Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
2.
Nat Immunol ; 13(11): 1092-100, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23001146

RESUMEN

Germinal centers (GCs) are sites of intense B cell proliferation and are central for T cell-dependent antibody responses. However, the role of c-Myc, a key cell-cycle regulator, in this process has been questioned. Here we identified c-Myc(+) B cell subpopulations in immature and mature GCs and found, by genetic ablation of Myc, that they had indispensable roles in the formation and maintenance of GCs. The identification of these functionally critical cellular subsets has implications for human B cell lymphomagenesis, which originates mostly from GC B cells and frequently involves MYC chromosomal translocations. As these translocations are generally dependent on transcription of the recombining partner loci, the c-Myc(+) GC subpopulations may be at a particularly high risk for malignant transformation.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Linfocitos B/metabolismo , Ciclo Celular/genética , Centro Germinal/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Animales , Linfocitos B/inmunología , Linfocitos B/patología , Ciclo Celular/inmunología , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Eliminación de Gen , Regulación de la Expresión Génica/inmunología , Genes Reporteros , Sitios Genéticos , Centro Germinal/inmunología , Centro Germinal/patología , Proteínas Fluorescentes Verdes , Linfoma/genética , Linfoma/metabolismo , Linfoma/patología , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-myc/deficiencia , Proteínas Proto-Oncogénicas c-myc/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología , Translocación Genética
3.
Immunity ; 43(6): 1075-86, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26620760

RESUMEN

Phosphatidylinositol 3' OH kinase (PI3K) signaling and FOXO transcription factors play opposing roles at several B cell developmental stages. We show here abundant nuclear FOXO1 expression in the proliferative compartment of the germinal center (GC), its dark zone (DZ), and PI3K activity, downregulating FOXO1, in the light zone (LZ), where cells are selected for further differentiation. In the LZ, however, FOXO1 was expressed in a fraction of cells destined for DZ reentry. Upon FOXO1 ablation or induction of PI3K activity, GCs lost their DZ, owing at least partly to downregulation of the chemokine receptor CXCR4. Although this prevented proper cyclic selection of cells in GCs, somatic hypermutation and proliferation were maintained. Class switch recombination was partly lost due to a failure of switch region targeting by activation-induced deaminase (AID).


Asunto(s)
Linfocitos B/inmunología , Diferenciación Celular/inmunología , Factores de Transcripción Forkhead/inmunología , Centro Germinal/inmunología , Fosfatidilinositol 3-Quinasas/inmunología , Animales , Linfocitos B/citología , Separación Celular , Cromatografía Liquida , Citidina Desaminasa/inmunología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteína Forkhead Box O1 , Regulación de la Expresión Génica/inmunología , Centro Germinal/citología , Cambio de Clase de Inmunoglobulina/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Mutantes , Reacción en Cadena de la Polimerasa , Hipermutación Somática de Inmunoglobulina/inmunología , Espectrometría de Masas en Tándem
4.
Cell ; 139(3): 573-86, 2009 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-19879843

RESUMEN

Previous work has shown that mature B cells depend upon survival signals delivered to the cells by their antigen receptor (BCR). To identify the molecular nature of this survival signal, we have developed a genetic approach in which ablation of the BCR is combined with the activation of specific, BCR dependent signaling cascades in mature B cells in vivo. Using this system, we provide evidence that the survival of BCR deficient mature B cells can be rescued by a single signaling pathway downstream of the BCR, namely PI3K signaling, with the FOXO1 transcription factor playing a central role.


Asunto(s)
Linfocitos B/citología , Linfocitos B/metabolismo , Supervivencia Celular , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Animales , Ratones , Ratones Noqueados , Transducción de Señal
5.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33419925

RESUMEN

Affinity maturation depends on how efficiently germinal centers (GCs) positively select B cells in the light zone (LZ). Positively selected GC B cells recirculate between LZs and dark zones (DZs) and ultimately differentiate into plasmablasts (PBs) and memory B cells (MBCs). Current understanding of the GC reaction presumes that cMyc-dependent positive selection of LZ B cells is a competitive affinity-dependent process; however, this cannot explain the production of GC-derived lower-affinity MBCs or retention of GC B cells with varied affinities. Here, by combining single-cell/bulk RNA sequencing and flow cytometry, we identified and characterized temporally and functionally distinct positively selected cMyc+ GC B cell subpopulations. cMyc+ LZ B cell subpopulations enriched with either higher- or lower-affinity cells diverged soon after permissive positive selection. The former subpopulation contained PB precursors, whereas the latter comprised less proliferative MBC precursors and future DZ entrants. The overall affinity of future DZ entrants was enhanced in the LZ through preferential proliferation of higher-affinity cells. Concurrently, lower-affinity cells were retained in GCs and protected from apoptosis. These findings redefine positive selection as a dynamic process generating three distinct B cell fates and elucidate how positive selection ensures clonal diversity for broad protection.


