Biological Significance of B Cell Receptor Mediated Regulation of Autophagy in Chronic Lymphocytic Leukemia

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4130-4130
Author(s):  
Lindsay Smith ◽  
Elizabeth Hogg ◽  
Angus Haynes ◽  
Jonathan C Strefford ◽  
Francesco Forconi ◽  
...  

Abstract Chronic lymphocytic leukaemia (CLL) is characterised by an accumulation of B cells which is broadly split into two groups representing a progressive IGHV unmutated (U-CLL) and a more indolent IGHV mutated (M-CLL) disease. Activation of the B cell receptor (BCR) by antigen/autoantigen engagement is crucial for CLL cell survival, disease progression and resistance to therapy, however further research is required to better understand how BCR signalling impacts on CLL biology. Autophagy is known to play a role in tumorigenesis and resistance to therapy in solid tumors, however whether autophagy has a role in CLL biology and how it is regulated has not been fully investigated. Autophagy is important for normal B cell development and is known to be regulated by various drug treatments in vitro in CLL samples. A previous study showed that activation of the BCR on murine splenic B cells with soluble or bead immobilised (BI) anti-IgM induced autophagy and subsequent apoptosis, however, the role of BCR-induced autophagy has not been explored in B cell malignancies and particularly CLL. Firstly, we assessed basal protein expression of key autophagy markers LC3BII, and ATG3 in CLL samples and age-matched normal donor B cells (NDB). CLL cells expressed significantly more LC3BII (p=.014, n=57) and ATG3 (p=.04, n=58) compared with NDB (n=8), with a greater LC3BII protein expression in U-CLL compared to M-CLL (p=.039, n=57), indicating more autophagy occurs in U-CLL. Furthermore basal increases in autophagy markers GABARAPL2 (LC3B family member) (p=.0004, n=34) and ATG4A (p=.04, n=20) at the RNA level were significantly associated with the ability of CLL cells to flux calcium (>10%) in response to anti-IgM. This indicated a possible role of the BCR in the regulation of autophagy in CLL samples and a possible association with progressive disease. Activation of the CLL BCR with BI anti-IgM significantly induced expression of autophagy markers ATG3 (p=.002, n=22), LC3BII (p<.0001, n=23) and p62 (p=.0011, n=9) at the protein level and ATG3 (p=.04, n=8) and GABARAPL2 (p=.03, n=6) at the RNA level in a time dependent manner. Next, CLL samples were treated with BI anti-IgM in the presence or absence of the autophagy inhibitor hydroxychloroquine (HCQ) to confirm the increase in autophagic flux indicated at the RNA level. Addition of HCQ in combination with BI anti-IgM significantly increased accumulation of LC3BII protein expression compared with HCQ or anti-IgM alone (p=.01 and p=.02 respectively, n=5) indicating an increase in autophagic flux. Treatment with ibrutinib or R406 prevented BI anti-IgM dependent increases in LC3BII (p=.02 and p=.009 respectively, n=9) and p62 expression (p=.02 and p=.006 respectively, n=9). Confirming the role of the BCR in the regulation of autophagy in CLL samples and indicating that inhibition of autophagy may contribute to the clinical responses seen with these BCR kinase inhibitors. Finally we showed that BI anti-IgM increased CLL cell viability compared to BI isotype control (p=.002, n=7), whilst the addition of HCQ significantly reversed this effect (p=.001, n=7). More importantly treatment with BI anti-IgM protected CLL cells against fludarabine induced cell death (p=.0004, n=3) which again was reversed by the addition of HCQ (p=.05, n=3). These data emphasise the importance of BCR signalling in the regulation of autophagy and its impact on therapy resistance and suggest a possible role for autophagy inhibitors in the treatment of CLL. Disclosures Strefford: Roche: Research Funding. Steele:Portola Pharmaceuticals: Other: Travel bursary to ASH 2015; Janssen: Other: Travel bursary to EHA 2015.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 375-375 ◽  
Author(s):  
Fatima Talab ◽  
Victoria Thompson ◽  
John C Allen ◽  
Ke Lin ◽  
Joseph R Slupsky

Abstract Abstract 375 B cell receptor (BCR) signaling promotes survival of the malignant clone in chronic lymphocytic leukaemia (CLL) through its ability to stimulate NFkB pathway signaling. In lymphoid cells, antigen receptor stimulation of this pathway is achieved by engaging the Carma-1 – Bcl10 – MALT1 (CBM) complex for eventual activation of I-kB kinases (IKKs). In B cells, protein kinase C beta (PKCbeta) is an important mediator of CBM complex activation. However, in CLL cells we found that PKCs do not appear to have a role in BCR-mediated NFkB pathway signaling, despite high expression levels of PKCbeta, because the presence of specific inhibitors of this kinase (LY379196 and bisindolylmaleimide-I) has no effect on the induction of IKK phosphorylation during BCR crosslinking. Examination of CBM complex expression suggests an explanation for this phenomenon; the expression levels of Carma-1 and MALT-1 are largely similar in CLL and normal B cells, but the expression of Bcl10 is much reduced in CLL cells. These findings, taken together with the established role of Bcl10 in the pathway of BCR-induced NFkB activation, suggest that CLL cells may employ a different mechanism to activate this pathway during BCR stimulation. Tyrosine kinases are known to play a role in BCR-induced IKK activation in CLL cells because compounds like dasatinib and PP2 inhibit NFkB pathway activation by BCR. One possible tyrosine kinase is c-Abl because we have shown this protein to be overexpressed in CLL cells, where it plays a role in activation of the NFkB pathway. To investigate the role of c-Abl in BCR-induced IKK activation, we used the inhibitor imatinib and found that the presence of this compound partially inhibited IKK phosphorylation in BCR-stimulated CLL cells. However, imatinib can also inhibit Lck, a T cell-specific src-family tyrosine kinase that is expressed by CLL cells. To differentiate between Lck- and c-Abl-mediated BCR signals we used the specific inhibitor 4-amino-5-(4-phenoxyphenyl)-7H-pyrrolo[3,2d] pyrimidin-7-yl-cyclopentane (Lck-i). We found that the presence of this compound in CLL cell cultures undergoing BCR stimulation almost completely inhibited the induction of IKK activation. Investigation of Lck-i specificity revealed this compound did not inhibit either c-Abl or Lyn at the concentration used to inhibit Lck in CLL cell cultures. Further investigation of the effects of Lck-i showed that this compound was also effective in inhibiting BCR-induced activation of the Akt and ERK signaling pathways. Taken together, these data suggest a major role for Lck in BCR-mediated signaling in CLL cells, and question the existing paradigm on the importance of Lyn. Disclosures: No relevant conflicts of interest to declare.


2006 ◽  
Vol 26 (24) ◽  
pp. 9364-9376 ◽  
Author(s):  
Renren Wen ◽  
Yuhong Chen ◽  
Li Bai ◽  
Guoping Fu ◽  
James Schuman ◽  
...  

ABSTRACT Phospholipase Cγ2 (PLCγ2) is a critical signaling effector of the B-cell receptor (BCR). Here we show that PLCγ2 deficiency impedes early B-cell development, resulting in an increase of B220+ CD43+ BP-1+ CD24hi pre-BCR+ large pre-B cells. PLCγ2 deficiency impairs pre-BCR-mediated functions, leading to enhanced interleukin-7 (IL-7) signaling and elevated levels of RAGs in the selected large pre-B cells. Consequently, PLCγ2 deficiency renders large pre-B cells susceptible to transformation, resulting in dramatic acceleration of Myc-induced lymphomagenesis. PLCγ2 −/− Eμ-Myc transgenic mice mainly develop lymphomas of B220+ CD43+ BP-1+ CD24hi pre-BCR+ large pre-B-cell origin, which are uncommon in wild-type Eμ-Myc transgenics. Furthermore, lymphomas from PLCγ2 −/− Eμ-Myc transgenic mice exhibited a loss of p27Kip1 and often displayed alterations in Arf or p53. Thus, PLCγ2 plays an important role in pre-BCR-mediated early B-cell development, and its deficiency leads to markedly increased pools of the most at-risk large pre-B cells, which display hyperresponsiveness to IL-7 and express high levels of RAGs, making them prone to secondary mutations and Myc-induced malignancy.


Blood ◽  
2009 ◽  
Vol 113 (19) ◽  
pp. 4566-4574 ◽  
Author(s):  
James Schuman ◽  
Yuhong Chen ◽  
Andrew Podd ◽  
Mei Yu ◽  
Hong-Hsing Liu ◽  
...  

Abstract The kinase TAK1 is essential for T-cell receptor (TCR)–mediated nuclear factor κB (NF-κB) activation and T-cell development. However, the role of TAK1 in B-cell receptor (BCR)–mediated NF-κB activation and B-cell development is not clear. Here we show that B-cell–specific deletion of TAK1 impaired the transition from transitional type 2 to mature follicular (FO) B cells and caused a marked decrease of marginal zone (MZ) B cells. TAK1-deficient B cells exhibited an increase of BCR-induced apoptosis and impaired proliferation in response to BCR ligation. Importantly, TAK1-deficient B cells failed to activate NF-κB after BCR stimulation. Thus, TAK1 is critical for B-cell maturation and BCR-induced NF-κB activation.


1996 ◽  
Vol 184 (6) ◽  
pp. 2079-2084 ◽  
Author(s):  
Shiaoching Gong ◽  
Mercedes Sanchez ◽  
Michel C. Nussenzweig

The pre-B cell receptor is a key checkpoint regulator in developing B cells. Early events that are controlled by the pre-B cell receptor include positive selection for cells express membrane immunoglobulin heavy chains and negative selection against cells expressing truncated immunoglobulins that lack a complete variable region (Dμ). Positive selection is known to be mediated by membrane immunoglobulin heavy chains through Igα-Igβ, whereas the mechanism for counterselection against Dμ has not been determined. We have examined the role of the Igα-Igβ signal transducers in counterselection against Dμ using mice that lack Igβ. We found that Dμ expression is not selected against in developing B cells in Igβ mutant mice. Thus, the molecular mechanism for counterselection against Dμ in pre-B cells resembles positive selection in that it requires interaction between mDμ and Igα-Igβ.


2001 ◽  
Vol 21 (19) ◽  
pp. 6640-6650 ◽  
Author(s):  
Daniel Krappmann ◽  
Alina Patke ◽  
Vigo Heissmeyer ◽  
Claus Scheidereit

ABSTRACT Antigen receptor signaling is known to activate NF-κB in lymphocytes. While T-cell-receptor-induced NF-κB activation critically depends on novel protein kinase C θ (PKCθ), the role of novel PKCs in B-cell stimulation has not been elucidated. In primary murine splenic B cells, we found high expression of the novel PKCs δ and ɛ but only weak expression of the θ isoform. Rottlerin blocks phorbol ester (phorbol myristate acetate [PMA])- or B-cell receptor (BCR)-mediated NF-κB and c-Jun N-terminal kinase (JNK) activation in primary B and T cells to a similar extent, suggesting that novel PKCs are positive regulators of signaling in hematopoietic cells. Mouse 70Z/3 pre-B cells have been widely used as a model for NF-κB activation in B cells. Similar to the situation in splenic B cells, rottlerin inhibits BCR and PMA stimulation of NF-κB in 70Z/3 cells. A derivative of 70Z/3 cells, 1.3E2 cells, are defective in NF-κB activation due to the lack of the IκB kinase (IKKγ) protein. Ectopic expression of IKKγ can rescue NF-κB activation in response to lipopolysaccharides (LPS) and interleukin-1β (IL-1β), but not to PMA. In addition, PMA-induced activation of the mitogen-activated protein kinase JNK is blocked in 1.3E2 cells, suggesting that an upstream component common to both pathways is either missing or mutated. Analysis of various PKC isoforms revealed that exclusively PKCθ was absent in 1.3E2 cells while it was expressed in 70Z/3 cells. Stable expression of either novel PKCθ or -δ but not classical PKCβII in 1.3E2 IKKγ-expressing cells rescues PMA activation of NF-κB and JNK signaling, demonstrating a critical role of novel PKCs for B-cell activation.


2000 ◽  
Vol 14 (1) ◽  
pp. 11-16 ◽  
Author(s):  
Yuji Yamanashi ◽  
Toshiki Tamura ◽  
Toshihide Kanamori ◽  
Hidehiro Yamane ◽  
Hideo Nariuchi ◽  
...  

Antigenic stimulation of the B-cell receptor (BCR) is a central event in the immune response. In contrast, antigen bound to IgG negatively regulates signals from the BCR by cross-linking it to the inhibitory receptor FcγRIIB. Here we show that upon cross-linking of BCR or BCR with FcγRIIB, the rasGAP-associated protein p62dok is prominently tyrosine phosphorylated in a Lyn-dependent manner. Inactivation of the dok gene by homologous recombination has shown that upon BCR cross-linking, p62dok suppresses MAP kinase and is indispensable for FcγRIIB-mediated negative regulation of cell proliferation. We propose that p62dok, a downstream target of many PTKs, plays a negative role in various signaling situations.


2008 ◽  
Vol 320 (1-2) ◽  
pp. 93-100 ◽  
Author(s):  
Masaharu Terashima ◽  
Mai Takahashi ◽  
Makoto Shimoyama ◽  
Yoshinori Tanigawa ◽  
Takeshi Urano ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document