Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
1.
J Exp Med ; 218(7)2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-33951726

RESUMEN

The pioneer transcription factor (TF) PU.1 controls hematopoietic cell fate by decompacting stem cell heterochromatin and allowing nonpioneer TFs to enter otherwise inaccessible genomic sites. PU.1 deficiency fatally arrests lymphopoiesis and myelopoiesis in mice, but human congenital PU.1 disorders have not previously been described. We studied six unrelated agammaglobulinemic patients, each harboring a heterozygous mutation (four de novo, two unphased) of SPI1, the gene encoding PU.1. Affected patients lacked circulating B cells and possessed few conventional dendritic cells. Introducing disease-similar SPI1 mutations into human hematopoietic stem and progenitor cells impaired early in vitro B cell and myeloid cell differentiation. Patient SPI1 mutations encoded destabilized PU.1 proteins unable to nuclear localize or bind target DNA. In PU.1-haploinsufficient pro-B cell lines, euchromatin was less accessible to nonpioneer TFs critical for B cell development, and gene expression patterns associated with the pro- to pre-B cell transition were undermined. Our findings molecularly describe a novel form of agammaglobulinemia and underscore PU.1's critical, dose-dependent role as a hematopoietic euchromatin gatekeeper.


Asunto(s)
Agammaglobulinemia/genética , Cromatina/genética , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Adolescente , Adulto , Linfocitos B/fisiología , Diferenciación Celular/genética , Línea Celular , Niño , Preescolar , Células Dendríticas/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Células HEK293 , Hematopoyesis/genética , Células Madre Hematopoyéticas/fisiología , Humanos , Lactante , Linfopoyesis/genética , Masculino , Mutación/genética , Células Precursoras de Linfocitos B/fisiología , Células Madre/fisiología , Adulto Joven
2.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33850015

RESUMEN

Central B cell tolerance, the process restricting the development of many newly generated autoreactive B cells, has been intensely investigated in mouse cells while studies in humans have been hampered by the inability to phenotypically distinguish autoreactive and nonautoreactive immature B cell clones and the difficulty in accessing fresh human bone marrow samples. Using a human immune system mouse model in which all human Igκ+ B cells undergo central tolerance, we discovered that human autoreactive immature B cells exhibit a distinctive phenotype that includes lower activation of ERK and differential expression of CD69, CD81, CXCR4, and other glycoproteins. Human B cells exhibiting these characteristics were observed in fresh human bone marrow tissue biopsy specimens, although differences in marker expression were smaller than in the humanized mouse model. Furthermore, the expression of these markers was slightly altered in autoreactive B cells of humanized mice engrafted with some human immune systems genetically predisposed to autoimmunity. Finally, by treating mice and human immune system mice with a pharmacologic antagonist, we show that signaling by CXCR4 is necessary to prevent both human and mouse autoreactive B cell clones from egressing the bone marrow, indicating that CXCR4 functionally contributes to central B cell tolerance.


Asunto(s)
Tolerancia Central/fisiología , Células Precursoras de Linfocitos B/metabolismo , Receptores CXCR4/metabolismo , Animales , Autoanticuerpos/metabolismo , Autoantígenos/inmunología , Autoinmunidad/inmunología , Linfocitos B/inmunología , Médula Ósea/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/genética , Tolerancia Central/inmunología , Femenino , Humanos , Tolerancia Inmunológica/genética , Recién Nacido , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Fenotipo , Células Precursoras de Linfocitos B/fisiología , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores CXCR4/inmunología , Receptores CXCR4/fisiología , Transducción de Señal/genética
3.
Mol Immunol ; 128: 150-164, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33129017

RESUMEN

During mammalian lymphoid development, Notch signaling is necessary at multiple stages of T lymphopoiesis, including lineage commitment, and later stages of T cell effector differentiation. In contrast, outside of a defined role in the development of splenic marginal zone B cells, there is conflicting evidence regarding whether Notch signaling plays functional roles in other B cell sub-populations. Complement receptor 2 (CR2) modulates BCR-signaling and is tightly regulated throughout differentiation. During B lymphopoiesis, CR2 is detected on immature and mature B cells with high surface expression on marginal zone B cells. Here, we have explored the possibility that Notch regulates human CR2 transcriptional activity using in vitro models including a co-culture system, co-transfection gene reporters and chromatin accessibility assays. We provide evidence that Notch signaling regulates CR2 promoter activity in a mature B cell line, as well as the induction of endogenous CR2 mRNA in a non-expressing pre-B cell line. The dynamics of endogenous gene activation suggests additional unidentified factors are required to mediate surface CR2 expression on immature and mature B lineage cells.


