scholarly journals The transcriptional coactivator Maml1 is required for Notch2-mediated marginal zone B-cell development

Blood ◽  
2007 ◽  
Vol 110 (10) ◽  
pp. 3618-3623 ◽  
Author(s):  
Lizi Wu ◽  
Ivan Maillard ◽  
Makoto Nakamura ◽  
Warren S. Pear ◽  
James D. Griffin

Abstract Signaling mediated by various Notch receptors and their ligands regulates diverse biological processes, including lymphoid cell fate decisions. Notch1 is required during T-cell development, while Notch2 and the Notch ligand Delta-like1 control marginal zone B (MZB) cell development. We previously determined that Mastermind-like (MAML) transcriptional coactivators are required for Notchinduced transcription by forming ternary nuclear complexes with Notch and the transcription factor CSL. The 3 MAML family members (MAML1-MAML3) are collectively essential for Notch activity in vivo, but whether individual MAMLs contribute to the specificity of Notch functions is unknown. Here, we addressed this question by studying lymphopoiesis in the absence of the Maml1 gene. Since Maml1−/− mice suffered perinatal lethality, hematopoietic chimeras were generated with Maml1−/−, Maml1+/−, or wild-type fetal liver progenitors. Maml1 deficiency minimally affected T-cell development, but was required for the development of MZB cells, similar to the phenotype of Notch2 deficiency. Moreover, the number of MZB cells correlated with Maml1 gene dosage. Since all 3 Maml genes were expressed in MZB cells and their precursors, these results suggest that Maml1 is specifically required for Notch2 signaling in MZB cells.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2294-2294
Author(s):  
Tomofusa Fukuyama ◽  
Fayçal Boussouar ◽  
Lawryn H. Kasper ◽  
Jan M. van Deursen ◽  
Paul K. Brindle

Abstract Defining the epigenetic mechanisms (e.g. chromatin modifications) that underlie T cell fate decisions is a major challenge. The transcriptional coactivators CREB binding protein (CBP) and the closely related p300 comprise a two-member family of histone/protein acetyltransferases that interact with over 50 T lymphocyte-essential transcriptional regulators. Rather than having distinct regulatory roles, CBP and p300 are often thought to confer utilitarian transactivation and histone modifying functions to transcription factors that mediate T cell fate. In contrast to this view, we show here that CBP acts uniquely in conventional T cell development. Inactivation of CBP, but not p300, starting at the double negative stage of T cell development yielded thymocytes with partial activation of an effector/memory- or innate-T cell program. CD8SP thymocytes from CBP mutant mice expressed genes that define professional CD8 cells such as Il-2/Il-15 receptor β chain, granzyme A, interferon γ (Ifnγ), Fas ligand, perforin, and the chemokine receptors Ccr5, and Cxcr3. CD4SP thymocytes from CBP mutant mice also expressed effector genes such as Ifnγ, Il-4, and Ccr5. In addition, CD8SP and CD4SP thymocytes from CBP mutant mice produced Ifnγ protein when the cells were stimulated with phorbol ester and ionomycin. Mechanistically, loss of CBP acted cell non-autonomously to induce the expression of the CD8 T cell master regulatory transcription factor eomesodermin (Eomes). This suggests that CBP in thymocytes or T cells controls an extracellular factor that helps demarcate conventional naïve T cell development in the thymus from effector/memory T cell differentiation in the periphery.


Blood ◽  
2010 ◽  
Vol 115 (6) ◽  
pp. 1137-1144 ◽  
Author(s):  
Namita Saran ◽  
Marcin Łyszkiewicz ◽  
Jens Pommerencke ◽  
Katrin Witzlau ◽  
Ramin Vakilzadeh ◽  
...  

Abstract T-cell development in the thymus depends on continuous supply of T-cell progenitors from bone marrow (BM). Several extrathymic candidate progenitors have been described that range from multipotent cells to lymphoid cell committed progenitors and even largely T-lineage committed precursors. However, the nature of precursors seeding the thymus under physiologic conditions has remained largely elusive and it is not known whether there is only one physiologic T-cell precursor population or many. Here, we used a competitive in vivo assay based on depletion rather than enrichment of classes of BM-derived precursor populations, thereby only minimally altering physiologic precursor ratios to assess the contribution of various extrathymic precursors to T-lineage differentiation. We found that under these conditions multiple precursors, belonging to both multipotent progenitor (MPP) and common lymphoid progenitor (CLP) subsets have robust T-lineage potential. However, differentiation kinetics of different precursors varied considerably, which might ensure continuous thymic output despite gated importation of extrathymic precursors. In conclusion, our data suggest that the thymus functions to impose T-cell fate on any precursor capable of filling the limited number of progenitor niches.


2010 ◽  
Vol 207 (3) ◽  
pp. 623-635 ◽  
Author(s):  
David R. Gibb ◽  
Mohey El Shikh ◽  
Dae-Joong Kang ◽  
Warren J. Rowe ◽  
Rania El Sayed ◽  
...  

The proteolytic activity of a disintegrin and metalloproteinase 10 (ADAM10) regulates cell-fate decisions in Drosophila and mouse embryos. However, in utero lethality of ADAM10−/− mice has prevented examination of ADAM10 cleavage events in lymphocytes. To investigate their role in B cell development, we generated B cell–specific ADAM10 knockout mice. Intriguingly, deletion of ADAM10 prevented development of the entire marginal zone B cell (MZB) lineage. Additionally, cleavage of the low affinity IgE receptor, CD23, was profoundly impaired, but subsequent experiments demonstrated that ADAM10 regulates CD23 cleavage and MZB development by independent mechanisms. Development of MZBs is dependent on Notch2 signaling, which requires proteolysis of the Notch2 receptor by a previously unidentified proteinase. Further experiments revealed that Notch2 signaling is severely impaired in ADAM10-null B cells. Thus, ADAM10 critically regulates MZB development by initiating Notch2 signaling. This study identifies ADAM10 as the in vivo CD23 sheddase and an important regulator of B cell development. Moreover, it has important implications for the treatment of numerous CD23- and Notch-mediated pathologies, ranging from allergy to cancer.


1999 ◽  
Vol 11 (1) ◽  
pp. 23-37 ◽  
Author(s):  
Mariëlle C. Haks ◽  
Mariëtte A. Oosterwegel ◽  
Bianca Blom ◽  
Hergen M. Spits ◽  
Ada M Kruisbeek

2008 ◽  
Vol 205 (11) ◽  
pp. 2507-2513 ◽  
Author(s):  
Katsuto Hozumi ◽  
Carolina Mailhos ◽  
Naoko Negishi ◽  
Ken-ichi Hirano ◽  
Takashi Yahata ◽  
...  

The thymic microenvironment is required for T cell development in vivo. However, in vitro studies have shown that when hematopoietic progenitors acquire Notch signaling via Delta-like (Dll)1 or Dll4, they differentiate into the T cell lineage in the absence of a thymic microenvironment. It is not clear, however, whether the thymus supports T cell development specifically by providing Notch signaling. To address this issue, we generated mice with a loxP-flanked allele of Dll4 and induced gene deletion specifically in thymic epithelial cells (TECs). In the thymus of mutant mice, the expression of Dll4 was abrogated on the epithelium, and the proportion of hematopoietic cells bearing the intracellular fragment of Notch1 (ICN1) was markedly decreased. Corresponding to this, CD4 CD8 double-positive or single-positive T cells were not detected in the thymus. Further analysis showed that the double-negative cell fraction was lacking T cell progenitors. The enforced expression of ICN1 in hematopoietic progenitors restored thymic T cell differentiation, even when the TECs were deficient in Dll4. These results indicate that the thymus-specific environment for determining T cell fate indispensably requires Dll4 expression to induce Notch signaling in the thymic immigrant cells.


2009 ◽  
Vol 182 (6) ◽  
pp. 3390-3397 ◽  
Author(s):  
Niklas Beyersdorf ◽  
Attila Braun ◽  
Timo Vögtle ◽  
David Varga-Szabo ◽  
Ronmy Rivera Galdos ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 73-73
Author(s):  
Kazuaki Yokoyama ◽  
Nozomi Yokoyama ◽  
Kiyoko Izawa ◽  
Ai Kotani ◽  
Ratanakanit Harnprasopwat ◽  
...  

