Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 14(12): 823, 2023 12 13.
Article in English | MEDLINE | ID: mdl-38092743

ABSTRACT

The molecular mechanisms induced by hypoxia are misunderstood in non-small cell lung cancer (NSCLC), and above all the hypoxia and RASSF1A/Hippo signaling relationship. We confirmed that human NSCLC (n = 45) as their brain metastases (BM) counterpart are hypoxic since positive with CAIX-antibody (target gene of Hypoxia-inducible factor (HIF)). A severe and prolonged hypoxia (0.2% O2, 48 h) activated YAP (but not TAZ) in Human Bronchial Epithelial Cells (HBEC) lines by downregulating RASSF1A/kinases Hippo (except for NDR2) regardless their promoter methylation status. Subsequently, the NDR2-overactived HBEC cells exacerbated a HIF-1A, YAP and C-Jun-dependent-amoeboid migration, and mainly, support BM formation. Indeed, NDR2 is more expressed in human tumor of metastatic NSCLC than in human localized NSCLC while NDR2 silencing in HBEC lines (by shRNA) prevented the xenograft formation and growth in a lung cancer-derived BM model in mice. Collectively, our results indicated that NDR2 kinase is over-active in NSCLC by hypoxia and supports BM formation. NDR2 expression is thus a useful biomarker to predict the metastases risk in patients with NSCLC, easily measurable routinely by immunohistochemistry on tumor specimens.


Subject(s)
Brain Neoplasms , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Humans , Mice , Brain Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Epithelial Cells/metabolism , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lung Neoplasms/pathology
2.
Target Oncol ; 17(4): 407-431, 2022 07.
Article in English | MEDLINE | ID: mdl-35906513

ABSTRACT

Malignant pleural mesothelioma is a rare and aggressive neoplasm, which has primarily been attributed to the exposure to asbestos fibers (83% of cases); yet, despite a ban of using asbestos in many countries, the incidence of malignant pleural mesothelioma failed to decline worldwide. While little progress has been made in malignant pleural mesothelioma diagnosis, bevacizumab at first, then followed by double immunotherapy (nivolumab plus ipilumumab), were all shown to improve survival in large phase III randomized trials. The morphological analysis of the histological subtyping remains the primary indicator for therapeutic decision making at an advanced disease stage, while a platinum-based chemotherapy regimen combined with pemetrexed, either with or without bevacizumab, is still the main treatment option. Consequently, malignant pleural mesothelioma still represents a significant health concern owing to poor median survival (12-18 months). Given this context, both diagnosis and therapy improvements require better knowledge of the molecular mechanisms underlying malignant pleural mesothelioma's carcinogenesis and progression. Hence, the Hippo pathway in malignant pleural mesothelioma initiation and progression has recently received increasing attention, as the aberrant expression of its core components may be closely related to patient prognosis. The purpose of this review was to provide a critical analysis of our current knowledge on these topics, the main focus being on the available evidence concerning the role of each Hippo pathway's member as a promising biomarker, enabling detection of the disease at earlier stages and thus improving prognosis.


Subject(s)
Asbestos , Lung Neoplasms , Mesothelioma, Malignant , Mesothelioma , Pleural Neoplasms , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/therapeutic use , Humans , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Mesothelioma/pathology , Pemetrexed/therapeutic use , Pleural Neoplasms/drug therapy , Pleural Neoplasms/pathology
3.
Int J Cancer ; 150(11): 1889-1904, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35262190

ABSTRACT

The Hippo pathway effector YAP is dysregulated in malignant pleural mesothelioma (MPM). YAP's target genes include the secreted growth factor amphiregulin (AREG), which is overexpressed in a wide range of epithelial cancers and plays an elusive role in MPM. We assayed the expression of YAP and AREG in MPM pathology samples and that of AREG additionally in plasma samples of patients from the randomized phase 3 IFCT-0701 Mesothelioma Avastin Cisplatin Pemetrexed Study (MAPS) using immunohistochemistry and ELISA assays, respectively. MPM patients frequently presented high levels of tumor AREG (64.3%), a high cytosolic AREG expression being predictive of a better prognosis with longer median overall and progression-free survival. Surprisingly, tumor AREG cytosolic expression was not correlated with secreted plasma AREG. By investigating the AREG metabolism and function in MPM cell lines H2452, H2052, MSTO-211H and H28, in comparison with the T47D ER+ breast cancer cell line used as a positive control, we confirm that AREG is important for cell invasion, growth without anchorage, proliferation and apoptosis in mesothelioma cells. Yet, most of these MPM cell lines failed to correctly execute AREG posttranslational processing by metalloprotease ADAM17/tumor necrosis factor-alpha-converting enzyme (TACE) and extracell secretion. The favorable prognostic value of high cytosolic AREG expression in MPM patients could therefore be sustained by default AREG posttranslational processing and release. Thus, the determination of mesothelioma cell AREG content could be further investigated as a prognostic marker for MPM patients and used as a stratification factor in future clinical trials.