Asunto(s)
Complemento C3d/genética , Células Precursoras de Linfocitos B/fisiología , Regiones Promotoras Genéticas/genética , Receptores de Complemento 3d/genética , Receptores Notch/genética , Transducción de Señal/genética , Transcripción Genética/genética , Linfocitos B/fisiología , Diferenciación Celular/genética , Línea Celular , Línea Celular Tumoral , Cromatina/genética , Técnicas de Cocultivo/métodos , Humanos , Células K562 , Activación de Linfocitos/genética , Linfopoyesis/genética
4.
Sci Rep ; 10(1): 15193, 2020 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-32938995

RESUMEN

SHOC2 scaffold protein has been mainly related to oncogenic ERK signaling through the RAS-SHOC2-PP1 phosphatase complex. In leukemic cells however, SHOC2 upregulation has been previously related to an increased 5-year event-free survival of pediatric pre-B acute lymphoid leukemia, suggesting that SHOC2 could be a potential prognostic marker. To address such paradoxical function, our study investigated how SHOC2 impact leukemic cells drug response. Our transcriptome analysis has shown that SHOC2 can modulate the DNA-damage mediated by p53. Notably, upon genetic inhibition of SHOC2 we observed a significant impairment of p53 expression, which in turn, leads to the blockage of key apoptotic molecules. To confirm the specificity of DNA-damage related modulation, several anti-leukemic drugs has been tested and we did confirm that the proposed mechanism impairs cell death upon daunorubicin-induced DNA damage of human lymphoid cells. In conclusion, our study uncovers new insights into SHOC2 function and reveals that this scaffold protein may be essential to activate a novel mechanism of p53-induced cell death in pre-B lymphoid cells.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Leucemia Linfoide/metabolismo , Células Precursoras de Linfocitos B/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Antineoplásicos/uso terapéutico , Apoptosis , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Daunorrubicina/uso terapéutico , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Leucemia Linfoide/diagnóstico , Leucemia Linfoide/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas , Proteína p53 Supresora de Tumor/genética , Proteínas ras/metabolismo
5.
Nat Immunol ; 20(10): 1393-1403, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31477919

RESUMEN

In B lymphopoiesis, activation of the pre-B cell antigen receptor (pre-BCR) is associated with both cell cycle exit and Igk recombination. Yet how the pre-BCR mediates these functions remains unclear. Here, we demonstrate that the pre-BCR initiates a feed-forward amplification loop mediated by the transcription factor interferon regulatory factor 4 and the chemokine receptor C-X-C motif chemokine receptor 4 (CXCR4). CXCR4 ligation by C-X-C motif chemokine ligand 12 activates the mitogen-activated protein kinase extracellular-signal-regulated kinase, which then directs the development of small pre- and immature B cells, including orchestrating cell cycle exit, pre-BCR repression, Igk recombination and BCR expression. In contrast, pre-BCR expression and escape from interleukin-7 have only modest effects on B cell developmental transcriptional and epigenetic programs. These data show a direct and central role for CXCR4 in orchestrating late B cell lymphopoiesis. Furthermore, in the context of previous findings, our data provide a three-receptor system sufficient to recapitulate the essential features of B lymphopoiesis in vitro.


Asunto(s)
Linfocitos B/inmunología , Cadenas kappa de Inmunoglobulina/genética , Células Precursoras de Linfocitos B/fisiología , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores CXCR4/metabolismo , Animales , Puntos de Control del Ciclo Celular , Células Cultivadas , Quimiocina CXCL12/metabolismo , Femenino , Factores Reguladores del Interferón/genética , Linfopoyesis , Masculino , Ratones , Receptores de Antígenos de Linfocitos B/genética , Receptores CXCR4/genética , Recombinación Genética
6.
J Immunol ; 202(12): 3423-3433, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-31085591