Abstract Abstract 73 Interleukin-7 (IL7) is essential for T cell development and homeostasis. Dysregulation of signals that control normal T-cell development has been implicated in the onset of T-cell acute lymphoblastic leukemia (T-ALL). By analogy to activating mutations in the Notch pathways, we hypothesized that any mutations in the IL7 signaling axis might also contribute to T-ALL. Direct sequencing of human IL7 receptor a chain (hIL7RA) gene in a panel of 16 T-ALL cell lines identified two types of mutations in two different cell lines. One was an insertion mutation of 4 amino acids (LSRC) in the transmembrane region (INS, Fig.1A) from DND-41, a gamma-delta TCR+ T-ALL cell line, and the other was a truncated, loss-of-function, mutation in the cytoplasmic region from MOLT-4. We demonstrated that hIL7RA-INS mutant spontaneously formed a homodimer and constitutively activated downstream signals including Stat family members (1, 3 and 5), Akt and Erk via Jak1, but not Jak3. Next, we investigated oncogenic activity of hIL7RA-INS in primary hematopoietic progenitor cells. To this aim, lin− E.14 Balb/c fetal liver (FL) cells were retrovirally transduced with hIL7RA-INS in parallel with hIL7RA-wild type (WT), and then tested for their cytokine dependence in vitro. As expected, only hIL7RA-INS-transduced lin−FL cells showed abrogation of cytokine dependence. hIL7RA-transduced lin−FL cells were also transplanted into lethally irradiated syngeneic mice. Within 7–9 weeks after transplantation of lin−FL cells transduced with hIL7RA-INS, but not with hIL7RA-WT, recipient mice developed well-tolerated myelo- and lymphoproliferative disorders, characterized by marked leukocytosis, systemic lymphadenopathy and splenomegaly (Fig.1B). Notably, concomitant increase in hIL7RA+gamma-delta TCR+ T cells and decrease in B cells were observed in peripheral blood (Fig.1C). Histological examination of bone marrow, spleen and liver specimens from diseased mice revealed moderate to severe myeloid hyperplasia, disrupted splenic architecture by disseminated mature myeloid cells and infiltration of both myeloid and mononuclear cells into hepatic parenchyma, respectively. In addition, recipient mice for hIL7RA-INS-transduced lin−FL cells frequently manifested ruffled fur as well as mononuclear cell infiltration into salivary gland and pericardium, suggesting an autoimmune-like disorder. However, during median follow-up of 11 weeks, these recipient mice did not develop either overt leukemia or lymphoma, indicating that additional transforming events are required for evolution to aggressive hematological malignancies. These in vivo findings highlighted the possibility that aberrant signals via IL7RA in hematopoietic stem/progenitor cells might preferentially stimulate myelopoiesis over lymphopoiesis, and also confirmed the essential role of IL7RA in gamma-delta TCR+ T cell development, previously shown by IL7RA-knockout mice. Taken together, we speculated that dysregulated IL7RA signaling axis might be involved in the onset of T-ALL, especially with gamma-delta TCR+ phenotype. Finally, the present study, together with the recent report (JEM 208:901, 2011), emphasizes the significance of the sequential Notch-IL7RA pathways in the pathogenesis of T-ALL as well as the dominant role of the IL7RA/Jak1 axis in IL7 proliferative signal. Disclosures: No relevant conflicts of interest to declare.


1996 ◽  
Vol 62 (7) ◽  
pp. 994-1001 ◽  
Author(s):  
Jan G.M.C. Damoiseaux ◽  
Leo J.J. Beijleveld ◽  
Henk-Jan Schuurman ◽  
Peter J.C. van Breda Vriesman

PLoS ONE ◽  
2012 ◽  
Vol 7 (12) ◽  
pp. e52949 ◽  
Author(s):  
Afonso Rocha Martins Almeida ◽  
Sílvia Arroz-Madeira ◽  
Diogo Fonseca-Pereira ◽  
Hélder Ribeiro ◽  
Reena Lasrado ◽  
...  

Blood ◽  
2005 ◽  
Vol 106 (1) ◽  
pp. 193-200 ◽  
Author(s):  
Rafik Terra ◽  
Isabelle Louis ◽  
Richard Le Blanc ◽  
Sophie Ouellet ◽  
Juan Carlos Zúñiga-Pflücker ◽  
...  

In the thymus, 2 types of Lin–Sca-1+ (lineage-negative stem cell antigen-1–positive) progenitors can generate T-lineage cells: c-Kithi interleukin-7 receptor α–negative (c-KithiIL-7Rα–) and c-KitloIL-7Rα+. While c-KithiIL-7Rα– progenitors are absent, c-KitloIL-7Rα+ progenitors are abundant in the lymph nodes (LNs). c-KitloIL-7Rα+ progenitors undergo abortive T-cell commitment in the LNs and become arrested in the G1 phase of the cell cycle because they fail both to up-regulate c-myb, c-myc, and cyclin D2 and to repress junB, p16INK4a, and p21Cip1/WAF. As a result, development of LN c-KitloIL-7Rα+ progenitors is blocked at an intermediate CD44+CD25lo development stage in vivo, and LN-derived progenitors fail to generate mature T cells when cultured with OP9-DL1 stromal cells. LN stroma can provide key signals for T-cell development including IL-7, Kit ligand, and Delta-like–1 but lacks Wnt4 and Wnt7b transcripts. LN c-KitloIL-7Rα+ progenitors are able to generate mature T cells when cultured with stromal cells producing wingless-related MMTV integration site 4 (Wnt4) or upon in vivo exposure to oncostatin M whose signaling pathway intersects with Wnt. Thus, supplying Wnt signals to c-KitloIL-7Rα+ progenitors may be sufficient to transform the LN into a primary T-lymphoid organ. These data provide unique insights into the essence of a primary T-lymphoid organ and into how a cryptic extrathymic T-cell development pathway can be amplified.


Sign in / Sign up

Export Citation Format

Share Document