Asunto(s)
Linfocitos B/metabolismo , Centro Germinal/inmunología , Animales , Apoptosis , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Selección Clonal Mediada por Antígenos , Femenino , Humanos , Ganglios Linfáticos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Plasmáticas , Receptores de Antígenos de Linfocitos B/genética
6.
Cell ; 131(1): 146-59, 2007 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-17923094

RESUMEN

MiR-150 is a microRNA (miRNA) specifically expressed in mature lymphocytes, but not their progenitors. A top predicted target of miR-150 is c-Myb, a transcription factor controlling multiple steps of lymphocyte development. Combining loss- and gain-of-function gene targeting approaches for miR-150 with conditional and partial ablation of c-Myb, we show that miR-150 indeed controls c-Myb expression in vivo in a dose-dependent manner over a narrow range of miRNA and c-Myb concentrations and that this dramatically affects lymphocyte development and response. Our results identify a key transcription factor as a critical target of a stage-specifically expressed miRNA in lymphocytes and suggest that this and perhaps other miRNAs have evolved to control the expression of just a few critical target proteins in particular cellular contexts.


Asunto(s)
Linfocitos B/fisiología , Diferenciación Celular/fisiología , Regulación de la Expresión Génica , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-myb/metabolismo , Regiones no Traducidas 3' , Animales , Linfocitos B/citología , Muerte Celular , Células Cultivadas , Marcación de Gen , Genes Reporteros , Humanos , Sistema Inmunológico/fisiología , Ratones , Ratones Noqueados , MicroARNs/genética , Proteínas Proto-Oncogénicas c-myb/genética , Linfocitos T/fisiología
7.
Nat Immunol ; 9(4): 405-14, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18327259

RESUMEN

The genomic region encoding the miR-17-92 microRNA (miRNA) cluster is often amplified in lymphoma and other cancers, and cancer cells carrying this amplification have higher expression of miRNA in this cluster. Retroviral expression of miR-17-92 accelerates c-Myc-induced lymphoma development, but precisely how higher expression of miR-17-92 promotes lymphomagenesis remains unclear. Here we generated mice with higher expression of miR-17-92 in lymphocytes. These mice developed lymphoproliferative disease and autoimmunity and died prematurely. Lymphocytes from these mice showed more proliferation and less activation-induced cell death. The miR-17-92 miRNA suppressed expression of the tumor suppressor PTEN and the proapoptotic protein Bim. This mechanism probably contributed to the lymphoproliferative disease and autoimmunity of miR-17-92-transgenic mice and contributes to lymphoma development in patients with amplifications of the miR-17-92 coding region.


Asunto(s)
Enfermedades Autoinmunes/genética , Linfocitos/inmunología , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/inmunología , MicroARNs/biosíntesis , MicroARNs/genética , Animales , Enfermedades Autoinmunes/patología , Muerte Celular/genética , Muerte Celular/inmunología , Proliferación Celular , Células Cultivadas , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Células Jurkat , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Linfocitos/metabolismo , Linfoma/genética , Linfoma/inmunología , Trastornos Linfoproliferativos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/fisiología
8.
Immunity ; 30(1): 80-91, 2009 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-19144316

RESUMEN

Foxp3(+) regulatory T (Treg) cells limit pathogenic immune responses to self-antigens and foreign antigens. An essential role for microRNA (miRNA) in the maintenance and function of Treg cells, revealed by the Treg cell-specific Dicer ablation, raised a question as to a specific miRNA contribution. We found that Foxp3 controlled the elevated miR155 expression required for maintaining Treg cell proliferative activity and numbers under nonlymphopenic conditions. Moreover, miR155 deficiency in Treg cells resulted in increased suppressor of cytokine signaling 1 (SOCS1) expression accompanied by impaired activation of signal transducer and activator of transcription 5 (STAT5) transcription factor in response to limiting amounts of interleukin-2. Our studies suggest that Foxp3-dependent regulation of miR155 maintains competitive fitness of Treg cell subsets by targeting SOCS1, and they provide experimental support for a proposed role for miRNAs in ensuring the robustness of cellular phenotypes.