RESUMEN

Induction of programmed DNA damage and its recognition and repair are fundamental for B cell development. The ssDNA-binding protein SSB1 has been described in human cells as essential for the recognition and repair of DNA damage. To study its relevance for B cells, we recently developed Ssb1 -/- and conditional Ssb1 -/- mice. Although SSB1 loss did not affect B cell development, Ssb1 -/- cells exhibited compensatory expression of its homolog SSB2. We have now generated Ssb2 -/- mice and show in this study that SSB2 is also dispensable for B cell development and DNA damage response activation. In contrast to the single loss of Ssb1 or Ssb2, however, combined SSB1/2 deficiency caused a defect in early B cell development. We relate this to the sensitivity of B cell precursors as mature B cells largely tolerated their loss. Toxicity of combined genetic SSB1/2 loss can be rescued by ectopic expression of either SSB1 or SSB2, mimicked by expression of SSB1 ssDNA-binding mutants, and attenuated by BCL2-mediated suppression of apoptosis. SSB1/2 loss in B cell precursors further caused increased exposure of ssDNA associated with disruption of genome fragile sites, inefficient cell cycle progression, and increased DNA damage if apoptosis is suppressed. As such, our results establish SSB1/2 as safeguards of B cell development and unveil their differential requirement in immature and mature B lymphocytes.


Asunto(s)
Linfocitos B/fisiología , Proteínas de Unión al ADN/metabolismo , Células Precursoras de Linfocitos B/fisiología , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Apoptosis , Diferenciación Celular , Células Cultivadas , Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN/genética , Genoma/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/genética
8.
Apoptosis ; 24(1-2): 145-156, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30578463

RESUMEN

Arginase has therapeutic potential as a cytotoxic agent in some cancers, but this is unclear for precursor B acute lymphoblastic leukaemia (pre-B ALL), the commonest form of childhood leukaemia. We compared arginase cytotoxicity with asparaginase, currently used in pre-B ALL treatment, and characterised the forms of cell death induced in a pre-B ALL cell line 697. Arginase and asparaginase both efficiently killed 697 cells and mature B lymphoma cell line Ramos, but neither enzyme killed normal lymphocytes. Arginase depleted cellular arginine, and arginase-treated media induced cell death, blocked by addition of arginine or arginine-precursor citrulline. Asparaginase depleted both asparagine and glutamine, and asparaginase-treated media induced cell death, blocked by asparagine, but not glutamine. Both enzymes induced caspase cleavage and activation, chromatin condensation and phosphatidylserine exposure, indicating apoptosis. Both arginase- and asparaginase-induced death were blocked by caspase inhibitors, but with different sensitivities. BCL-2 overexpression inhibited arginase- and asparaginase-induced cell death, but did not prevent arginase-induced cytostasis, indicating a different mechanism of growth arrest. An autophagy inhibitor, chloroquine, had no effect on the cell death induced by arginase, but doubled the cell death induced by asparaginase. In conclusion, arginase causes death of lymphoblasts by arginine-depletion induced apoptosis, via mechanism distinct from asparaginase. Therapeutic implications for childhood ALL include: arginase might be used as treatment (but antagonised by dietary arginine and citrulline), chloroquine may enhance efficacy of asparaginase treatment, and partial resistance to arginase and asparaginase may develop by BCL-2 expression. Arginase or asparaginase might potentially be used to treat Burkitt lymphoma.


Asunto(s)
Apoptosis/efectos de los fármacos , Arginasa/farmacología , Asparaginasa/farmacología , Células Precursoras de Linfocitos B/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Arginasa/fisiología , Arginasa/uso terapéutico , Asparaginasa/fisiología , Asparaginasa/uso terapéutico , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Niño , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Células Precursoras de Linfocitos B/fisiología
9.
Front Immunol ; 9: 2490, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30450096

RESUMEN

Receptor tyrosine kinase-like orphan receptor 2 (ROR2) has been identified as a highly relevant tumor-associated antigen in a variety of cancer indications of high unmet medical need, including renal cell carcinoma and osteosarcoma, making it an attractive target for targeted cancer therapy. Here, we describe the de novo discovery of fully human ROR2-specific antibodies and potent antibody drug conjugates (ADCs) derived thereof by combining antibody discovery from immune libraries of human immunoglobulin transgenic animals using the Transpo-mAb mammalian cell-based IgG display platform with functional screening for internalizing antibodies using a secondary ADC assay. The discovery strategy entailed immunization of transgenic mice with the cancer antigen ROR2, harboring transgenic IgH and IgL chain gene loci with limited number of fully human V, D, and J gene segments. This was followed by recovering antibody repertoires from the immunized animals, expressing and screening them as full-length human IgG libraries by transposon-mediated display in progenitor B lymphocytes ("Transpo-mAb Display") for ROR2 binding. Individual cellular "Transpo-mAb" clones isolated by single cell sorting and capable of expressing membrane-bound as well as secreted human IgG were directly screened during antibody discovery, not only for high affinity binding to human ROR2, but also functionally as ADCs using a cytotoxicity assay with a secondary anti-human IgG-toxin-conjugate. Using this strategy, we identified and validated 12 fully human, monoclonal anti-human ROR2 antibodies with nanomolar affinities that are highly potent as ADCs and could be promising candidates for the therapy of human cancer. The screening for functional and internalizing antibodies during the early phase of antibody discovery demonstrates the utility of the mammalian cell-based Transpo-mAb Display platform to select for functional binders and as a powerful tool to improve the efficiency for the development of therapeutically relevant ADCs.