Subject(s)
Lung Neoplasms , Mesothelioma, Malignant , Mesothelioma , Pleural Neoplasms , Amphiregulin/genetics , Cell Line, Tumor , Humans , Lung Neoplasms/genetics , Mesothelioma/metabolism , Pleural Neoplasms/drug therapy , Pleural Neoplasms/pathology
4.
Sci Rep ; 11(1): 11239, 2021 05 27.
Article in English | MEDLINE | ID: mdl-34045576

ABSTRACT

Lung cancer patients frequently develop brain metastases (BM). Despite aggressive treatment including neurosurgery and external-radiotherapy, overall survival remains poor. There is a pressing need to further characterize factors in the microenvironment of BM that may confer resistance to radiotherapy (RT), such as hypoxia. Here, hypoxia was first evaluated in 28 biopsies from patients with non­small cell lung cancer (NSCLC) BM, using CA-IX immunostaining. Hypoxia characterization (pimonidazole, CA-IX and HIF-1α) was also performed in different preclinical NSCLC BM models induced either by intracerebral injection of tumor cells (H2030-Br3M, H1915) into the cortex and striatum, or intracardial injection of tumor cells (H2030-Br3M). Additionally, [18F]-FMISO-PET and oxygen-saturation-mapping-MRI (SatO2-MRI) were carried out in the intracerebral BM models to further characterize tumor hypoxia and evaluate the potential of Hypoxia-image-guided-RT (HIGRT). The effect of RT on proliferation of BM ([18F]-FLT-PET), tumor volume and overall survival was determined. We showed that hypoxia is a major yet heterogeneous feature of BM from lung cancer both preclinically and clinically. HIGRT, based on hypoxia heterogeneity observed between cortical and striatal metastases in the intracerebrally induced models, showed significant potential for tumor control and animal survival. These results collectively highlight hypoxia as a hallmark of BM from lung cancer and the value of HIGRT in better controlling tumor growth.


Subject(s)
Brain Neoplasms/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Lung Neoplasms/diagnostic imaging , Radiotherapy, Image-Guided , Tumor Hypoxia , Aged , Animals , Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/radiotherapy , Carcinoma, Non-Small-Cell Lung/secondary , Cell Line, Tumor , Humans , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Magnetic Resonance Imaging , Middle Aged , Rats , Registries
5.
Biomed Res Int ; 2020: 2701345, 2020.
Article in English | MEDLINE | ID: mdl-32351987

ABSTRACT

By allowing insured communication between cancer cells themselves and with the neighboring stromal cells, tunneling nanotubes (TNTs) are involved in the multistep process of cancer development from tumorigenesis to the treatment resistance. However, despite their critical role in the biology of cancer, the study of the TNTs has been announced challenging due to not only the absence of a specific biomarker but also the fragile and transitory nature of their structure and the fact that they are hovering freely above the substratum. Here, we proposed to review guidelines to follow for studying the structure and functionality of TNTs in tumoral neuroendocrine cells (PC12) and nontumorigenic human bronchial epithelial cells (HBEC-3, H28). In particular, we reported how crucial is it (i) to consider the culture conditions (culture surface, cell density), (ii) to visualize the formation of TNTs in living cells (mechanisms of formation, 3D representation), and (iii) to identify the cytoskeleton components and the associated elements (categories, origin, tip, and formation/transport) in the TNTs. We also focused on the input of high-resolution cell imaging approaches including Stimulated Emission Depletion (STED) nanoscopy, Transmitted and Scanning Electron Microscopies (TEM and SEM). In addition, we underlined the important role of the organelles in the mechanisms of TNT formation and transfer between the cancer cells. Finally, new biological models for the identification of the TNTs between cancer cells and stromal cells (liquid air interface, ex vivo, in vivo) and the clinical considerations will also be discussed.