Asunto(s)
Factores de Transcripción Forkhead/fisiología , MicroARNs , Proteínas Supresoras de la Señalización de Citocinas/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Animales , Secuencia de Bases , Citometría de Flujo , Factores de Transcripción Forkhead/farmacología , Humanos , Inmunohistoquímica , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Alineación de Secuencia , Proteína 1 Supresora de la Señalización de Citocinas
9.
Sci Immunol ; 9(94): eadk0092, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38579014

RESUMEN

The transition from immunoglobulin M (IgM) to affinity-matured IgG antibodies is vital for effective humoral immunity. This is facilitated by germinal centers (GCs) through affinity maturation and preferential maintenance of IgG+ B cells over IgM+ B cells. However, it is not known whether the positive selection of the different Ig isotypes within GCs is dependent on specific transcriptional mechanisms. Here, we explored IgG1+ GC B cell transcription factor dependency using a CRISPR-Cas9 screen and conditional mouse genetics. We found that MIZ1 was specifically required for IgG1+ GC B cell survival during positive selection, whereas IgM+ GC B cells were largely independent. Mechanistically, MIZ1 induced TMBIM4, an ancestral anti-apoptotic protein that regulated inositol trisphosphate receptor (IP3R)-mediated calcium (Ca2+) mobilization downstream of B cell receptor (BCR) signaling in IgG1+ B cells. The MIZ1-TMBIM4 axis prevented mitochondrial dysfunction-induced IgG1+ GC cell death caused by excessive Ca2+ accumulation. This study uncovers a unique Ig isotype-specific dependency on a hitherto unidentified mechanism in GC-positive selection.


Asunto(s)
Linfocitos B , Inmunoglobulina G , Proteínas de la Membrana , Animales , Ratones , Centro Germinal , Inmunoglobulina G/metabolismo , Inmunoglobulina M/metabolismo , Transducción de Señal , Proteínas de la Membrana/metabolismo
10.
bioRxiv ; 2023 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-37609190

RESUMEN

To increase antibody affinity against pathogens, positively selected GC-B cells initiate cell division in the light zone (LZ) of germinal centres (GCs). Among those, higher-affinity clones migrate to the dark zone (DZ) and vigorously proliferate by relying on oxidative phosphorylation (OXPHOS). However, it remains unknown how positively selected GC-B cells adapt their metabolism for cell division in the glycolysis-dominant, cell cycle arrest-inducing, hypoxic LZ microenvironment. Here, we show that microRNA (miR)-155 mediates metabolic reprogramming during positive selection to protect high-affinity clones. Transcriptome examination and mass spectrometry analysis revealed that miR-155 regulates H3K36me2 levels by directly repressing hypoxia-induced histone lysine demethylase, Kdm2a. This is indispensable for enhancing OXPHOS through optimizing the expression of vital nuclear mitochondrial genes under hypoxia. The miR-155-Kdm2a interaction is crucial to prevent excessive production of reactive oxygen species and apoptosis. Thus, miR-155-mediated epigenetic regulation promotes mitochondrial fitness in high-affinity clones, ensuring their expansion and consequently affinity maturation.

