Role of Hematopoietic-Specific Protein 1 (HS1) in Apoptosis in B-Chronic Lymphocytic Leukemia.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2809-2809
Author(s):  
Livio Trentin ◽  
Antonella Contri ◽  
Anna Maria Brunati ◽  
Federica Frezzato ◽  
Martina Frasson ◽  
...  

Abstract B-cell chronic lymphocytic leukemia (B-CLL) is the most common leukemia in adults and is characterized by the accumulation of clonal CD5+ B lymphocytes. Several protein kinase pathways have been claimed to be involved in the regulation of apoptosis and cell survival. We previously demonstrated that Src kinase Lyn is overexpressed at the protein level in leukemic cells as compared to normal B lymphocytes with substantial amount of the kinase anomalously present in the cytosol. Moreover, most of Lyn is constitutively active in resting leukemic cells and is poorly responsive to BCR engagement. The finding that B CLL cells contained cytosolic Lyn fraction and are defective in programmed cell death suggest that the tyrosine phosphorlation of specific cytosolic targets might account, at least in part, for cell resistance to apoptosis. The 75 KDa HS1 protein is one of the major substrate of Lyn kinase upon BCR cross-linking that plays a crucial role in BCR- induced apoptosis in the mouse B lymphoma cell line WEHI-231. A recent study demonstrates that most HS1 protein was constitutively phosphorylated in B CLL patients with poor prognosis whereas only a fraction was phosphorylated in patients with good prognoses. In the present study, the relative HS1 protein levels were measured by Western blot analysis in 50 CLL patients belonging to different clinical stages. The relative HS1 protein levels were compared with corresponding levels in normal peripheral blood and with Jurkat cells. For normal B cells, the mean ± SD for HS1: actin ratio was 0,88 ± 0,10. There was considerable variation in the levels of HS1/actin ratio in CLL cells, which ranged from 0,49 to 2,50. Thus, compared to normal B cells, 15 CLL patients had a HS1 level which fell within the mean ± 1SD HS1 levels for normal B cells, while 9 patients had lower levels and 26 patients had higher levels. When assessed by flow cytometry, HS1 expression was normally distributed among CLL cells in individual patients and the mean levels correlated with those obtained by Western blotting. A difference in the levels of HS1 was also observed between mutated and unmutated patients. Using confocal microscopy and subcellular cell fractionation, we observed that HS1 protein was abnormally distributed in malignant cells as compare with normal B cells: a 4–7% aliquot of HS1 was anomalously present in the nucleus of leukemic cells. When primary CLL cells were in vitro treated whith dexamethazone, cyclosporin A, chlorambucil, or fludarabine the HS1 levels decreased correlating with the sensitivity of these cells to undergo apoptosis. Using a polyclonal antiserum against HS1 a major cleavage product of the apparent molecular weight of 64 KDa and one minor product of approximately 46 Kda was detected in B CLL cells cultured for 24 hours whith drugs. These findings suggest that HS1 plays a pivotal role in the regulation of cell survival of leukemic B cells and suggest that HS1 might represent a target for the development of new drugs to be used in vivo in these patients.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2927-2927 ◽  
Author(s):  
Jerome Paggetti ◽  
Franziska Haderk ◽  
Martina Seiffert ◽  
Bassam Janji ◽  
Yeoun Jin Kim ◽  
...  