Asunto(s)
Anticuerpos Monoclonales Humanizados/aislamiento & purificación , Anticuerpos Monoclonales/aislamiento & purificación , Inmunoconjugados/aislamiento & purificación , Neoplasias/terapia , Células Precursoras de Linfocitos B/fisiología , Animales , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Humanos , Inmunización , Inmunoconjugados/farmacología , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/genética , Inmunotoxinas/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/inmunología , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/inmunología , Análisis de la Célula Individual , Exones VDJ/genética
10.
Front Immunol ; 9: 2150, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30294329

RESUMEN

Humoral immunity depends on intrinsic B cell developmental programs guided by systemic signals that convey physiologic needs. Aberrant cues or their improper interpretation can lead to immune insufficiency or a failure of tolerance and autoimmunity. The means by which such systemic signals are conveyed remain poorly understood. Hence, further insight is essential to understanding and treating autoimmune diseases and to the development of improved vaccines. ST6Gal-1 is a sialyltransferase that constructs the α2,6-sialyl linkage on cell surface and extracellular glycans. The requirement for functional ST6Gal-1 in the development of humoral immunity is well documented. Canonically, ST6Gal-1 resides within the intracellular ER-Golgi secretory apparatus and participates in cell-autonomous glycosylation. However, a significant pool of extracellular ST6Gal-1 exists in circulation. Here, we segregate the contributions of B cell intrinsic and extrinsic ST6Gal-1 to B cell development. We observed that B cell-intrinsic ST6Gal-1 is required for marginal zone B cell development, while B cell non-autonomous ST6Gal-1 modulates B cell development and survival at the early transitional stages of the marrow and spleen. Exposure to extracellular ST6Gal-1 ex vivo enhanced the formation of IgM-high B cells from immature precursors, and increased CD23 and IgM expression. Extrinsic sialylation by extracellular ST6Gal-1 augmented BAFF-mediated activation of the non-canonical NF-kB, p38 MAPK, and PI3K/AKT pathways, and accelerated tyrosine phosphorylation after B cell receptor stimulation. in vivo, systemic ST6Gal-1 did not influence homing of B cells to the spleen but was critical for their long-term survival and systemic IgG levels. Circulatory ST6Gal-1 levels respond to inflammation, infection, and malignancy in mammals, including humans. In turn, we have shown previously that systemic ST6Gal-1 regulates inflammatory cell production by modifying bone marrow myeloid progenitors. Our data here point to an additional role of systemic ST6Gal-1 in guiding B cell development, which supports the concept that circulating ST6Gal-1 is a conveyor of systemic cues to guide the development of multiple branches of immune cells.


Asunto(s)
Diferenciación Celular/inmunología , Células Precursoras de Linfocitos B/fisiología , Sialiltransferasas/metabolismo , Traslado Adoptivo , Animales , Supervivencia Celular/inmunología , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Precursoras de Linfocitos B/trasplante , Cultivo Primario de Células , Sialiltransferasas/genética , Bazo/citología , Bazo/inmunología , Irradiación Corporal Total , beta-D-Galactósido alfa 2-6-Sialiltransferasa
11.
Front Immunol ; 9: 2074, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30271408