11.
Blood Cancer Discov ; 4(1): 78-97, 2023 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-36346827

RESUMEN

Genomic profiling revealed the identity of at least 5 subtypes of diffuse large B-cell lymphoma (DLBCL), including the MCD/C5 cluster characterized by aberrations in MYD88, BCL2, PRDM1, and/or SPIB. We generated mouse models harboring B cell-specific Prdm1 or Spib aberrations on the background of oncogenic Myd88 and Bcl2 lesions. We deployed whole-exome sequencing, transcriptome, flow-cytometry, and mass cytometry analyses to demonstrate that Prdm1- or Spib-altered lymphomas display molecular features consistent with prememory B cells and light-zone B cells, whereas lymphomas lacking these alterations were enriched for late light-zone and plasmablast-associated gene sets. Consistent with the phenotypic evidence for increased B cell receptor signaling activity in Prdm1-altered lymphomas, we demonstrate that combined BTK/BCL2 inhibition displays therapeutic activity in mice and in five of six relapsed/refractory DLBCL patients. Moreover, Prdm1-altered lymphomas were immunogenic upon transplantation into immuno-competent hosts, displayed an actionable PD-L1 surface expression, and were sensitive to antimurine-CD19-CAR-T cell therapy, in vivo. SIGNIFICANCE: Relapsed/refractory DLBCL remains a major medical challenge, and most of these patients succumb to their disease. Here, we generated mouse models, faithfully recapitulating the biology of MYD88-driven human DLBCL. These models revealed robust preclinical activity of combined BTK/BCL2 inhibition. We confirmed activity of this regimen in pretreated non-GCB-DLBCL patients. See related commentary by Leveille et al., p. 8. This article is highlighted in the In This Issue feature, p. 1.


Asunto(s)
Linfoma de Células B Grandes Difuso , Factor 88 de Diferenciación Mieloide , Humanos , Ratones , Animales , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Linfocitos B , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/terapia , Células Plasmáticas/metabolismo , Células Plasmáticas/patología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/uso terapéutico
12.
Cancers (Basel) ; 14(18)2022 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-36139665

RESUMEN

During the anti-tumour response to breast cancer, the primary tumour, the peripheral blood, and the lymph nodes each play unique roles. Immunological features at each site reveal evidence of continuous immune cross-talk between them before, during and after treatment. As such, immune responses to breast cancer are found to be highly dynamic and truly systemic, integrating three distinct immune sites, complex cell-migration highways, as well as the temporal dimension of disease progression and treatment. In this review, we provide a connective summary of the dynamic immune environment triad of breast cancer. It is critical that future studies seek to establish dynamic immune profiles, constituting multiple sites, that capture the systemic immune response to breast cancer and define patient-selection parameters resulting in more significant overall responses and survival rates for breast cancer patients.

13.
Front Immunol ; 12: 661678, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33868314

RESUMEN

Germinal centers (GCs) are essential sites for the production of high-affinity antibody secreting plasma cells (PCs) and memory-B cells (MBCs), which form the framework of vaccination. Affinity maturation and permissive selection in GCs are key for the production of PCs and MBCs, respectively. For these purposes, GCs positively select "fit" cells in the light zone of the GC and instructs them for one of three known B cell fates: PCs, MBCs and persistent GC-B cells as dark zone entrants. In this review, we provide an overview of the positive selection process and discuss its mechanisms and how B cell fates are instructed.


Asunto(s)
Anticuerpos/inmunología , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Centro Germinal/inmunología , Memoria Inmunológica/inmunología , Células Plasmáticas/inmunología , Animales , Anticuerpos/metabolismo , Afinidad de Anticuerpos/inmunología , Linfocitos B/metabolismo , Centro Germinal/citología , Centro Germinal/metabolismo , Humanos , Células Plasmáticas/metabolismo , Receptores de Antígenos de Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos B/metabolismo
14.
Front Mol Biosci ; 8: 673051, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34124156

RESUMEN

Lymph nodes (LNs) are highly organized secondary lymphoid organs, and reflective of immune responses to infection, injuries, or the presence of cancer. Extensive molecular and morphological analyses of immune and stromal features in tumors and LNs of breast cancer patients have revealed novel patterns indicative of disease progression. Within LNs, there are dynamic structures called germinal centers (GCs), that act as the immunological hubs for B cell development and generation of affinity matured memory B and antibody-producing plasma cells. Acting as a bridge between systemic and local immunity, associations are observed between the frequency of GCs within cancer-free LNs, the levels of stromal tumor infiltrating lymphocytes, and cancer progression. Scattered throughout the tumor microenvironment (TME) or aggregated in clusters forming tertiary lymphoid structures (TLS), the occurrence of tumor infiltrating B cells (TIL-Bs) has been linked mostly to superior disease trajectories in solid cancers. Recent TIL-Bs profiling studies have revealed a plethora of different TIL-B populations, their functional roles, and whether they are derived from GC reactions in the LN, and/or locally from GC-like structures within the TME remains to be investigated. However, parallels between the immunogenic nature of LNs as a pre-metastatic niche, TIL-B populations within the TME, and the presence of TLS will help to decipher local and widespread TIL-Bs responses and their influence on cancer progression to the lymphatics. Therapies that enhance TIL-Bs responses in the LN GC and/or in GC-like structures in the TME are thus emerging management strategies for breast and other cancer patients.