Abstract Chronic lymphocytic leukemia (CLL), the most common hematologic malignancy in Western countries, is mostly affecting the elderly over 65 year-old. CLL is characterized by the accumulation of mature but non-functional B lymphocytes of clonal origin in the blood and the primary lymphoid organs. CLL was previously considered as a relatively static disease resulting from the accumulation of apoptosis-resistant but quiescent B lymphocytes. However, recent studies using heavy water labeling indicated that CLL is in fact a very dynamic disease with alternation of proliferation phases and peripheral circulation. A focus on the trafficking of CLL cells in vivo has shown that leukemic cells circulate between the blood and the lymphoid organs but have a preference for the bone marrow. Recent next-generation sequencing of CLL cells indicated the presence of different genetic subclones. This intraclonal heterogeneity observed in CLL subpopulations may be in part determined by the interactions that leukemic cells entertain with their microenvironment when B cells migrate into the lymph nodes and the bone marrow. Indeed, tumor-stroma interactions are not only providing signals necessary for leukemic cells survival but may also influence the clonal architecture and evolution. One of these interactions involves CLL-derived exosomes. Here, we show that CLL-exosomes efficiently transfer nucleic acids, including functional microRNAs, and proteins, including MHC-Class II molecules and B-cell specific proteins, to bone marrow mesenchymal stem cells and endothelial cells. CLL-exosomes also activate signaling pathways, including PI3K and NF-κB pathways, in these stromal cells. As a consequence, gene expression is strongly modified indicating a switch towards a cancer-associated fibroblast phenotype. Functionally, exosome-stimulated stromal cells show a striking actin cytoskeleton remodeling characterized by the formation of stress fibers, and enhanced proliferation, motility and angiogenic properties. We also identified several proteins synthesized and secreted by stromal cells that promote leukemic cell adhesion and survival ex vivo. To confirm the involvement of CLL-exosomes in CLL pathology in vivo, MEC-1-eGFP cells were subcutaneously injected into immunocompromised NSG mice together with CLL-exosomes. We observed a significant increase in tumor size and a reduction in survival of exosome-treated animals. Flow cytometry analysis of selected organs indicated an enrichment in leukemic cells in the kidney, providing a potential explanation to the renal failures observed in CLL patients. In conclusion, the communication between CLL cells and stromal cells may be a critical factor influencing CLL progression by promoting leukemic cell survival. This study demonstrates the crucial role of exosomes as mediators of the communication between leukemic cells and their microenvironment. Exosomes could thus represent a suitable target for therapeutic intervention in CLL. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1983 ◽  
Vol 62 (4) ◽  
pp. 767-774 ◽  
Author(s):  
LA Fernandez ◽  
JM MacSween ◽  
GR Langley

Abstract The mechanism of the hypogammaglobulinemia in patients with chronic lymphocytic leukemia (CLL) was studied by determining the generation of specific immunoglobulin-secreting cells in response to mitogen and antigen stimulation in culture. Normal peripheral blood B lymphocytes from 18 normal subjects cocultured with equal numbers of autologous T cells generated cells secreting 2,542 +/- 695 IgG, 2,153 +/- 615 IgA, and 2,918 +/- 945 IgM. Normal B lymphocytes cocultured with normal allogeneic T cells generated similar numbers. However, B lymphocytes from patients with chronic lymphocytic leukemia cocultured with T cells from the same patient generated only 0.5% as many cells secreting IgG and 11% and 23% as many secreting IgA and IgM, respectively. The reason for this markedly defective generation of immunoglobulin-secreting cells was investigated by evaluating T-helper, T-suppressor, and B-cell function using B cells from tonsil and T and B cells from peripheral blood of normal and leukemic individuals. T cells from patients with chronic lymphocytic leukemia provided somewhat greater help than did normal T cells to normal peripheral blood B cells and normal help to tonsil B cells, whether stimulated with mitogen or antigen. T cells from patients with chronic lymphocytic leukemia did not demonstrate increased suppressor function compared to normals with B cells from normal peripheral blood. The hypogammaglobulinemia in these patients therefore was associated with a markedly defective generation of immunoglobulin secreting cells, and as there was normal or increased T- cell helper activity without excessive suppressor activity, it seems likely that this was due to an intrinsic B-cell defect.


Author(s):  
Wafaa Ahmed El- Neanaey ◽  
Rania Shafik Swelem ◽  
Omar Mohamed Ghallab ◽  
Sara Mohamed Abu-Shelou

Background: The present work aimed to investigate the expression of CD160/ CD200 in CLL and other mature B-cell neoplasms (MBN) and their use as an additional diagnostic tool for differentiating CLL from other MBN. Materials and Methods: Using flow cytometry, we detected the expression of CD160 &CD200 on B-cells from 30 CLL patients, 30 other MBN patients in addition to 20 controls. CDs160/200 measurements were determined as a percentage expression (≥20% was considered positive) and as a ratio of the mean fluorescence intensities (MFIR) of leukemic cells/controls and were considered positive when the ratios were ≥2 and 20, respectively. Results: 90% and 100% of the CLL group expressed CDs160/200 in comparison to 60% and 63.3% of other MBN (p=0.007, p<0.001), respectively. By MFIR, 96.7% and 50% of our CLL group expressed CDs160/200 in comparison to 76.7% and 30% of other MBN, respectively. CDs160/ 200 were not expressed on the controls. Positive co-expression of CD160 and CD200 was found in 90% of the CLL cases, 60% of HCL patients and only in 40% of B-NHL. However, double negative expression of both markers was found only in 24% of the B-NHL patients. Conclusion: CD160 with CD200 can be used as additional diagnostic markers to the available routine panel to differentiate between B-CLL and other non-specified B-NHL patients.


Cancers ◽  
2020 ◽  
Vol 12 (8) ◽  
pp. 2124 ◽  
Author(s):  
Brigitte Bauvois ◽  
Elodie Pramil ◽  
Ludovic Jondreville ◽  
Elise Chapiro ◽  
Claire Quiney ◽  
...  

The resistance to apoptosis of chronic lymphocytic leukemia (CLL) cells partly results from the deregulated production of survival signals from leukemic cells. Despite the development of new therapies in CLL, drug resistance and disease relapse still occur. Recently, neutrophil gelatinase-associated lipocalin (NGAL), a secreted glycoprotein, has been suggested to have a critical role in the biology of tumors. Thus, we investigated the relevance of NGAL in CLL pathogenesis, analyzed the expression of its cellular receptor (NGAL-R) on malignant B cells and tested whether CLL cells are resistant to apoptosis through an autocrine process involving NGAL and NGAL-R. We observed that NGAL concentrations were elevated in the serum of CLL patients at diagnosis. After treatment (and regardless of the therapeutic regimen), serum NGAL levels normalized in CLL patients in remission but not in relapsed patients. In parallel, NGAL and NGAL-R were upregulated in leukemic cells from untreated CLL patients when compared to normal peripheral blood mononuclear cells (PBMCs), and returned to basal levels in PBMCs from patients in remission. Cultured CLL cells released endogenous NGAL. Anti-NGAL-R antibodies enhanced NGAL-R+ leukemia cell death. Conversely, recombinant NGAL protected NGAL-R+ CLL cells against apoptosis by activating a STAT3/Mcl-1 signaling pathway. Our results suggest that NGAL and NGAL-R, overexpressed in untreated CLL, participate in the deregulation of the apoptotic machinery in CLL cells, and may be potential therapeutic clues for CLL treatment.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4368-4368
Author(s):  
Federica Frezzato ◽  
Flavia Raggi ◽  
Filippo Severin ◽  
Veronica Martini ◽  
Valentina Trimarco ◽  
...  

Abstract INTRODUCTION We recently found that the Heat Shock Protein of 70kDa (HSP70), an ATP-dependent chaperone that is induced by cellular stress and protects cells against various apoptotic stimuli, was particularly overexpressed in neoplastic B cells from Chronic Lymphocytic Leukemia (CLL) vs normal B lymphocytes. HSP70 responds to a wide variety of physiological and environmental stress signals, thus allowing cells to survive to lethal conditions. The primary responsible for the transcription of HSP70 is the heat shock factor 1 (HSF1), being the major regulator of HSP70 expression. In response to stress, HSF1 becomes phosphorylated, forms homotrimers, binds DNA and activates heat shock gene transcription. Considering that the search for molecules involved in the apoptosis resistance and increased survival of B cells from CLL is still ongoing, with this as a background, we were aimed at studying and targeting HSP70 or players related to it (i.e. HSF1) in view of their clinical, prognostic and therapeutical relevance in CLL. METHODS HSP70/HSF1 axis was analysed in freshly isolated leukemic B cells from CLL patients. Expression levels of HSP70, HSF1 and HSF1-Ser326 were assessed by Western blotting analysis with specific antibodies and the obtained expression data have been correlated with clinical features of the patients. HSP70 subcellular localization has been determined by confocal microscopy and cell fractionation. HSP70 expression and localization was also assessed by immunohistochemistry in lymph nodes from CLL patients. Leukemic B cells from 15 CLL therapy-free patients were treated with different concentrations of: i) Zafirlukast, an oral leukotriene receptor antagonist used to prevent asthma symptoms and acting also as HSP70 inhibitor and ii) Fisetin, a dietary flavonoid acting as anti-inflammatory and anti-carcinogen, that inhibits HSF1 activity through the block of its binding to the HSP70 promoter. Apoptosis induction in CLL cells was evaluated by Annexin V/Propidium Iodide flow cytometry test and by the presence of cleaved PARP observed in Western blotting. RESULTS We found that HSP70 and HSF1 proteins were overexpressed in leukemic vs normal B cells and correlated to poor prognosis. In particular, IGHV unmutated or ZAP70 positive patients presented higher levels of HSP70 and HSF1 with respect to patients with a favorable prognosis. Moreover, the two proteins presented a positive correlation (p<0.0001, r=0.84; Pearson's correlation) thus hypothesizing a positive loop feedback for their expression. We found that, in CLL, HSF1 was constitutively phosphorylated at activatory Ser326, thus being positively regulated, in a large part of our patients. In addition, patients presenting a higher phosphorylation of HSF1 at Ser326 were mostly ZAP70 positive patients. We also observed an abnormal constitutive nuclear localization of HSP70 in leukemic cells. On the basis of these results and the pro-survival role played by HSP70 and HSF1, we analyzed the effects of their inhibition in leukemic cells of our patients by using two inhibitors of this axis, Zafirlukast and Fisetin. Both inhibitors have been proven to be effective in inducing a dose-dependent cell apoptosis in CLL B cells. CONCLUSIONS HSP70 overexpression is involved in a diminished response to treatment by promoting the adaptation of tumor cells to changes (i.e. toxic conditions) currently induced by chemotherapy thus revealing critical roles for HSP70 in cancer initiation and progression. It has been shown that HSP70 depletion results in an increased sensitivity to chemotherapy. For this reason, and considering its prognostic implications and functional role in cancers, including CLL, HSP70 represents an interesting target for antileukemic therapies. In this context, our results suggest: i) an involvement of HSP70/HSF1 axis in the pathogenesis of CLL; ii) an input for further studies that consider the possible involvement of ZAP70 in HSP70/HSF1 axis in CLL; iii) the putative usage of Zafirlukast, which is a drug already available for clinical use and in the targeting of HSF1 in CLL. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1983 ◽  
Vol 62 (4) ◽  
pp. 767-774 ◽  
Author(s):  
LA Fernandez ◽  
JM MacSween ◽  
GR Langley

The mechanism of the hypogammaglobulinemia in patients with chronic lymphocytic leukemia (CLL) was studied by determining the generation of specific immunoglobulin-secreting cells in response to mitogen and antigen stimulation in culture. Normal peripheral blood B lymphocytes from 18 normal subjects cocultured with equal numbers of autologous T cells generated cells secreting 2,542 +/- 695 IgG, 2,153 +/- 615 IgA, and 2,918 +/- 945 IgM. Normal B lymphocytes cocultured with normal allogeneic T cells generated similar numbers. However, B lymphocytes from patients with chronic lymphocytic leukemia cocultured with T cells from the same patient generated only 0.5% as many cells secreting IgG and 11% and 23% as many secreting IgA and IgM, respectively. The reason for this markedly defective generation of immunoglobulin-secreting cells was investigated by evaluating T-helper, T-suppressor, and B-cell function using B cells from tonsil and T and B cells from peripheral blood of normal and leukemic individuals. T cells from patients with chronic lymphocytic leukemia provided somewhat greater help than did normal T cells to normal peripheral blood B cells and normal help to tonsil B cells, whether stimulated with mitogen or antigen. T cells from patients with chronic lymphocytic leukemia did not demonstrate increased suppressor function compared to normals with B cells from normal peripheral blood. The hypogammaglobulinemia in these patients therefore was associated with a markedly defective generation of immunoglobulin secreting cells, and as there was normal or increased T- cell helper activity without excessive suppressor activity, it seems likely that this was due to an intrinsic B-cell defect.


Blood ◽  
2003 ◽  
Vol 101 (5) ◽  
pp. 1962-1969 ◽  
Author(s):  
Luisa Granziero ◽  
Paola Circosta ◽  
Cristina Scielzo ◽  
Elisa Frisaldi ◽  
Stefania Stella ◽  
...  

Growth and survival of chronic B-cell tumors are favored by the malignant cell's capacity to respond to selected microenvironmental stimuli provided by nontumoral bystander cells. To investigate which mechanisms operate in these crosstalks and whether they are malignancy-related or reproduce the mechanisms used by normal B cells we have studied the expression and functional role of semaphorin CD100 (now called Sema4D) in chronic lymphocytic leukemia (CLL) cells and normal CD5+ B cells. We demonstrate here that (1) leukemic and normal CD5+ B lymphocytes uniformly express CD100; (2) the CD100 high-affinity receptor Plexin-B1 is expressed by bone marrow stromal cells, follicular dendritic cells, and activated T lymphocytes, and is thus available to CD100+ lymphocytes in different specific microenvironments; and (3) upon interaction between CD100 and Plexin-B1 both CLL and normal CD5+ B cells increase their proliferative activity and extend their life span. These findings establish that Plexin-B1 is an easily accessible receptor for CD100 within the immune system. The encounter of CD100+ leukemic cells with Plexin-B1 may promote the proliferation and survival of malignant cells. The crosstalk operated by the CD100/Plexin-B1 interaction is not malignancy related but reproduces a mechanism used by normal CD5+ B cells.


Blood ◽  
2002 ◽  
Vol 100 (8) ◽  
pp. 2973-2979 ◽  
Author(s):  
Anne J. Novak ◽  
Richard J. Bram ◽  
Neil E. Kay ◽  
Diane F. Jelinek

B-cell chronic lymphocytic leukemia (B-CLL) is defined by the accumulation of CD5+ B cells in the periphery and bone marrow. This disease is not characterized by highly proliferative cells but rather by the presence of leukemic cells with significant resistance to apoptosis and, therefore, prolonged survival. B-lymphocyte stimulator (BLyS) is a newly identified tumor necrosis factor (TNF) family member shown to be critical for maintenance of normal B-cell development and homeostasis and it shares significant homology with another TNF superfamily member, APRIL. The striking effects of BLyS on normal B-cell maintenance and survival raises the possibility that it may be involved in pathogenesis and maintenance of hematologic malignancies, including B-CLL. In this study, we investigated the status of APRIL and BLyS expression, as well as their receptors, in this disease. All B-CLL patient cells studied expressed one or more of 3 known receptors for BLyS; however, the pattern of expression was variable. In addition, we demonstrate for the first time that B-CLL cells from a subset of patients aberrantly express BLyS and APRIL mRNA, whereas these molecules were not detectable in normal B cells. Furthermore, we provide in vitro evidence that BLyS protects B-CLL cells from apoptosis and enhances cell survival. Because these molecules are key regulators of B-cell homeostasis and tumor progression, leukemic cell autocrine expression of BLyS and APRIL may be playing an important role in the pathogenesis of this disease.


Blood ◽  
1981 ◽  
Vol 57 (2) ◽  
pp. 324-327 ◽  
Author(s):  
P Rambotti ◽  
S Davis

Abstract Lactic dehydrogenase (LDH) was quantitated and the isozyme pattern studied in lymphocyte subpopulations from normal people and patients with chronic lymphocytic leukemia (CLL). Normal T lymphocytes differed from normal B lymphocytes in having greater total LDH activity (597.2 versus 252.1). Total LDH activity in CLL T cells (347.1) was lower than normal T cells., but not significantly different than normal B cells. Total LDH activity in CLL B cells (124.6) was lower then normal B cells and normal T cells. The isozyme pattern of normal T lymphocytes showed a higher activity in the LDH-1 band (26.7% versus 5.4%) but showed a lower activity in LDH-5 band (4.3% versus 16.3%) compared to normal B cells. Chronic lymphocytic leukemia T cells could be distinguished from CLL B cells by a high LDH-5 band (22.3% versus 7.6%) and from normal T cells by a high LDH-5 band (22.3% versus 4.3%) and a low LDH-1 band (7.3% versus 26.7%). CLL B cells could be distinguished from normal B cells by a low LDH-5 band (7.6% versus 16.3%). Thus, the LDH isozyme pattern distinguishes normal T lymphocytes from normal B lymphocytes, and normal T and B lymphocytes from CLL T and B lymphocytes.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2965-2965 ◽  
Author(s):  
Anu Cherukuri ◽  
Edward Kadel ◽  
Sang H. Lee ◽  
Cheryl Goldbeck ◽  
Carla Heise ◽  
...  

Abstract CD40 and CD40 ligand (CD40L) interaction is a key regulator of B-chronic lymphocytic leukemia (CLL) survival. CD40 activation leads to binding with tumor necrosis factor receptor-associated factors (TRAFs) and the subsequent activation of multiple downstream signaling pathways involved in cellular proliferation and survival. We have generated a novel fully human IgG1 anti-CD40 antagonistic monoclonal antibody, CHIR-12.12, using XenoMouse® mice (Abgenix, Inc). CHIR-12.12 blocks CD40L binding to CD40 and inhibits CD40L-induced proliferation/survival of normal human B cells, primary CLL cells, and primary non-Hodgkin’s lymphoma (NHL) cells. We have also demonstrated that it has highly potent antibody-dependent cellular cytotoxicity (ADCC) against primary CLL and non-Hodgkin’s lymphoma cells. We have now investigated its effects on primary CLL cell survival. Soluble human CD40L prolongs primary CLL cell survival in culture, and treatment with CHIR-12.12 inhibits this survival when measured 48–72 hours after addition of CHIR-12.12. CD40L-mediated survival is associated with activation and phosphorylation of Akt, p38 MAPK, ERK, and IkB kinases a and b. Additionally, the anti-apoptotic proteins Mcl-1, Bcl-xl, and XIAP are induced, and markers of apoptosis (cleaved PARP and Caspase-3) are reduced. In contrast, CHIR-12.12 treatment of CD40L-stimulated primary CLL cells ex vivo inhibited downstream phosphorylation of Akt, p38 MAPK, ERK, and IkB kinases (IKK) a and b. Additionally, CHIR-12.12 treatment resulted in induction of cleaved caspase-3 and PARP, and reduction of XIAP, Mcl-1, and Bcl-xl expression, ultimately leading to CLL cell apoptosis. These results demonstrate that CHIR-12.12 inhibits CD40L-mediated signaling pathways and cell survival and could be a potential therapeutic treatment for CLL. CHIR-12.12 is currently in a Phase I clinical study for CLL.


Sign in / Sign up

Export Citation Format

Share Document