Subject(s)
Cell Communication , Electron Microscope Tomography , Microscopy, Electron, Scanning , Microtubules , Neoplasms , Animals , Humans , Microtubules/metabolism , Microtubules/ultrastructure , Neoplasms/metabolism , Neoplasms/ultrastructure , PC12 Cells , Rats
6.
Endocr Relat Cancer ; 26(6): 615-628, 2019 06.
Article in English | MEDLINE | ID: mdl-30978702

ABSTRACT

Breast cancer (BC) is the primary cause of cancer-related mortality among women. Patients who express the estrogen receptor (ER), which mediates the tumorigenic effects of estrogens, respond to antihormonal therapy. Loss of ER expression or acquired resistance to E2 is associated with aggressive malignant phenotypes, which lead to relapse. These BC subtypes overexpress syndecan-1 (SDC1), a transmembrane heparan sulfate proteoglycan that mediates angiogenesis as well as the proliferation and invasiveness of cancer cells. We showed here that the activation of ER-alpha (ERα) by estrogens induces downregulation of SDC1 expression in ER(+) MCF7 cells but not in T47D cells. Loss of ERα expression, induced by RNA interference or a selective ER downregulator, led to subsequent SDC1 overexpression. E2-dependent downregulation of SDC1 expression required de novo protein synthesis and was antagonized by treatment with BAY 11-7085, an irreversible inhibitor of IκBα phosphorylation, which inhibits the activation of NFκB. Downregulation of SDC1 expression required ERα and activation of IKK, but was independent to downstream transcriptional regulators of NFκB. BAY 11-7085 prevented E2-mediated phosphorylation of ERα on Ser118, increasing its proteasomal degradation, suggesting that IKK stabilized E2-activated ERα, leading to subsequent downregulation of SDC1 expression. Our results showed that sustained ER signaling inhibits SDC1 expression. Such antagonism elucidates the inverse correlation between SDC1 and ER expression in ER(+) BC as well as the overexpression of SDC1 in hormone receptor-negative BC subtypes with the most aggressive phenotypes. These results identify SDC1 as an attractive therapeutic target for BC as well as for other endocrine-associated cancers.


Subject(s)
Breast Neoplasms/metabolism , Cell Proliferation , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Gene Expression Regulation, Neoplastic , Syndecan-1/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Estrogen Receptor alpha/genetics , Female , Humans , Signal Transduction , Syndecan-1/metabolism , Tumor Cells, Cultured
7.
J Exp Clin Cancer Res ; 38(1): 158, 2019 Apr 12.
Article in English | MEDLINE | ID: mdl-30979377

ABSTRACT

BACKGROUND: RASSF1A, a tumor suppressor gene, is frequently inactivated in lung cancer leading to a YAP-dependent epithelial-mesenchymal transition (EMT). Such effects are partly due to the inactivation of the anti-migratory RhoB GTPase via the inhibitory phosphorylation of GEF-H1, the GDP/GTP exchange factor for RhoB. However, the kinase responsible for RhoB/GEF-H1 inactivation in RASSF1A-depleted cells remained unknown. METHODS: NDR1/2 inactivation by siRNA or shRNA effects on epithelial-mesenchymal transition, invasion, xenograft formation and growth in SCID-/- Beige mice, apoptosis, proliferation, cytokinesis, YAP/TAZ activation were investigated upon RASSF1A loss in human bronchial epithelial cells (HBEC). RESULTS: We demonstrate here that depletion of the YAP-kinases NDR1/2 reverts migration and metastatic properties upon RASSF1A loss in HBEC. We show that NDR2 interacts directly with GEF-H1 (which contains the NDR phosphorylation consensus motif HXRXXS/T), leading to GEF-H1 phosphorylation. We further report that the RASSF1A/NDR2/GEF-H1/RhoB/YAP axis is involved in proper cytokinesis in human bronchial cells, since chromosome proper segregation are NDR-dependent upon RASSF1A or GEF-H1 loss in HBEC. CONCLUSION: To summarize, our data support a model in which, upon RASSF1A silencing, NDR2 gets activated, phosphorylates and inactivates GEF-H1, leading to RhoB inactivation. This cascade induced by RASSF1A loss in bronchial cells is responsible for metastasis properties, YAP activation and cytokinesis defects.


Subject(s)
Cell Movement/genetics , Cytokinesis/genetics , Gene Silencing , Genes, Suppressor , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/genetics , Animals , Biomarkers, Tumor , Cell Cycle Proteins , Cell Line, Tumor , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Heterografts , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Mice , Neoplasm Metastasis , Nuclear Proteins/metabolism , Phenotype , Phosphorylation , Prognosis , Transcription Factors/metabolism , rhoB GTP-Binding Protein/metabolism
8.
Mol Cell Endocrinol ; 476: 185-197, 2018 11 15.
Article in English | MEDLINE | ID: mdl-29753772

