scholarly journals Chronic Lymphocytic Leukemic B Cells But Not Normal B Cells Are Rescued From Apoptosis by Contact With Normal Bone Marrow Stromal Cells

Blood ◽  
1998 ◽  
Vol 91 (7) ◽  
pp. 2387-2396 ◽  
Author(s):  
L. Lagneaux ◽  
A. Delforge ◽  
D. Bron ◽  
C. De Bruyn ◽  
P. Stryckmans

Abstract The leukemic B lymphocytes from chronic lymphocytic leukemic (CLL) patients have a long survival in vivo, although ex vivo they rapidly die by apoptosis. To further investigate the mechanism of this, we have studied the influence of bone marrow stromal cells from normal subjects on apoptosis of B-CLL cells and normal umbilical cord blood (UCB) B lymphocytes. After 48 hours of incubation in medium alone, leukemic and normal B cells showed, respectively, 22 ± 3% and 31 ± 5% of apoptosis. Cocultures with stromal cells reduced the percentage of leukemic cells undergoing apoptosis (8 ± 2%, P< .0005) and prevented the loss of bcl-2 protein expression. In contrast, stromal cells slightly increased normal B-cell apoptosis (37 ± 6%). Direct contact between leukemic cells and stromal cells was found to be essential for inhibition of leukemic cell apoptosis; indeed, separation of leukemic cells from stromal cells by microporous membrane increased spontaneous apoptosis, and comparable results were obtained with stromal cell conditioned medium. The difference in behavior observed between normal and leukemic B cells plated on stromal cells can be explained by the fact that only a few normal B cells adhere to stromal cells in comparison with B-CLL cells. B-CLL cell adhesion to stromal cells is mediated by β1 and β2 integrins acting simultaneously. Contact between B-CLL cells and bone marrow stromal cells seems to play a major role in the accumulation and survival of B-CLL cells in the bone marrow.

Blood ◽  
1998 ◽  
Vol 91 (7) ◽  
pp. 2387-2396 ◽  
Author(s):  
L. Lagneaux ◽  
A. Delforge ◽  
D. Bron ◽  
C. De Bruyn ◽  
P. Stryckmans

The leukemic B lymphocytes from chronic lymphocytic leukemic (CLL) patients have a long survival in vivo, although ex vivo they rapidly die by apoptosis. To further investigate the mechanism of this, we have studied the influence of bone marrow stromal cells from normal subjects on apoptosis of B-CLL cells and normal umbilical cord blood (UCB) B lymphocytes. After 48 hours of incubation in medium alone, leukemic and normal B cells showed, respectively, 22 ± 3% and 31 ± 5% of apoptosis. Cocultures with stromal cells reduced the percentage of leukemic cells undergoing apoptosis (8 ± 2%, P< .0005) and prevented the loss of bcl-2 protein expression. In contrast, stromal cells slightly increased normal B-cell apoptosis (37 ± 6%). Direct contact between leukemic cells and stromal cells was found to be essential for inhibition of leukemic cell apoptosis; indeed, separation of leukemic cells from stromal cells by microporous membrane increased spontaneous apoptosis, and comparable results were obtained with stromal cell conditioned medium. The difference in behavior observed between normal and leukemic B cells plated on stromal cells can be explained by the fact that only a few normal B cells adhere to stromal cells in comparison with B-CLL cells. B-CLL cell adhesion to stromal cells is mediated by β1 and β2 integrins acting simultaneously. Contact between B-CLL cells and bone marrow stromal cells seems to play a major role in the accumulation and survival of B-CLL cells in the bone marrow.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2587-2587
Author(s):  
Yang Yang ◽  
Baohua Sun ◽  
Saradhi Mallampati ◽  
Zhen Cai ◽  
Xiaoping Sun

