B-cell chronic lymphocytic leukemia (CLL) is a heterogenous and incurable B-cell malignancy. CLL cells migrate and accumulate in different sites including the peripheral blood (PB), bone marrow (BM) and lymph nodes (LN) in vivo, but undergo apoptosis in vitro. Therefore, we hypothesized that CLL cells at these sites are different and receive different microenvironmental signals that regulate their proliferation/survival and migration. Most reports on the microenvironmental influence on CLL cells have used in vitro models consisting of stromal and CLL cells. However, in this study, to better understand the influence of site-specific microenvironments in vivo, gene expression patterns of CLL cells obtained from PB, BM and LN were investigated. CLL cells were isolated from patients’ PB (PB-CLL, n= 20), BM (BM-CLL, n=14) and LN (LN-CLL, n=15) and used to determine the gene expression patterns by microarray analysis. In addition, we also included PB-CLL cases from our previous study (n=40) to further validate the findings of this study. Significant Analyses of Microarray (SAM) revealed differential expression of more than 500 genes among these three sites. To understand the potential roles of these differentially-expressed genes and their association with relevant functional pathways in CLL, Gene Set Enrichment Analysis (GSEA) was performed. The validation of pathway specific genes was further confirmed by quantitative real time PCR. Among the pathways identified, the most active pathways associated with the migration and proliferation/survival of CLL cells, namely chemokine-signaling, BCR signaling, BAFF/APRIL-signaling, and NFκB-signaling pathways, were selected for further analyses. We hypothesized that chemokines and their receptors mediate the migration of CLL cells between PB and LN or BM, and that molecules of the BCR, BAFF/APRIL and NFκB pathways regulate proliferation/survival. To determine the role of chemokines and their receptors in CLL cell migration, we studied the expression of 52 chemokine/chemokine receptors and found that PB-CLL cells significantly (p<0.005) overexpressed CXCR4 and CCR7 compared to BM-CLL and LN-CLL cells. The ligands CCL21 and CXCL13 were significantly overexpressed (p<0.005 and p<0.01 respectively) in LN-CLL. These results indicate that PB-CLL cells express distinct chemokine receptors which may lead them to home to BM or LN and receive stimuli to form proliferation centers. Based on GSEA analysis, the stimuli for proliferation/survival for CLL cells in the LN and BM are provided by Syk and Btk (BCR signaling), BAFF and TRAF2 (BAFF/APRIL signaling), and several targets of the NFκB pathways. Syk and Btk were significantly overexpressed in LN-CLL (p<0.05) and PB-CLL (p<0.005) compared to BM-CLL, with the highest expression in LN-CLL, suggesting chronic activation of CLL cells in lymph node. Similarly, BAFF and TRAF2 were significantly overexpressed (p<0.03) in LN-CLL compared to PB-CLL and BM-CLL. Furthermore, the NFκB pathway, which is important for the proliferation and survival, also showed distinct association in different CLL-cell compartments. The RELA, NFκB1, NFκB2, TNFAIP3 and NFκB regulators such as NFκBIA, NFκBIE were also significantly (p<0.01) overexpressed in PB-CLL and BM-CLL compared to LN-CLL with highest expression in BM-CLL. Whereas few NFκB associated genes such as NFκB1L1 and RelB were significantly (p<0.02) expressed in LN-CLL cells. Thus, differentially-expressed NFkB genes among PB-CLL, BM-CLL and LN-CLL cells indicate that these different CLL cells utilize different NFκB molecules for proliferation/survival. Together, our results show that CLL cells from different in vivo microenvironments such as PB, BM and LN exhibit differential gene expression patterns, and many of the genes are involved in regulation of migration and proliferation/survival. Furthermore, LN-CLL cells expressing chemokine ligands, BCR, BAFF and NFκB signaling molecules attract other cells including more CLL cells to form an optimal microenvironment which provide prosurvival and proliferative signals to CLL cells.

Disclosures: No relevant conflicts of interest to declare.

(This work was supported by the CLL Foundation, Houston, TX).

Author notes

Corresponding author

Sign in via your Institution