33 research outputs found

    Regulating TRAIL Receptor-Induced Cell Death at the Membrane: A Deadly Discussion

    Get PDF
    The use of TRAIL/APO2L and monoclonal antibodies targeting TRAIL receptors for cancer therapy holds great promise, due to their ability to restore cancer cell sensitivity to apoptosis in association with conventional chemotherapeutic drugs in a large variety of tumors. TRAIL-induced cell death is tightly regulated right from the membrane and at the DISC (Death-Inducing Signaling Complex) level. The following patent and literature review aims to present and highlight recent findings of the deadly discussion that determines tumor cell fate upon TRAIL engagement

    The " Mendelian Gene " and the " Molecular Gene " : Two Relevant Concepts of Genetic Units

    Get PDF
    International audienceWe focus here on two prevalent meanings of the word gene in research articles. On one hand, the gene, named here “molecular gene”, is a stretch of DNA that is transcribed and codes for an RNA or a polypeptide with a known or presumed function (as in “gene network'), whose exact spatial delimitation on the chromosome remains a matter of debate, especially in cases with alternative splicing, antisense transcripts, etc. On the other hand, the gene, called here “Mendelian gene”, is a segregating genetic unit which is detected through phenotypic differences associated with different alleles at the same locus (as in “gene flow”). We show that the “Mendelian gene” concept is still extensively used today in biology research and is sometimes confused with the “molecular gene”. We try here to clarify the distinction between both concepts. Efforts to delineate the beginning and the end of the DNA sequence corresponding to the “Mendelian gene” and the “molecular gene” reveal that both entities do not always match. We argue that both concepts are part of two relevant frameworks for explaining the biological world

    TRAIL promotes membrane blebbing, detachment and migration of cells displaying a dysfunctional intrinsic pathway of apoptosis

    Get PDF
    Recently, tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL/Apo2L) has been shown to be a potential candidate for cancer therapy. TRAIL induces apoptosis in various cancer cells but not in normal tissues. Here we show that HCT116 and SW480 cells with a deficient mitochondrial apoptotic pathway were resistant to TRAIL-induced apoptosis, whereas HCT116 and SW480 cells with a functional mitochondrial apoptotic pathway underwent apoptosis upon exposure to TRAIL. Surprisingly, TRAIL induced phenotypic changes in cells with a dysfunctional mitochondrial apoptotic pathway, including membrane blebbing and a transient loss of adhesion properties to the substratum. Accordingly, TRAIL stimulated the ability of these cells to migrate. This behavior was the consequence of a transient TRAIL-induced ROCK1 cleavage. In addition, we report that Bax-deficient HCT116 cells exposed to TRAIL for a prolonged period lost their sensitivity to TRAIL as a result of downregulation of TRAIL receptor expression, and became resistant to combination of TRAIL and other drugs such as MG-132 and bortezomib. These findings may have important consequences for TRAIL anti-cancer therap

    TRAIL-R4 Promotes Tumor Growth and Resistance to Apoptosis in Cervical Carcinoma HeLa Cells through AKT

    Get PDF
    International audienceBACKGROUND: TRAIL/Apo2L is a pro-apoptotic ligand of the TNF family that engages the apoptotic machinery through two pro-apoptotic receptors, TRAIL-R1 and TRAIL-R2. This cell death program is tightly controlled by two antagonistic receptors, TRAIL-R3 and TRAIL-R4, both devoid of a functional death domain, an intracellular region of the receptor, required for the recruitment and the activation of initiator caspases. Upon TRAIL-binding, TRAIL-R4 forms a heteromeric complex with the agonistic receptor TRAIL-R2 leading to reduced caspase-8 activation and apoptosis. METHODOLOGY/PRINCIPAL FINDINGS: We provide evidence that TRAIL-R4 can also exhibit, in a ligand independent manner, signaling properties in the cervical carcinoma cell line HeLa, through Akt. Ectopic expression of TRAIL-R4 in HeLa cells induced morphological changes, with cell rounding, loss of adherence and markedly enhanced cell proliferation in vitro and tumor growth in vivo. Disruption of the PI3K/Akt pathway using the pharmacological inhibitor LY294002, siRNA targeting the p85 regulatory subunit of phosphatidylinositol-3 kinase, or by PTEN over-expression, partially restored TRAIL-mediated apoptosis in these cells. Moreover, the Akt inhibitor, LY294002, restituted normal cell proliferation index in HeLa cells expressing TRAIL-R4. CONCLUSIONS/SIGNIFICANCE: Altogether, these results indicate that, besides its ability to directly inhibit TRAIL-induced cell death at the membrane, TRAIL-R4 can also trigger the activation of signaling pathways leading to cell survival and proliferation in HeLa cells. Our findings raise the possibility that TRAIL-R4 may contribute to cervical carcinogenesis

    TRAIL-R4 mediated resistance to TRAIL induced apoptosis : use of chemotherapy and TRAIL mimetics to overcome it

