Role of the LFA-1 Molecule in B Cell Differentiation

Author(s):  
Gail Abendroth Bishop ◽  
Geoffrey Haughton
PLoS ONE ◽  
2018 ◽  
Vol 13 (12) ◽  
pp. e0208343 ◽  
Author(s):  
Lucy Cooper ◽  
Lauren Hailes ◽  
Amania Sheikh ◽  
Colby Zaph ◽  
Gabrielle T. Belz ◽  
...  

2015 ◽  
Vol 2015 ◽  
pp. 1-20 ◽  
Author(s):  
Pier Paolo Piccaluga ◽  
Claudio Agostinelli ◽  
Fabio Fuligni ◽  
Simona Righi ◽  
Claudio Tripodo ◽  
...  

The interferon-inducible DNA sensor IFI16 is involved in the modulation of cellular survival, proliferation, and differentiation. In the hematopoietic system, IFI16 is consistently expressed in the CD34+ stem cells and in peripheral blood lymphocytes; however, little is known regarding its regulation during maturation of B- and T-cells. We explored the role of IFI16 in normal B-cell subsets by analysing its expression and relationship with the major transcription factors involved in germinal center (GC) development and plasma-cell (PC) maturation.IFI16mRNA was differentially expressed in B-cell subsets with significant decrease inIFI16mRNA in GC and PCs with respect to naïve and memory subsets.IFI16mRNA expression is inversely correlated with a few master regulators of B-cell differentiation such asBCL6, XBP1, POU2AF1, andBLIMP1. In contrast,IFI16expression positively correlated withSTAT3, REL, SPIB, RELA, RELB, IRF4, STAT5B, andSTAT5A. ARACNE algorithm indicated a direct regulation ofIFI16byBCL6,STAT5B, andRELB, whereas the relationship betweenIFI16and the other factors is modulated by intermediate factors. In addition, analysis of the CD40 signaling pathway showed thatIFI16gene expression directly correlated with NF-κB activation, indicating that IFI16 could be considered an upstream modulator of NF-κB in human B-cells.


Author(s):  
Mingzeng Zhang ◽  
Shigeru Iwata ◽  
Maiko Hajime ◽  
Naoaki Ohkubo ◽  
Yasuyuki Todoroki ◽  
...  

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 844-844
Author(s):  
Yiguo Hu ◽  
Linghong Kong ◽  
Kevin Staples ◽  
Kevin Mills ◽  
John G. Monroe ◽  
...  

Abstract The BCR-ABL oncogene induces human Philadelphia-positive (Ph+) B-cell acute lymphoblastic leukemia (B-ALL) and chronic myeloid leukemia (CML) that advances to acute phase of CML called blast crisis. In this acute phase, CML patients can develop either B-ALL or acute myeloid leukemia. In B-ALL, differentiation of leukemic cells are blocked at pro-/pre-B stage, and the underlying mechanism is unknown. We hypothesize that this blockade of B-cell differentiation may be important for the development of B-ALL induced by BCR-ABL, and if so, promotion of B-leukemic cell differentiation would create a novel therapeutic strategy for B-ALL. To test this hypothesis, we first compared the percentages of IgM+ B-leukemic cells in BALB/c and C57BL/6 (B6) mice with BCR-ABL-induced B-ALL, because we have previously found that B-ALL develops more quickly in BALB/c mice than in B6 mice (Li et al, J. Exp. Med.189:1399–1412, 1999). We expressed BCR-ABL in bone marrow (BM) using retroviral transduction and transplantation in these two different strains of inbred mice to induce B-ALL. There were significantly more peripheral blood B220+ B cells in BALB/c B-ALL mice than those in B6 mice, correlating to faster B-ALL in BALB/c mice than in B6 mice. Among these B220+ cells, IgM+ cells were much less in BALB/c mice than in B6 mice. We also compared rearrangement of the B cell antigen receptor (BCR) heavy chains (m chains) between BALB/c and B6 backgrounds using BCR-ABL-expressing pro-B cell lines isolated from the B-ALL mice. Normal m chains rearrangement was found in B6 leukemic cells, but not in BALB/c leukemic cells. These results indicate that more differentiated B-leukemic cells are associated with less aggressive disease. To further demonstrate the role of blockade of B-cell differentiation in B-ALL development, we induced B-leukemic cell differentiation by co-expression of BCR-ABL and intact immunoregulatory tyrosine activation motifs (ITAM) contained in immunoglobulin (Ig)_/Igß complexes in BM cells of B-ALL mice, comparing to expression of BCR-ABL alone. We treated these mice with imatinib (orally, 100 mg/kg, twice a day). The treated mice with B-ALL induced by co-expression of BCR-ABL and ITAM lived three-week longer than those with B-ALL induced by BCR-ABL only, with some mice in long-term remission. Prolonged survival was associated with 50% increased B220+/IgM+ B-leukemic cells in peripheral blood of the mice. Taken together, our results demonstrate that blockade of B-cell differentiation is critical for the development of B-ALL induced by BCR-ABL, and provide a rationale for combination therapy of B-ALL with imatinib and induction of leukemic cell differentiation.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1416-1416
Author(s):  
Grazia Fazio ◽  
Chiara Palmi ◽  
Greta Giordano Attianese ◽  
Andrea Biondi ◽  
Antonius Rolink ◽  
...  