RESUMEN

To date there has not been a study directly comparing relative Igκ rearrangement frequencies obtained from genomic DNA (gDNA) and cDNA and since each approach has potential biases, this is an important issue to clarify. Here we used deep sequencing to compare the unbiased gDNA and RNA Igκ repertoire from the same pre-B cell pool. We find that ~20% of Vκ genes have rearrangement frequencies ≥2-fold up or down in RNA vs. DNA libraries, including many members of the Vκ3, Vκ4, and Vκ6 families. Regression analysis indicates Ikaros and E2A binding are associated with strong promoters. Within the pre-B cell repertoire, we observed that individual Vκ genes rearranged at very different frequencies, and also displayed very different Jκ usage. Regression analysis revealed that the greatly unequal Vκ gene rearrangement frequencies are best predicted by epigenetic marks of enhancers. In particular, the levels of newly arising H3K4me1 peaks associated with many Vκ genes in pre-B cells are most predictive of rearrangement levels. Since H3K4me1 is associated with long range chromatin interactions which are created during locus contraction, our data provides mechanistic insight into unequal rearrangement levels. Comparison of Igκ rearrangements occurring in pro-B cells and pre-B cells from the same mice reveal a pro-B cell bias toward usage of Jκ-distal Vκ genes, particularly Vκ10-96 and Vκ1-135. Regression analysis indicates that PU.1 binding is the highest predictor of Vκ gene rearrangement frequency in pro-B cells. Lastly, the repertoires of iEκ-/- pre-B cells reveal that iEκ actively influences Vκ gene usage, particularly Vκ3 family genes, overlapping with a zone of iEκ-regulated germline transcription. These represent new roles for iEκ in addition to its critical function in promoting overall Igκ rearrangement. Together, this study provides insight into many aspects of Igκ repertoire formation.


Asunto(s)
Linfocitos B/fisiología , Cadenas Ligeras de Inmunoglobulina/genética , Células Precursoras de Linfocitos B/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , ADN Complementario/genética , Epigénesis Genética , Reordenamiento Génico de Cadena Ligera de Linfocito B , Genoma , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Región Variable de Inmunoglobulina/genética , Cadenas kappa de Inmunoglobulina/genética , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo
12.
J Immunol ; 201(11): 3258-3268, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30373855

RESUMEN

The B cell survival cytokine BAFF has been linked with the pathogenesis of systemic lupus erythematosus (SLE). BAFF binds distinct BAFF-family surface receptors, including the BAFF-R and transmembrane activator and CAML interactor (TACI). Although originally characterized as a negative regulator of B cell activation, TACI signals are critical for class-switched autoantibody (autoAb) production in BAFF transgenic mice. Consistent with this finding, a subset of transitional splenic B cells upregulate surface TACI expression and contribute to BAFF-driven autoAb. In the current study, we interrogated the B cell signals required for transitional B cell TACI expression and Ab production. Surprisingly, despite established roles for dual BCR and TLR signals in autoAb production in SLE, signals downstream of these receptors exerted distinct impacts on transitional B cell TACI expression and autoAb titers. Whereas loss of BCR signals prevented transitional B cell TACI expression and resulted in loss of serum autoAb across all Ig isotypes, lack of TLR signals exerted a more limited impact restricted to autoAb class-switch recombination without altering transitional B cell TACI expression. Finally, in parallel with the protective effect of TACI deletion, loss of BAFF-R signaling also protected against BAFF-driven autoimmunity. Together, these findings highlight how multiple signaling pathways integrate to promote class-switched autoAb production by transitional B cells, events that likely impact the pathogenesis of SLE and other BAFF-dependent autoimmune diseases.


Asunto(s)
Autoanticuerpos/metabolismo , Glomerulonefritis por IGA/inmunología , Lupus Eritematoso Sistémico/inmunología , Glicoproteínas de Membrana/metabolismo , Células Precursoras de Linfocitos B/fisiología , Receptores de Antígenos de Linfocitos B/metabolismo , Receptor Toll-Like 7/metabolismo , Animales , Factor Activador de Células B/genética , Factor Activador de Células B/metabolismo , Receptor del Factor Activador de Células B/genética , Modelos Animales de Enfermedad , Humanos , Cambio de Clase de Inmunoglobulina , Activación de Linfocitos , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptor Cross-Talk , Receptores de Antígenos de Linfocitos B/genética , Transducción de Señal , Receptor Toll-Like 7/genética , Proteína Activadora Transmembrana y Interactiva del CAML/genética , Proteína Activadora Transmembrana y Interactiva del CAML/metabolismo
13.
Adv Immunol ; 139: 51-92, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30249334

RESUMEN

Activation-induced cytidine deaminase (AID) expression in the germinal center response drives the immunoglobulin class-switch recombination and V(D)J hypermutation necessary for efficacious, high-affinity antibody responses. That AID is expressed in developing lymphocytes is less well known, but represents an evolutionarily conserved pattern of lymphocyte development that is represented in all vertebrate species. Here we review the role of early, developmentally regulated AID expression in mice and humans and its role in establishing the first B-cell tolerance checkpoint. This newly recognized component of central tolerance requires coordinate signaling by poly- or autoreactive B-cell antigen receptors and endosomal Toll-like receptors. These signals synergize to upregulate AID expression in immature and transitional B cells to levels that approach that of germinal center B cells with the result of caspase 3-mediated cell death. In this review, we discuss the origins and mechanism of this interesting collaboration between adaptive and innate receptors to purge the primary B-cell repertoire of self-reactivity and how it may be related to receptor editing, the other major mechanism for central tolerance.