    No full text
    La protéine TRAIL (TNF Related Apoptosis Inducing Ligand) suscite un grand intérêt en thérapie anticancéreuse. Contrairement à la plupart des traitements couramment utilisés en clinique, cette protéine induit sélectivement la mort par apoptose de nombreuses cellules cancéreuses. Cette cytokine exerce son activité cytotoxique en se liant à des récepteurs transmembranaires exprimés à la surface de la cellule cible. Par un jeu d’interactions protéiques, la fixation de TRAIL sur ces récepteurs agonistes (TRAIL-R1 et TRAIL-R2) conduit à l’activation de l’apoptose. L’expression de deux récepteurs antagonistes, TRAIL-R3 et TRAIL-R4, par les cellules cancéreuses, permet aux cellules cibles d’échapper à l’apoptose induite par TRAIL. Nous montrons que ces deux récepteurs font intervenir des mécanismes moléculaires distincts. Leur expression pouvant potentiellement représenter un frein à l’utilisation clinique de TRAIL, nous avons étudié l’effet de la surexpression de l’un d’entre eux, TRAIL-R4 sur l’efficacité des stratégies thérapeutiques associant TRAIL aux chimiothérapies conventionnelles. Les résultats obtenus montrent également que la résistance induite par TRAIL-R4 peut être contournée in vitro et in vivo en associant TRAIL à des agents chimiothérapeutiques. D’un point de vue moléculaire, nous avons montré que la sensibilisation à TRAIL 1) implique une augmentation du recrutement et de l’activation de la caspase-8 au sein du DISC de TRAIL, 2) ne nécessite pas la voie mitochondriale, et 3) est négativement régulée de manière coopérative par c-FLIP, un inhibiteur sélectif de la caspase-8. De manière intéressante, comme les anticorps agonistes actuellement testés en clinique, de petits peptides agonistes de TRAIL-R2, développés en collaboration avec une équipe de chimiste, permettent de contourner la résistance induite par TRAIL-R4, offrant des perspectives thérapeutiques intéressantes. Les récepteurs TRAIL-R3 et TRAIL-R4 sont donc des inhibiteurs de TRAIL. Nos travaux démontrent cependant, que les stratégies associant TRAIL à des agents chimiothérapeutiques, ou l'utilisation d'agonistes TRAIL-R2 permet de contourner la résistance induite par les récepteurs antagonistes de TRAIL et donc d’éliminer ces cellules cancéreuses.TRAIL (TNF Related Apoptosis Inducing Ligand) is a very promising cytokine for cancer therapy. Contrary to current treatments, this protein is able to selectively kill cancer cells, whilst sparing healthy cells. TRAIL induces apoptosis following binding to one of its two different agonistic membrane receptors, TRAIL-R1 and TRAIL-R2. However, expression of one of its two antagonistic receptors, TRAIL-R3 and TRAIL-R4, on cancer cells can impair cancer cell killing by TRAIL. We have shown that these receptors inhibit TRAIL-induced cell death differentially. As these receptors can represent a brake for the use of TRAIL in cancer therapy, we investigated the effect of the expression of one of them, TRAIL-R4 on the efficacy of the different therapeutic strategies associating TRAIL and conventional therapeutic drugs. We show that acquired resistance to TRAIL following expression of TRAIL-R4 can be overcome in vitro and in vivo by combining TRAIL with chemotherapeutic agents. From a molecular point of view, we could demonstrate that sensitization to TRAIL 1) occurs mainly through an increase of caspase-8 recruitment and activation within the TRAIL DISC, 2) is independent of the mitochondrial pathway and 3) is negatively regulated, in a cooperative manner by c-FLIP, a caspase-8 selective inhibitor. Interestingly, like agonistic receptors currently tested in clinic, small agonistic peptides targeting TRAIL-R2, engineered in collaboration with a team of chemists, afford cancer cell killing regardless of TRAIL-R4 expression, providing novel therapeutic perspectives. TRAIL-R3 and TRAIL-R4 should thus be considered as TRAIL inhibitors. Our results demonstrate however that strategies aiming at combining TRAIL with chemotherapeutic agents or the use of TRAIL-R1 or TRAIL-R2 agonists could be effective treatments to eradicate cancer cells that express TRAIL antagonistic receptors

    Regulating TRAIL receptor-induced cell death at the membrane : a deadly discussion.: TRAIL - A deadly DISCussion

    No full text
    Article Open access plusInternational audienceThe use of TRAIL/APO2L and monoclonal antibodies targeting TRAIL receptors for cancer therapy holds great promise, due to their ability to restore cancer cell sensitivity to apoptosis in association with conventional chemotherapeutic drugs in a large variety of tumors. TRAIL-induced cell death is tightly regulated right from the membrane and at the DISC (Death-Inducing Signaling Complex) level. The following patent and literature review aims to present and highlight recent findings of the deadly discussion that determines tumor cell fate upon TRAIL engagement

    Regulating TRAIL receptor-induced cell death at the membrane : a deadly discussion.: TRAIL - A deadly DISCussion

    No full text
    Article Open access plusInternational audienceThe use of TRAIL/APO2L and monoclonal antibodies targeting TRAIL receptors for cancer therapy holds great promise, due to their ability to restore cancer cell sensitivity to apoptosis in association with conventional chemotherapeutic drugs in a large variety of tumors. TRAIL-induced cell death is tightly regulated right from the membrane and at the DISC (Death-Inducing Signaling Complex) level. The following patent and literature review aims to present and highlight recent findings of the deadly discussion that determines tumor cell fate upon TRAIL engagement

    Résistance à l'apoptose induite par TRAIL-R4 (sensibilisation des cellules tumorales par la chimiothérapie ou des mimétiques de TRAIL)

    No full text
    La protéine TRAIL (TNF Related Apoptosis Inducing Ligand) suscite un grand intérêt en thérapie anticancéreuse. Contrairement à la plupart des traitements couramment utilisés en clinique, cette protéine induit sélectivement la mort par apoptose de nombreuses cellules cancéreuses. Cette cytokine exerce son activité cytotoxique en se liant à des récepteurs transmembranaires exprimés à la surface de la cellule cible. Par un jeu d interactions protéiques, la fixation de TRAIL sur ces récepteurs agonistes (TRAIL-R1 et TRAIL-R2) conduit à l activation de l apoptose. L expression de deux récepteurs antagonistes, TRAIL-R3 et TRAIL-R4, par les cellules cancéreuses, permet aux cellules cibles d échapper à l apoptose induite par TRAIL. Nous montrons que ces deux récepteurs font intervenir des mécanismes moléculaires distincts. Leur expression pouvant potentiellement représenter un frein à l utilisation clinique de TRAIL, nous avons étudié l effet de la surexpression de l un d entre eux, TRAIL-R4 sur l efficacité des stratégies thérapeutiques associant TRAIL aux chimiothérapies conventionnelles. Les résultats obtenus montrent également que la résistance induite par TRAIL-R4 peut être contournée in vitro et in vivo en associant TRAIL à des agents chimiothérapeutiques. D un point de vue moléculaire, nous avons montré que la sensibilisation à TRAIL 1) implique une augmentation du recrutement et de l activation de la caspase-8 au sein du DISC de TRAIL, 2) ne nécessite pas la voie mitochondriale, et 3) est négativement régulée de manière coopérative par c-FLIP, un inhibiteur sélectif de la caspase-8. De manière intéressante, comme les anticorps agonistes actuellement testés en clinique, de petits peptides agonistes de TRAIL-R2, développés en collaboration avec une équipe de chimiste, permettent de contourner la résistance induite par TRAIL-R4, offrant des perspectives thérapeutiques intéressantes. Les récepteurs TRAIL-R3 et TRAIL-R4 sont donc des inhibiteurs de TRAIL. Nos travaux démontrent cependant, que les stratégies associant TRAIL à des agents chimiothérapeutiques, ou l'utilisation d'agonistes TRAIL-R2 permet de contourner la résistance induite par les récepteurs antagonistes de TRAIL et donc d éliminer ces cellules cancéreuses.TRAIL (TNF Related Apoptosis Inducing Ligand) is a very promising cytokine for cancer therapy. Contrary to current treatments, this protein is able to selectively kill cancer cells, whilst sparing healthy cells. TRAIL induces apoptosis following binding to one of its two different agonistic membrane receptors, TRAIL-R1 and TRAIL-R2. However, expression of one of its two antagonistic receptors, TRAIL-R3 and TRAIL-R4, on cancer cells can impair cancer cell killing by TRAIL. We have shown that these receptors inhibit TRAIL-induced cell death differentially. As these receptors can represent a brake for the use of TRAIL in cancer therapy, we investigated the effect of the expression of one of them, TRAIL-R4 on the efficacy of the different therapeutic strategies associating TRAIL and conventional therapeutic drugs. We show that acquired resistance to TRAIL following expression of TRAIL-R4 can be overcome in vitro and in vivo by combining TRAIL with chemotherapeutic agents. From a molecular point of view, we could demonstrate that sensitization to TRAIL 1) occurs mainly through an increase of caspase-8 recruitment and activation within the TRAIL DISC, 2) is independent of the mitochondrial pathway and 3) is negatively regulated, in a cooperative manner by c-FLIP, a caspase-8 selective inhibitor. Interestingly, like agonistic receptors currently tested in clinic, small agonistic peptides targeting TRAIL-R2, engineered in collaboration with a team of chemists, afford cancer cell killing regardless of TRAIL-R4 expression, providing novel therapeutic perspectives. TRAIL-R3 and TRAIL-R4 should thus be considered as TRAIL inhibitors. Our results demonstrate however that strategies aiming at combining TRAIL with chemotherapeutic agents or the use of TRAIL-R1 or TRAIL-R2 agonists could be effective treatments to eradicate cancer cells that express TRAIL antagonistic receptors.DIJON-BU Doc.électronique (212319901) / SudocSudocFranceF
    corecore