ABSTRACT

The loss of estrogen receptor α (ERα) expression in breast cancer constitutes a major hallmark of tumor progression to metastasis and is generally correlated to a strong increase in Hyaluronic Acid (HA) turnover. The aim of our study was to search for a putative link between these two major events of breast cancer progression in the estrogen receptor-positive (ER+) MCF7 breast cancer cell line. The increase in HA turnover was performed by stable overexpression of the standard CD44 (CD44S) isoform and also by treatment with exogenous Hyaluronidase (Hyal) to allow an increase in HA catabolism. Stable overexpression of CD44S in MCF7 cells was correlated to a decrease in ESR1 gene expression, which did not lead to alteration of estrogen response. Moreover, our results showed that the exposure to exogenous Hyal stimulates the proliferation and strongly decreases the expression of ERα whatever the expression level of CD44 in the MCF7 cell line. The culture in the presence of Hyal led to the decrease in estrogens responsiveness and to hormonal therapy resistance. The effect on growth is correlated to the activation of MAPK/ERK and PI3K/Akt signaling pathways while the Hyal-induced down-regulation of ESR1 gene expression involves the activation of PI3K/Akt and NF-κB signaling pathways. Many of our data suggest that the effects of Hyal described here could be related to the activation of TLR signaling. Taken together, our results demonstrate that the increase in HA degradation could be involved in breast cancer progression and in resistance to hormonal therapy.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Hyaluronic Acid/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inflammation/pathology , MCF-7 Cells , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Toll-Like Receptors/metabolism
9.
Reproduction ; 153(6): 797-808, 2017 06.
Article in English | MEDLINE | ID: mdl-28348070

ABSTRACT

Albeit devoid of intrinsic catalytic activity, the transmembrane heparan sulphate proteoglycan syndecan 1 plays critical roles in cellular processes such as extracellular matrix crosstalk, cytoskeletal organization, cell spreading, proliferation and differentiation. During the ovarian cycle, the expression of syndecan 1 in granulosa cells shows cyclic variation suggesting that it might fulfil specific roles in follicle development. To investigate its physiological roles on granulosa cells, syndecan 1 was overexpressed in human granulosa cell line KGN which retains features of granulosa cells from small antral follicle such as estradiol (E2) synthesis and low expression of functional FSH receptor (FSHR). We demonstrated that overexpression of syndecan 1 in immature granulosa cells (KGN-SDC1) induces a profound alteration in their intrinsic characteristics including enhanced spreading and attachment, both associated with a reduced growth rate. Flow cytometry analysis revealed that syndecan 1 overexpression increases the percentage of KGN cells in quiescent phase. This partial cell cycle exit is concordant with downregulated levels of CCND1 and CDK4 and upregulated expression of CDK inhibitor CDKN1A In parallel both unstimulated and FSH-induced E2 synthesis are reduced in KGN-SDC1 through both repression of CYP19A1 and FSHR mRNA associated with decreased levels of potential regulators NR5A1 and ESR2 Additionally, we provide evidence that transient cAMP accumulation reduction in cells overexpressing syndecan 1 is accompanied by an increase in cAMP-hydrolysing PDE activity. Our results demonstrated that syndecan 1 might regulate differentiation of granulosa cells and follicular development by means of various mechanisms involving morphological changes, control of signalling pathways and alterations in gene expressions.Free French abstract: A French translation of this abstract is freely available at http://www.reproduction-online.org/content/153/6/797/suppl/DC1.Reproduction.


Subject(s)
Cell Proliferation/drug effects , Follicle Stimulating Hormone/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Granulosa Cell Tumor/pathology , Syndecan-1/metabolism , Aromatase/metabolism , Cell Cycle/drug effects , Cell Differentiation/drug effects , Female , Granulosa Cell Tumor/drug therapy , Granulosa Cell Tumor/metabolism , Humans , Middle Aged , Receptors, FSH/metabolism , Signal Transduction , Tumor Cells, Cultured
10.
Biochim Biophys Acta ; 1830(6): 3466-75, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23500014

ABSTRACT

BACKGROUND: During the pre-pubertal life, the cessation of Sertoli cell proliferation and the onset of differentiation are associated with a shift in the FSH-mediated signaling leading to inhibition of the ERK-mitogenic pathway and to a concomitant sensitization of cAMP/PKA pathway. METHODS: To highlight the role of cell proteoglycans (PGs) in the shift of FSH signaling, both FSH-induced cAMP production and ERK1/2 inactivation were studied in untreated and sodium chlorate PG-depleted cultured Sertoli cells from 20day-old rats. RESULTS: Depletion of cell membrane PGs by sodium chlorate reduced FSH-, but not cholera toxin-stimulated cAMP production as well as basal ERK phosphorylation through an okadaic acid (OA)-sensitive mechanism. Involvement of PP2A was further substantiated by a marked decrease in membrane- associated PP2A activity under SC conditions and by the OA-induced restoration of PKA-dependent ERK inactivation in SC-treated cells. CONCLUSIONS: In 20-day-old rat Sertoli cells, transmembrane cell PGs, through tethering/activation of PP2A activity exerts regulatory control on both FSH receptor/Gs coupling and ERK phosphorylation. GENERAL SIGNIFICANCE: Besides their antiproliferative roles, cell PGs such as syndecan-1, could be involved in the increase in cAMP response to FSH occurring in Sertoli cells at the time of transition between proliferative and differentiated states.


