12 research outputs found

    Signalisation androgénique dans les cellules de Sertoli

    No full text
    Sertoli cells (SC) have essential roles in the androgen regulation of spermatogenesis, via the androgen receptor (AR)-mediated signaling. This work aimed at identifying the molecular mechanisms related to the androgenic regulation of the AR and its molecular partners in Sertoli cells during different testicular developmental stages. We first characterized and studied a novel murine mature immortalized Sertoli cell line, called ST38c, which harbors substantial expression of endogenous AR, conserves its androgen-dependent transcriptional activation and exhibits agonist-dependent transcriptional and posttranslational regulation, as well as posttranslational stability.We used this cellular model in order to test the hypothesis that anti MĂŒllerian Hormone (AMH) suppression in mature Sertoli cells would be directly androgen and AR mediated.Next, we hypothesized that the physiological androgen resistance in the neonate would also be related to the differential expression of several AR coregulators. Therefore, we analysed the differential expression and contribution of two AR co-regulators (SRC-2 and HBO1) during human testicular ontogeny, as well as in pathologies associated with androgen action or AR impairment (such as androgen insensitivity syndromes, congenital hypogonadotropic hypogonadism).Using in vitro transfection assays, we showed that SRC-2 is an AR coactivator while HBO1 is an AR corepressor in Sertoli cell models. We provided the cartography of SRC-2 and HBO1 expression during human testicular postnatal different stages and showed that SRC-2 presented a stable expression contrasting with the progressive evolution profile of the AR signaling, suggesting that Sertoli SRC-2 expression was independent of the androgen signaling. Interestingly, HBO1 and AR presented a temporal and positively correlated maturation profile, suggesting that HBO1 expression would be related to a functional AR signaling in the Sertoli cell. Moreover HBO1 expression is induced by androgens in the presence of the AR. Unlike SRC-2, HBO1 is not only expressed in Sertoli cells, but also in spermatogonia, being an interesting germ cell marker.Finally, we also studied AR and AMH immunoexpression in posptubertal cases of 5-α reductase type 2 deficiency and minimal androgen receptor resistance, in respect with the spermatogenesis status of seminiferous tubules, and androgen induced AMH suppression in order to assess the differential contribution of testosterone versus dihydrotestosterone and gather more information about the fertility perspectives in this particular pathologies.Les cellules de Sertoli (CS) jouent des rĂŽles essentiels pour la rĂ©gulation de la spermatogenĂšse, via la signalisation modulĂ©e par le rĂ©cepteur aux androgĂšnes (RA). Les objectifs de ce travail ont Ă©tĂ© d’identifier les mĂ©canismes molĂ©culaires de la rĂ©gulation androgĂ©nique et des rĂŽles des partenaires molĂ©culaires dans la rĂ©gulation androgĂ©nique des cellules de Sertoli pendant le dĂ©veloppement testiculaire, du fƓtus Ă  l’ñge adulte, pendant diffĂ©rentes stades du dĂ©veloppement. Nous avons caractĂ©risĂ© et Ă©tudiĂ© une nouvelle lignĂ©e immortalisĂ©e, mature, de CS, ST38c, prĂ©sentant une expression substantielle de RA endogĂšne, une activation transcriptionnelle du RA induite par les androgĂšnes, ainsi qu’une rĂ©gulation et une stabilisation de la protĂ©ine RA par des mĂ©canismes post traductionnels. Ce modĂšle a Ă©tĂ© utilisĂ© afin de tester l’hypothĂšse que la suppression de l’hormone antimĂŒllĂ©rienne (AMH) est modulĂ©e directement par les androgĂšnes, via la RA, dans les cellules matures de Sertoli.En parallĂšle, nous avons testĂ© l’hypothĂšse que la rĂ©sistance physiologique aux androgĂšnes du nouveau-nĂ© est liĂ©e Ă  l’expression diffĂ©rentielle de quelques corĂ©gulateurs du RA. Ainsi, nous avons analysĂ© l’expression et la contribution de deux corĂ©gulateurs du RA (SRC-2 et HBO1) pendant l’ontogenĂšse testiculaire humaine et pour des pathologies liĂ©es au disfonctionnement de l’action des androgĂšnes ou du RA (syndromes d’insensibilitĂ© aux androgĂšnes, hypogonadisme hypogonadotrophique congĂ©nital).Nous avons dĂ©montrĂ©, aprĂšs des essais de transfection in vitro, que SRC-2 est un coactivateur, alors que HBO1 est in corepresseur du RA dans un modĂšle de cellules de Sertoli. Nous avons cartographiĂ© l’expression testiculaire humaine de SRC-2 et HBO1, pendant diffĂ©rentes stades du dĂ©veloppement prĂ© et postnatal et nous avons dĂ©montrĂ© que SRC-2 prĂ©sente une expression stable, contrastant avec le profil d’expression diffĂ©rentielle, progressive avec l’ñge de RA dans la cellule de Sertoli, suggĂ©rant que l’expression du SRC-2 est indĂ©pendante de la signalisation androgĂšnique. Par contre, HBO1 et le RA prĂ©sentent un profil de maturation et d’expression temporelle corrĂ©lĂ©, suggĂ©rant que l’expression du HBO1 serait liĂ©e Ă  une signalisation du RA fonctionnelle dans les cellules de Sertoli. Nous avons aussi dĂ©montrĂ© que l’expression du HBO1 est induite par les androgĂšnes en prĂ©sence du RA. Contrairement au SRC-2, HBO1 est non seulement exprimĂ© dans les cellules de Sertoli, mais aussi dans les spermatogonies, pouvant reprĂ©senter un marqueur potentiellement intĂ©ressant des cellules germinales.Enfin, nous avons aussi Ă©tudiĂ© l’immuno-expression testiculaire de RA et AMH chez des patients post pubĂšres avec un syndrome de dĂ©ficit en 5α-rĂ©ductase type 2, et insensibilitĂ© minime aux androgĂšnes (MAIS), afin d’étudier la contribution des androgĂšnes (testostĂ©rone versus dihydrotĂ©stosterone) pour la spĂ©rmatogenĂšse, la rĂ©pression de l’AMH ainsi que pour mieux comprendre les perspectives de fertilitĂ© chez ces patients

