7 research outputs found

    Androgen-dependent apoptosis in male germ cells is regulated through the proto-oncoprotein Cbl

    Get PDF
    The proto-oncoprotein Cbl is known to control several signaling processes. It is highly expressed in the testis, and because spermatogenesis is androgen dependent, we investigated the androgen dependency expression of Cbl through its testicular sublocalization and its expression levels in rats that were exposed to the antiandrogen flutamide or were hypophysectomized. We report the androgen dependency of Cbl as it localizes in pachytene spermatocytes during androgen-dependent stages, is down-regulated upon flutamide exposure, and is up-regulated with testosterone in hypophysectomized rats. Coculture experiments showed the key control exerted by the Sertoli cell on Cbl activity. As flutamide induces germ cell apoptosis, we investigate members of the Bcl-2 family upon flutamide exposure. We show that the proapoptotic Bcl-2 family member Bim mirrored Cbl expression through a posttranscriptional process. We also show that in Cbl knockout mouse testes, the imbalance between the high expression of Bim and Smac/Diablo and antiapoptotic factors such as cellular inhibitor of apoptosis 2 favors a survival process, which makes these mice unresponsive to androgen withdrawal and could explain their hypofertility

    A protective role against oxidative stress for the E3-ubiquitin ligase c-Cbl : usefulness as a prognostic marker for carcinomas

    No full text
    Le travail prĂ©sentĂ© a portĂ© sur l’analyse in vivo du proto-oncogĂšne c-cbl, dont la forme connue est c-Cbl (p120cbl). Il s’agit d’une E3-Ubiquitine ligase et un poly-adaptateur molĂ©culaire. Nous avons montrĂ© l’androgĂ©no-dĂ©pendance de l’expression de c-Cbl dans les cellules germinales testiculaires et les cellules Ă©pithĂ©liales de la prostate de rats et de souris. Nous avons montrĂ© la rĂ©gulation anti-apoptotique exercĂ©e in vivo par la c-Cbl dans la prostate par comparaison des souris c-cbl invalidĂ©es ou non pour c-cbl (KO ou WT). L’effet exercĂ© par c-cbl dans le testicule est pro-apoptotique (J.Cell Biol, 2005), que nous avons ultĂ©rieurement attribuĂ© Ă  une nouvelle isoforme testiculaire de c-Cbl (Δ-c-Cbl). La comparaison des MEF KO et WT aprĂšs induction d’apoptose par l’étoposide, a confortĂ© l’effet anti-apoptotique exercĂ©e in vivo par c-Cbl dans la prostate. Elle a aussi montrĂ© la forte apoptose des MEF KO au peroxyde d’hydrogĂšne : c-Cbl peut ĂȘtre considĂ©rĂ©e comme un protecteur du stress oxydant. L’intensitĂ© du stress oxydant associĂ© aux cancers et leur forte rĂ©sistance Ă  l’apoptose sont des propriĂ©tĂ©s qui pourraient ĂȘtre reliĂ©es Ă  c-Cbl. L’analyse in situ effectuĂ©e Ă  partir de tumeurs congelĂ©es et de Tissue Microarrays (TMA) a montrĂ© une expression Ă©levĂ©e de c-Cbl dans certains cancers, dont l’intensitĂ© pourrait correspondre Ă  la gravitĂ© de l’atteinte anatomo-pathologique. La protĂ©ine c-Cbl est apparue ĂȘtre un marqueur d’agressivitĂ© du cancer de la prostate, probablement de l’ovaire, de l’utĂ©rus, du cerveau, du poumon, du colon et du rectum. Nous la considĂ©rons aussi comme une cible thĂ©rapeutique car, protecteur du stress oxydant, elle prendrait part Ă  la rĂ©sistance Ă  l’apoptose des cellules tumorales. Un brevet a Ă©tĂ© dĂ©posĂ© (2009, co-inventeurs : S.Yakoub et al). Un article rapportant ces rĂ©sultats est en cours de soumission (S. Yakoub et al)This work has focused on the in vivo analysis of the proto-oncogene c-cbl, coding for c-Cbl (p120cbl). We demonstrated the androgen-dependency of c-Cbl in the testicular germ cells and the prostatic epithelial cells of rats and mice. We then identified the anti-apoptotic regulation exerted by p120cbl in the prostate, comparing mouse c-cbl KO and WT, unlike this exerted in the testis (J.Cell Biol, 2005). We reported this difference to the high expression in testis of a new c-Cbl isoform, Δc-Cbl. The comparison of MEF KO and WT allowed confirming the anti-apoptotic regulation to etoposide exerted by c-Cbl. A very high apoptotic effect was observed in MEF KO with H2O2: c-Cbl is a strong stress oxidative protector. Knowing the intensity of oxidative stress in several cancers and their particular resistance to apoptosis as well, the in situ analysis of these malignancies was made from frozen tumours and tissue microassays (TMA). c-Cbl was indeed highly expressed and its intensity appears to reflect the aggressiveness of the pathology. c-Cbl could then be considered as a marker of severity of prostate cancer but probably also ovary, uterus, brain, lung, colon and rectum. It can also be considered as a therapeutic target involved in resistance to apoptosis as a stress oxidative protector. A patent was filed in the United States (2009, co-inventors: S. Yakoub et al

    Down-regulation of DcR2 sensitizes androgen-dependent prostate cancer LNCaP cells to TRAIL-induced apoptosis.

    Get PDF
    International audienceUNLABELLED: ABSTRACT: BACKGROUND: Dysregulation of many apoptotic related genes and androgens are critical in the development, progression, and treatment of prostate cancer. The differential sensitivity of tumour cells to TRAIL-induced apoptosis can be mediated by the modulation of surface TRAIL receptor expression related to androgen concentration. Our previous results led to the hypothesis that downregulation of TRAIL-decoy receptor DcR2 expression following androgen deprivation would leave hormone sensitive normal prostate cells vulnerable to the cell death signal generated by TRAIL via its pro-apoptotic receptors. We tested this hypothesis under pathological conditions by exploring the regulation of TRAIL-induced apoptosis related to their death and decoy receptor expression, as also to hormonal concentrations in androgen-sensitive human prostate cancer, LNCaP, cells. RESULTS: In contrast to androgen-insensitive PC3 cells, decoy (DcR2) and death (DR5) receptor protein expression was correlated with hormone concentrations and TRAIL-induced apoptosis in LNCaP cells. Silencing of androgen-sensitive DcR2 protein expression by siRNA led to a significant increase in TRAIL-mediated apoptosis related to androgen concentration in LNCaP cells. CONCLUSIONS: The data support the hypothesis that hormone modulation of DcR2 expression regulates TRAIL-induced apoptosis in LNCaP cells, giving insight into cell death induction in apoptosis-resistant hormone-sensitive tumour cells from prostate cancer. TRAIL action and DcR2 expression modulation are potentially of clinical value in advanced tumour treatment
    corecore