Abstract The PAX5/TEL chimeric gene was cloned from the translocation t(9;12)(q11;p13) in an ALL patient. Recent data indicate that the PAX5/TEL fusion defines the cytogenetic entity dic(9;12)(p13;p13), which accounts for about 1% of childhood ALL, almost exclusively B-progenitor ALL. PAX5/TEL is likely to be an aberrant transcription factor, resulting from joining the 5′ region of PAX5 (a transcription factor essential for B cell development) to the 3′ region of TEL/ETV6, containing the Ets-family DNA binding domain. We have cloned the FLAG-full length chimeric PAX5/TEL cDNA in the retroviral vector pMSCV-IRES-GFP (MigR1) to transduce target cells. We have demonstrated a specific nuclear localization of the chimeric protein in NIH3T3 by immunofluorescence analysis. Moreover, we observed a PAX5/TEL dependent decrease of the cellular growth rate in IL-3 dependent murine proB Ba/F3 cells. We further investigated the function of the PAX5/TEL chimeric protein as a potential oncoprotein in murine preBI cells, as a more physiological model. Murine PAX5 −/− preBI cells and wild type preBI cells were purified as B220+/c-KIT+ cells from mouse fetal liver and they were cultured on OP9 and DL1-OP9 stroma cells in presence of IL-7. The OP9 stroma supports B cell proliferation and survival; the DL1-OP9 stroma expresses Delta-like1, one of the Notch ligands, and it’s important to support T cell development. Both PAX5 −/− preBI cells and wild type preBI cells were transduced with the retroviral construct pMSCV-PAX5/TEL-IRES-GFP to analyze cell proliferation, differentiation and growth-dependence on IL-7. Wild type preBI cells expressing PAX5/TEL showed down modulation of CD19 when cultured on OP9 stroma in presence of IL-7; an inverse correlation was observed between the levels of expression of GFP and of CD19. The down modulation of CD19 can be involved in driving the preBI cell into differentiation block. A possible explanation of CD19 repression can rely on a potential competition between PAX5/TEL and endogenous PAX5 to bind PAX5 consensus region on DNA. On OP9 stroma, PAX5/TEL preBI cells are resistant to TGFbeta anti-proliferative and apoptotic effects, with a three-fold increased growth rate than control cells. Although the specific mechanism of PAX5/TEL disruption of TGFbeta signalling pathway remains to be investigated, we propose the TGFbeta resistance by PAX5/TEL as a way to evade the immunosurveillance. PAX5/TEL-preBI cells cultured on DL1-OP9 showed a different phenotype, with up-regulation of c-KIT and down-regulation of CD44. PAX5−/− preBI cells infected with PAX5TEL and grown on OP9 were CD19 negative even in the presence of PAX5TEL. On DL1-OP9 stroma, PAX5TEL cells were able to differentiate maintaining the developmental plasticity of PAX5 −/− preBI cells. These preliminary results indicate a role of PAX5/TEL as a transcription factor, potentially with a suppressor function, down regulating CD19 expression, thus suggesting a function on B cell differentiation. The chimera is able to interfere with TGFbeta pathway, inducing resistance and conferring an advantage in cell survival, evading the immunosurveillance. PAX5TEL do not replace PAX5 functions in PAX5−/− cells, it cannot activate PAX5 target genes as CD19, important for restoring B cell differentiation. Further analyeis are needed to better evaluate the role of PAX5/TEL protein, both in vivo and in vitro models.


1984 ◽  
Vol 14 (11) ◽  
pp. 1021-1027 ◽  
Author(s):  
Malcolm K. Brenner ◽  
Margaret E. North ◽  
Hakikat R. Chadda ◽  
Christine A. Newton ◽  
Mirek Malkovsky ◽  
...  

2009 ◽  
Vol 46 (8-9) ◽  
pp. 1736-1743 ◽  
Author(s):  
Cécile Tonnelle ◽  
Marilyne Dijon ◽  
Thomas Moreau ◽  
Céline Garulli ◽  
Florence Bardin ◽  
...  

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1201-1201
Author(s):  
Ramesh C Nayak ◽  
Mark Jordan Althoff ◽  
Ashley Wellendorf ◽  
Fatima Mohmoud ◽  
Maria Diaz-Meco ◽  
...  

Abstract The molecular mechanisms that control leukemic transformation in B-cell acute lymphoblastic leukemia remain unclear. Leukemic stem cells/progenitors (LSC/P) hijack common pathways utilized by hematopoietic stem cells/progenitors (HSC/P) while they also activate other unique pathways that result in increased self-renewal and differentiation arrest susceptible of therapeutic intervention. A major regulator of HSC self-renewal vs differentiation is the distribution of polarized cell fate determinants. Atypical protein kinase C (aPKC)-ζ and aPKCi in humans or its homologue aPKCλ in mice are the catalytic components of the apical polarity complex PAR3-PAR6. aPKCs have also been argued to function as tumor suppressors. We have previously shown that the genetic deficiency of aPKC activity is dispensable for normal HSC/P activity (Sengupta A et al., PNAS 2011). We had found that primary human and murine BCR-ABL+ LSC/P have ~2-3 fold increased activation of aPKCλ/i. To investigate the possible role of aPKCs in LSC/P activity, we utilized a lymphoid blast-crisis chronic myelogenous leukemia model and evaluated the role aPKCζ and aPKCλ inducible deficiency in leukemic progression. After leukemia induction resulting from doxcycycline withdrawal in Scl-tTA/TRE-BCR-ABL mice that were also transgenic for Mx1Cre, aPKCζ-/- and/or aPKCλflox/flox, leukemic mice (dox-off) were treated with polyI:C to induce deletion of aPKCλ. aPKCλΔ/Δ LSC (Lin-cKit+Sca1+CD135-CD34-) and progenitors (CFU-C) of transplanted recipient mice were 40 and 87% reduced, respectively, suggesting a critical role of aPKCλ in the maintenance of LSC/P. Upon serial transplantation of bone marrow (BM) from CML animals, all WT and aPKCζ-/- recipient mice died by 6-10 weeks of B-ALL. However, mice transplanted with aPKCλΔ/Δ or aPKCζ-/-;aPKCλΔ/Δ BM cells did not die after serial transplantation followed for over one year, despite maintaining leukemic B-cells in circulation and hematopoietic tissues with catalytically active BCR-ABL expression. Leukemic ProB/PreB cells deficient in aPKCλ activity showed ~67% reduced proliferation and 2-fold higher apoptosis than WT and aPKCζ deficient ProB/PreB cells while non-leukemic aPKCλ deficient B-cell lymphopoiesis was grossly normal. Similarly, shRNA-mediated knock-down of aPKCi, in CML patient derived LSC/P led to reduced proliferation, increased apoptosis and enhanced differentiation indicating that aPKCi is also required for the survival and maintenance of human LSC/P. Strikingly, the B-cell differentiation program was restored in aPKCλΔ/Δ mice with expansion of IgM expressing B cells. Downstream, the activation of CrkL, MEK/ERK and Myc signaling pathways were attenuated in absence of aPKCλ in leukemic B-progenitor cells. Unlike non-leukemic B-cell progenitors, the transcriptome of aPKCλ-deficient leukemic B-progenitors showed up-regulation of the B-lymphoid differentiation gene network (Pax5, Ebf1, Ikzf1, Ikzf2, Ikzf3, Rag1 & Rag2). Interestingly, Myc protein expression was reduced and the cell fate determinant Numb mislocalized in aPKCλΔ/Δ leukemic B-cell progenitors, with predominant nuclear distribution. The mRNA/protein expression of Satb2, a chromatin modifier controlling Myc and Numb expression as well as B-cell differentiation, was significantly reduced in aPKCλ-deficient leukemic B-cell progenitors while the expression of its antagonist Satb1 was unchanged. Chromatin immunoprecipitation analysis of leukemic B-cell progenitors showed ~16-200 fold increased enrichment of Satb2 binding to promoter sequences of Cdkn2a and B-cell differentiation genes Pax5 and Ebf1 over non-leukemic B-cell progenitors. Finally, forced expression of Satb2 in aPKCλΔ/Δ deficient LSC/P rescued leukemic proliferation and B-cell differentiation arrest in vivo indicating that Satb2, downstream of aPKCλ, is required for BCR-ABL+ LSC/P activity. Taken together, these data indicate that aPKCλacts as an oncogene in leukemic B-cell progenitors. Loss of aPKCλ reduces LSC/proB proliferation and survival, and restores B-cell differentiation through combined reduction of MAPK activation, Numb mislocalization and differential expression/activity of the chromatin modifier Satb2. This data defines the role of aPKCλ in LSC/P activity and identifies aPKCλ as a new target for pharmacological intervention dispensable for normal lympho-hematopoiesis. Disclosures Cancelas: US Army Medical Research and Material Command (Award W81XWH-15-C-0047) / Department of Defense: Research Funding; Terumo BCT: Research Funding; Cellphire, Inc.: Membership on an entity's Board of Directors or advisory committees; Cerus Corporation: Research Funding; Citra Labs, Inc.: Research Funding; Leukemia & Lymphoma Society of North America: Research Funding; William & Lawrence Hughes Foundation: Research Funding; Haemonetics, Inc.: Research Funding; National Institutes of Health: Research Funding; New Health Sciences, Inc.: Membership on an entity's Board of Directors or advisory committees.


Sign in / Sign up

Export Citation Format

Share Document