    Signalisation androgénique dans les cellules de Sertoli

    No full text
    Les cellules de Sertoli (CS) jouent des rĂŽles essentiels pour la rĂ©gulation de la spermatogenĂšse, via la signalisation modulĂ©e par le rĂ©cepteur aux androgĂšnes (RA). Les objectifs de ce travail ont Ă©tĂ© d identifier les mĂ©canismes molĂ©culaires de la rĂ©gulation androgĂ©nique et des rĂŽles des partenaires molĂ©culaires dans la rĂ©gulation androgĂ©nique des cellules de Sertoli pendant le dĂ©veloppement testiculaire, du fƓtus Ă  l Ăąge adulte, pendant diffĂ©rentes stades du dĂ©veloppement. Nous avons caractĂ©risĂ© et Ă©tudiĂ© une nouvelle lignĂ©e immortalisĂ©e, mature, de CS, ST38c, prĂ©sentant une expression substantielle de RA endogĂšne, une activation transcriptionnelle du RA induite par les androgĂšnes, ainsi qu une rĂ©gulation et une stabilisation de la protĂ©ine RA par des mĂ©canismes post traductionnels. Ce modĂšle a Ă©tĂ© utilisĂ© afin de tester l hypothĂšse que la suppression de l hormone antimĂŒllĂ©rienne (AMH) est modulĂ©e directement par les androgĂšnes, via la RA, dans les cellules matures de Sertoli.En parallĂšle, nous avons testĂ© l hypothĂšse que la rĂ©sistance physiologique aux androgĂšnes du nouveau-nĂ© est liĂ©e Ă  l expression diffĂ©rentielle de quelques corĂ©gulateurs du RA. Ainsi, nous avons analysĂ© l expression et la contribution de deux corĂ©gulateurs du RA (SRC-2 et HBO1) pendant l ontogenĂšse testiculaire humaine et pour des pathologies liĂ©es au disfonctionnement de l action des androgĂšnes ou du RA (syndromes d insensibilitĂ© aux androgĂšnes, hypogonadisme hypogonadotrophique congĂ©nital).Nous avons dĂ©montrĂ©, aprĂšs des essais de transfection in vitro, que SRC-2 est un coactivateur, alors que HBO1 est in corepresseur du RA dans un modĂšle de cellules de Sertoli. Nous avons cartographiĂ© l expression testiculaire humaine de SRC-2 et HBO1, pendant diffĂ©rentes stades du dĂ©veloppement prĂ© et postnatal et nous avons dĂ©montrĂ© que SRC-2 prĂ©sente une expression stable, contrastant avec le profil d expression diffĂ©rentielle, progressive avec l Ăąge de RA dans la cellule de Sertoli, suggĂ©rant que l expression du SRC-2 est indĂ©pendante de la signalisation androgĂšnique. Par contre, HBO1 et le RA prĂ©sentent un profil de maturation et d expression temporelle corrĂ©lĂ©, suggĂ©rant que l expression du HBO1 serait liĂ©e Ă  une signalisation du RA fonctionnelle dans les cellules de Sertoli. Nous avons aussi dĂ©montrĂ© que l expression du HBO1 est induite par les androgĂšnes en prĂ©sence du RA. Contrairement au SRC-2, HBO1 est non seulement exprimĂ© dans les cellules de Sertoli, mais aussi dans les spermatogonies, pouvant reprĂ©senter un marqueur potentiellement intĂ©ressant des cellules germinales.Enfin, nous avons aussi Ă©tudiĂ© l immuno-expression testiculaire de RA et AMH chez des patients post pubĂšres avec un syndrome de dĂ©ficit en 5a-rĂ©ductase type 2, et