Subject(s)
MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 3/metabolism , Protein Phosphatase 2/metabolism , Receptors, FSH/metabolism , Sertoli Cells/metabolism , Syndecan-1/metabolism , Animals , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Male , Rats , Rats, Sprague-Dawley , Receptors, FSH/agonists , Second Messenger Systems/physiology , Sheep
11.
Mol Cancer Ther ; 11(5): 1203-13, 2012 May.
Article in English | MEDLINE | ID: mdl-22411898

ABSTRACT

We assessed the prognostic and predictive value of ß-tubulin III (TUBB3) expression, as determined by immunohistochemistry, in 412 non-small cell lung cancer (NSCLC) specimens from early-stage patients who received neoadjuvant chemotherapy (paclitaxel- or gemcitabine-based) in a phase III trial (IFCT-0002). We also correlated TUBB3 expression with K-Ras and EGF receptor (EGFR) mutations in a subset of 208 cryopreserved specimens. High TUBB3 protein expression was associated with nonsquamous cell carcinomas (P < 0.001) and K-Ras mutation (P < 0.001). The 127 (30.8%) TUBB3-negative patients derived more than 1 year of overall survival advantage, with more than 84 months median overall survival versus 71.7 months for TUBB3-positive patients [HR, 1.58; 95% confidence interval (CI), 1.11-2.25)]. This prognostic value was confirmed in multivariate analysis (adjusted HR for death, 1.51; 95% CI, 1.04-2.21; P = 0.031) with a bootstrapping validation procedure. TUBB3 expression was associated with nonresponse to chemotherapy (adjusted HR, 1.31; 95% CI, 1.01-1.70; P = 0.044) but had no predictive value (taxane vs. gemcitabine). Taking account of these clinical findings, we further investigated TUBB3 expression in isogenic human bronchial cell lines only differing by K-Ras gene status and assessed the effect of K-Ras short interfering RNA (siRNA) mediated depletion, cell hypoxia, or pharmacologic inhibitors of K-Ras downstream effectors, on TUBB3 protein cell content. siRNA K-Ras knockdown, inhibition of RAF/MEK (MAP-ERK kinase) and phosphoinositide 3-kinase (PI3K)/AKT signaling, and hypoxia were shown to downregulate TUBB3 expression in bronchial cells. This study is the first one to identify K-Ras mutations as determinant of TUBB3 expression, a chemoresistance marker. Our in vitro data deserve studies combining standard chemotherapy with anti-MEK or anti-PI3K drugs in patients with TUBB3-overexpressing tumors.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Tubulin/metabolism , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/mortality , Male , Middle Aged , Mutation , Neoadjuvant Therapy , Prognosis , Proto-Oncogene Proteins p21(ras)/genetics , Survival Analysis , Treatment Outcome , Tubulin/genetics
12.
Reprod Biomed Online ; 21(1): 56-65, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20457540

ABSTRACT

Gonadotrophin-releasing hormone (GnRH) agonists and antagonists have been widely used to prevent premature LH surge during ovarian stimulation. However, studies have shown a significantly lower serum oestradiol concentration on the day of human chorionic gonadotrophin administration for cycles using GnRH antagonist. This study compared aromatase gene expression in granulosa lutein cells from 50 women randomly assigned to receive either GnRH agonist (group 1, n=28) or GnRH antagonist (group 2, n=22). The cellular mechanism involved in the observed effects was also investigated. GnRH antagonist treatment significantly affected serum oestradiol concentration (1894+/-138 versus 1074+/-63 pg/ml; P < or = 0.001), follicular-fluid oestradiol concentration in large follicles (18,565+/-2467 versus 10,184+/-1993 pg/ml; P < or = 0.05), aromatase activity (9600+/-1179 versus 5376+/-997 fmol/10(6) cells/h; P < or = 0.05) and mRNA aromatase/mRNA glyceraldehyde 3-phosphate dehydrogenase (15+/-3 versus 6+/-1; P < 0.05). Protein kinase C (PKC) activity in granulosa lutein cells from the GnRH antagonist group was 2.5-fold higher than in the GnRH agonist group. In-vitro experiments showed that selective down-regulation of PKC was only observed in GnRH-desensitized granulosa lutein cells. This report suggests that, in granulosa lutein cells, the modulation of the FSH-induced protein kinase A pathway by PKC was different in agonist versus antagonist cycles.


