27 research outputs found

    Glucocorticoids promote breast cancer metastasis

    Get PDF
    Diversity within or between tumours and metastases (known as intra-patient tumour heterogeneity) that develops during disease progression is a serious hurdle for therapy(1-3). Metastasis is the fatal hallmark of cancer and the mechanisms of colonization, the most complex step in the metastatic cascade(4), remain poorly defined. A clearer understanding of the cellular and molecular processes that underlie both intra-patient tumour heterogeneity and metastasis is crucial for the success of personalized cancer therapy. Here, using transcriptional profiling of tumours and matched metastases in patient-derived xenograft models in mice, we show cancer-site-specific phenotypes and increased glucocorticoid receptor activity in distant metastases. The glucocorticoid receptor mediates the effects of stress hormones, and of synthetic derivatives of these hormones that are used widely in the clinic as anti-inflammatory and immunosuppressive agents. We show that the increase in stress hormones during breast cancer progression results in the activation of the glucocorticoid receptor at distant metastatic sites, increased colonization and reduced survival. Our transcriptomics, proteomics and phospho-proteomics studies implicate the glucocorticoid receptor in the activation of multiple processes in metastasis and in the increased expression of kinase ROR1, both of which correlate with reduced survival. The ablation of ROR1 reduced metastatic outgrowth and prolonged survival in preclinical models. Our results indicate that the activation of the glucocorticoid receptor increases heterogeneity and metastasis, which suggests that caution is needed when using glucocorticoids to treat patients with breast cancer who have developed cancer-related complications.Peer reviewe

    PI3K inhibition circumvents resistance to SHP2 blockade in metastatic triple-negative breast cancer.

    Get PDF
    The protein tyrosine phosphatase SHP2 activates oncogenic pathways downstream of most receptor tyrosine kinases (RTK) and has been implicated in various cancer types, including the highly aggressive subtype of triple-negative breast cancer (TNBC). Although allosteric inhibitors of SHP2 have been developed and are currently being evaluated in clinical trials, neither the mechanisms of the resistance to these agents, nor the means to circumvent such resistance have been clearly defined. The PI3K signaling pathway is also hyperactivated in breast cancer and contributes to resistance to anticancer therapies. When PI3K is inhibited, resistance also develops for example via activation of RTKs. We therefore assessed the effect of targeting PI3K and SHP2 alone or in combination in preclinical models of metastatic TNBC. In addition to the beneficial inhibitory effects of SHP2 alone, dual PI3K/SHP2 treatment decreased primary tumor growth synergistically, blocked the formation of lung metastases, and increased survival in preclinical models. Mechanistically, transcriptome and phospho-proteome analyses revealed that resistance to SHP2 inhibition is mediated by PDGFRβ-evoked activation of PI3K signaling. Altogether, our data provide a rationale for co-targeting of SHP2 and PI3K in metastatic TNBC

    Involvement of netrin-1 dependence receptors in colorectal tumorigenesis

    No full text
    Les récepteurs à dépendance ont la particularité d’induire deux signalisations différentes en fonction de la disponibilité de leur ligand. En présence de leur ligand, ils initient une signalisation positive favorisant notamment la survie cellulaire tandis qu’en leur absence, ils induisent la mort de la cellule par apoptose. Ils créent donc un état de dépendance de la cellule vis-à-vis du ligand pour survivre. Cette dualité fonctionnelle leur confère un rôle central dans le maintien de l’homéostasie tissulaire chez l’adulte et particulièrement dans l’intestin et le colon. En ce sens, l’apoptose induite par les récepteurs à dépendance serait un mécanisme de sauvegarde limitant l’échappement tumoral en permettant l’élimination des cellules proliférant ou migrant dans un environnement dépourvu d’une quantité suffisante de ligand. Un avantage sélectif potentiel pour une cellule tumorale serait donc de perdre cette induction de la mort cellulaire, soit via l’altération des récepteurs soit via un gain excessif d’expression du ligand. Ainsi, les travaux présentés montrent qu’un gain d’expression du ligand netrine-1 par la cellule tumorale elle-même caractérisée une fraction des tumeurs colorectales dérivées de pathologies inflammatoires chroniques et que la perte de l’expression des récepteurs à dépendance fixant la nétrine- 1 est observée dans le reste des cancers colorectaux. En particulier, nous avons montré que l’expression du gène qui code pour le récepteur de la netrin-1 UNC5C est altérée par méthylation dans 70% des cancers colorectaux, et que ce gène comporte également des mutations ponctuelles germinales dans des formes familiales de cancer colorectal, chez lesquels aucune autre mutation n’avait été décrite. Nous avons ainsi montré que les récepteurs à dépendance agissent comme des suppresseurs de tumeur conditionnels à l’absence de nétrine-1 et que l’altération de leur signalisation apoptotique constitue un avantage sélectif qui favorise la progression des cancers colorectauxDependence receptors share the common property of inducing two types of signaling according to their ligand availability. In the presence of their ligand, they induce a positive signal allowing cell survival whereas in its absence, they induce cell death by apoptosis. These receptors thus create cellular states of dependence on the ligands. Dependence receptors dual functionality plays a major role both during the embryonic development and in the regulation of tissue homeostasis in adult, particularly in intestine and colon. Indeed, apoptosis induced by dependence receptors would be a safeguard mechanism that restricts tumor escape by eliminating cells that would proliferate or migrate in settings of limited ligand availability. A potential selective advantage for a tumor cell would then be to loose this apoptotic signaling, either through a loss of the receptor apoptotic function or through an autocrine gain of the ligand expression. We show here than an over-expression of the ligand netrin-1 by a tumoral cell characterize a fraction of colorectal tumor derived from chronic inflammatory pathology, while the others tumors have lost the expression of netrin-1 dependence receptors. In particular, we have shown that the expression of UNC5C gene is down-regulated due to methylation of the promoter in 70% of cases, and different variants of the gene are associated with a familial form of colorectal cancer. Dependence receptors behave as conditional tumor suppressors in absence of their ligand, and the alteration of their apoptotic signaling is a selective advantage for tumoral cells and favors cancer progressio