insensibilitĂ© minime aux androgĂšnes (MAIS), afin d Ă©tudier la contribution des androgĂšnes (testostĂ©rone versus dihydrotĂ©stosterone) pour la spĂ©rmatogenĂšse, la rĂ©pression de l AMH ainsi que pour mieux comprendre les perspectives de fertilitĂ© chez ces patients.Sertoli cells (SC) have essential roles in the androgen regulation of spermatogenesis, via the androgen receptor (AR)-mediated signaling. This work aimed at identifying the molecular mechanisms related to the androgenic regulation of the AR and its molecular partners in Sertoli cells during different testicular developmental stages. We first characterized and studied a novel murine mature immortalized Sertoli cell line, called ST38c, which harbors substantial expression of endogenous AR, conserves its androgen-dependent transcriptional activation and exhibits agonist-dependent transcriptional and posttranslational regulation, as well as posttranslational stability.We used this cellular model in order to test the hypothesis that anti MĂŒllerian Hormone (AMH) suppression in mature Sertoli cells would be directly androgen and AR mediated.Next, we hypothesized that the physiological androgen resistance in the neonate would also be related to the differential expression of several AR coregulators. Therefore, we analysed the differential expression and contribution of two AR co-regulators (SRC-2 and HBO1) during human testicular ontogeny, as well as in pathologies associated with androgen action or AR impairment (such as androgen insensitivity syndromes, congenital hypogonadotropic hypogonadism).Using in vitro transfection assays, we showed that SRC-2 is an AR coactivator while HBO1 is an AR corepressor in Sertoli cell models. We provided the cartography of SRC-2 and HBO1 expression during human testicular postnatal different stages and showed that SRC-2 presented a stable expression contrasting with the progressive evolution profile of the AR signaling, suggesting that Sertoli SRC-2 expression was independent of the androgen signaling. Interestingly, HBO1 and AR presented a temporal and positively correlated maturation profile, suggesting that HBO1 expression would be related to a functional AR signaling in the Sertoli cell. Moreover HBO1 expression is induced by androgens in the presence of the AR. Unlike SRC-2, HBO1 is not only expressed in Sertoli cells, but also in spermatogonia, being an interesting germ cell marker.Finally, we also studied AR and AMH immunoexpression in posptubertal cases of 5-a reductase type 2 deficiency and minimal androgen receptor resistance, in respect with the spermatogenesis status of seminiferous tubules, and androgen induced AMH suppression in order to assess the differential contribution of testosterone versus dihydrotestosterone and gather more information about the fertility perspectives in this particular pathologies.PARIS11-SCD-Bib. Ă©lectronique (914719901) / SudocSudocFranceF