Asunto(s)
Linfocitos B/fisiología , Citidina Desaminasa/metabolismo , Centro Germinal/inmunología , Células Precursoras de Linfocitos B/fisiología , Animales , Formación de Anticuerpos , Citidina Desaminasa/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Tolerancia Inmunológica/genética , Activación de Linfocitos , Ratones , Receptor Cross-Talk , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo
14.
Front Immunol ; 9: 2053, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30250473

RESUMEN

In response to external stimuli, naïve B cells proliferate and take on a range of fates important for immunity. How their fate is determined is a topic of much recent research, with candidates including asymmetric cell division, lineage priming, stochastic assignment, and microenvironment instruction. Here we manipulate the generation of plasmablasts from B lymphocytes in vitro by varying CD40 stimulation strength to determine its influence on potential sources of fate control. Using long-term live cell imaging, we directly measure times to differentiate, divide, and die of hundreds of pairs of sibling cells. These data reveal that while the allocation of fates is significantly altered by signal strength, the proportion of siblings identified with asymmetric fates is unchanged. In contrast, we find that plasmablast generation is enhanced by slowing times to divide, which is consistent with a hypothesis of competing timed stochastic fate outcomes. We conclude that this mechanistically simple source of alternative fate regulation is important, and that useful quantitative models of signal integration can be developed based on its principles.


Asunto(s)
Linfocitos B/fisiología , Células Plasmáticas/fisiología , Células Precursoras de Linfocitos B/fisiología , Animales , Relojes Biológicos , Antígenos CD40/metabolismo , Diferenciación Celular , División Celular , Linaje de la Célula , Células Cultivadas , Femenino , Inmunización , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Procesos Estocásticos
15.
Eur J Immunol ; 48(6): 975-989, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29505092

RESUMEN

Hematopoietic stem cells and lineage-uncommitted progenitors are able to home to the bone marrow upon transplantation and reconstitute the host with hematopoietic progeny. Expression of miR221 in B-lineage committed preBI-cells induces their capacity to home to the bone marrow. However, the molecular mechanisms underlying miR221-controlled bone marrow homing and retention remain poorly understood. Here, we demonstrate, that miR221 regulates bone marrow retention of such B-cell precursors by targeting PTEN, thus enhancing PI3K signaling in response to the chemokine CXCL12. MiR221-enhanced PI3K signaling leads to increased expression of the anti-apoptotic protein Bcl2 and VLA4 integrin-mediated adhesion to VCAM1 in response to CXCL12 in vitro. Ablation of elevated PI3K activity abolishes the retention of miR221 expressing preBI-cells in the bone marrow. These results suggest that amplification of PI3K signaling by miR221 could be a general mechanism for bone marrow residence, shared by miR221-expressing hematopoietic cells.


Asunto(s)
Linfocitos B/fisiología , MicroARNs/genética , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Células Precursoras de Linfocitos B/fisiología , Animales , Médula Ósea/fisiología , Adhesión Celular , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Quimiocina CXCL12/inmunología , Integrina alfa4beta1/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-bcl-2/genética , Transducción de Señal
16.
J Allergy Clin Immunol ; 142(2): 630-646, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29391254

RESUMEN

BACKGROUND: Roifman syndrome is a rare inherited disorder characterized by spondyloepiphyseal dysplasia, growth retardation, cognitive delay, hypogammaglobulinemia, and, in some patients, thrombocytopenia. Compound heterozygous variants in the small nuclear RNA gene RNU4ATAC, which is necessary for U12-type intron splicing, were identified recently as driving Roifman syndrome. OBJECTIVE: We studied 3 patients from 2 unrelated kindreds harboring compound heterozygous or homozygous stem II variants in RNU4ATAC to gain insight into the mechanisms behind this disorder. METHODS: We systematically profiled the immunologic and hematologic compartments of the 3 patients with Roifman syndrome and performed RNA sequencing to unravel important splicing defects in both cell lineages. RESULTS: The patients exhibited a dramatic reduction in B-cell numbers, with differentiation halted at the transitional B-cell stage. Despite abundant B-cell activating factor availability, development past this B-cell activating factor-dependent stage was crippled, with disturbed minor splicing of the critical mitogen-activated protein kinase 1 signaling component. In the hematologic compartment patients with Roifman syndrome demonstrated defects in megakaryocyte differentiation, with inadequate generation of proplatelets. Platelets from patients with Roifman syndrome were rounder, with increased tubulin and actin levels, and contained increased α-granule and dense granule markers. Significant minor intron retention in 354 megakaryocyte genes was observed, including DIAPH1 and HPS1, genes known to regulate platelet and dense granule formation, respectively. CONCLUSION: Together, our results provide novel molecular and cellular data toward understanding the immunologic and hematologic features of Roifman syndrome.