    Involvement of netrin-1 dependence receptors in colorectal tumorigenesis

    No full text
    Les récepteurs à dépendance ont la particularité d’induire deux signalisations différentes en fonction de la disponibilité de leur ligand. En présence de leur ligand, ils initient une signalisation positive favorisant notamment la survie cellulaire tandis qu’en leur absence, ils induisent la mort de la cellule par apoptose. Ils créent donc un état de dépendance de la cellule vis-à-vis du ligand pour survivre. Cette dualité fonctionnelle leur confère un rôle central dans le maintien de l’homéostasie tissulaire chez l’adulte et particulièrement dans l’intestin et le colon. En ce sens, l’apoptose induite par les récepteurs à dépendance serait un mécanisme de sauvegarde limitant l’échappement tumoral en permettant l’élimination des cellules proliférant ou migrant dans un environnement dépourvu d’une quantité suffisante de ligand. Un avantage sélectif potentiel pour une cellule tumorale serait donc de perdre cette induction de la mort cellulaire, soit via l’altération des récepteurs soit via un gain excessif d’expression du ligand. Ainsi, les travaux présentés montrent qu’un gain d’expression du ligand netrine-1 par la cellule tumorale elle-même caractérisée une fraction des tumeurs colorectales dérivées de pathologies inflammatoires chroniques et que la perte de l’expression des récepteurs à dépendance fixant la nétrine- 1 est observée dans le reste des cancers colorectaux. En particulier, nous avons montré que l’expression du gène qui code pour le récepteur de la netrin-1 UNC5C est altérée par méthylation dans 70% des cancers colorectaux, et que ce gène comporte également des mutations ponctuelles germinales dans des formes familiales de cancer colorectal, chez lesquels aucune autre mutation n’avait été décrite. Nous avons ainsi montré que les récepteurs à dépendance agissent comme des suppresseurs de tumeur conditionnels à l’absence de nétrine-1 et que l’altération de leur signalisation apoptotique constitue un avantage sélectif qui favorise la progression des cancers colorectauxDependence receptors share the common property of inducing two types of signaling according to their ligand availability. In the presence of their ligand, they induce a positive signal allowing cell survival whereas in its absence, they induce cell death by apoptosis. These receptors thus create cellular states of dependence on the ligands. Dependence receptors dual functionality plays a major role both during the embryonic development and in the regulation of tissue homeostasis in adult, particularly in intestine and colon. Indeed, apoptosis induced by dependence receptors would be a safeguard mechanism that restricts tumor escape by eliminating cells that would proliferate or migrate in settings of limited ligand availability. A potential selective advantage for a tumor cell would then be to loose this apoptotic signaling, either through a loss of the receptor apoptotic function or through an autocrine gain of the ligand expression. We show here than an over-expression of the ligand netrin-1 by a tumoral cell characterize a fraction of colorectal tumor derived from chronic inflammatory pathology, while the others tumors have lost the expression of netrin-1 dependence receptors. In particular, we have shown that the expression of UNC5C gene is down-regulated due to methylation of the promoter in 70% of cases, and different variants of the gene are associated with a familial form of colorectal cancer. Dependence receptors behave as conditional tumor suppressors in absence of their ligand, and the alteration of their apoptotic signaling is a selective advantage for tumoral cells and favors cancer progressio