15.
Elife ; 92020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-33136000

RESUMEN

Plasma cells (PCs) are essential for protection from infection, and at the origin of incurable cancers. Current studies do not circumvent the limitations of removing PCs from their microenvironment and confound formation and maintenance. Also, the investigation of PC population dynamics has mostly relied on nucleotide analog incorporation that does not label quiescent cells, a property of most PCs. The main impediment is the lack of tools to perform specific genetic manipulation in vivo. Here we characterize a genetic tool (JchaincreERT2) in the mouse that permits first-ever specific genetic manipulation in PCs in vivo, across immunoglobulin isotypes. Using this tool, we found that splenic and bone marrow PC numbers remained constant over-time with the decay in genetically labeled PCs being compensated by unlabeled PCs, supporting homeostatic population turnover in these tissues. The JchaincreERT2 tool paves the way for an in-depth mechanistic understanding of PC biology and pathology in vivo, in their microenvironment.


Asunto(s)
Homeostasis , Isotipos de Inmunoglobulinas/genética , Células Plasmáticas/inmunología , Animales , Médula Ósea/inmunología , Células de la Médula Ósea/inmunología , Isotipos de Inmunoglobulinas/inmunología , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos , Bazo/citología , Bazo/inmunología
16.
J Exp Med ; 217(7)2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32407433

RESUMEN

Memory B cells (MBCs) are key for protection from reinfection. However, it is mechanistically unclear how germinal center (GC) B cells differentiate into MBCs. MYC is transiently induced in cells fated for GC expansion and plasma cell (PC) formation, so-called positively selected GC B cells. We found that these cells coexpressed MYC and MIZ1 (MYC-interacting zinc-finger protein 1 [ZBTB17]). MYC and MIZ1 are transcriptional activators; however, they form a transcriptional repressor complex that represses MIZ1 target genes. Mice lacking MYC-MIZ1 complexes displayed impaired cell cycle entry of positively selected GC B cells and reduced GC B cell expansion and PC formation. Notably, absence of MYC-MIZ1 complexes in positively selected GC B cells led to a gene expression profile alike that of MBCs and increased MBC differentiation. Thus, at the GC positive selection stage, MYC-MIZ1 complexes are required for effective GC expansion and PC formation and to restrict MBC differentiation. We propose that MYC and MIZ1 form a module that regulates GC B cell fate.


Asunto(s)
Linfocitos B/citología , Diferenciación Celular , Centro Germinal/citología , Memoria Inmunológica , Animales , Linfocitos B/metabolismo , Ciclo Celular/genética , Proliferación Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Citidina Desaminasa/metabolismo , Ratones Noqueados , Unión Proteica , Proteínas Inhibidoras de STAT Activados/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Regulación hacia Arriba/genética
17.
Cell Rep ; 11(5): 715-26, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25921526

RESUMEN

Diffuse large B cell lymphoma (DLBCL) is a complex disease comprising diverse subtypes and genetic profiles. Possibly because of the prevalence of genetic alterations activating canonical NF-κB activity, a role for oncogenic lesions that activate the alternative NF-κB pathway in DLBCL has remained elusive. Here, we show that deletion/mutation of TRAF3, a negative regulator of the alternative NF-κB pathway, occurs in ∼15% of DLBCLs and that it often coexists with BCL6 translocation, which prevents terminal B cell differentiation. Accordingly, in a mouse model constitutive activation of the alternative NF-κB pathway cooperates with BCL6 deregulation in DLBCL development. This work demonstrates a key oncogenic role for the alternative NF-κB pathway in DLBCL development.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , FN-kappa B/metabolismo , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Supervivencia Celular , Proteínas de Unión al ADN/deficiencia , Humanos , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma de Células B Grandes Difuso/patología , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6 , Transducción de Señal , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/metabolismo , Quinasa de Factor Nuclear kappa B
18.
J Exp Med ; 208(13): 2733-46, 2011 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-22143888