Asunto(s)
Linfocitos B/fisiología , Plaquetas/fisiología , Cardiomiopatías/genética , Síndromes de Inmunodeficiencia/genética , Megacariocitos/fisiología , Discapacidad Intelectual Ligada al Cromosoma X/genética , Proteína Quinasa 1 Activada por Mitógenos/genética , Osteocondrodisplasias/genética , Células Precursoras de Linfocitos B/fisiología , ARN Nuclear Pequeño/genética , Enfermedades de la Retina/genética , Adolescente , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Niño , Preescolar , Humanos , Lactante , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Linaje , Enfermedades de Inmunodeficiencia Primaria , Empalme de Proteína/genética , Transducción de Señal/genética , Secuenciación del Exoma
17.
J Immunol ; 199(7): 2408-2420, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28807996

RESUMEN

Klhl6 belongs to the KLHL gene family, which is composed of an N-terminal BTB-POZ domain and four to six Kelch motifs in tandem. Several of these proteins function as adaptors of the Cullin3 E3 ubiquitin ligase complex. In this article, we report that Klhl6 deficiency induces, as previously described, a 2-fold reduction in mature B cells. However, we find that this deficit is centered on the inability of transitional type 1 B cells to survive and to progress toward the transitional type 2 B cell stage, whereas cells that have passed this step generate normal germinal centers (GCs) upon a T-dependent immune challenge. Klhl6-deficient type 1 B cells showed a 2-fold overexpression of genes linked with cell proliferation, including most targets of the anaphase-promoting complex/cyclosome complex, a set of genes whose expression is precisely downmodulated upon culture of splenic transitional B cells in the presence of BAFF. These results thus suggest a delay in the differentiation process of Klhl6-deficient B cells between the immature and transitional stage. We further show, in the BL2 Burkitt's lymphoma cell line, that KLHL6 interacts with Cullin3, but also that it binds to HBXIP/Lamtor5, a protein involved in cell-cycle regulation and cytokinesis. Finally, we report that KLHL6, which is recurrently mutated in B cell lymphomas, is an off-target of the normal somatic hypermutation process taking place in GC B cells in both mice and humans, thus leaving open whether, despite the lack of impact of Klhl6 deficiency on GC B cell expansion, mutants could contribute to the oncogenic process.


Asunto(s)
Linfocitos B/fisiología , Proteínas Portadoras/fisiología , Centro Germinal/citología , Animales , Linfocitos B/inmunología , Linfoma de Burkitt/patología , Proteínas Portadoras/genética , Diferenciación Celular , Línea Celular , Proliferación Celular , Centro Germinal/inmunología , Humanos , Linfoma de Células B/genética , Linfoma de Células B/patología , Ratones , Mutación , Células Precursoras de Linfocitos B/fisiología
18.
J Immunol ; 199(5): 1706-1715, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28739882

RESUMEN

CD79a and CD79b proteins associate with Ig receptors as integral signaling components of the B cell Ag receptor complex. To study B cell development in zebrafish, we isolated orthologs of these genes and performed in situ hybridization, finding that their expression colocalized with IgH-µ in the kidney, which is the site of B cell development. CD79 transgenic lines were made by linking the promoter and upstream regulatory segments of CD79a and CD79b to enhanced GFP to identify B cells, as demonstrated by PCR analysis of IgH-µ expression in sorted cells. We crossed these CD79-GFP lines to a recombination activating gene (Rag)2:mCherry transgenic line to identify B cell development stages in kidney marrow. Initiation of CD79:GFP expression in Rag2:mCherry+ cells and the timing of Ig H and L chain expression revealed simultaneous expression of both IgH-µ- and IgL-κ-chains, without progressing through the stage of IgH-µ-chain alone. Rag2:mCherry+ cells without CD79:GFP showed the highest Rag1 and Rag2 mRNAs compared with CD79a and CD79b:GFP+ B cells, which showed strongly reduced Rag mRNAs. Thus, B cell development in zebrafish does not go through a Raghi CD79+IgH-µ+ pre-B cell stage, different from mammals. After the generation of CD79:GFP+ B cells, decreased CD79 expression occurred upon differentiation to Ig secretion, as detected by alteration from membrane to secreted IgH-µ exon usage, similar to in mammals. This confirmed a conserved role for CD79 in B cell development and differentiation, without the requirement of a pre-B cell stage in zebrafish.