    Inhibition of endothelial cell apoptosis by Netrin-1 during angiogenesis

    Get PDF
    International audienceNetrin-1 was recently proposed to play an important role in embryonic and pathological angiogenesis. However, data reported led to the apparently contradictory conclusions that netrin-1 is either a pro- or an antiangiogenic factor. Here, we reconcile these opposing observations by demonstrating that netrin-1 acts as a survival factor for endothelial cells, blocking the proapoptotic effect of the dependence receptor UNC5B and its downstream death signaling effector, the serine/threonine kinase DAPK. The netrin-1 effect on blood vessel development is mimicked by caspase inhibitors in ex vivo assays, and the inhibition of caspase activity, the silencing of the UNC5B receptor, and the silencing of DAPK are each sufficient to rescue the vascular sprouting defects induced by netrin-1 silencing in zebrafish. Thus, the proapoptotic effect of unbound UNC5B, and the survival effect of netrin-1 on endothelial cells finely tune the angiogenic process

    Dependence receptor TrkC is a putative colon cancer tumor suppressor

    No full text
    The TrkC neurotrophin receptor belongs to the functional dependence receptor family, members of which share the ability to induce apoptosis in the absence of their ligands. Such a trait has been hypothesized to confer tumor-suppressor activity. Indeed, cells that express these receptors are thought to be dependent on ligand availability for their survival, a mechanism that inhibits uncontrolled tumor cell proliferation and migration. TrkC is a classic tyrosine kinase receptor and therefore generally considered to be a proto-oncogene. We show here that TrkC expression is down-regulated in a large fraction of human colorectal cancers, mainly through promoter methylation. Moreover, we show that TrkC silencing by promoter methylation is a selective advantage for colorectal cell lines to limit tumor cell death. Furthermore, reestablished TrkC expression in colorectal cancer cell lines is associated with tumor cell death and inhibition of in vitro characteristics of cell transformation, as well as in vivo tumor growth. Finally, we provide evidence that a mutation of TrkC detected in a sporadic cancer is a loss-of-proapoptotic function mutation. Together, these data support the conclusion that TrkC is a colorectal cancer tumor suppressor

    Dependence receptor TrkC is a putative colon cancer tumor suppressor

    No full text
    The TrkC neurotrophin receptor belongs to the functional dependence receptor family, members of which share the ability to induce apoptosis in the absence of their ligands. Such a trait has been hypothesized to confer tumor-suppressor activity. Indeed, cells that express these receptors are thought to be dependent on ligand availability for their survival, a mechanism that inhibits uncontrolled tumor cell proliferation and migration. TrkC is a classic tyrosine kinase receptor and therefore generally considered to be a proto-oncogene. We show here that TrkC expression is down-regulated in a large fraction of human colorectal cancers, mainly through promoter methylation. Moreover, we show that TrkC silencing by promoter methylation is a selective advantage for colorectal cell lines to limit tumor cell death. Furthermore, reestablished TrkC expression in colorectal cancer cell lines is associated with tumor cell death and inhibition of in vitro characteristics of cell transformation, as well as in vivo tumor growth. Finally, we provide evidence that a mutation of TrkC detected in a sporadic cancer is a loss-of-proapoptotic function mutation. Together, these data support the conclusion that TrkC is a colorectal cancer tumor suppressor

    Nuclear accumulation of MKL1 in luminal breast cancer cells impairs genomic activity of ERα and is associated with endocrine resistance

    No full text
    International audienceEstrogen receptor (ERα) is central in driving the development of hormone-dependent breast cancers. A major challenge in treating these cancers is to understand and overcome endocrine resistance. The Megakaryoblastic Leukemia 1 (MKL1, MRTFA) protein is a master regulator of actin dynamic and cellular motile functions, whose nuclear translocation favors epithelial-mesenchymal transition. We previously demonstrated that nuclear accumulation of MKL1 in estrogen-responsive breast cancer cell lines promotes hormonal escape. In the present study, we confirm through tissue microarray analysis that nuclear immunostaining of MKL1 is associated with endocrine resistance in a cohort of breast cancers and we decipher the underlining mechanisms using cell line models. We show through gene expression microarray analysis that the nuclear accumulation of MKL1 induces dedifferentiation leading to a mixed luminal/basal phenotype and suppresses estrogen-mediated control of gene expression. Chromatin immunoprecipitation of DNA coupled to high-throughput sequencing (ChIP-Seq) shows a profound reprogramming in ERα cistrome associated with a massive loss of ERα binding sites (ERBSs) generally associated with lower ERα-binding levels. Novel ERBSs appear to be associated with EGF and RAS signaling pathways. Collectively, these results highlight a major role of MKL1 in the loss of ERα transcriptional activity observed in certain cases of endocrine resistances, thereby contributing to breast tumor cells malignancy
    corecore