Subject(s)
Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/agonists , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Ovulation Induction/methods , Triptorelin Pamoate/therapeutic use , Adult , Aromatase/biosynthesis , Aromatase/genetics , Down-Regulation , Estradiol/blood , Female , Follicular Fluid/metabolism , Gonadotropin-Releasing Hormone/therapeutic use , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Humans , Ovarian Follicle/metabolism , Pregnancy , Pregnancy Rate , Protein Kinase C/metabolism
13.
J Endocrinol ; 197(1): 45-54, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18372231

ABSTRACT

Cessation of rat testicular Sertoli cells proliferation around days 15-20 post partum is associated in vitro with the highest rise in rolipram-sensitive cAMP-catabolizing phosphodiesterase-4 activities (PDE4s) triggered by FSH during the early postnatal period. The transient nature of FSH-induced increase in PDE4s suggests concomitant changes in both PKA-mediated activation and subsequent deactivation of these activities. In this study, we demonstrated that the deactivation of FSH-stimulated particulate, but not soluble, PDE4s in cultured Sertoli cells from 20-day-old rats was inhibited by phosphoprotein phosphatase (PP) inhibitors, okadaïc acid, and calyculin A. Moreover, the deactivation of FSH-stimulated particulate PDE4s was timely related with the gonadotropin-induced increase in both particulate PP2A activity and particulate PP2A catalytic subunit immunoreactive expression independently of any transcriptional regulation of that subunit. Both the FSH-induced increase in recruitment/activation of particulate PP2A and the subsequent deactivation of particulate PDE4 were abolished when Sertoli cell proteoglycans (PGs) synthesis was altered by sodium chlorate. Sodium chlorate effect was developmentally regulated as evidenced by its ability to silence particulate PDE4 deactivation only in non-proliferating (from 20- to 30-day-old rats) but not in proliferating (from 10-day-old rats) Sertoli cells. All these data suggested that PGs could be involved in the FSH-induced recruitment/activation of PP2A. Particularly, developmentally regulated transmembrane syndecans, the most abundant PGs in Sertoli cells, by targeting PP2A at the membrane level could allow developmental control of activated particulate PDE4s and, potentially, other signaling phosphoproteins, including the FSH receptor, during the early postnatal period.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Follicle Stimulating Hormone/pharmacology , Protein Phosphatase 2/physiology , Proteoglycans/biosynthesis , Sertoli Cells/metabolism , Animals , Cells, Cultured , Male , Rats , Rats, Sprague-Dawley , Sodium Cholate/pharmacology
14.
Biol Reprod ; 76(5): 794-803, 2007 May.
Article in English | MEDLINE | ID: mdl-17215491

ABSTRACT

Major changes in the cAMP-dependent signal transduction pathway triggered by FSH take place during transition of rat Sertoli cells from proliferative to the quiescent/terminally differentiated state. Using Sertoli cell cultures isolated from 10-, 20-, and 30-day-old rats, we recorded a specific increase in PDE4 activity in both the soluble and particulate subcellular fractions of 20-day-old Sertoli cells, which also displayed the highest cAMP response to FSH and the highest FSH-induced increase in PDE4 activity in both subcellular compartments. RT-PCR and immunoblotting experiments showed that almost all the PDE4D isoforms, known as the main cAMP-regulated rolipram-sensitive PDE in Sertoli cells, were expressed throughout the early postpartum period, whereas only the short PDE4D isoforms (PDE4D1 and PDE4D2) were transcriptionally regulated by FSH. Unexpectedly, the immunoblot data also revealed that the soluble PDE4 activities were mainly related to the long PDE4D isoforms and that short PDE4D1 was predominantly particulate. The subcellular distribution and expression of PDE4D proteins were unaffected by the developmental status of the Sertoli cells. Only the expression of short PDE4D1 appeared to be upregulated by FSH and only in 20-day-old Sertoli cells, which suggests phenotype-dependent differential regulation of Pde4d1 mRNA translation. Resensitization of the cAMP response to FSH in 20-day-old Sertoli cells was also associated with the highest FSH-induced transient increase in both soluble and particulate PDE4 activities, which suggests developmental changes in the PKA-mediated upregulation of the catalytic activities of long PDE4D. Such alterations may be involved in the phenotype-dependent alterations in FSH receptor coupling with its associated G proteins in rat Sertoli cells.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/biosynthesis , Aging/physiology , Follicle Stimulating Hormone/pharmacology , Sertoli Cells/enzymology , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Animals , Blotting, Western , Cyclic Nucleotide Phosphodiesterases, Type 4 , DNA Primers , Electrophoresis, Polyacrylamide Gel , Female , Isoenzymes/biosynthesis , Isoenzymes/genetics , Kinetics , Male , Phosphodiesterase Inhibitors/pharmacology , Protein Isoforms/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Rolipram/pharmacology , Sertoli Cells/drug effects , Stimulation, Chemical , Subcellular Fractions/metabolism
15.
Biochim Biophys Acta ; 1770(4): 638-48, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17261351