Abstract Abstract 2587 Acute lymphoblastic leukemia (ALL) is one of the fastest-growing hematological malignancies affecting patients with all ages, particularly children. Significant advances have been made in recent years in our understanding of the disease and the development of new therapies, which have led to a greatly improved outcome. Nevertheless, in a significant number of patients with ALL, the disease relapse and become resistant to treatment, causing death of the patients. Increasing evidence suggests that relapse of the disease and resistant to treatment are largely attributed to the protection of the leukemic cells by various components in the microenvironment, such as bone marrow stromal cells. However, the cross-talk between leukemic cells and their microenvironment remains poorly understood. Therefore, better understanding the mechanisms underlying the protection of ALL cells by the microenvironment is of ultimate importance in developing new therapies targeting such protection and eventually eradicating all the leukemic cells to cure the disease. In this study, we used a coculture system with leukemic cells and bone marrow stromal cells (MSC) to mimic the in vivo interaction between the two cell types to explore the molecular events that might be responsible for the protection of ALL cells from Ara-C induced apoptosis. We cocultured human primary ALL cells with hTERT-immortalized normal human MSC and evaluated ALL cell apoptosis by FACS after staining with Annexin V and propidium iodide. In all 8 cases, the MSC provided significant protection of ALL cells from both spontaneous and Ara-C induced apoptosis. For example, the mean Ara-C induced apoptosis of ALL cells cultured without MCS was 42.7% (range, 27–54%), whereas it was 19.1% (range, 8–27%) with MSC. Similar results were obtained with human leukemia cell lines Reh, SEMK2 and RS4.11. We also found that the murine MSC line M210B4 could provide similar protection to ALL cells, whether the ALL cells are primary or cell lines. The reduced apoptosis in the coculture were confirmed by Western blot which showed that MSC could protect ALL cells from Caspase-3 and PARP cleavage. Furthermore, our results showed no significant Ara-C induced reduction in S phase when cocultured with MSC. This phenomenon was associated with decreased cyclinA and CDK2 expression. In addition, we found that cocultured with MSC resulted in phosphorylation of AKT in ALL cells and PI3K inhibitor LY294002 specifically inhibited MSC-induced activation of AKT and promoted ALL cell apoptosis. In addition, beta-catenin and c-myc had increased expression in ALL cells cocultured with MSC, suggesting that Wnt pathway could play a role in MSC-mediated protection. To identify candidate molecules potentially involved in the protection of ALL cells by MSC, we performed gene expression microarray analyses with ALL cells exposed to Ara-C in presence or absence of MSC. Our data indicated that several signaling pathways might be involved in this process, including apoptosis signaling and cell cycle checkpoint control, which confirmed above findings. The top expressed genes identified in the microarray studies were confirmed by RT-PCR. Collectively, our results demonstrated that MSC can protect ALL cells from Ara-C induced apoptosis by multiple signaling pathways, such as those involving PI3K/AKT and Wnt signaling. Hence, targeting these pathways may become potential novel therapeutic strategies to disrupt the support of the microenvironment to ALL cells and to eventually eradicate leukemic cells. Disclosures: No relevant conflicts of interest to declare.


2015 ◽  
Vol 6 (1) ◽  
Author(s):  
Rikke K. Andersen ◽  
Walid Zaher ◽  
Kenneth H. Larsen ◽  
Nicholas Ditzel ◽  
Katharina Drews ◽  
...  

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3794-3794
Author(s):  
Saradhi Mallampati ◽  
Baohua Sun ◽  
Yun Gong ◽  
Enze Wang ◽  
M. James You ◽  
...  