    : AR stabilization in a novel murine Sertoli cell line

    No full text
    Mature Sertoli cells (SC) are critical mediators of androgen regulation of spermatogenesis, via the androgen receptor (AR) signaling. Available immortalized SC lines loose AR expression or androgen responsiveness, hampering the study of endogenous AR regulation in SC. We have established and characterized a novel clonal mouse immortalized SC line, ST38c. These cells express some SC specific genes (sox9, wt1, tjp1, clu, abp, inhbb), but not fshr, yet more importantly, maintain substantial expression of endogenous AR as determined by PCR, immunocytochemistry, testosterone binding assays and Western blots. Microarrays allowed identification of some (146) but not all (rhox5, spinlw1), androgen-dependent, SC expressed target genes. Quantitative Real-Time PCR validated regulation of five up-regulated and two down-regulated genes. We show that AR undergoes androgen-dependent transcriptional activation as well as agonist-dependent posttranslational stabilization in ST38c cells. This cell line constitutes a useful experimental tool for future investigations on the molecular and cellular mechanisms of androgen receptor signaling in SC function

    Testicular histological and immunohistochemical aspects in a post-pubertal patient with 5 alpha-reductase type 2 deficiency: case report and review of the literature in a perspective of evaluation of potential fertility of these patients.

    Get PDF
    International audienceBACKGROUND: Testicular morphology and immunohistochemical studies have never been reported in genetically documented adult patients with 5 alpha-reductase type 2 deficiency (5α-R2 deficiency). CASE PRESENTATION: We describe the testicular histopathology of a 17-year-old XY subject with 5α-R2 deficiency caused by the recurrent homozygous Gly115Asp loss of function mutation of the SRD5A2 gene.We also performed an immunohistochemical analysis in order to further study the relationship between seminiferous tubules structure, Sertoli cell differentiation and androgenic signaling impairment in this case. We thus evaluated the testicular expression of the anti-MĂŒllerian hormone (AMH), androgen receptor (AR) and 3ÎČ-hydroxysteroid dehydrogenase (3ÎČHSD). Histological analysis revealed a heterogeneous aspect with a majority (92%) of seminiferous tubules (ST) presenting a mature aspect but containing only Sertoli cells and devoid of germ cells and spermatogenesis. Focal areas of immature ST (8%) were also found. Testicular AR and 3ÎČHSD expression were detected in adult male control, 5α-R2 deficiency and CAIS subjects. However, AMH expression was heterogeneous (detectable only in few AR negative prepubertal ST, but otherwise repressed) in the 5α-R2 deficiency, conversely to normal adult testis in which AMH was uniformly repressed and to an adult CAIS testis in which AMH was uniformly and strongly expressed. CONCLUSION: Intratesticular testosterone can repress AMH by itself, independently of its metabolism into dihydrotestosterone. We also compare our results to the few post pubertal cases of 5α-R2 deficiency with available histological testicular description, reported in the literature. We will discuss these histological findings, in the more general context of evaluating the fertility potential of these patients if they were raised as males and were azoospermic

    RADTHYR: an open-label, single-arm, prospective multicenter phase II trial of Radium-223 for the treatment of bone metastases from radioactive iodine refractory differentiated thyroid cancer

    No full text
    International audiencePurpose: This is the first prospective trial evaluating the efficacy of alpha emitter Radium-223 in patients with bone metastases from radioactive iodine (RAI) refractory (RAIR) differentiated thyroid cancer.Methods: RADTHYR is a multicenter, single-arm prospective Simon two-stage phase II trial (NCT02390934). The primary objective was to establish the efficacy of three administrations of 55 kBq/kg of Radium-223 by 18F-FDG PET/CT according to PERCIST criteria. Secondary objectives were to establish the efficacy of six administrations of Radium-223 by 18F-FDG PET/CT, 99mTc-HMDP bone scan and 18FNa PET/CT, clinical benefits, changes in serum bone markers, thyroglobulin levels, and safety.Results: Ten patients were enrolled between July 2015 and December 2017 (4 M; median age 74 years). Prior to Radium-223 administration, patients received a median RAI cumulative activity of 15 GBq (7.4-35.6), external radiation therapy (n = 9), bone surgery (n = 8), cimentoplasty (n = 5), and cryoablation (n = 2). 18F-FDG PET/CT showed stable disease (SD) in 4/10 and progressive disease (PD) in 6/10 cases after three administrations and SD in 4/10, PD in 5/10 cases, and 1/10 non-evaluable (NE) case after six administrations. After six injections, 99mTc-HMDP bone scan showed SD in 9 cases and was NE in 1 case; 18FNa PET/CT showed SD in 8 cases, partial response (PR) in 1 case, and was NE in 1 case. No significant clinical benefits were reported during the study. A skeletal event occurred in 6 patients (median time without skeletal event of 12.1 months). Seventy-seven adverse events were reported during treatment (7 of grade 3-4). Three patients developed an acute myeloid, a promyelocytic, and a chronic myeloid leukemia after the last Radium-223 administration considered as drug-related.Conclusion: The trial was stopped after interim analysis for lack of response of bone metastases from RAIR thyroid cancer to Radium-223. Severe hematological toxicity was observed in patients heavily pretreated with RAI and external radiation

    The cholesterol-derived metabolite dendrogenin A functionally reprograms breast adenocarcinoma and undifferentiated thyroid cancer cells

    No full text
    International audienceDendrogenin A (DDA) is a tumor suppressor mammalian cholesterol-derived metabolite and a new class of ligand of the Liver X receptor (LXR), which displays tumor cell differentiation. In human MCF7 breast adenocarcinoma cells, DDA-induced cell differentiation was associated with an increased accumulation of neutral lipids and proteins found in milk indicating that DDA re-activates some functions of lactating cells. Active iodide transport occurs in the normal lactating mammary cells through the sodium/iodide symporter (NIS) and iodide (I) is secreted into milk to be used by the nursing newborn for thyroid hormones biosynthesis. In the present study, we assessed whether DDA may induce other characteristic of lactating cells such as NIS expression and iodine uptake in MCF7 breast cancer cells and extended this study to the papillary B-CPAP and undifferentiated anaplastic 8505c thyroid cancer cells. Moreover, we evaluated DDA impact on the expression of thyroid specific proteins involved in thyroid hormone biogenesis. We report here that DDA induces NIS expression in MCF7 cells and significantly increases the uptake of 131-I by acting through the LXR. In addition, DDA induces phenotypic, molecular and functional characteristics of redifferentiation in the two human thyroid carcinoma cell lines and the uptake of 131-I in the undifferentiated 8505c cells was associated with a strong expression of all the specific proteins involved in thyroid hormone biosynthesis, TSH receptor, thyroperoxidase and thyroglobulin. 131-I incorporation in the 8505c cells was stimulated by DDA as well as by the synthetic LXR ligand, GW3965. Together these data show that the re-differentiation of breast and thyroid cancer cells by DDA, is associated with the recovery of functional NIS expression and involves an LXR-dependent mechanism. These results open new avenues of research for the diagnosis of thyroid cancers as well as the development of new therapeutic approaches for radioiodine refractory thyroid cancers

    The Cholesterol-5,6-Epoxide Hydrolase: A Metabolic Checkpoint in Several Diseases

    No full text
    International audienceCholesterol-5,6-epoxides (5,6-ECs) are oxysterols (OS) that have been linked to several pathologies including cancers and neurodegenerative diseases. 5,6-ECs can be produced from cholesterol by several mechanisms including reactive oxygen species, lipoperoxidation, and cytochrome P450 enzymes. 5,6-ECs exist as two different diastereoisomers: 5,6α-EC and 5,6ÎČ-EC with different metabolic fates. They can be produced as a mixture or as single products of epoxidation. The epoxide ring of 5,6α-EC and 5,6ÎČ-EC is very stable and 5,6-ECs are prone to hydration by the cholesterol-5,6-epoxide hydrolase (ChEH) to give cholestane-3ÎČ,5α,6ÎČ-triol, which can be further oxidized into oncosterone. 5,6α-EC is prone to chemical and enzymatic conjugation reactions leading to bioactive compounds such as dendrogenins, highlighting the existence of a new metabolic branch on the cholesterol pathway centered on 5,6α-EC. We will summarize in this chapter current knowledge on this pathway which is controlled by the ChEH

    Clinical outcomes 1 year after empiric 131I therapy for hyperthyroid disorders

    No full text
    International audienceRadioiodine is a therapeutic option in Europe for Graves' disease (GD) and toxic multinodular goiter (MNG).PURPOSE:To compare empiric and calculated I activities using 2013 EANM recommendations. To look for predictive factors of therapeutic response to an empiric activity of I. To assess clinical situations favoring calculated treatment modalities.PATIENTS AND METHODS:Prospective monocentric study of clinical outcomes at 1 year follow-up in 86 patients with GD and MNG who received empiric I therapeutic activities (348-939 MBq). Differences between empiric and calculated activities were confronted to clinical outcomes. Physicians were not aware of the calculated activity at the time of prescription.RESULTS:One year after treatment, 9% (5/57) of GD patients and 7% (2/29) of MNG patients were still in a hyperthyroid state. Thyroid volume was reduced by 67% for GD and by 50% for MNG. In GD, empiric I activities were higher than calculated ones (564±131 vs. 316±319 MBq, P<0.001) in 93% (53/57) of patients. Pretherapeutic thyroid volume (>26 ml for GD; >40 ml for MNG) was associated with persistent hyperthyroidism.CONCLUSION:Empirically administered I for GD and MNG was associated with very high efficacy in thyroid function control and no side effects. Thyroid volume reduction did not preclude treatment efficacy. Activity calculation could be a useful method for treating patients with GD and thyroid volumes higher than 26 ml or patients with MNG and thyroid volumes higher than 40 ml. A selective approach based on pretherapeutic thyroid volume and radioiodine biokinetics might improve treatment success

    Insertion of synthetic lesions on patient data: a method for evaluating clinical performance differences between PET systems

    No full text
    Abstract Background Performance assessment of positron emission tomography (PET) scanners is crucial to guide clinical practice with efficiency. We have already introduced and experimentally evaluated a simulation method allowing the creation of a controlled ground truth for system performance assessment. In the current study, the goal was to validate the method using patient data and demonstrate its relevance to assess PET performances accuracy in clinical conditions. Methods Twenty-four patients were recruited and sorted into two groups according to their body mass index (BMI). They were administered with a single dose of 2 MBq/kg 18F-FDG and scanned using clinical protocols consecutively on two PET systems: the Discovery-IQ (DIQ) and the Discovery-MI (DMI). For each BMI group, sixty synthetic lesions were dispatched in three subgroups and inserted at relevant anatomical locations. Insertion of synthetic lesions (ISL) was performed at the same location into the two consecutive exams. Two nuclear medicine physicians evaluated individually and blindly the images by qualitatively and semi-quantitatively reporting each detected lesion and agreed on a consensus. We assessed the inter-system detection rates of synthetic lesions and compared it to an initial estimate of at least 1.7 more targets detected on the DMI and the detection rates of natural lesions. We determined the inter-reader variability, evaluated according to the inter-observer agreement (IOA). Adequate inter-reader variability was found for IOA above 80%. Differences in standardized uptake value (SUV) metrics were also studied. Results In the BMI ≀ 25 group, the relative true positive rate (RTPR) for synthetic and natural lesions was 1.79 and 1.83, respectively. In the BMI > 25 group, the RTPR for synthetic and natural lesions was 2.03 and 2.27, respectively. For each BMI group, the detection rate using ISL was consistent to our estimate and with the detection rate measured on natural lesions. IOA above 80% was verified for any scenario. SUV metrics showed a good agreement between synthetic and natural lesions. Conclusions ISL proved relevant to evaluate performance differences between PET scanners. Using these synthetically modified clinical images, we can produce a controlled ground truth in a realistic anatomical model and exploit the potential of PET scanner for clinical purposes
    corecore