ABSTRACT

FSH-induced upregulation of cAMP-PDE4 activities was decreased in cultured Sertoli cells when alteration of cell proteoglycans (PGs) metabolism was simultaneously induced either by para-nitrophenyl beta-d-xyloside (PNPX) or by sodium chlorate. This effect was restricted to the particulate PDE4 activities and its timing was consistent with the half-life of Sertoli cell PGs. It did not result from alterations in Pde4d variants expression, the major FSH-regulated PDE4 in Sertoli cells. Moreover, lack of changes in the particulate levels of major immunoreactive 75 kDa and 90 kDa PDE4D proteins, corresponding likely to short PDE4D1 and long PDE4D3/D8/D9 isoforms respectively, suggested that the decrease in FSH-stimulated of PDE4 activities in chlorate- and PNPX-treated cells at the end of the 24-h incubation period resulted from the increased reversal of the activated particulate PDE4(D) activities back to unstimulated levels. By controlling FSH-stimulated particulate PDE4 inactivation through a still unknown mechanism (sustained activation of PKA or reduction of phosphoprotein phosphatase activities), cell PGs could be involved in the alteration of cAMP response to FSH accompanying the transition of Sertoli cells from proliferative to non-proliferative differentiated state.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Follicle Stimulating Hormone/pharmacology , Proteoglycans/biosynthesis , Sertoli Cells/metabolism , Signal Transduction , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Animals , Cells, Cultured , Chlorates/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4 , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Glycosides/pharmacology , Isoenzymes/metabolism , Male , Phosphodiesterase Inhibitors/pharmacology , Protein Biosynthesis/drug effects , Rats , Rats, Sprague-Dawley , Rolipram/pharmacology , Sertoli Cells/drug effects , Signal Transduction/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Time Factors
16.
Mol Endocrinol ; 18(10): 2553-69, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15256532

ABSTRACT

We have analyzed the ontogeny and putative mechanisms of transregulation of LH receptor (LHR) and transcription factor GATA-4, coexpressed during the adrenocortical tumorigenesis of prepubertally gonadectomized transgenic (TG) mice expressing the inhibin alpha-subunit promoter/simian virus 40 T-antigen (inhalpha/Tag) transgene. The onset of adrenal LHR mRNA and protein expression coincided with that of GATA-4 at the age of 4 months and preceded the appearance of discernible adrenal tumors at about 6 months. In situ hybridization and double-immunohistochemistry demonstrated colocalization of the LHR and GATA-4 messages and proteins in the adrenal cortex. A GATA-4 expression plasmid cotransfected with a murine LHR promoter-driven luciferase reporter plasmid, containing a consensus GATA-binding site, induced a dose-dependent significant transactivation of the LHR promoter in nonsteroidogenic human embryonic kidney 293, steroidogenic murine mLTC-1 Leydig cells and in murine adrenal Y-1 cells. The Calpha1 cells derived from an Inhalpha/Tag adrenal tumor did not show this response, apparently due to their high endogenous GATA-4 expression. However, an additional link between GATA-4 and LHR in Calpha1 cells was provided upon the LH/human chorionic gonadotropin stimulation of LHR promoter activity; mutations or deletion of the consensus GATA-4 binding site of the LHR promoter abolished this transactivation. EMSAs further proved GATA-4 binding to the putative consensus GATA recognition site. Our results demonstrate direct interrelationship between LHR and GATA-4 expression during adrenocortical tumorigenesis of the inhalpha/Tag mice. There is apparently a positive and reciprocal feed-forward amplification link between LHR and GATA-4 expression. This mechanism gradually and in synergy with Tag expression leads to formation of the LH-dependent adrenocortical tumors.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Antigens, Polyomavirus Transforming/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Inhibins/genetics , Receptors, LH/genetics , Transcription Factors/genetics , Animals , Base Sequence , Blotting, Northern , Cell Line, Tumor , DNA Primers , GATA4 Transcription Factor , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Mutagenesis, Site-Directed , Orchiectomy , Reverse Transcriptase Polymerase Chain Reaction , Simian virus 40/genetics
17.
Mol Cell Endocrinol ; 193(1-2): 137-43, 2002 Jul 31.
Article in English | MEDLINE | ID: mdl-12161013