Asunto(s)
Linfocitos B/fisiología , Antígenos CD79/metabolismo , Proteínas de Peces/metabolismo , Riñón/fisiología , Células Precursoras de Linfocitos B/fisiología , Pez Cebra/inmunología , Animales , Animales Modificados Genéticamente , Antígenos CD79/genética , Diferenciación Celular , Clonación Molecular , Proteínas de Unión al ADN/genética , Proteínas de Peces/genética , Genes Reporteros/genética , Proteínas Fluorescentes Verdes/genética , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/metabolismo , Cadenas Ligeras de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/metabolismo , Activación de Linfocitos , Transgenes/genética , Proteínas de Pez Cebra/genética
19.
EMBO Rep ; 18(9): 1604-1617, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28705801

RESUMEN

Precursor B lymphocytes expand upon expression of a pre-B cell receptor (pre-BCR), but then transit into a resting state in which immunoglobulin light chain gene recombination is initiated. This bi-phasic sequence is orchestrated by the IL-7 receptor (IL-7R) and pre-BCR signaling, respectively, but little is known about microRNAs fine-tuning these events. Here, we show that pre-B cells lacking miR-15 family functions exhibit prolonged proliferation due to aberrant expression of the target genes cyclin E1 and D3. As a consequence, they fail to trigger the transcriptional reprogramming normally accompanying their differentiation, resulting in a developmental block at the pre-B cell stage. Intriguingly, our data indicate that the miR-15 family is suppressed by both IL-7R and pre-BCR signaling, suggesting it is actively integrated into the regulatory circuits of developing B cells. These findings identify the miR-15 family as a novel element required to promote the switch from pre-B cell proliferation to differentiation.


Asunto(s)
Diferenciación Celular , Proliferación Celular , MicroARNs/inmunología , MicroARNs/metabolismo , Células Precursoras de Linfocitos B/fisiología , Animales , Linfocitos B/inmunología , Ciclina D3/genética , Ciclina E/genética , Activación de Linfocitos , Linfopoyesis , Ratones , MicroARNs/genética , Proteínas Oncogénicas/genética , Receptores de Antígenos de Linfocitos B/genética , Receptores de Interleucina-7/genética , Transducción de Señal
20.
Proc Natl Acad Sci U S A ; 114(20): E3954-E3963, 2017 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-28461481

RESUMEN

Most tissue-resident macrophages (Mφs) are believed to be derived prenatally and are assumed to maintain themselves throughout life by self-proliferation. However, in adult mice we identified a progenitor within bone marrow, early pro-B cell/fraction B, that differentiates into tissue Mφs. These Mφ precursors have non-rearranged B-cell receptor genes and coexpress myeloid (GR1, CD11b, and CD16/32) and lymphoid (B220 and CD19) lineage markers. During steady state, these precursors exit bone marrow, losing Gr1, and enter the systemic circulation, seeding the gastrointestinal system as well as pleural and peritoneal cavities but not the brain. While in these tissues, they acquire a transcriptome identical to embryonically derived tissue-resident Mφs. Similarly, these Mφ precursors also enter sites of inflammation, gaining CD115, F4/80, and CD16/32, and become indistinguishable from blood monocyte-derived Mφs. Thus, we have identified a population of cells within the bone marrow early pro-B cell compartment that possess functional plasticity to differentiate into either tissue-resident or inflammatory Mφs, depending on microenvironmental signals. We propose that these precursors represent an additional source of Mφ populations in adult mice during steady state and inflammation.


Asunto(s)
Activación de Macrófagos/fisiología , Macrófagos/inmunología , Células Precursoras de Linfocitos B/fisiología , Animales , Linfocitos B/fisiología , Médula Ósea , Células de la Médula Ósea/fisiología , Homeostasis/fisiología , Inflamación/inmunología , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Células Precursoras de Linfocitos B/inmunología , Células Precursoras de Linfocitos B/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA