Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Autoimmun ; 138: 103057, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37224732

RESUMEN

The forkhead family transcription factor (FOXP3) is an essential regulator for the development of regulatory T cells (Tregs) and orchestrates both suppressive function and Treg lineage identity. Stable expression of FOXP3 enables Tregs to maintain immune homeostasis and prevent autoimmunity. However, under pro-inflammatory conditions, FOXP3 expression in Tregs can become unstable, leading to loss of suppressive function and conversion into pathogenic T effector cells. Therefore, the success of adoptive cell therapy with chimeric antigen receptor (CAR) Tregs is highly dependent on the stability of FOXP3 expression to ensure the safety of the cell product. To warrant the stable expression of FOXP3 in CAR-Treg products, we have developed an HLA-A2-specific CAR vector that co-expresses FOXP3. The transduction of isolated human Tregs with the FOXP3-CAR led to an increase in the safety and efficacy of the CAR-Treg product. In a hostile microenvironment, under pro-inflammatory and IL-2-deficient conditions, FOXP3-CAR-Tregs showed a stable expression of FOXP3 compared to Control-CAR-Tregs. Furthermore, additional exogenous expression of FOXP3 did not induce phenotypic alterations and dysfunctions such as cell exhaustion, loss of functional Treg characteristics or abnormal cytokine secretion. In a humanized mouse model, FOXP3-CAR-Tregs displayed an excellent ability to prevent allograft rejection. Furthermore, FOXP3-CAR-Tregs revealed coherent Treg niche-filling capabilities. Overexpression of FOXP3 in CAR-Tregs has thereby the potential to increase the efficacy and reliability of cellular products, promoting their clinical use in organ transplantation and autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes , Receptores Quiméricos de Antígenos , Animales , Humanos , Ratones , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/terapia , Enfermedades Autoinmunes/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Reproducibilidad de los Resultados , Linfocitos T Reguladores
2.
Int J Mol Sci ; 24(6)2023 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-36982860

RESUMEN

IgM is the first antibody to emerge during phylogeny, ontogeny, and immune responses and serves as a first line of defense. Effector proteins interacting with the Fc portion of IgM, such as complement and its receptors, have been extensively studied for their functions. IgM Fc receptor (FcµR), identified in 2009, is the newest member of the FcR family and is intriguingly expressed by lymphocytes only, suggesting the existence of distinct functions as compared to the FcRs for switched Ig isotypes, which are expressed by various immune and non-hematopoietic cells as central mediators of antibody-triggered responses by coupling the adaptive and innate immune responses. Results from FcµR-deficient mice suggest a regulatory function of FcµR in B cell tolerance, as evidenced by their propensity to produce autoantibodies of both IgM and IgG isotypes. In this article, we discuss conflicting views about the cellular distribution and potential functions of FcµR. The signaling function of the Ig-tail tyrosine-like motif in the FcµR cytoplasmic domain is now formally shown by substitutional experiments with the IgG2 B cell receptor. The potential adaptor protein associating with FcµR and the potential cleavage of its C-terminal cytoplasmic tail after IgM binding are still enigmatic. Critical amino acid residues in the Ig-like domain of FcµR for interacting with the IgM Cµ4 domain and the mode of interaction are now defined by crystallographic and cryo-electron microscopic analyses. Some discrepancies on these interactions are discussed. Finally, elevated levels of a soluble FcµR isoform in serum samples are described as the consequence of persistent B cell receptor stimulation, as seen in chronic lymphocytic leukemia and probably in antibody-mediated autoimmune disorders.


Asunto(s)
Receptores de Antígenos de Linfocitos B , Receptores Fc , Animales , Ratones , Inmunoglobulina M , Receptores Fc/metabolismo , Isoformas de Proteínas
3.
Front Immunol ; 13: 842340, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35371049

RESUMEN

The generation, differentiation, survival and activation of B cells are coordinated by signals emerging from the B cell antigen receptor (BCR) or its precursor, the pre-BCR. The adaptor protein SLP65 (also known as BLNK) is an important signaling factor that controls pre-B cell differentiation by down-regulation of PI3K signaling. Here, we investigated the mechanism by which SLP65 interferes with PI3K signaling. We found that SLP65 induces the activity of the small GTPase RHOA, which activates PTEN, a negative regulator of PI3K signaling, by enabling its translocation to the plasma membrane. The essential role of RHOA is confirmed by the complete block in early B cell development in conditional RhoA-deficient mice. The RhoA-deficient progenitor B cells showed defects in activation of immunoglobulin gene rearrangement and fail to survive both in vitro and in vivo. Reconstituting the RhoA-deficient cells with RhoA or Foxo1, a transcription factor repressed by PI3K signaling and activated by PTEN, completely restores the survival defect. However, the defect in differentiation can only be restored by RhoA suggesting a unique role for RHOA in B cell generation and selection. In full agreement, conditional RhoA-deficient mice develop increased amounts of autoreactive antibodies with age. RHOA function is also required at later stage, as inactivation of RhoA in peripheral B cells or in a transformed mature B cell line resulted in cell loss. Together, these data show that RHOA is the key signaling factor for B cell development and function by providing a crucial SLP65-activated link between BCR signaling and activation of PTEN. Moreover, the identified essential role of RHOA for the survival of transformed B cells offers the opportunity for targeting B cell malignancies by blocking RHOA function.


Asunto(s)
Proteínas de Unión al GTP Monoméricas , Células Precursoras de Linfocitos B , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Ratones , Proteínas de Unión al GTP Monoméricas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Células Precursoras de Linfocitos B/metabolismo , Receptores de Antígenos de Linfocitos B/genética , Proteína de Unión al GTP rhoA
4.
Analyst ; 146(15): 4744-4747, 2021 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-34226908

RESUMEN

We have developed a series of monovalent fluorophore-conjugated affinity probes based on the hapten 3-nitro-4-hydroxy-5-iodophenylacetyl (NIP), which is widely used as a model antigen to study B lymphocytes and the functional principles of B cell antigen receptors (BCRs). We successfully used them in flow-cytometry, confocal and super-resolution microscopy techniques.


Asunto(s)
Colorantes Fluorescentes , Microscopía , Antígenos , Linfocitos B , Haptenos
5.
EMBO Rep ; 22(9): e51328, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34323351

RESUMEN

Antigen-specific B-cell responses require endosomal trafficking to regulate antigen uptake and presentation to helper T cells, and to control expression and signaling of immune receptors. However, the molecular composition of B-cell endosomal trafficking pathways and their specific roles in B-cell responses have not been systematically investigated. Here, we report high-throughput identification of genes regulating B-cell receptor (BCR)-mediated antigen internalization using genome-wide functional screens. We show that antigen internalization depends both on constitutive, clathrin-mediated endocytosis and on antigen-induced, clathrin-independent endocytosis mediated by endophilin A2. Although endophilin A2-mediated endocytosis is dispensable for antigen presentation, it is selectively required for metabolic support of B-cell proliferation, in part through regulation of iron uptake. Consequently, endophilin A2-deficient mice show defects in GC B-cell responses and production of high-affinity IgG. The requirement for endophilin A2 highlights a unique importance of clathrin-independent intracellular trafficking in GC B-cell clonal expansion and antibody responses.


Asunto(s)
Clatrina , Endocitosis , Animales , Linfocitos B , Endosomas , Centro Germinal , Ratones
6.
Cancers (Basel) ; 13(3)2021 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-33530306

RESUMEN

Resistance of tumor cells to chemoradiotherapy represents a fundamental problem in clinical oncology. The underlying mechanisms are actively debated. Here we show that blocking inflammatory cytokine receptor signaling via STAT3 re-sensitized treatment-refractory cancer cells and abolished tumor growth in a xenograft mouse model when applied together with chemoradiotherapy. STAT3 executed treatment resistance by triggering the expression of RBPJ, the key transcriptional regulator of the NOTCH pathway. The mandatory RBPJ interaction partner, NOTCH intracellular domain, was provided by tumor cell-intrinsic expression of NOTCH ligands that caused tonic NOTCH proteolysis. In fact, NOTCH inhibition phenocopied the effect of blocking STAT3 signaling. Moreover, genetic profiling of rectal cancer patients revealed the importance of the STAT3/NOTCH axis as NOTCH expression correlated with clinical outcome. Our data uncovered an unprecedented signal alliance between inflammation and cellular development that orchestrated resistance to chemoradiotherapy. Clinically, our findings allow for biomarker-driven patient stratification and offer novel treatment options.

7.
Eur J Immunol ; 50(12): 1912-1928, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32671844

RESUMEN

Antigen recognition by B-cell antigen receptors (BCRs) activates distinct intracellular signaling pathways that control the differentiation fate of activated B lymphocytes. BCR-proximal signaling enzymes comprise protein tyrosine kinases, phosphatases, and plasma membrane lipid-modifying enzymes, whose function is furthermore coordinated by catalytically inert adaptor proteins. Here, we show that an additional class of enzymatic activity provided by guanine-nucleotide exchange factors (GEFs) of the Vav family controls BCR-proximal Ca2+ mobilization, cytoskeletal actin reorganization, and activation of the PI3 kinase/Akt pathway. Whereas Vav1 and Vav3 supported all of those signaling processes to different extents in a human B-cell model system, Vav2 facilitated Actin remodeling, and activation of Akt but did not promote Ca2+ signaling. On BCR activation, Vav1 was directly recruited to the phosphorylated BCR and to the central adaptor protein SLP65 via its Src homology 2 domain. Pharmacological inhibition or genetic inactivation of the substrates of Vav GEFs, small G proteins of the Rho/Rac family, impaired BCR-induced Ca2+ mobilization, probably because phospholipase Cγ2 requires activated Rac proteins for optimal activity. Our findings show that Vav family members are key relays of the BCR signalosome that differentially control distinct signaling pathways both in a catalysis-dependent and -independent manner.


Asunto(s)
Proteínas Proto-Oncogénicas c-vav/metabolismo , Receptores de Antígenos de Linfocitos B/inmunología , Transducción de Señal/inmunología , Linfocitos B/inmunología , Calcio/inmunología , Diferenciación Celular/inmunología , Línea Celular Tumoral , Humanos , Fosforilación/inmunología , Dominios Homologos src/inmunología
8.
J Allergy Clin Immunol ; 146(3): 642-651.e5, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32113980

RESUMEN

BACKGROUND: Allergic inflammation is driven by IgE-producing plasma cells (PCs), which are required for IgE-mediated activation of mast cells and basophils. Repeated antigen encounter elicits a memory IgE response with elevated serum IgE titers and accumulation of IgE-producing PCs. However, the cellular compartment and molecular signals that underlie the immunologic memory of IgE responses remain unclear. OBJECTIVE: With this study we aimed at clarifying whether inactivation of the cytoplasmic immunoglobulin tail tyrosine (ITT) motif in transmembrane IgE (mIgE) impairs the memory IgE response in mice. METHODS: We generated mice with an inactivated mIgE-ITT motif and analyzed serum IgE levels as well as the generation of IgE-producing germinal center B cells and PCs subsequent to primary and secondary infection with helminths. In vitro cultures were used to study the mIgE-ITT-controlled expression of mIgE on the surface of PCs. Systemic mast cell activation was determined by serum Mcpt1 ELISA in response to ovalbumin challenge. RESULTS: mIgE-ITT-mutant mice showed an impaired memory IgE response subsequent to helminth infection. Furthermore, sensitization and challenge of mIgE-ITT-mutant mice with ovalbumin resulted in diminished serum IgE titers and reduced mast cell activation. The mIgE-ITT motif was required for optimal cell surface expression of mIgE B-cell antigen receptors but not for intracellular IgE expression in PCs. CONCLUSION: These results indicate that the mIgE B-cell antigen receptor plays a critical role in establishing or maintaining the population of IgE-producing PCs during memory IgE responses.


Asunto(s)
Inmunoglobulina E/inmunología , Memoria Inmunológica , Células Plasmáticas/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Tricostrongiloidiasis/inmunología , Alérgenos/inmunología , Animales , Quimasas/inmunología , Femenino , Masculino , Mastocitos/inmunología , Ratones Transgénicos , Ovalbúmina/inmunología , Trichostrongyloidea
9.
Nat Commun ; 10(1): 4403, 2019 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-31562305

RESUMEN

Specialized epitope tags are widely used for detecting, manipulating or purifying proteins, but often their versatility is limited. Here, we introduce the ALFA-tag, a rationally designed epitope tag that serves a remarkably broad spectrum of applications in life sciences while outperforming established tags like the HA-, FLAG®- or myc-tag. The ALFA-tag forms a small and stable α-helix that is functional irrespective of its position on the target protein in prokaryotic and eukaryotic hosts. We characterize a nanobody (NbALFA) binding ALFA-tagged proteins from native or fixed specimen with low picomolar affinity. It is ideally suited for super-resolution microscopy, immunoprecipitations and Western blotting, and also allows in vivo detection of proteins. We show the crystal structure of the complex that enabled us to design a nanobody mutant (NbALFAPE) that permits efficient one-step purifications of native ALFA-tagged proteins, complexes and even entire living cells using peptide elution under physiological conditions.


Asunto(s)
Epítopos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Anticuerpos de Dominio Único/metabolismo , Células 3T3 , Animales , Células COS , Chlorocebus aethiops , Epítopos/química , Epítopos/genética , Proteínas Fluorescentes Verdes/genética , Células HeLa , Humanos , Ratones , Microscopía Fluorescente , Mutación , Unión Proteica , Proteínas/genética , Proteínas/metabolismo , Proteínas Recombinantes de Fusión/genética , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/genética
10.
Nat Commun ; 10(1): 820, 2019 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-30778055

RESUMEN

Stimulation of the B cell antigen receptor (BCR) triggers signaling pathways that promote the differentiation of B cells into plasma cells. Despite the pivotal function of BCR in B cell activation, the organization of the BCR on the surface of resting and antigen-activated B cells remains unclear. Here we show, using STED super-resolution microscopy, that IgM-containing BCRs exist predominantly as monomers and dimers in the plasma membrane of resting B cells, but form higher oligomeric clusters upon stimulation. By contrast, a chronic lymphocytic leukemia-derived BCR forms dimers and oligomers in the absence of a stimulus, but a single amino acid exchange reverts its organization to monomers in unstimulated B cells. Our super-resolution microscopy approach for quantitatively analyzing cell surface proteins may thus help reveal the nanoscale organization of immunoreceptors in various cell types.


Asunto(s)
Linfocitos B/metabolismo , Membrana Celular/metabolismo , Microscopía Fluorescente/métodos , Receptores de Antígenos de Linfocitos B/metabolismo , Linfoma de Burkitt/patología , Línea Celular Tumoral , Humanos , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/metabolismo , Inmunoglobulina M/metabolismo , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Multimerización de Proteína , Receptores de Antígenos de Linfocitos B/genética
11.
J Exp Med ; 215(5): 1327-1336, 2018 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-29636373

RESUMEN

Ubiquitously expressed Cbl-interacting protein of 85 kD (CIN85) is a multifunctional adapter molecule supposed to regulate numerous cellular processes that are critical for housekeeping as well as cell type-specific functions. However, limited information exists about the in vivo roles of CIN85, because only conditional mouse mutants with cell type-specific ablation of distinct CIN85 isoforms in brain and B lymphocytes have been generated so far. No information is available about the roles of CIN85 in humans. Here, we report on primary antibody deficiency in patients harboring a germline deletion within the CIN85 gene on the X chromosome. In the absence of CIN85, all immune cell compartments developed normally, but B lymphocytes showed intrinsic defects in distinct effector pathways of the B cell antigen receptor, most notably NF-κB activation and up-regulation of CD86 expression on the cell surface. These results reveal nonredundant functions of CIN85 for humoral immune responses.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Anticuerpos/metabolismo , Cromosomas Humanos X/genética , Eliminación de Gen , Células Germinativas/metabolismo , Síndromes de Inmunodeficiencia/genética , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Linfocitos B/inmunología , Calcio/metabolismo , Humanos , Inmunoglobulina G/sangre , Síndromes de Inmunodeficiencia/sangre , Inmunofenotipificación , Activación de Linfocitos/inmunología , Masculino , FN-kappa B/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Hermanos , Transducción de Señal
12.
Immunol Rev ; 283(1): 150-160, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29664567

RESUMEN

The generation of memory B cells (MBCs) that have undergone immunoglobulin class switching from IgM, which dominates primary antibody responses, to other immunoglobulin isoforms is a hallmark of immune memory. Hence, humoral immunological memory is characterized by the presence of serum immunoglobulins of IgG subtypes known as the γ-globulin fraction of blood plasma proteins. These antibodies reflect the antigen experience of B lymphocytes and their repeated triggering. In fact, efficient protection against a previously encountered pathogen is critically linked to the production of pathogen-specific IgG molecules even in those cases where the primary immune response required cellular immunity, for example, T cell-mediated clearance of intracellular pathogens such as viruses. Besides IgG, also IgA and IgE can provide humoral immunity depending on the microbe's nature and infection route. The molecular mechanisms underlying the preponderance of switched immunoglobulin isotypes during memory antibody responses are a matter of active and controversial debate. Here, we summarize the phenotypic characteristics of distinct MBC subpopulations and discuss the decisive roles of different B cell antigen receptor isotypes for the functional traits of class-switched B cell populations.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Inmunidad Humoral , Memoria Inmunológica , Receptores de Antígenos de Linfocitos B/metabolismo , Animales , Formación de Anticuerpos/inmunología , Antígenos/inmunología , Antígenos/metabolismo , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Humanos , Inmunidad Humoral/genética , Inmunidad Mucosa/genética , Inmunidad Mucosa/inmunología , Inmunoglobulina A/genética , Inmunoglobulina A/inmunología , Inmunoglobulina E/genética , Inmunoglobulina E/inmunología , Memoria Inmunológica/genética , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos B/genética , Transducción de Señal
13.
Sci Rep ; 8(1): 4244, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29523808

RESUMEN

The B cell antigen receptor (BCR) employs enzymatically inactive adaptor proteins to facilitate activation of intracellular signaling pathways. In animal model systems, adaptor proteins of the growth factor receptor-bound 2 (Grb2) family have been shown to serve critical functions in lymphocytes. However, the roles of Grb2 and the Grb2-related adaptor protein (GRAP) in human B lymphocytes remain unclear. Using TALEN-mediated gene targeting, we show that in human B cells Grb2 and GRAP amplify signaling by the immunoglobulin tail tyrosine (ITT) motif of mIgE-containing BCRs and furthermore connect immunoreceptor tyrosine-based activation motif (ITAM) signaling to activation of the Ras-controlled Erk MAP kinase pathway. In contrast to mouse B cells, BCR-induced activation of Erk in human B cells is largely independent of phospholipase C-É£ activity and diacylglycerol-responsive members of Ras guanine nucleotide releasing proteins. Together, our results demonstrate that Grb2 family adaptors are critical regulators of ITAM and ITT signaling in naïve and IgE-switched human B cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Linfocitos B/metabolismo , Proteína Adaptadora GRB2/metabolismo , Sistema de Señalización de MAP Quinasas , Receptores de Antígenos de Linfocitos B/metabolismo , Línea Celular , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Fosfolipasas de Tipo C/metabolismo
14.
Eur J Immunol ; 48(3): 441-453, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29150831

RESUMEN

Immunoglobulin E (IgE) antibodies are key mediators of allergic reactions. Due to their potentially harmful anaphylactic properties, their production is tightly regulated. The membrane-bound isoform of IgE (mIgE), which is an integral component of the B cell antigen receptor, has been shown to be critical for the regulation of IgE responses in mice. In primate species including humans, mIgE can be expressed in two isoforms that are produced by alternative splicing of the primary ε Ig heavy chain transcript, and differ in the absence or presence of an extracellular membrane-proximal domain (EMPD) consisting of 52 amino acids. However, the function of the EMPD remains unclear. Here, we demonstrate that the EMPD restricts surface expression of mIgE-containing BCRs in human and murine B cells. The EMPD does not interfere with BCR assembly but acts as an autonomous endoplasmic reticulum retention domain. Limited surface expression of EMPD-containing mIgE-BCRs caused impaired activation of intracellular signaling cascades and hence represents a regulatory mechanism that may control the production of potentially anaphylactic IgE antibodies in primate species.


Asunto(s)
Linfocitos B/inmunología , Inmunoglobulina E/química , Receptores de Antígenos de Linfocitos B/química , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Apoptosis/inmunología , Linfocitos B/citología , Línea Celular Tumoral , Retículo Endoplásmico/inmunología , Evolución Molecular , Espacio Extracelular/inmunología , Humanos , Inmunoglobulina E/genética , Inmunoglobulina E/inmunología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Primates/genética , Primates/inmunología , Dominios Proteicos , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología , Homología de Secuencia de Aminoácido , Transducción de Señal
16.
Immunol Lett ; 178: 27-30, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27345519
17.
Curr Top Microbiol Immunol ; 393: 107-121, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26362935

RESUMEN

Activated B lymphocytes preserve their antigen experience by differentiating into long-lived pools of antibody-secreting plasma cells or various types of memory B cells (MBCs). The former population constantly produces serum immunoglobulins with sufficient specificity and affinity to thwart infections with recurrent pathogens. By contrast, memory B cell populations retain their antigen receptors on the cell surface and hence need pathogen-induced differentiation steps before they can actively contribute to host defense. The terminal differentiation of MBCs into antibody-secreting plasma cells is hallmarked by the absence of the lag phase characteristic for primary antibody responses. Moreover, secondary antibody responses are predominantly driven by MBCs that bear an antigen receptor of the IgG class on their surface although IgM-positive memory populations exist as well. These fundamental principles of B cell memory were enigmatic for decades. Only recently, we have begun to understand the underlying mechanisms. This review summarizes our current understanding of how different subpopulations of MBCs are generated during primary immune responses and how their functional heterogeneity on antigen recall is controlled by different signaling capabilities of B cell antigen receptor (BCR) isotypes and by the nature of the antigen.


Asunto(s)
Linfocitos B/inmunología , Memoria Inmunológica , Receptores de Antígenos de Linfocitos B/inmunología , Animales , Formación de Anticuerpos , Humanos , Receptores de Antígenos de Linfocitos B/genética
18.
Nat Commun ; 6: 8575, 2015 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-26815242

RESUMEN

Secondary antibody responses are marked by faster kinetics, improved antibody affinity and a switch from IgM to other immunoglobulin isotypes, most notably IgG, compared with primary responses. These changes protect from reinfection and represent the principle of most vaccination strategies. Yet, the molecular mechanisms that underlie B-cell memory responses are unclear. Here we show, by inactivating the immunoglobulin tail tyrosine (ITT) signalling motif of membrane-bound IgG1 in the mouse, that the ITT facilitates maintenance and reactivation of IgG-switched memory B cells in vivo. The ITT motif equips IgG-switched cells with enhanced BCR signalling capacity, which supports their competitiveness in secondary immune reactions and drives the formation of IgG-secreting plasma cells even in the absence of T-cell help. Our results demonstrate that ITT signalling promotes the vigorous production of IgG antibodies and thus provide a molecular basis for humoral immunological memory.


Asunto(s)
Linfocitos B/metabolismo , Inmunoglobulina G/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Animales , Formación de Anticuerpos , Calcio/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Ratones , Ratones Mutantes , Fosforilación , Receptores de Antígenos de Linfocitos B/genética , Transducción de Señal/fisiología
19.
Nat Commun ; 5: 5456, 2014 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-25413232

RESUMEN

The vigorous response of IgG-switched memory B cells to recurring pathogens involves enhanced signalling from their B-cell antigen receptors (BCRs). However, the molecular signal amplification mechanisms of memory-type BCRs remained unclear. Here, we identify the immunoglobulin tail tyrosine (ITT) motif in the cytoplasmic segments of membrane-bound IgGs (mIgGs) as the principle signal amplification device of memory-type BCRs in higher vertebrates and decipher its signalling microanatomy. We show that different families of protein tyrosine kinases act upstream and downstream of the ITT. Spleen tyrosine kinase (Syk) activity is required for ITT phosphorylation followed by recruitment of the adaptor protein Grb2 into the mIgG-BCR signalosome. Grb2 in turn recruits Bruton's tyrosine kinase (Btk) to amplify BCR-induced Ca(2+) mobilization. This molecular interplay of kinases and adaptors increases the antigen sensitivity of memory-type BCRs, which provides a cell-intrinsic trigger mechanism for the rapid reactivation of IgG-switched memory B cells on antigen recall.


Asunto(s)
Linfocitos B/metabolismo , Proteína Adaptadora GRB2/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptores de Antígenos de Linfocitos B/química , Receptores de Antígenos de Linfocitos B/metabolismo , Tirosina/metabolismo , Agammaglobulinemia Tirosina Quinasa , Secuencias de Aminoácidos , Animales , Calcio/metabolismo , Femenino , Proteína Adaptadora GRB2/genética , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/genética , Inmunoglobulina G/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Tirosina Quinasas/genética , Receptores de Antígenos de Linfocitos B/genética , Transducción de Señal , Tirosina/genética
20.
Immunol Lett ; 160(2): 109-12, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24852107

RESUMEN

B lymphocyte development in the mouse begins with the generation of long-term reconstituting, pluripotent hematopoietic stem cells, over multipotent myeloid/lymphoid progenitors and common lymphoid progenitors to B-lineage committed pro/pre B and pre B cells, which first express pre B cell receptors and then immunoglobulins, B cell receptors, to generate the repertoires of peripheral B cells. This development is influenced and guided by cells of non-hematopoietic and hematopoietic origins. We review here some of the recent developments, and our contributions in this fascinating field of developmental immunology.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...