Abstract Development and progression of leukemia requires interaction of leukemia-initiating cells with their bone marrow niches. The niches serve as the nursery and shelter for the leukemic cells, which can result in drug resistance, disease recurrence, and minimal residual disease, the most important causes for the death of patients with leukemia. Therefore, obliteration of the interaction between the leukemic cells and their niches is of utmost importance in eradicating leukemic cells during therapy to cure the disease. However, little is currently known of the molecular mechanisms underlying the interaction of the two types of cells. Sox4, a SRY-related HMG-box containing transcription factor that is vital during development, plays an important role in leukemia. Published mouse studies demonstrated that increased expression of Sox4 was associated with leukemogenesis. We determined the expression levels of Sox4 by real-time RT-PCR in 100 human leukemic samples and found high levels of expression in B- and T-ALL, but not in AML, CML, CLL, Sezary syndrome, or T cell prolymphocytic leukemia. In accordance, 7 of the 8 ALL cell lines (the exception was 697) we tested showed high expression levels of Sox4, but AML cell lines, normal mature B cells, T cells, and bone marrow CD34+ cells had low levels of expression. Since the majority of clinical B-ALL cases correspond to the pre-B cell stage, we investigated the role of Sox4 in a pre-B cell line (Nalm6) by lentivirus-mediated RNAi. Remarkably, knockdown of Sox4 in Nalm6 cells caused 70% reduction in the formation of leukemic cell clusters under the monolayer of co-cultured M2-10B4 bone marrow stromal cells, a phenomenon known as pseudo-emperipolesis. Similar results were obtained with ex vivo cultured bone marrow cells from conditional Sox4 knockout mice that displayed B cell developmental arrest at the transition from pro-B to pre-B cell stage and an absence of pre-B cells. These findings suggested that Sox4 is required for the interaction of the developing B cells or leukemic cells with bone marrow stromal cells, a component of the bone marrow niche. Since CXCR4/SDF1-mediated “homing” is known to be required for pseudo-emperipolesis, we tested the effect of Sox4 on Nalm6 cell migration toward SDF1 gradient and found that Sox4 did not affect the migration, suggesting that Sox4 is not acting through “homing”. Instead, our data indicated that the role of Sox4 in the interaction of leukemic cells with stromal cells is most likely mediated by its ability in enhancing the adhesion of the leukemic cells because we found that lentivirus-medicated overexpression of Sox4 in the 697 B cell line caused the suspension cells to display a spindle and adhesive morphology. In addition, 21% of the putative Sox4 downstream genes that we identified by multiple sets of gene expression microarray experiments are known to be involved in cell adhesion. Moreover, we found that the changes in gene expression profile of leukemic cells upon Sox4 knockdown or overexpression significantly overlap with the changes in response to the presence of bone marrow stromal cells in co-culture, indicating that Sox4 pathways are involved in leukemic cell response to stromal cell signaling. Based on these findings we hypothesize that deletion of Sox4 abolishes the interaction between the developing lymphocytes and their niches during lymphopoiesis. Conversely, overexpression of Sox4 may enforce these cells to over-interact with the niches so that they are overexposed to local growth factor stimuli. If superimposed with other genetic and/or epigenetic changes in the developing lymphocytes, such over-interaction may result in the development of leukemia. In case of established leukemia, such over-interaction may lead to the enhanced protection of leukemic cells by their niches. Therefore, the role of Sox4 in the interaction of developing lymphocytes or leukemic cells with their niches is like “rooting into the soil” of a growing tree, abbreviated as “rooting”.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1779-1779
Author(s):  
Marta Crespo ◽  
Eva Calpe ◽  
Noelia Purroy ◽  
Cecilia Carpio ◽  
Pau Abrisqueta ◽  
...  

Abstract Abstract 1779 Chronic lymphocytic leukemia (CLL) is a lymphoproliferative disorder characterized by the accumulation and proliferation of monoclonal CD5+ B lymphocytes in peripheral blood, secondary lymphoid tissues, and bone marrow. In CLL, high expression of ZAP-70 is a strong prognostic factor that identifies patients with higher probability of progression and shorter survival. Increasing evidence shows that the microenvironment plays a relevant role in the natural history of this disease, providing CLL cells with proliferative and anti-apoptotic signals. In this sense, despite that the role of ZAP-70 in the biology of CLL has not been fully elucidated, it is well known that it is associated with enhanced response to several microenvironmental stimuli. Herein, we aimed to analyze the effect of ZAP-70 on the interaction of B-cells with the microenvironment in an in vivo xenograft model of disseminated B-cell leukemia. For this, we stably transfected Raji B-cells with a vector expressing a GFP-ZAP-70 fusion protein or GFP only as a control. Cells were then intravenously injected into 7- to 9-weeks old female C.B-17 SCID mice (Raji GFP n=12; Raji GFP-ZAP-70 n=9). This xenograft model develops hind legs paralysis at around 14 to 19 days after cell injection due to central nervous system (CNS) infiltration, which precedes death by 1 to 2 days; therefore, paralysis was considered the end point of the study. The infiltration of the CNS was not influenced by ZAP-70 expression; thus, median survival was of 16 days for both Raji GFP and Raji GFP-ZAP-70-injected mice. Analysis of the infiltration of Raji B-cells in the different organs by flow cytometry and immunohistochemistry disclosed a significantly increased infiltration in the bone marrow of Raji GFP-ZAP-70 mice compared to Raji GFP mice (67% ± 5.76% vs 2.9% ± 1.49%; P <.001). Based on that, we analyzed the in vitro migrative capacity of cells toward SDF-1α, the main chemokine regulating lymphocyte trafficking into the bone marrow. We observed that, despite that Raji GFP-ZAP-70 and Raji GFP transfectants had similar surface expression of CXCR4, GFP-ZAP-70 cells had higher migration toward SDF-1α compared to GFP cells (migration index: 34.6 ± 4.11 vs 9.9 ± 1.47; P =.0079). Of note, B-cells expressing ZAP-70 also migrated more toward bone marrow stromal cells than B-cells non-expressing ZAP-70 (migration index: 27.1 ± 1.6 and 9.43 ± 0.77, respectively; P =.028). This increased migrative capacity independent of CXCR4 expression could be explained by the observation that Raji GFP-ZAP-70 cells showed enhanced response to CXCR4 stimulation by SDF-1α, evidenced by increased phosphorylation of ZAP-70, Akt and ERK1/2 proteins. Accordingly, we found that primary CLL subclones with high expression ZAP-70 also had increased migration toward SDF-1α. The relevance of the CXCR4/SDF-1α axis in the migrative capacity of Raji B-cells was demonstrated by the blockage of CXCR4 with a mAb, which resulted in reductions of more than 85% in the in vitro migration to both SDF-1α and to bone marrow stromal cells in both cell lines. To evaluate this axis in the xenograft model, SCID mice were injected with malignant B-cells treated with anti-CXCR4 mAb (Raji GFP n=5; Raji GFP-ZAP-70 n= 8) or isotypic control antibody (Raji GFP n=5; Raji GFP-ZAP-70 n= 8). In mice injected with Raji-GFP cells, CXCR4 blockage resulted in a delayed onset of the disease (median survival: 15 days vs 69 days; p<0.01), having these cells restored their CXCR4 expression. Surprisingly, CXCR4 blockage in Raji GFP-ZAP-70 cells produced different effects, with 7 out of 8 mice remaining asymptomatic without detectable malignant B-cells during follow-up (> 100 days). This suggests that ZAP-70-positive malignant B-cells developed dependence on CXCR4-derived survival signals. In summary, we showed that ZAP-70 is responsible for an enhanced cellular infiltration into the bone marrow caused by an amplified response to CXCR4 as regards signaling and migration. Interestingly, ZAP-70-positive malignant B-cells apparently became addicted to survival signals derived from CXCR4 as a consequence of the enhanced signaling. Further elucidation of the role of ZAP-70 in signaling from the microenvironment, particularly from the CXCR4/SDF-1α axis, will contribute to enlighten the biology behind the adverse clinical impact of high ZAP-70 expression in CLL, and will also provide a rationale for the development of novel therapies. Disclosures: No relevant conflicts of interest to declare.


2015 ◽  
Vol 24 (7) ◽  
pp. 1205-1220 ◽  
Author(s):  
Hai Van Thi Do ◽  
Wan Ting Loke ◽  
Irene Kee ◽  
Vivienne Liang ◽  
Sebastian J. David ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document