Abstract

BackgroundDysregulation in calcium (Ca2+) signaling is a hallmark of chronic lymphocytic leukemia (CLL). While the role of the B cell receptor (BCR) Ca2+ pathway has been associated with disease progression, the importance of the newly described constitutive Ca2+ entry (CE) pathway is less clear. In addition, we hypothesized that these differences reflect modifications of the CE pathway and Ca2+ actors such as Orai1, transient receptor potential canonical (TRPC) 1, and stromal interaction molecule 1 (STIM1), the latter being the focus of this study.MethodsAn extensive analysis of the Ca2+ entry (CE) pathway in CLL B cells was performed including constitutive Ca2+ entry, basal Ca2+ levels, and store operated Ca2+ entry (SOCE) activated following B cell receptor engagement or using Thapsigargin. The molecular characterization of the calcium channels Orai1 and TRPC1 and to their partner STIM1 was performed by flow cytometry and/or Western blotting. Specific siRNAs for Orai1, TRPC1 and STIM1 plus the Orai1 channel blocker Synta66 were used. CLL B cell viability was tested in the presence of an anti-STIM1 monoclonal antibody (mAb, clone GOK) coupled or not with an anti-CD20 mAb, rituximab. The Cox regression model was used to determine the optimal threshold and to stratify patients.ResultsSeeking to explore the CE pathway, we found in untreated CLL patients that an abnormal CE pathway was (i) highly associated with the disease outcome; (ii) positively correlated with basal Ca2+ concentrations; (iii) independent from the BCR-PLCγ2-InsP3R (SOCE) Ca2+ signaling pathway; (iv) supported by Orai1 and TRPC1 channels; (v) regulated by the pool of STIM1 located in the plasma membrane (STIM1PM); and (vi) blocked when using a mAb targeting STIM1PM. Next, we further established an association between an elevated expression of STIM1PM and clinical outcome. In addition, combining an anti-STIM1 mAb with rituximab significantly reduced in vitro CLL B cell viability within the high STIM1PM CLL subgroup.ConclusionsThese data establish the critical role of a newly discovered BCR independent Ca2+ entry in CLL evolution, provide new insights into CLL pathophysiology, and support innovative therapeutic perspectives such as targeting STIM1 located at the plasma membrane.

Highlights

  • Dysregulation in calcium (Ca2+) signaling is a hallmark of chronic lymphocytic leukemia (CLL)

  • Cells from patients with progressive disease As deregulation in Ca2+ signaling is an important hallmark of B cells (B-CLL) cells, and suspected to vary during CLL disease progression [2], Ca2+ entry in the absence of B cell receptor (BCR) engagement, designated as CE, was evaluated in resting B-CLL cells

  • According to this dichotomy, the KaplanMeier log-rank analysis revealed, for those CE+ CLL patients (n = 16), a significant difference with regards to parameters associated with disease outcome such as progression free survival (PFS) (P = 0.001; Fig. 1b), treatment free survival (TFS) (P = 0.003; Fig. 1c) and lymphocyte doubling time (LDT) (P = 0.02; Fig. 1d)

Read more

Summary

Introduction

Dysregulation in calcium (Ca2+) signaling is a hallmark of chronic lymphocytic leukemia (CLL). We hypothesized that these differences reflect modifications of the CE pathway and Ca2+ actors such as Orai, transient receptor potential canonical (TRPC) 1, and stromal interaction molecule 1 (STIM1), the latter being the focus of this study. There is long standing evidence that CLL B cells (B-CLL) present an altered calcium (Ca2+) signaling pathway, which evolves with disease progression [2, 3]. (STIM1) and plasma-membrane Orai channel are believed to be the main molecular actors of store operated Ca2+ entry (SOCE) in lymphocytes [5,6,7,8].

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call