11), interrupt maturation and prevent differentiation of transitional B cells into marginal zone and follicular B cells. Mature B-cell subsets, immune responses, and memory B-cell and plasma cell development are disturbed by mutations affecting Toll-like receptor signaling, B-cell antigen receptor coreceptors (eg, CD19), or enzymes responsible for immunoglobulin class-switch recombination. Transgenic mouse models helped to identify key regulatory mechanisms, such as receptor editing and clonal anergy, preventing the activation of B cells expressing antibodies recognizing autoantigens. Nevertheless, the combination of susceptible genetic backgrounds with the rescue of self-reactive B cells by T cells allows the generation of autoreactive clones found in patients with many autoimmune diseases and even in those with primary immunodeficiencies. The rapid progress of functional genomic research is expected to foster the development of new tools that specifically target dysfunctional B lymphocytes to treat autoimmunity, B-cell malignancies, and immunodeficiency.


Asunto(s)
Subgrupos de Linfocitos B/patología , Linfocitos B/patología , Síndromes de Inmunodeficiencia/patología , Receptores de Antígenos de Linfocitos B/inmunología , Agammaglobulinemia Tirosina Quinasa , Animales , Anticuerpos/inmunología , Subgrupos de Linfocitos B/inmunología , Linfocitos B/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Regulación de la Expresión Génica , Humanos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Memoria Inmunológica , Ratones , Ratones Transgénicos , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/inmunología , Receptores de Antígenos de Linfocitos B/genética , Transducción de Señal , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...