RESUMEN

Immunoglobulin heavy chain (IgH) class-switch recombination (CSR) replaces initially expressed Cµ (IgM) constant regions (C(H)) exons with downstream C(H) exons. Stimulation of B cells with anti-CD40 plus interleukin-4 induces CSR from Cµ to Cγ1 (IgG1) and Cε (IgE), the latter of which contributes to the pathogenesis of atopic diseases. Although Cε CSR can occur directly from Cµ, most mature peripheral B cells undergo CSR to Cε indirectly, namely from Cµ to Cγ1, and subsequently to Cε. Physiological mechanisms that influence CSR to Cγ1 versus Cε are incompletely understood. In this study, we report a role for B cell developmental maturity in IgE CSR. Based in part on a novel flow cytometric IgE CSR assay, we show that immature B cells preferentially switch to IgE versus IgG1 through a mechanism involving increased direct CSR from Cµ to Cε. Our findings suggest that IgE dysregulation in certain immunodeficiencies may be related to impaired B cell maturation.


Asunto(s)
Linfocitos B/inmunología , Cambio de Clase de Inmunoglobulina/fisiología , Inmunoglobulina E/inmunología , Cadenas epsilon de Inmunoglobulina/inmunología , Cadenas mu de Inmunoglobulina/inmunología , Recombinación Genética/fisiología , Animales , Inmunodeficiencia Variable Común/genética , Inmunodeficiencia Variable Común/inmunología , Inmunoglobulina E/genética , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Cadenas epsilon de Inmunoglobulina/genética , Cadenas mu de Inmunoglobulina/genética , Ratones , Ratones Noqueados
19.
Cancer Cell ; 18(6): 580-9, 2010 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-21156282

RESUMEN

Diffuse large B cell lymphoma (DLBCL) comprises disease entities with distinct genetic profiles, including germinal center B cell (GCB)-like and activated B cell (ABC)-like DLBCLs. Major differences between these two subtypes include genetic aberrations leading to constitutive NF-κB activation and interference with terminal B cell differentiation through BLIMP1 inactivation, observed in ABC- but not GCB-DLBCL. Using conditional gain-of-function and/or loss-of-function mutagenesis in the mouse, we show that constitutive activation of the canonical NF-κB pathway cooperates with disruption of BLIMP1 in the development of a lymphoma that resembles human ABC-DLBCL. Our work suggests that both NF-κB signaling, as an oncogenic event, and BLIMP1, as a tumor suppressor, play causal roles in the pathogenesis of ABC-DLBCL.


Asunto(s)
Linfoma de Células B Grandes Difuso/etiología , FN-kappa B/metabolismo , Factores de Transcripción/fisiología , Animales , Proliferación Celular , Centro Germinal/fisiología , Quinasa I-kappa B/genética , Ratones , Mutación , Células Plasmáticas/fisiología , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Factores de Transcripción/genética
20.
Science ; 324(5932): 1334-8, 2009 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-19498172

RESUMEN

A central challenge for improving autoimmune therapy is preventing inflammatory pathology without inducing generalized immunosuppression. T helper 17 (TH17) cells, characterized by their production of interleukin-17, have emerged as important and broad mediators of autoimmunity. Here we show that the small molecule halofuginone (HF) selectively inhibits mouse and human TH17 differentiation by activating a cytoprotective signaling pathway, the amino acid starvation response (AAR). Inhibition of TH17 differentiation by HF is rescued by the addition of excess amino acids and is mimicked by AAR activation after selective amino acid depletion. HF also induces the AAR in vivo and protects mice from TH17-associated experimental autoimmune encephalomyelitis. These results indicate that the AAR pathway is a potent and selective regulator of inflammatory T cell differentiation in vivo.


Asunto(s)
Aminoácidos/metabolismo , Piperidinas/farmacología , Quinazolinonas/farmacología , Subgrupos de Linfocitos T/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Factor de Transcripción Activador 4/metabolismo , Aminoácidos/farmacología , Animales , Autoinmunidad/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Factor 2 Eucariótico de Iniciación/metabolismo , Expresión Génica , Humanos , Interleucina-17/biosíntesis , Interleucina-17/genética , Linfopoyesis/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Fosforilación , Piperidinas/uso terapéutico , Proteínas Serina-Treonina Quinasas/metabolismo , Quinazolinonas/uso terapéutico , Transducción de Señal , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA