Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
1.
Nat Immunol ; 20(10): 1393-1403, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31477919

RESUMO

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.


Assuntos
Linfócitos B/imunologia , Cadeias kappa de Imunoglobulina/genética , Células Precursoras de Linfócitos B/fisiologia , Receptores de Antígenos de Linfócitos B/metabolismo , Receptores CXCR4/metabolismo , Animais , Pontos de Checagem do Ciclo Celular , Células Cultivadas , Quimiocina CXCL12/metabolismo , Feminino , Fatores Reguladores de Interferon/genética , Linfopoese , Masculino , Camundongos , Receptores de Antígenos de Linfócitos B/genética , Receptores CXCR4/genética , Recombinação Genética
2.
Nat Immunol ; 18(3): 321-333, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28135254

RESUMO

The FcµR receptor for the crystallizable fragment (Fc) of immunoglobulin M (IgM) can function as a cell-surface receptor for secreted IgM on a variety of cell types. We found here that FcµR was also expressed in the trans-Golgi network of developing B cells, where it constrained transport of the IgM-isotype BCR (IgM-BCR) but not of the IgD-isotype BCR (IgD-BCR). In the absence of FcµR, the surface expression of IgM-BCR was increased, which resulted in enhanced tonic BCR signaling. B-cell-specific deficiency in FcµR enhanced the spontaneous differentiation of B-1 cells, which resulted in increased serum concentrations of natural IgM and dysregulated homeostasis of B-2 cells; this caused the spontaneous formation of germinal centers, increased titers of serum autoantibodies and excessive accumulation of B cells. Thus, FcµR serves as a critical regulator of B cell biology by constraining the transport and cell-surface expression of IgM-BCR.


Assuntos
Linfócitos B/fisiologia , Imunoglobulina M/metabolismo , Células Precursoras de Linfócitos B/fisiologia , Receptores de Antígenos de Linfócitos B/metabolismo , Receptores Fc/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Citocinas/metabolismo , Feminino , Regulação da Expressão Gênica , Imunoglobulina M/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Antígenos de Linfócitos B/genética , Transdução de Sinais , Células Th1/imunologia , Células Th2/imunologia
3.
Nat Immunol ; 13(12): 1196-204, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23064439

RESUMO

The genome is folded into domains located in compartments that are either transcriptionally inert or transcriptionally permissive. Here we used genome-wide strategies to characterize domains during B cell development. Structured interaction matrix analysis showed that occupancy by the architectural protein CTCF was associated mainly with intradomain interactions, whereas sites bound by the histone acetyltransferase p300 or the transcription factors E2A or PU.1 were associated with intra- and interdomain interactions that are developmentally regulated. We identified a spectrum of genes that switched nuclear location during early B cell development. In progenitor cells, the transcriptionally inactive locus encoding early B cell factor (Ebf1) was sequestered at the nuclear lamina, which thereby preserved their multipotency. After development into the pro-B cell stage, Ebf1 and other genes switched compartments to establish new intra- and interdomain interactions associated with a B lineage-specific transcription signature.


Assuntos
Linfócitos B/fisiologia , Linhagem da Célula , Núcleo Celular/genética , Linfopoese , Células Precursoras de Linfócitos B/fisiologia , Animais , Linfócitos B/citologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fator de Ligação a CCCTC , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Lâmina Nuclear/metabolismo , Células Precursoras de Linfócitos B/citologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética , Transativadores/genética , Transativadores/metabolismo , Transcrição Gênica , Fatores de Transcrição de p300-CBP/genética
4.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33850015

RESUMO

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.


Assuntos
Tolerância Central/fisiologia , Células Precursoras de Linfócitos B/metabolismo , Receptores CXCR4/metabolismo , Animais , Autoanticorpos/metabolismo , Autoantígenos/imunologia , Autoimunidade/imunologia , Linfócitos B/imunologia , Medula Óssea/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Diferenciação Celular/genética , Tolerância Central/imunologia , Feminino , Humanos , Tolerância Imunológica/genética , Recém-Nascido , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Fenótipo , Células Precursoras de Linfócitos B/fisiologia , Receptores de Antígenos de Linfócitos B/metabolismo , Receptores CXCR4/imunologia , Receptores CXCR4/fisiologia , Transdução de Sinais/genética
5.
Nat Immunol ; 10(4): 403-11, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19252491

RESUMO

Immature B cells developing in the bone marrow are found in the parenchyma and sinusoids. The mechanisms that control the positioning of B cells in the sinusoids are not understood. Here we show that the integrin alpha(4)beta(1) (VLA-4) and its ligand VCAM-1 were required, whereas the chemokine receptor CXCR4 was dispensable, for sinusoidal retention of B cells. Instead, cannabinoid receptor 2 (CB2), a Galpha(i) protein-coupled receptor upregulated in immature B cells, was required for sinusoidal retention. Using two-photon microscopy, we found immature B cells entering and crawling in sinusoids; these immature B cells were displaced by CB2 antagonism. Moreover, CB2-deficient mice had a lower frequency of immunoglobulin lambda-chain-positive B cells in the peripheral blood and spleen. Our findings identify unique requirements for the retention of B cells in the bone marrow sinusoidal niche and suggest involvement of CB2 in the generation of the B cell repertoire.


Assuntos
Medula Óssea/fisiologia , Células Precursoras de Linfócitos B/fisiologia , Receptor CB2 de Canabinoide/fisiologia , Animais , Medula Óssea/imunologia , Movimento Celular/fisiologia , Subunidades alfa de Proteínas de Ligação ao GTP/fisiologia , Cadeias lambda de Imunoglobulina/sangue , Cadeias lambda de Imunoglobulina/imunologia , Integrina alfa4beta1/imunologia , Integrina alfa4beta1/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Precursoras de Linfócitos B/citologia , Células Precursoras de Linfócitos B/imunologia , Receptor CB2 de Canabinoide/genética , Receptor CB2 de Canabinoide/imunologia , Receptores CXCR4/imunologia , Receptores CXCR4/metabolismo , Baço/imunologia , Regulação para Cima , Molécula 1 de Adesão de Célula Vascular/imunologia , Molécula 1 de Adesão de Célula Vascular/fisiologia
6.
J Immunol ; 202(12): 3423-3433, 2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-31085591

RESUMO

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.


Assuntos
Linfócitos B/fisiologia , Proteínas de Ligação a DNA/metabolismo , Células Precursoras de Linfócitos B/fisiologia , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Animais , Apoptose , Diferenciação Celular , Células Cultivadas , Dano ao DNA , Reparo do DNA , Proteínas de Ligação a DNA/genética , Genoma/genética , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Supressoras da Sinalização de Citocina/genética
7.
J Immunol ; 201(11): 3258-3268, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30373855

RESUMO

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.


Assuntos
Autoanticorpos/metabolismo , Glomerulonefrite por IGA/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Glicoproteínas de Membrana/metabolismo , Células Precursoras de Linfócitos B/fisiologia , Receptores de Antígenos de Linfócitos B/metabolismo , Receptor 7 Toll-Like/metabolismo , Animais , Fator Ativador de Células B/genética , Fator Ativador de Células B/metabolismo , Receptor do Fator Ativador de Células B/genética , Modelos Animais de Doenças , Humanos , Switching de Imunoglobulina , Ativação Linfocitária , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptor Cross-Talk , Receptores de Antígenos de Linfócitos B/genética , Transdução de Sinais , Receptor 7 Toll-Like/genética , Proteína Transmembrana Ativadora e Interagente do CAML/genética , Proteína Transmembrana Ativadora e Interagente do CAML/metabolismo
8.
Proc Natl Acad Sci U S A ; 114(20): E3954-E3963, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28461481

RESUMO

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.


Assuntos
Ativação de Macrófagos/fisiologia , Macrófagos/imunologia , Células Precursoras de Linfócitos B/fisiologia , Animais , Linfócitos B/fisiologia , Medula Óssea , Células da Medula Óssea/fisiologia , Homeostase/fisiologia , Inflamação/imunologia , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/imunologia , Células Precursoras de Linfócitos B/imunologia , Células Precursoras de Linfócitos B/metabolismo
9.
Apoptosis ; 24(1-2): 145-156, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30578463

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Arginase/farmacologia , Asparaginase/farmacologia , Células Precursoras de Linfócitos B/efeitos dos fármacos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Arginase/fisiologia , Arginase/uso terapêutico , Asparaginase/fisiologia , Asparaginase/uso terapêutico , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Criança , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Células Precursoras de Linfócitos B/fisiologia
10.
Eur J Immunol ; 48(6): 975-989, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29505092

RESUMO

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.


Assuntos
Linfócitos B/fisiologia , MicroRNAs/genética , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/metabolismo , Células Precursoras de Linfócitos B/fisiologia , Animais , Medula Óssea/fisiologia , Adesão Celular , Diferenciação Celular , Movimento Celular , Células Cultivadas , Quimiocina CXCL12/imunologia , Integrina alfa4beta1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-bcl-2/genética , Transdução de Sinais
11.
EMBO Rep ; 18(9): 1604-1617, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28705801

RESUMO

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.


Assuntos
Diferenciação Celular , Proliferação de Células , MicroRNAs/imunologia , MicroRNAs/metabolismo , Células Precursoras de Linfócitos B/fisiologia , Animais , Linfócitos B/imunologia , Ciclina D3/genética , Ciclina E/genética , Ativação Linfocitária , Linfopoese , Camundongos , MicroRNAs/genética , Proteínas Oncogênicas/genética , Receptores de Antígenos de Linfócitos B/genética , Receptores de Interleucina-7/genética , Transdução de Sinais
12.
J Immunol ; 198(10): 3978-3988, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28381640

RESUMO

B cell development in Justy mutant mice is blocked due to a precursor mRNA splicing defect that depletes the protein GON4-like (GON4L) in B cell progenitors. Genetic and biochemical studies have suggested that GON4L is a transcriptional regulator that coordinates cell division with differentiation, but its role in B cell development is unknown. To understand the function of GON4L, we characterized B cell differentiation, cell cycle control, and mitotic gene expression in GON4L-deficient B cell progenitors from Justy mice. We found that these cells established key aspects of the transcription factor network that guides B cell development and proliferation and rearranged the IgH gene locus. However, despite intact IL-7 signaling, GON4L-deficient pro-B cell stage precursors failed to undergo a characteristic IL-7-dependent proliferative burst. These cells also failed to upregulate genes required for mitotic division, including those encoding the G1/S cyclin D3 and E2F transcription factors and their targets. Additionally, GON4L-deficient B cell progenitors displayed defects in DNA synthesis and passage through the G1/S transition, contained fragmented DNA, and underwent apoptosis. These phenotypes were not suppressed by transgenic expression of prosurvival factors. However, transgenic expression of cyclin D3 or other regulators of the G1/S transition restored pro-B cell development from Justy progenitor cells, suggesting that GON4L acts at the beginning of the cell cycle. Together, our findings indicate that GON4L is essential for cell cycle progression and division during the early stages of B cell development.


Assuntos
Mitose , Proteínas Nucleares/deficiência , Proteínas Nucleares/fisiologia , Células Precursoras de Linfócitos B/fisiologia , Animais , Linfócitos B/fisiologia , Ciclo Celular , Proteínas de Ciclo Celular , Divisão Celular , Proliferação de Células , Proteínas Correpressoras , Ciclina D3/genética , Proteínas de Ligação a DNA , Fator de Transcrição E2F4/genética , Fator de Transcrição E2F4/metabolismo , Regulação da Expressão Gênica , Interleucina-7/imunologia , Interleucina-7/metabolismo , Camundongos , Proteínas Nucleares/genética
13.
J Immunol ; 199(7): 2408-2420, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28807996

RESUMO

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.


Assuntos
Linfócitos B/fisiologia , Proteínas de Transporte/fisiologia , Centro Germinativo/citologia , Animais , Linfócitos B/imunologia , Linfoma de Burkitt/patologia , Proteínas de Transporte/genética , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Centro Germinativo/imunologia , Humanos , Linfoma de Células B/genética , Linfoma de Células B/patologia , Camundongos , Mutação , Células Precursoras de Linfócitos B/fisiologia
14.
J Immunol ; 199(5): 1706-1715, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28739882

RESUMO

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.


Assuntos
Linfócitos B/fisiologia , Antígenos CD79/metabolismo , Proteínas de Peixes/metabolismo , Rim/fisiologia , Células Precursoras de Linfócitos B/fisiologia , Peixe-Zebra/imunologia , Animais , Animais Geneticamente Modificados , Antígenos CD79/genética , Diferenciação Celular , Clonagem Molecular , Proteínas de Ligação a DNA/genética , Proteínas de Peixes/genética , Genes Reporter/genética , Proteínas de Fluorescência Verde/genética , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/metabolismo , Cadeias Leves de Imunoglobulina/genética , Cadeias Leves de Imunoglobulina/metabolismo , Ativação Linfocitária , Transgenes/genética , Proteínas de Peixe-Zebra/genética
15.
J Immunol ; 198(4): 1565-1574, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28062693

RESUMO

B cell development and Ig rearrangement are governed by cell type- and developmental stage-specific transcription factors. PU.1 and Spi-B are E26-transformation-specific transcription factors that are critical for B cell differentiation. To determine whether PU.1 and Spi-B are required for B cell development in the bone marrow, Spi1 (encoding PU.1) was conditionally deleted in B cells by Cre recombinase under control of the Mb1 gene in Spib (encoding Spi-B)-deficient mice. Combined deletion of Spi1 and Spib resulted in a lack of mature B cells in the spleen and a block in B cell development in the bone marrow at the small pre-B cell stage. To determine target genes of PU.1 that could explain this block, we applied a gain-of-function approach using a PU.1/Spi-B-deficient pro-B cell line in which PU.1 can be induced by doxycycline. PU.1-induced genes were identified by integration of chromatin immunoprecipitation-sequencing and RNA-sequencing data. We found that PU.1 interacted with multiple sites in the Igκ locus, including Vκ promoters and regions located downstream of Vκ second exons. Induction of PU.1 induced Igκ transcription and rearrangement. Upregulation of Igκ transcription was impaired in small pre-B cells from PU.1/Spi-B-deficient bone marrow. These studies reveal an important role for PU.1 in the regulation of Igκ transcription and rearrangement and a requirement for PU.1 and Spi-B in B cell development.


Assuntos
Linfócitos B/fisiologia , Diferenciação Celular , Regulação da Expressão Gênica , Cadeias Leves de Imunoglobulina/genética , Células Precursoras de Linfócitos B/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Transcrição Gênica , Animais , Doxiciclina/farmacologia , Ativação Linfocitária/imunologia , Camundongos , Células Precursoras de Linfócitos B/efeitos dos fármacos , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Transativadores/deficiência , Transativadores/genética
16.
J Allergy Clin Immunol ; 142(2): 630-646, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29391254

RESUMO

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.


Assuntos
Linfócitos B/fisiologia , Plaquetas/fisiologia , Cardiomiopatias/genética , Síndromes de Imunodeficiência/genética , Megacariócitos/fisiologia , Deficiência Intelectual Ligada ao Cromossomo X/genética , Proteína Quinase 1 Ativada por Mitógeno/genética , Osteocondrodisplasias/genética , Células Precursoras de Linfócitos B/fisiologia , RNA Nuclear Pequeno/genética , Doenças Retinianas/genética , Adolescente , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Criança , Pré-Escolar , Humanos , Lactente , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Linhagem , Doenças da Imunodeficiência Primária , Processamento de Proteína/genética , Transdução de Sinais/genética , Sequenciamento do Exoma
17.
Immunol Rev ; 263(1): 192-209, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25510278

RESUMO

Pre-B cells within the bone marrow represent the normal counterpart for most acute lymphoblastic leukemia (ALL). During normal early B-cell development, survival and proliferation signals are dominated by cytokines, particularly interleukin-7 (IL-7) for murine developing B cells. With expression of a functional pre-B-cell receptor (BCR), cytokine signaling is attenuated and the tonic/autonomous pre-BCR signaling pathway provides proliferation as well as differentiation signals. In this review, we first describe checkpoint mechanisms during normal B-cell development and then discuss how genetic lesions in these pathways function as oncogenic mimicries and allow transformed pre-B cells to bypass checkpoint control. We focus on cytokine receptor signaling that is mimicked by activating lesions in receptor subunits or downstream mediators as well as aberrant activation of non-B lymphoid cytokine receptors. Furthermore, we describe the molecular switch from cytokine receptor to pre-BCR signaling, how this pathway is of particular importance for certain ALL subtypes, and how pre-BCR signaling is engaged by genetic lesions, such as BCR-ABL1. We discuss the transcriptional control mechanisms downstream of both cytokine- and pre-BCR signaling and how normal checkpoint control mechanisms are circumvented in pre-B ALL. Finally, we highlight new therapeutic concepts for targeted inhibition of oncogenic cytokine or pre-BCR signaling pathways.


Assuntos
Proteínas de Fusão bcr-abl/metabolismo , Receptores de Células Precursoras de Linfócitos B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Células Precursoras de Linfócitos B/fisiologia , Animais , Antineoplásicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Citocinas/metabolismo , Proteínas de Fusão bcr-abl/genética , Humanos , Camundongos , Terapia de Alvo Molecular , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos
18.
EMBO J ; 33(3): 217-28, 2014 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-24442639

RESUMO

TopBP1 was initially identified as a topoisomerase II-ß-binding protein and it plays roles in DNA replication and repair. We found that TopBP1 is expressed at high levels in lymphoid tissues and is essential for early lymphocyte development. Specific abrogation of TopBP1 expression resulted in transitional blocks during early lymphocyte development. These defects were, in major part, due to aberrant V(D)J rearrangements in pro-B cells, double-negative and double-positive thymocytes. We also show that TopBP1 was located at sites of V(D)J rearrangement. In TopBP1-deficient cells, γ-H2AX foci were found to be increased. In addition, greater amount of γ-H2AX product was precipitated from the regions where TopBP1 was localized than from controls, indicating that TopBP1 deficiency results in inefficient DNA double-strand break repair. The developmental defects were rescued by introducing functional TCR αß transgenes. Our data demonstrate a novel role for TopBP1 as a crucial factor in V(D)J rearrangement during the development of B, T and iNKT cells.


Assuntos
Proteínas de Transporte/genética , Reparo do DNA , DNA/genética , Linfócitos/fisiologia , Recombinação V(D)J/imunologia , Animais , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Imunoprecipitação da Cromatina , Quebras de DNA de Cadeia Dupla , Dano ao DNA , Expressão Gênica , Histonas/genética , Histonas/metabolismo , Linfócitos/imunologia , Camundongos , Camundongos Knockout , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/fisiologia , Células Precursoras de Linfócitos B/imunologia , Células Precursoras de Linfócitos B/fisiologia , Deleção de Sequência , Organismos Livres de Patógenos Específicos , Transgenes , Recombinação V(D)J/genética
19.
Eur J Immunol ; 47(5): 911-920, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28294314

RESUMO

Deregulated expression of c-myc and bcl-xL is long known to generate transformed B cells in humans and mice. We overexpressed these genes to induce in vitro and in vivo differentiation of fetal liver-derived mouse pre-BI cells to B1-lineage pre-BII-like, immature and mature B-cell lines, and to Ig-secreting cells. In vitro, doxycycline-controlled c-myc/bcl-xL-overexpressing CD19+ CD93+ c-kikt+ IgM- pre-BI cells differentiate to and survive as CD19+ CD93+ c-kit- IgM+ immature B1 cells. Timed CpG stimulation of these oncogene-overexpressing pre-B or immature B1 cells generates either CD19+ CD93low c-kit- IgM- SLC- pre-BII-like or IgM+ MHCII+ CD73+ CD80+ CD40+ mature B1-cell lines and IgM-secreting B1 cells in vitro and fixes their state of differentiation. All cell lines are clonable, but a majority of immature and mature B1-cell clones eventually reach a nonproliferating, surviving G0 -state. Transplanted in vivo, c-myc/bcl-xL-overexpressing pre-B cells expand to mature B1 cells, and to IgM- and IgA-secreting plasmablasts and plasma cells. Within 2 months, plasmablasts have expanded most prominently in BM and spleen, indicating that the host selectively expanded development of these transformed plasma cells. The sIgM+ B1-cell lines and clones offer the possibility to study their roles in the development of B1-Ab repertoires, of B1-cell-mediated autoimmune diseases and of B1-cell malignancies.


Assuntos
Linfócitos B/fisiologia , Células Precursoras de Linfócitos B , Animais , Linfócitos B/imunologia , Diferenciação Celular , Linhagem Celular , Genes myc , Imunoglobulina A/imunologia , Imunoglobulina M/imunologia , Fígado/citologia , Fígado/imunologia , Camundongos , Plasmócitos/imunologia , Plasmócitos/fisiologia , Células Precursoras de Linfócitos B/imunologia , Células Precursoras de Linfócitos B/fisiologia , Baço/citologia , Baço/imunologia , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
20.
J Immunol ; 196(9): 3525-31, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-27022196

RESUMO

Mice overexpressing B cell activating factor of the TNF family (BAFF) develop systemic autoimmunity characterized by class-switched anti-nuclear Abs. Transmembrane activator and CAML interactor (TACI) signals are critical for BAFF-mediated autoimmunity, but the B cell developmental subsets undergoing TACI-dependent activation in settings of excess BAFF remain unclear. We report that, although surface TACI expression is usually limited to mature B cells, excess BAFF promotes the expansion of TACI-expressing transitional B cells. TACI(+) transitional cells from BAFF-transgenic mice are characterized by an activated, cycling phenotype, and the TACI(+) cell subset is specifically enriched for autoreactivity, expresses activation-induced cytidine deaminase and T-bet, and exhibits evidence of somatic hypermutation. Consistent with a potential contribution to BAFF-mediated humoral autoimmunity, TACI(+) transitional B cells from BAFF-transgenic mice spontaneously produce class-switched autoantibodies ex vivo. These combined findings highlight a novel mechanism through which BAFF promotes humoral autoimmunity via direct, TACI-dependent activation of transitional B cells.


Assuntos
Autoanticorpos/biossíntese , Fator Ativador de Células B/metabolismo , Células Precursoras de Linfócitos B/imunologia , Proteína Transmembrana Ativadora e Interagente do CAML/metabolismo , Animais , Autoanticorpos/imunologia , Autoimunidade , Fator Ativador de Células B/genética , Subpopulações de Linfócitos B/imunologia , Ativação Linfocitária , Camundongos , Camundongos Transgênicos , Células Precursoras de Linfócitos B/fisiologia , Proteína Transmembrana Ativadora e Interagente do CAML/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA