scholarly journals Notch 1–Deficient Common Lymphoid Precursors Adopt a B Cell Fate in the Thymus

2001 ◽  
Vol 194 (7) ◽  
pp. 1003-1012 ◽  
Author(s):  
Anne Wilson ◽  
H. Robson MacDonald ◽  
Freddy Radtke

We have recently reported that Notch 1, a member of the Notch multigene family, is essential for the development of murine T cells. Using a mouse model in which Notch 1 is inactivated in bone marrow (BM) precursors we have shown that B cells instead of T cells are found in the thymus of BM chimeras. However, it is not clear whether these B cells develop by default from a common lymphoid precursor due to the absence of Notch 1 signaling, or whether they arise as a result of perturbed migration of BM-derived B cells and/or altered homeostasis of normal resident thymic B cells. In this report we show that Notch 1–deficient thymic B cells resemble BM B cells in phenotype and turnover kinetics and are located predominantly in the medulla and corticomedullary junction. Peripheral blood lymphocyte analysis shows no evidence of recirculating Notch1−/− BM B cells. Furthermore, lack of T cell development is not due to a failure of Notch1−/− precursors to home to the thymus, as even after intrathymic reconstitution with BM cells, B cells instead of T cells develop from Notch 1–deficient precursors. Taken together, these results provide evidence for de novo ectopic B cell development in the thymus, and support the hypothesis that in the absence of Notch 1 common lymphoid precursors adopt the default cell fate and develop into B cells instead.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3151-3151
Author(s):  
Jalal Taneera ◽  
Emma Smith ◽  
Mikael Sigvardsson ◽  
Emil Hansson ◽  
Urban Lindahl ◽  
...  

Abstract Notch activation has been suggested to promote T cell development at the expense of B cell commitment at the level of a common lymphoid progenitor prior to B cell commitment. Here, we explored the possibility that Notch activation might be able to switch the fate of already committed B cell progenitors towards T cell development upon Notch activation. To address this we overexpressed constitutively activated Notch-3 (N3IC) in B cell progenitors purified from transgenic mice in which human CD25 is expressed under control of the λ5 promoter. Strikingly, whereas untransduced and control transduced B220+λ5+CD3− B cell progenitors gave rise exclusively to B cells, CD4+ and CD8+ T cells but no B cells were derived from N3IC-transduced cells when transplanted into sublethally irradiated NOD-SCID mice. Gene expression profiling demonstrated that untransduced B220+ λ5+CD3− B cell progenitors expressed λ5 and CD19 but not the T cell specific genes GATA-3, lck and pTα, whereas CD3+ T cells derived from N3IC-transduced B220+λ5+CD3−cells failed to express λ5 and CD19, but were positive for GATA-3, lck and pTα expression as well as a and b T cell rearrangement. Furthermore, DJ rearrangements were detected at very low levels in CD3+ cells isolated from normal non-transduced BM, but were more abundant in the N3IC-transduced CD3+ BM cells. Noteworthy, N3IC-transduced B220+λ5+CD3−CD19+ proB cell progenitors failed to generate B as well as T cells, whereas N3IC-transduced B220+λ5+CD3−CD19− pre-proB cells produced exclusively T cells, even when evaluated at low cell numbers. In conclusion Notch activation can switch committed B cell progenitors from a B cell to a T cell fate, but this plasticity is lost at the Pro-B cell stage, upon upregulation of CD19 expression.


Blood ◽  
2016 ◽  
Vol 128 (7) ◽  
pp. e10-e19 ◽  
Author(s):  
Tiago F. Brazão ◽  
Jethro S. Johnson ◽  
Jennifer Müller ◽  
Andreas Heger ◽  
Chris P. Ponting ◽  
...  

AbstractLong noncoding RNAs (lncRNAs) are potentially important regulators of cell differentiation and development, but little is known about their roles in B lymphocytes. Using RNA-seq and de novo transcript assembly, we identified 4516 lncRNAs expressed in 11 stages of B-cell development and activation. Most of these lncRNAs have not been previously detected, even in the closely related T-cell lineage. Comparison with lncRNAs previously described in human B cells identified 185 mouse lncRNAs that have human orthologs. Using chromatin immunoprecipitation-seq, we classified 20% of the lncRNAs as either enhancer-associated (eRNA) or promoter-associated RNAs. We identified 126 eRNAs whose expression closely correlated with the nearest coding gene, thereby indicating the likely location of numerous enhancers active in the B-cell lineage. Furthermore, using this catalog of newly discovered lncRNAs, we show that PAX5, a transcription factor required to specify the B-cell lineage, bound to and regulated the expression of 109 lncRNAs in pro-B and mature B cells and 184 lncRNAs in acute lymphoblastic leukemia.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1883-1883
Author(s):  
Oleg Kolupaev ◽  
Michelle West ◽  
Bruce R. Blazar ◽  
Stephen Tilley ◽  
James Coghill ◽  
...  

Abstract Background. Chronic-graft-versus-host disease (cGvHD) continues to be a major complication following allogeneic hematopoietic stem cell transplantation (HSCT). Despite significant progress, mechanisms underlying development of the pathology are yet to be fully understood. Recent studies utilizing mouse models and patient samples have demonstrated a critical role for B cells in GvHD pathogenesis. Bone marrow (BM)-derived B cells can produce auto-reactive antibodies causing tissue fibrosis and multiorgan cGvHD. Impaired B cell homeostasis in the periphery, activation due to abnormally high levels of B cell-activating factor (BAFF), increased survival of auto-reactive B cells and aberrant BCR signaling are shown to be important for disease progression in cGvHD patients. Murine models also highlighted the critical role of germinal center reactions, particularly interactions between T follicular helper (Tfh) cells and B cells for generation of auto-antibodies which are responsible for triggering immune responses and cell-mediated toxicity. A growing body of evidence has emerged highlighting the fact that BM itself is a target organ during acute GvHD (aGvHD) with recent work suggesting a role for donor CD4+ T cells in BM specific aGvHD. Our group has shown that patients with higher numbers of BM B cell precursors were less likely to develop cGvHD after allogeneic HSCT (Fedoriw et al., 2012). These observations indicate clinical relevance of impaired BM B lymphopoiesis for cGvHD development. Methods. In order to investigate the effect of cGvHD on BM B cell development, we used the well-characterized major mismatch B6 into B10.BR model of systemic cGvHD. Recipient mice were treated with cyclophosphamide on day -3 and -2, irradiated with 700 cGy on day -1, and injected with 107 T cell depleted (TCD) BM with or without total splenic T cells (0.5-1x105). Mice were monitored for 30 days, and BM and spleen was harvested and analyzed using flow cytometry. Results. Consistent with patient data, we observed a decrease in the frequency and number of donor-derived uncommitted common lymphoid progenitors (CLP) and B cell progenitors in the BM+ allogeneic T cells group (CLP: 0.17±0.03% vs. 0.06±0.01%, p <0.01; pro B: 2.2 ± 0.5% vs. 0.7 ± 0.3%, p<0.05; pre B: 15.3±1.8% vs. 6.3±2.4%, p<0.05; immature B cells: 5.7±0.7% vs. 2.1±0.7%, p<0.01) (Fig.1). As previously reported for this model, we also found a decrease in the frequency of follicular (FO) B cells (Flynn et al., 2014). We hypothesized that during cGvHD the B cell progenitor BM niche is affected by donor CD4+ T cells leading to impaired B lymphopoiesis. Bone marrow from BM+T cell animals had a significantly higher frequency of CD4+ cells compared to the control group (0.45±0.06% vs. 0.2±0.02%). Depletion of CD4+ T cells using anti-CD4 antibody during the first two weeks after transplant improved pathology scores and prevented weight loss in BM+T cells mice. We also observedpartial recovery of B cell progenitors and Lin-CD45-CD31-CD51+ osteoblasts (OB) in animals treated with anti-CD4 antibodies (pre B 3.5±1.1% vs. 20.4±4.5%, p<0.05; immature B: 1.9±0.9% vs. 3.5±0.3%; OB: 0.8±0.1% vs.1.2±0.2%). A recent study showed that activation and proliferation of conventional T cells in aGvHD model can be prevented by in vivo expansion of regulatory T cells (Tregs) using αDR3 antibody (4C12). We adopted this approach to determine whether Tregs can suppress the cytotoxic effect of donor CD4+ T cells in BM in cGvHD model. Animals that received T cells from 4C12-treated donors had an increase in survival and lower cGvHD pathology scores. These mice also had higher frequency of pro B, pre B, and immature B cells compared to the mice infused with T cells from isotype-treated donors. Conclusions. These studies demonstrate that BM development of B lymphocytes is impaired in a mouse model of systemic cGvHD. Our data suggests that donor-derived CD4+ T cells are involved in the destruction of hematopoietic niches in BM, particularly OB, which support B lymphopoiesis. Moreover, depletion of CD4+ T cells and infusion with in vivo expanded Tregs reduced the severity of cGvHD. Thus, Treg therapy in patients with cGvHD may be important for BM B cell development, and improvement of clinical outcomes. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 124-124
Author(s):  
Ivan Maillard ◽  
Laleh Talebian ◽  
Zhe Li ◽  
Yalin Guo ◽  
Daisuke Sugiyama ◽  
...  