ABSTRACT

The cytochrome P450 aromatase (P450arom) is the terminal enzyme responsible for the formation of estrogens from androgens. In the rat testis we have immunolocalized the P450arom not only in Leydig cells but also in germ cells and especially in elongated spermatids. Related to the stage of germ cell maturation, we have shown that the level of P450arom transcripts decreases, it is much more abundant in pachytene spermatocytes (PS) than in mature germ cells whereas the aromatase activity is two- to fourfold greater in spermatozoa when compared to younger germ cell preparations. In rat germ cells, the aromatase gene expression is not only under androgen and cyclic AMP control but also subjected to cytokine (TNFalpha) and growth factor (TGFbeta) regulation. In the bank-vole testis we have evidenced a positive correlation between a fully developed spermatogenesis and a strong immunoreactivity for both P450arom and estrogen receptor (ERbeta) not only in Sertoli cells but also in PS and round spermatids (RS). Therefore, the aromatase gene expression and its translation in a fully active protein in rodent germ cells evidence an additional site for estrogen production within the testis. Our recent data showing that human ejaculated spermatozoa expressed specific transcripts for P450arom reinforced the observations reported in germ cells of other mammalian species. Together with the widespread distribution of ERs in testicular cells these data bring enlightenment on the hormonal regulation of spermatogenesis.


Subject(s)
Aromatase/physiology , Estrogens/physiology , Testis/chemistry , Animals , Aromatase/biosynthesis , Aromatase/deficiency , Gene Expression Regulation , Germ Cells/chemistry , Germ Cells/enzymology , Humans , Leydig Cells/chemistry , Leydig Cells/enzymology , Male , Spermatogenesis , Testis/cytology , Testis/physiology
18.
Endocrinology ; 143(3): 909-19, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11861512

ABSTRACT

The gene for mouse relaxin-like factor (RLF), a member of the insulin/IGF/relaxin family of hormones, appears to be predominantly expressed in testicular Leydig cells. Mice deficient in RLF have revealed a role for this peptide in testicular descent, but the regulatory mechanisms of its function are still insufficiently characterized. In the present study we showed that the RLF promoter was active in both mLTC-1 Leydig cells and luteinized KK-1 granulosa tumor cells. Interestingly, the activity of the RLF promoter as well as the expression of endogenous RLF correlated with the amount of steroidogenic factor 1 (SF-1) expression in the four cell lines tested. The highest transcriptional activity (29-fold over promoterless plasmid) was detected in mLTC-1 using the 188-bp promoter fragment immediately 5' of the CAP site, containing three consensus sequences for SF-1 binding. However, the promoter fragments including the 188-bp promoter also showed significant SF-1-independent promoter activity in both mLTC-1 and KK-1 cells, 8-fold induced over the promoterless construct. Mutagenesis studies showed that all three SF-1-binding sites were needed to obtain maximal SF-1-dependent trans-activation. The most distal SF-1-binding site at position -144 to -136 showed the highest affinity toward SF-1, but the promoter fragments, including the SF-1-binding site at position -115 to -107, showed the strongest response to SF-1 in terms of transcriptional activation. Moreover, DAX-1 inhibited RLF promoter activity in mLTC-1 Leydig tumor cells and totally abolished SF-1-dependent RLF expression in nonsteroidogenic HEK-293 cells. DAX-1 especially inhibited binding of SF-1 to the binding motifs locating at positions -64 to -56 and -115 to -107, whereas no decrease was seen in the expression of SF-1. Taken together, these observations suggest that the 188-bp RLF promoter includes elements for both SF-1-dependent and -independent gene expression in steroidogenic cells. The data, furthermore, indicate differential binding affinities for the three SF-1 binding motifs toward SF-1, of which the motif locating at position -115 to -107 was the most critical for the promoter activity.


Subject(s)
DNA-Binding Proteins/genetics , Protein Biosynthesis , Proteins/genetics , Receptors, Retinoic Acid/genetics , Repressor Proteins , Transcription Factors/genetics , Animals , Base Sequence , DAX-1 Orphan Nuclear Receptor , Fushi Tarazu Transcription Factors , Gene Deletion , Homeodomain Proteins , Immunoblotting , Insulin , Leydig Cell Tumor/metabolism , Male , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear , Steroidogenic Factor 1 , Transfection , Tumor Cells, Cultured
19.
News Physiol Sci ; 15: 195-198, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11390909

ABSTRACT

Besides somatic cells, aromatase gene expression and its transduction in an active protein in germ cells provides evidence of an additional site for estrogen production within testes of some mammals. Together with the widespread distribution of estrogen receptors in testicular cells, these data illuminate the hormonal regulation of male reproductive function

SELECTION OF CITATIONS
SEARCH DETAIL
...