15 research outputs found

    Study of calcium entries deregulation in chronic lymphocytic leukemia B-cells : evidence for a new therapeutic target

    No full text
    La leucémie lymphoïde chronique (LLC) constitue l'hémopathie maligne la plus fréquente dans les pays occidentaux et résulte d’une accumulation de lymphocytes B (LB) monoclonaux matures porteurs de la glycoprotéine CD5. Les LB de LLC sont également caractérisées par une altération de l'homéostasie du calcium avec, d'une part, la mise en évidence de facteurs de survie contrôlés par le calcium tels que phospho-ERK, NFAT-2 et IL-10, et d'autre part par une progression de la maladie qui est associée à la réponse au calcium. Tout d'abord, afin de mieux comprendre l'impact de la molécule CD5 sur les dérégulations du calcium, il a été montré que l'introduction d'un plasmide d'expression pour CD5 dans les lignées de cellules B humaines s'accompagnait d'une entrée calcique à l’état basale. Ensuite, cette entrée, appelée entrée constitutive, a été recherchée dans les LB de LLC pour montrer qu'elle caractérisait les patients non traités en phase d'évolution. Comme pour les lignées de cellules B CD5, cette nouvelle voie d'entrée du calcium est autonome et indépendante de la voie classique de signalisation du LB de LLC : BCR-IP3R. L'étude des protéines responsables de cet influx a permis de mettre en évidence, premièrement trois partenaires STIM1, Orai1 et TRPC1, et deuxièmement l'importance de la fraction membranaire de STIM1 (STIMPM) puisque l'utilisation d'un anticorps monoclonal anti-STIM1 (Acm) est capable d’inhiber l'entrée constitutive du calcium qui à son tour agit sur la survie des LB, pour les patients STIMPM positifs, lorsque l'Acm anti-STIM1 est utilisé en association avec le rituximab, un Acm thérapeutique anti-CD20. Enfin, la modélisation de la partie Cterminale de STIM1 permet d'envisager plusieurs cibles potentielles pour le développement de nouveaux Acm anti-STIM1. En conclusion, la mise en place, par le clone malin de LLC, d'une entrée constitutive du calcium favorise son agressivité et constitue donc une nouvelle voie thérapeutique contrôlable par l'utilisation d’Acm anti-STIM1 ce qui ouvre de nouvelles perspectives comme outils diagnostiques et thérapeutiques.Chronic lymphocytic leukemia (CLL) is the most common hematological malignancy in Western countries and is a result of the accumulation of mature monoclonal B lymphocytes (B-CLL) carrying the CD5 glycoprotein. The B-CLL are also characterized by an alteration of calcium homeostasis with, on the one hand, the demonstration of calcium-controlled survival factors such as phospho-ERK, NFAT- 2 and IL-10, and on the other hand by a progression of the disease which is associated with the response to calcium. Initially, in order to better understand the impact of the CD5 molecule on calcium deregulations, it has been shown that the introduction of an expression plasmid for CD5 into human B cell lines was accompanied by a calcium entry in the basal state. Then, this entry, called constitutive entry, was sought in the B-CLL to show that it characterized untreated patients in the evolution phase.As with CD5 B cell lines, this new calcium entry pathway is autonomous and independent of the classical B-CLL signaling pathway: BCR-IP3R. The study of the proteins responsible for this influx made it possible to highlight, firstly three partners (STIM1, Orai1 and TRPC1), and secondly the importance of the membrane fraction of STIM1 (STIMPM) since the use of a human monoclonal antibody (mAb) anti-STIM1 is able to inhibit the constitutive entry of calcium which in turn acts on the survival of B-CLL, for STIMPM positive patients, when the anti-STIM1 mAb was used in combination with rituximab, a therapeutic anti-CD20 mAb. Finally, the modeling of the C-terminal part of STIM1 makes it possible to envisage several potential targets for the development of new anti-STIM1 mAbs. In conclusion, the introduction by the CLL malignant clone of a constitutive entry of calcium favors its aggressiveness and thus constitutes a new therapeutic pathway controllable by the use of anti-STIM1 mAb, which opens new perspectives like diagnostic and therapeutic tools

    Calcium signaling and cell fate: how can Ca (2+)signals contribute to wrong decisions for Chronic Lymphocytic Leukemic B lymphocyte outcome?

    No full text
    International audienceCa (2+)signaling is a key regulator of B lymphocyte cell fate and defects in this signaling pathway have been reported in numerous diseases such as Chronic lymphocytic leukemia (CLL). CLL is a B cell clonal disorder characterized by the accumulation of mature monoclonal CD5 (+)B cells. Although CLL could be considered to be a proliferative disease, most circulating CLL B cells are arrested in the G0 phase of the cell cycle and present both defects in calcium (Ca (2+)) homeostasis and signaling. The Ca (2+)response to antigen ligation is heterogeneous and related, in part, to defects arising from the incapacity to respond to B cell receptor (BCR) engagement (anergy), to the expression of T cell kinases (e.g. Zap70), and to the presence of negative feedback regulation by phosphatases (e.g. SHP-1). Anergic CD5 (+)CLL B cells are characterized by an elevated basal Ca (2+)level, IgM/CD79 downregulation, a constitutive activation of BCR pathway kinases, and an activation of the nuclear factor of activated T cells (NF-AT). Based on the Ca (2+)response, patients are classified into three groups: unresponders, responders with apoptosis, and responders with entry in the cell cycle. Moreover, internal and direct interaction between leukemic BCR-HCDR3 epitopes at the plasma membrane and interaction between Bcl-2 and the IP3-receptor at the endoplasmic reticulum are also suspected to interfere with the intracellular Ca (2+)homeostasis in CLL-B cells. As a whole, the Ca (2+)pathway is emerging to play a key role in malignant CLL-B survival, disease progression, and last but not least, in the therapeutic response

    Ofatumumab capacity to deplete B cells from chronic lymphocytic leukaemia is affected by C4 complement exhaustion.

    No full text
    International audienceThe management of patients with chronic lymphocytic leukaemia (CLL) has improved with the utilisation of ofatumumab as a novel anti-CD20 monoclonal antibody. However, as half of the patients fail to respond to the treatment, the aim of this study was to evaluate circulating CLL cell depletion and clinical response according to the context of complement activation and FcγRIIIA polymorphism in ten CLL patients with relapsed/refractory disease. At the end of the treatment, results indicated that circulating CD5(+) CD19(+) CLL cell depletion was major (50% decrease) in 4 of 10 patients and ineffective for the two other patients. No clinical modifications were observed following ofatumumab introduction. Ofatumumab administration leads to a rapid and important exhaustion of complement C4 levels in patients with initial lymphocytosis. C4 exhaustion was accelerated in a non-responder patient, and incomplete in two patients with partial circulating depletion. Moreover, delaying weekly to monthly ofatumumab injections improved CLL cell depletion in two patients. FcγRIIIA 158 polymorphism (FF n = 6 and VF n = 4) was not associated with major and/or partial circulating CLL cell depletion. In conclusion, ofatumumab induces an important C4 exhaustion that needs to be taken into account when treating CLL patients with ofatumumab

    Molecular Dynamics Simulations of Membrane-Bound STIM1 to Investigate Conformational Changes during STIM1 Activation upon Calcium Release.

    No full text
    International audienceCalcium is involved in important intracellular processes, such as intracellular signaling from cell membrane receptors to the nucleus. Typically, calcium levels are kept at less than 100 nM in the nucleus and cytosol, but some calcium is stored in the endoplasmic reticulum (ER) lumen for rapid release to activate intracellular calcium-dependent functions. Stromal interacting molecule 1 (STIM1) plays a critical role in early sensing of changes in the ER's calcium level, especially when there is a sudden release of stored calcium from the ER. Inactive STIM1, which has a bound calcium ion, is activated upon ion release. Following activation of STIM1, there is STIM1-assisted initiation of extracellular calcium entry through channels in the cell membrane. This extracellular calcium entering the cell then amplifies intracellular calcium-dependent actions. At the end of the process, ER levels of stored calcium are reestablished. The main focus of this work was to study the conformational changes accompanying homo- or heterodimerization of STIM1. For this purpose, the ER luminal portion of STIM1 (residues 58-236), which includes the sterile alpha motif (SAM) domain plus the calcium-binding EF-hand domains 1 and 2 attached to the STIM1 transmembrane region (TM), was modeled and embedded in a virtual membrane. Next, molecular dynamics simulations were performed to study the conformational changes that take place during STIM1 activation and subsequent protein-protein interactions. Indeed, the simulations revealed exposure of residues in the EF-hand domains, which may be important for dimerization steps. Altogether, understanding conformational changes in STIM1 can help in drug discovery when targeting this key protein in intracellular calcium functions

    Cx43 Present at the Leading Edge Membrane Governs Promigratory Effects of Osteoblast-Conditioned Medium on Human Prostate Cancer Cells in the Context of Bone Metastasis

    No full text
    International audienceAmong the different interacting molecules implicated in bone metastases, connexin43 (Cx43) may increase sensitivity of prostate cancer (PCa) cells to bone microenvironment, as suggested by our in silico and human tissue samples analyses that revealed increased level of Cx43 expression with PCa progression and a Cx43 specific expression in bone secondary sites. The goal of the present study was to understand how Cx43 influences PCa cells sensitivity and aggressiveness to bone microenvironment. By means of Cx43-overexpressing PCa cell lines, we revealed a Cx43-dependent promigratory effect of osteoblastic conditioned media (ObCM). This effect on directional migration relied on the presence of Cx43 at the plasma membrane and not on gap junctional intercellular communication and hemichannel functions. ObCM stimulation induced Rac1 activation and Cx43 interaction with cortactin in protrusions of migrating PCa cells. Finally, by transfecting two different truncated forms of Cx43 in LNCaP cells, we determined that the carboxy terminal (CT) part of Cx43 is crucial for the responsiveness of PCa cells to ObCM. Our study demonstrates that Cx43 level and its membrane localization modulate the phenotypic response of PCa cells to osteoblastic microenvironment and that its CT domain plays a pivotal role
    corecore