Abstract The family of core binding factors includes the DNA-binding subunits Runx1-3 and the common non-DNA binding partner CBFβ. Runx1 and CBFβ are essential for the emergence of hematopoietic stem cells during fetal development, but not for stem cell maintenance during later ontogeny. Runx1 is also required for megakaryocyte differentiation, B cell development, and for the DN2 to DN3 transition in thymocyte development. Runx2/CBFβ are critical for normal osteogenesis, and Runx3 for CD4 silencing in CD8+ T cells, but their contribution to other steps of hematopoietic development is unknown. To examine the collective role of core binding factors in hematopoiesis, we generated a hypomorphic Cbfb allele (Cbfbrss). CBFβ protein levels were reduced by approximately 2–3 fold in fetuses homozygous for the Cbfbrss allele (Cbfbrss/rss), and 3–4 fold in fetuses carrying one hypomorphic and one knockout allele (Cbfbrss/−). Cbfbrss/rss and Cbfbrss/− fetuses had normal erythroid and B cell development, and relatively mild abnormalities in megakaryocyte and granulocyte differentiation. In contrast, T cell development was very sensitive to an incremental reduction of CBFβ levels: mature thymocytes were decreased in Cbfbrss/rss fetuses, and virtually absent in Cbfbrss/−fetuses. We next assessed the development of Cbfbrss/rss and Cbfbrss/− fetal liver progenitors after transplantation to irradiated adult recipients, in competition with wild-type (wt) bone marrow cells. Wt, Cbfbrss/rss and Cbfbrss/− fetal progenitors replenished the erythroid, myeloid and B cell compartments equally well. The overall development of Cbfbrss/rss T cells was preserved, although CD4 expression was derepressed in double negative thymocytes. In Cbfbrss/− chimeras, mature thymocytes were entirely derived from competitor cells. Furthermore, the developmental block in Cbfbrss/− progenitors was present at the earliest stages of T cell development within the DN1 (ETP) and DN2 subsets. Our data define a critical CBFβ threshold for normal T cell development, and they situate an essential role of core binding factors during the earliest stages of T cell development. In addition, early thymopoiesis appeared more severely affected by reduced CBFβ dosage than by the lack of Runx1 (Ichikawa et al., Nat Med 2004; Growney et al., Blood 2005), suggesting that Runx2/3 may contribute to core binding factor activity in the T cell lineage.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 282-282
Author(s):  
Shuangmin Zhang ◽  
Yi Zheng ◽  
Richard Lang ◽  
Fukun Guo

Abstract Abstract 282 RhoA GTPase is an intracellular signal transducer capable of regulating a wide range of cell functions including cytoskeleton dynamics, proliferation, and survival. In lymphocytes, studies by using dominant negative mutant or C3 transferase expressing transgenic mice suggest that RhoA is involved in TCR and BCR signaling and related T cell functions such as polarization, migration, survival, and proliferation. To date, the physiological role of RhoA in lymphocyte development remains unclear. In this study, we have achieved T cell, B cell, and hematopoietic stem cell-specific deletion of RhoA by conditional gene targeting with CD2, CD19 and Mx1 promoter-driven Cre expression, respectively, in the RhoAloxP/loxP mice. First, we found that RhoA gene disruption in early T cells caused a drastic decrease in thymocyte cellularity, with the numbers of CD4−CD8− double negative (DN), CD4+CD8+ double positive (DP), CD4+CD8− single positive (SP), and CD4−CD8+ SP T cells decreased by 88.8% ± 6.0%, 99.4% ± 1.0%, 99.3% ± 1.2%, and 98.6% ± 2.0%, respectively. Among DN subpopulations, CD44+CD25− (DN1), CD44+CD25+ (DN2), CD44−CD25+ (DN3), and CD44−CD25− (DN4) cells were reduced by 91.7% ± 6.0%, 54.9% ± 27.7%, 50.9% ± 33.3%, and 96.7% ± 3.4%, respectively. Further, RhoA knockout led to a significant loss of DP thymocytes at the initial stage (CD69highTCRint) of positive selection, suggesting that RhoA is required for positive selection. The decreased thymocyte cellularity in mutant mice is associated with increased apoptosis of all thymic T lineages. RhoA deficiency also resulted in a perturbation in thymocyte cell cycle progression as manifested by increased BrdU incorporation in DN1 and DN2 cells and decreased BrdU incorporation in DN4 and DP cells. Concomitantly, RhoA-deficient thymocytes showed a 59.8% ± 26.3% reduction in proliferative potential in response to TCR crosslinking. Western blot analysis revealed that the activities of ZAP70, LAT, Akt, Erk, and p38 were impaired in RhoA-/- thymocytes. In periphery, spleens of the RhoA null mice contained 7.4% ± 8.0% of CD4+ T cells and 3.7% ± 2.7% of CD8+ T cells compared with that of wild type (WT) mice. Loss of peripheral mature T cells in mutant mice is reflected by a marked reduction of naive T cells, whereas effector and memory phenotype cells were marginally affected by RhoA deficiency. RhoA-deficient naïve T cells were more susceptible to apoptosis, suggesting that homeostatic defect of naïve T cells in RhoA-/- mice is attributed to impaired cell survival. Abrogation of RhoA caused an increased in vivo BrdU incorporation in naïve T cell compartments. Thus, RhoA deficiency induces naïve T cell homeostatic proliferation, possibly due to a compensatory effect of lymphopenia. In contrast to that in thymocytes, Erk was constitutively activated in RhoA-deficient splenic T cells. These observations implicate RhoA in the multiple stages of T cell development and the proper assembly of early TCR signaling complex. Second, deletion of RhoA in pre-proB cells had no effect on early B cell development in bone marrow but significantly inhibited late B cell development in spleen, resulting in 78.2% ± 13.6%, 78.6% ± 16.9%, and 93.2% ± 3.4% reduction in transitional, follicular, and marginal zone B cells, respectively. Plasma cells in spleen were decreased by 50.9 % ± 25.9% in RhoA null mice. However, we did not detect any changes in survival of in vivo RhoA-/- B cells or RhoA-/- B cells cultured in vitro with survival factor BAFF. Distinct from previously characterized Cdc42 knockout mice, BAFF-R expression was not altered in RhoA-/- B cells. Moreover, RhoA-/- B cells appeared to be normal in proliferation and Akt and Erk activation in response to BCR crosslinking. These data suggest that RhoA is important for late B cell development through regulation of differentiation but not cell survival or proliferation. Finally, deletion of RhoA from hematopoietic stem cells did not affect common lymphoid progenitor production, indicating that RhoA is not required for early lymphoid progenitor commitment. Taken together, these lineage-specific mouse genetic studies demonstrate that RhoA critically regulates T and B cell development by distinct cellular mechanisms at multiple stages of lymphopoiesis. Disclosures: No relevant conflicts of interest to declare.


1997 ◽  
Vol 155 (1) ◽  
pp. 165-170 ◽  
Author(s):  
R Kooijman ◽  
SC van Buul-Offers ◽  
LE Scholtens ◽  
RG Reijnen-Gresnigt ◽  
BJ Zegers

Treatment of mice with IGF-I stimulates T and B cell development. We showed that overexpression of IGF-II in transgenic FVB/N mice only stimulated T cell development. In the present study, we further addressed the in vivo effects of IGF-II in the absence of IGF-I to get more insight into the potential abilities of IGF-II to influence T and B cell development. To this end, we studied lymphocyte development in IGF-II transgenic Snell dwarf mice that are prolactin, GH and thyroid-stimulating hormone deficient and as a consequence show low serum IGF-I levels. We showed that T cell development was stimulated to the same extent as in IGF-II transgenic FVB/N mice. Furthermore, IGF-II increased the number of nucleated bone marrow cells and the number of immature B cells without having an effect on the number of mature B cells in spleen and bone marrow. Our data show that IGF-II has preferential effects on T cell development compared with B development, and that these preferential effects also occur in the absence of measurable IGF-I levels.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1465-1465
Author(s):  
Jason Mullenix ◽  
Kimi Y Kong ◽  
Kristin Severns Owens ◽  
Jason Rogers ◽  
Shannon FitzPatrick ◽  
...  

Abstract Abstract 1465 Poster Board I-488 The miR-23a microRNA (miRNAs) cluster inhibits both [ITALIC]in vitro[/ITALIC] and [ITALIC]in vivo[/ITALIC] B cell development. When murine hematopoietic progenitor cells expressing the 23a cluster miRNAs were cultured in B cell promoting conditions we observed over a five-fold decrease in the generation of CD19+ B cells compared to control cultures. Conversely, we observed over a five-fold increase in CD11b+ myeloid cells. When irradiated mice were transplanted with bone marrow expressing the miR-23a cluster we observed a two-fold decrease in bone marrow and splenic B cells, 8 weeks post-transplant compared to control mice. The miR-23a cluster codes for a single pri-transcript, which when processed yields three mature miRNAs: miR-23a, miR-27a, and miR-24-2. All three mature miRNAs are more abundant in myeloid cells compared to other hematopoietic cells. In vitro miR-24 alone is necessary and sufficient to inhibit B cell development. The promoter for the cluster contains conserved binding sites for the essential myeloid transcription factors PU.1 and C/EBP alpha. Chromatin immunoprecipitations demonstrated that PU.1 and C/EBP alpha are associated with the promoter in myeloid cells. In addition, C/EBP alpha is bound to several highly conserved regions upstream of the promoter. Both PU.1 and C/EBP alpha promote myeloid development at the expense of lymphopoiesis. Our work suggests that the miR-23a cluster may be a critical downstream target of PU.1 and C/EBP alpha in the specification of myeloid cell fate. Although miRNAs have been identified downstream of PU.1 and C/EBP alpha in mediating the development of monocytes and granulocytes, the 23a cluster is the first downstream miRNA target implicated in the regulating lymphoid cell fate acquisition. We are currently identifying targets of miR-24 that may mediate the inhibitory effect on B lymphopoiesis. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (12) ◽  
pp. 3917-3925 ◽  
Author(s):  
Stephen M. Jackson ◽  
Natessa Harp ◽  
Darshna Patel ◽  
Jeffrey Zhang ◽  
Savannah Willson ◽  
...  

Abstract To date, there is no consensus regarding the influence of different CD45 isoforms during peripheral B-cell development. Examining correlations between surface CD45RO expression and various physiologic processes ongoing during the germinal center (GC) reaction, we hypothesized that GC B cells, like T cells, that up-regulate surface RO should progressively acquire phenotypes commonly associated with activated, differentiating lymphocytes. GC B cells (IgD−CD38+) were subdivided into 3 surface CD45RO fractions: RO−, RO+/−, and RO+. We show here that the average number of mutations per IgVH transcript increased in direct correlation with surface RO levels. Conjunctional use of RO and CD69 further delineated low/moderately and highly mutated fractions. Activation-induced cytidine deaminase (AID) mRNA was slightly reduced among RO+ GC B cells, suggesting that higher mutation averages are unlikely due to elevated somatic mutation activity. Instead, RO+ GC B cells were negative for Annexin V, comprised mostly (93%) of CD77− centrocytes, and were enriched for CD69+ cells. Collectively, RO+ GC B cells occupy what seems to be a specialized niche comprised mostly of centrocytes that may be in transition between activation states. These findings are among the first to sort GC B cells into populations enriched for live mutated cells solely using a single extracellular marker.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 778-778
Author(s):  
Takahiro Maeda ◽  
Taha Merghoub ◽  
Lin Dong ◽  
Manami Maeda ◽  
Robin Hobbs ◽  
...  

Abstract LRF (Leukaemia/Lymphoma Related Factor, formerly described as Pokemon, FBI-1 and OCZF, encoded by the Zbtb7a gene) is a transcriptional repressor that belongs to the POK (POZ/BTB and Krüppel) protein family. We recently reported that LRF is a proto-oncogene, which is highly expressed in Non-Hodgkin’s Lymphoma tissues (Nature. 433, 278–85). To elucidate its function in fetal and adult lymphopoiesis, we analyzed LRF knockout mice (Zbtb7a−/−) and the conditional knockout mutants (Zbtb7a flox/−Mx-1cre+), respectively. Zbtb7a −/− mice are embryonic lethal due to severe anemia around 16.5 d.p.c. Absolute number of mature B cells was markedly decreased in 15.5 d.p.c Zbtb7a−/− fetal livers (FL), while total number of the earliest immature B cells (Lin-AA4.1+CD19−B220+) was comparable to wild type (WT) littermates. Flow-Sorted Zbtb7a−/− FL Hematopoietic Stem Cells (FL-HSCs) did not give rise to ProB cells in vitro in an OP9 cell culture system. Furthermore, competitive repopulation assay suggested that the defect in B cell development in Zbtb7a−/− FL was cell-autonomous. Conditional inactivation of LRF in adult mice resulted in a significant decrease of B220+ B cells in the peripheral blood (PB). Absolute numbers of both ProB and PreB cells in the BM were drastically reduced in Zbtb7a flox/−Mx-1cre+ mice after pIpC injections, while PreProB cells were rather accumulated. Zbtb7a flox/−Mx-1cre+ PreProB cells did not give rise to ProB cells in vitro in OP9 cell culture. Unexpectedly, Zbtb7a flox/−Mx-1cre+ PreProB cells ectopically expressed T cell genes (e.g. pTCRα, Notch1, Notch3, Hes1, Gata3, TCF1), while they lacked B-cell specific gene expression (e.g. E2A, Ebf1, Pax5, Rag, VpreB1). In agreement with this finding, Zbtb7a flox/−Mx-1cre+ PreProB cells efficiently differentiated into DP-T cells upon 6 days of culture on OP9-DL1 cells, which overexpress Notch1 ligand Delta-like1. We did not observe a gross defect in the T cell compartment in PB and Thymus of Zbtb7a flox/−Mx-1cre+ mice. However, we observed an accumulation of CD4/8 double positive (DP) T cells in their BM. DP-T cells consisted of nearly 30% of the BM mononuclear cells (MNCs) in Zbtb7a flox/−Mx-1cre+ mice. Since the phenotype of LRF conditional knockout mice is reminiscent of that of ICN1 (Intracellular domain of Notch1) overexpression in the mouse BM, we hypothesized that LRF could oppose Notch1 signaling pathway at the early lymphoid progenitor stage. We found that pTCRα, a Notch1 target gene, is highly up-regulated in Zbtb7a flox/−Mx-1cre+ CLPs and that LRF transcriptionally represses mouse pTCRα promoter activity. Our finding strongly indicates that LRF loss at the early lymphoid progenitor stage causes aberrant de-repression of T-cell specific genes, which results in the block of B cell development and generation of T cells in the BM. We therefore propose that LRF is essential in instructing the early lymphoid progenitors into B cell lineage by repressing T cell-instructive signal produced by Notch.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1961-1961
Author(s):  
Nagisa Sakurai ◽  
Manami Maeda ◽  
Sung-UK Lee ◽  
Toshiki Saito ◽  
Shigeru Chiba ◽  
...  

Abstract Abstract 1961 Poster Board I-984 LRF (Leukemia/Lymphoma Related Factor) is a transcriptional repressor originally identified as an interaction partner of the oncoprotein BCL6 (B cell Lymphoma 6). We previously found that LRF acts as a proto-oncogene by repressing tumor suppressor ARF (Alternative Reading Frame, also known as p19 in mice and p14 in humans) and is highly expressed in 60-80% of human Non-Hodgkin Lymphoma (NHL) cases (Maeda et al., Nature 2005). LRF was also found to be indispensable for hematopoietic stem cells (HSCs) to commit to the B cell lineage by opposing Notch function (Maeda et al., Science 2007). Considering that: 1) LRF is normally expressed in Germinal Center B cells (GCB) and overexpressed in NHL tissues and 2) LRF opposes Notch function to maintain normal B cell fate at HSC/progenitor levels, we explored the role of LRF in B cell development and its functional interaction with the Notch pathway in vivo. Upon T cell dependent (TD) immunization, GC formation was severely impaired in secondary lymphoid organs of B cell specific LRF conditional knockout mice (LRFflox/flox mb1-Cre+). While a GC reaction was robustly induced in control mice upon immunization, only few GCB cells were noted in secondary lymphoid organs of LRFflox/flox mb1-Cre+ mice. To assess functional significance of LRF loss in antigen response in vivo, titers of class-switched immunoglobulin (Ig) were measured in the serum; baseline serum titers of IgG1, IgG2b and IgG3 were perturbed, and the primary and secondary antibody response against the TD antigen was impaired in LRFflox/flox mb1-Cre+ mice. Absolute numbers of memory B cells and long-lived BM plasma cells were reduced in LRFflox/flox mb1-Cre+ mice 20 wk after immunization. To determine the cause of defective GC formation, apoptosis and proliferation of GCB cells were examined by FACS. While proportions of apoptotic (AnnexinV positive) GCB cells were similar, regardless of genotypes, LRF deficient GCB cells failed to proliferate upon antigen stimuli. Short-term kinetic analysis demonstrated 5-ethynyl-2'-deoxyuridine (EdU) incorporation was markedly decreased in LRF deficient GCB cells and that the proportion of GCB cells in S phase was reduced in LRFflox/flox mb1-Cre+ mice. In agreement with these findings, quantitative RT-PCR analysis in FACS-sorted GCB cells demonstrated up-regulation of p19Arf and p21, but not p53, mRNA levels in LRF deficient GCB cells. Up-regulation of p19Arf protein levels was also observed in Western Blots. Furthermore, microarray analysis and subsequent Gene Set Enrichment Analysis in FACS-sorted GCB cells showed signatures of defective proliferation, further implicating a critical role for LRF in GCB cell proliferation. Signals mediated by Notch2 are necessary for transitional B cells to commit to the marginal zone B cells (MZB). Inactivation of a component of the Notch pathway in mice resulted in no MZB development and increased follicular B cells (FOB). On the contrary, deletion of the MINT/SHARP gene, a suppressor of Notch signaling, lead to increase of MZB cells and concomitant reduction of FOB cells, indicating that Notch induces MZB cell fate at the transitional B cell stage. While B cell development in the BM was grossly normal, a reduction of FOB cells and a concomitant increase of MZB cells were observed in LRFflox/flox mb1-Cre+ mice. Since the phenotype was reminiscent of that seen in MINT/SHARP knockout mice and opposite to that observed in Notch2 knockout mice, we hypothesized that LRF antagonizes Notch2 mediated signal during the FOB vs. MZB fate determination process. To test this, LRF/Notch2 double knockout mice (LRFflox/flox Notch2flox/flox mb1-Cre+) were established and their mature B cell compartments analyzed. As expected, loss of the Notch2 gene led to an increase of FOB cells and decrease of MZB in LRFflox/flox mb1-Cre+ mice, suggesting that LRF regulates FOB vs. MZB fate in a Notch2 dependent manner. However, Notch2 deficiency did not restore GC formation in LRFflox/flox mb1-Cre+ mice. In summary, our genetic studies strongly indicate that the proto-oncogene LRF is required for normal mature B cell development and function via distinct mechanisms. We propose that LRF is necessary for mature B cell fate by blocking Notch2-mediated signals and plays a critical role in GCB cell proliferation via suppressing p19Arf mediated cell cycle arrests. Our findings provide a further rational for targeting LRF for the treatment of B cell malignancies as well as autoimmune diseases. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document