Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Oncogene ; 42(38): 2854-2867, 2023 09.
Article in English | MEDLINE | ID: mdl-37587334

ABSTRACT

Increasing evidence points towards a causal link between exposure to persistent organic pollutants (POPs) with increased incidence and aggressivity of various cancers. Among these POPs, dioxin and PCB-153 are widely found in our environment and represent a significant source of contamination. Dioxin exposure has already been linked to cancer such as non-Hodgkin's lymphoma, but remains to be more extensively investigated in other cancers. Potential implications of dioxin and PCB-153 in prostate cancer progression spurred us to challenge both ex vivo and in vivo models with low doses of these POPs. We found that dioxin or PCB-153 exposure increased hallmarks of growth and metastasis of prostate cancer cells ex vivo and in grafted NOD-SCID mice. Exposure induced histopathological carcinoma-like patterns in the Ptenpc-/- mice. We identified up-regulation of Acetyl-CoA Acetyltransferase-1 (ACAT1) involved in ketone bodies pathway as a potential target. Mechanistically, genetic inhibition confirmed that ACAT1 mediated dioxin effect on cell migration. Using public prostate cancer datasets, we confirmed the deregulation of ACAT1 and associated gene encoded ketone bodies pathway enzymes such as OXCT1, BDH1 and HMGCL in advanced prostate cancer. To further explore this link between dioxin and ACAT1 deregulation, we analyzed a unique prostate-tumour tissue collection from the USA veterans exposed to agent orange, known to be highly contaminated by dioxin because of industrial production. We found that ACAT1 histoscore is significantly increased in exposed patients. Our studies reveal the implication of dioxin and PCB-153 to induce a prometastatic programme in prostate tumours and identify ACAT1 deregulation as a key event in this process.


Subject(s)
Dioxins , Polychlorinated Dibenzodioxins , Prostatic Neoplasms , Male , Humans , Animals , Mice , Mice, Inbred NOD , Mice, SCID , Persistent Organic Pollutants , Dioxins/toxicity , Prostatic Neoplasms/chemically induced , Prostatic Neoplasms/genetics , Acetyltransferases
2.
Cells ; 10(9)2021 09 10.
Article in English | MEDLINE | ID: mdl-34572036

ABSTRACT

Prostate cancer is the most common cancer in aging men. Despite recent progress, there are still few effective treatments to cure its aggressive and metastatic stages. A better understanding of the molecular mechanisms driving disease initiation and progression appears essential to support the development of more efficient therapies and improve patient care. To do so, multiple research models, such as cell culture and mouse models, have been developed over the years and have improved our comprehension of the biology of the disease. Recently, a new model has been added with the use of the Drosophila accessory gland. With a high level of conservation of major signaling pathways implicated in human disease, this functional equivalent of the prostate represents a powerful, inexpensive, and rapid in vivo model to study epithelial carcinogenesis. The purpose of this review is to quickly overview the existing prostate cancer models, including their strengths and limitations. In particular, we discuss how the Drosophila accessory gland can be integrated as a convenient complementary model by bringing new understanding in the mechanisms driving prostate epithelial tumorigenesis, from initiation to metastatic formation.


Subject(s)
Disease Models, Animal , Drosophila/physiology , Genitalia, Male/pathology , Prostatic Neoplasms/pathology , Animals , Humans , Male
3.
Br J Pharmacol ; 178(16): 3277-3293, 2021 08.
Article in English | MEDLINE | ID: mdl-33080050

ABSTRACT

Liver X receptors (LXRs) are members of the nuclear receptor superfamily that are canonically activated by oxidized derivatives of cholesterol. Since the mid-90s, numerous groups have identified LXRs as endocrine receptors that are involved in the regulation of various physiological functions. As a result, when their expression is genetically modified in mice, phenotypic analyses reveal endocrine disorders ranging from infertility to diabetes and obesity, nervous system pathologies such Alzheimer's or Parkinson's disease, immunological disturbances, inflammatory response, and enhancement of tumour development. Based on such findings, it appears that LXRs could constitute good pharmacological targets to prevent and/or to treat these diseases. This review discusses the various aspects of LXR drug discovery, from the tools available for the screening of potential LXR modulators to the current situational analysis of the drugs in development. LINKED ARTICLES: This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc.


Subject(s)
Diabetes Mellitus , Parkinson Disease , Animals , Cholesterol , Liver X Receptors , Mice , Receptors, Cytoplasmic and Nuclear
4.
PLoS Biol ; 18(12): e3000948, 2020 12.
Article in English | MEDLINE | ID: mdl-33284790

ABSTRACT

Chronic inflammation is now a well-known precursor for cancer development. Infectious prostatitis are the most common causes of prostate inflammation, but emerging evidence points the role of metabolic disorders as a potential source of cancer-related inflammation. Although the widely used treatment for prostate cancer based on androgen deprivation therapy (ADT) effectively decreases tumor size, it also causes profound alterations in immune tumor microenvironment within the prostate. Here, we demonstrate that prostates of a mouse model invalidated for nuclear receptors liver X receptors (LXRs), crucial lipid metabolism and inflammation integrators, respond in an unexpected way to androgen deprivation. Indeed, we observed profound alterations in immune cells composition, which was associated with chronic inflammation of the prostate. This was explained by the recruitment of phagocytosis-deficient macrophages leading to aberrant hyporesponse to castration. This phenotypic alteration was sufficient to allow prostatic neoplasia. Altogether, these data suggest that ADT and inflammation resulting from metabolic alterations interact to promote aberrant proliferation of epithelial prostate cells and development of neoplasia. This raises the question of the benefit of ADT for patients with metabolic disorders.


Subject(s)
Immunity/physiology , Liver X Receptors/metabolism , Prostate/metabolism , Androgen Antagonists/immunology , Androgens/metabolism , Animals , Disease Models, Animal , Immunity/immunology , Liver X Receptors/genetics , Liver X Receptors/immunology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Neoplasms/etiology , Neoplasms/immunology , Neoplasms/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Receptors, Cytoplasmic and Nuclear/metabolism , Tumor Microenvironment
5.
Nat Commun ; 11(1): 2300, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32385236

ABSTRACT

One of the most important but less understood step of epithelial tumourigenesis occurs when cells acquire the ability to leave their epithelial compartment. This phenomenon, described as basal epithelial cell extrusion (basal extrusion), represents the first step of tumour invasion. However, due to lack of adequate in vivo model, implication of emblematic signalling pathways such as Ras/Mitogen-Activated Protein Kinase (MAPK) and phosphoinositide 3 kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signalling pathways, is scarcely described in this phenomenon. We have developed a unique model of basal extrusion in the Drosophila accessory gland. There, we demonstrate that both Ras/MAPK and PI3K/AKT/mTOR pathways are necessary for basal extrusion. Furthermore, as in prostate cancer, we show that these pathways are co-activated. This occurs through set up of Epidermal Growth Factor Receptor (EGFR) and Insulin Receptor (InR) dependent autocrine loops, a phenomenon that, considering human data, could be relevant for prostate cancer.


Subject(s)
Drosophila Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Drosophila , Exocrine Glands/metabolism , Male , Prostatic Neoplasms/metabolism , Signal Transduction/physiology
6.
Int J Mol Sci ; 21(10)2020 May 23.
Article in English | MEDLINE | ID: mdl-32456259

ABSTRACT

: The great majority of breast and prostate tumors are hormone-dependent cancers; hence, estrogens and androgens can, respectively, drive their developments, making it possible to use pharmacological therapies in their hormone-dependent phases by targeting the levels of steroid or modulating their physiological activity through their respective nuclear receptors when the tumors relapse. Unfortunately, at some stage, both breast and prostate cancers become resistant to pharmacological treatments that aim to block their receptors, estrogen (ER) or androgen (AR) receptors, respectively. So far, antiestrogens and antiandrogens used in clinics have been designed based on their structural analogies with natural hormones, 17-ß estradiol and dihydrotestosterone. Plants are a potential source of drug discovery and the development of new pharmacological compounds. The aim of this review article is to highlight the recent advances in the pharmacological modulation of androgen or estrogen levels, and their activity through their cognate nuclear receptors in prostate or breast cancer and the effects of some plants extracts.


Subject(s)
Androgens/metabolism , Antineoplastic Agents, Phytogenic/therapeutic use , Breast Neoplasms/metabolism , Estrogens/metabolism , Hormone Antagonists/therapeutic use , Plant Extracts/therapeutic use , Prostatic Neoplasms/metabolism , Animals , Breast Neoplasms/drug therapy , Female , Humans , Male , Plant Extracts/chemistry , Prostatic Neoplasms/drug therapy
7.
J Steroid Biochem Mol Biol ; 190: 173-182, 2019 06.
Article in English | MEDLINE | ID: mdl-30959154

ABSTRACT

Liver X receptors (LXRs) α (NR1H3) and ß (NR1H2) are nuclear receptors that have been involved in the regulation of many physiological processes, principally in the control of cholesterol homeostasis, as well as in the control of the cell death and proliferation balance. These receptors are thus promising therapeutic targets in various pathologies such as dyslipidemia, atherosclerosis, diabetes and/or cancers. These receptors are known to be activated by specific oxysterol compounds. The screening for LXR-specific ligands is a challenging process: indeed, these molecules should present a specificity towards each LXR-isoform. Because some natural products have significant effects in the regulation of the LXR-regulated homeostasis and are enriched in flavonoids, we have decided to test in cell culture the effects of 4 selected flavonoids (galangin, quercetin, apigenin and naringenin) on the modulation of LXR activity using double-hybrid experiments. In silico, molecular docking suggests specific binding pattern between agonistic and antagonistic molecules. Altogether, these results allow a better understanding of the ligand binding pocket of LXRα/ß. They also improve our knowledge about flavonoid mechanism of action, allowing the selection and development of better LXR selective ligands.


Subject(s)
Flavonoids/pharmacology , Liver X Receptors/agonists , Liver X Receptors/antagonists & inhibitors , Apigenin/pharmacology , Drug Evaluation, Preclinical , Flavanones/pharmacology , HeLa Cells , Humans , Liver X Receptors/metabolism , Molecular Docking Simulation , Quercetin/pharmacology , Structure-Activity Relationship
8.
Eur Urol Oncol ; 2(2): 198-206, 2019 03.
Article in English | MEDLINE | ID: mdl-31017097

ABSTRACT

BACKGROUND: Deregulation of cholesterol metabolism represents a hallmark of prostate cancer (PCa) and promotes its development. OBJECTIVE: To compare cholesterol metabolism on individual paired normal and tumour prostate tissues obtained from patients with PCa. DESIGN, SETTING, AND PARTICIPANTS: Between 2008 and 2012, normal and tumour paired tissue samples were collected from radical prostatectomy specimens from a cohort of 69 patients treated for localised PCa. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Tumour and normal tissues were subjected to gene analysis, sterol measurement, and immunohistochemistry. The Wilcoxon paired test and Spearman test were applied for comparison and correlation analyses, respectively. Principal component analysis was also carried out to investigate relationships between quantitative variables. RESULTS AND LIMITATIONS: Overall, cholesterol concentrations were not significantly different between tissue pairs. However, tumour samples were significantly associated with downregulated de novo cholesterol synthesis, but exhibited 54.7% overexpression of SCARB1 that could increase high-density lipoprotein uptake in PCa. Tumour tissues showed different trafficking of available cholesterol, with significantly lower ACAT1, and an altered efflux via APOE. Furthermore, cholesterol metabolism in tumour tissues was characterised by higher accumulation of 7α-hydroxycholesterol (OHC), 7ßOHC, and 7-ketosterol, and a lower level of 27OHC. CONCLUSIONS: Focusing on individually paired prostate tissues, our results highlighted several differences between normal and tumour samples linked to a metabolic shift in cholesterol flux. PCa samples exhibited a specific tissue signature characterised by higher SCARB1 expression, higher accumulation of OHC species, and clear downregulation of de novo cholesterol synthesis. PATIENT SUMMARY: Comparing normal and tumour tissues from the same prostates, our study identified a set of alterations in prostate cancer samples in terms of their use of cholesterol. These included higher cholesterol uptake, accumulation of oxidised cholesterol derivatives, and autonomous cellular production of cholesterol. Together, these data provide promising clinical targets to fight prostate cancer.


Subject(s)
Cholesterol/metabolism , Gene Regulatory Networks , Prostatic Neoplasms/surgery , Acetyl-CoA C-Acetyltransferase/genetics , Acetyl-CoA C-Acetyltransferase/metabolism , Aged , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Principal Component Analysis , Prostatectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism
9.
Int J Mol Sci ; 19(9)2018 Aug 28.
Article in English | MEDLINE | ID: mdl-30154328

ABSTRACT

Prostate cancer (PCa) incidence has been dramatically increasing these last years in westernized countries. Though localized PCa is usually treated by radical prostatectomy, androgen deprivation therapy is preferred in locally advanced disease in combination with chemotherapy. Unfortunately, PCa goes into a castration-resistant state in the vast majority of the cases, leading to questions about the molecular mechanisms involving the steroids and their respective nuclear receptors in this relapse. Interestingly, liver X receptors (LXRα/NR1H3 and LXRß/NR1H2) have emerged as new actors in prostate physiology, beyond their historical roles of cholesterol sensors. More importantly LXRs have been proposed to be good pharmacological targets in PCa. This rational has been based on numerous experiments performed in PCa cell lines and genetic animal models pointing out that using selective liver X receptor modulators (SLiMs) could actually be a good complementary therapy in patients with a castration resistant PCa. Hence, this review is focused on the interaction among the androgen receptors (AR/NR3C4), estrogen receptors (ERα/NR3A1 and ERß/NR3A2), and LXRs in prostate homeostasis and their putative pharmacological modulations in parallel to the patients' support.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Prostatic Neoplasms/etiology , Prostatic Neoplasms/metabolism , Androgens/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Disease Management , Endocrine Disruptors/adverse effects , Environmental Exposure/adverse effects , Estrogens/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lipid Metabolism , Liver X Receptors/genetics , Liver X Receptors/metabolism , Male , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Oxysterols/metabolism , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/therapy , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction
10.
Cell Cycle ; 15(10): 1352-62, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27050906

ABSTRACT

The mammalian target of rapamycin (mTOR) plays essential roles in the regulation of growth-related processes such as protein synthesis, cell sizing and metabolism in both normal and pathological growing conditions. These functions of mTOR are thought to be largely a consequence of its cytoplasmic activity in regulating translation rate, but accumulating data highlight supplementary role(s) for this serine/threonine kinase within the nucleus. Indeed, the nuclear activities of mTOR are currently associated with the control of protein biosynthetic capacity through its ability to regulate the expression of gene products involved in the control of ribosomal biogenesis and proliferation. Using primary murine embryo fibroblasts (MEFs), we observed that cells with overactive mTOR signaling displayed higher abundance for the growth-associated Npm1 protein, in what represents a novel mechanism of Npm1 gene regulation. We show that Npm1 gene expression is dependent on mTOR as demonstrated by treatment of wild-type and Pten inactivated MEFs cultured with rapamycin or by transient transfections of small interfering RNA directed against mTOR. In accordance, the mTOR kinase localizes to the Npm1 promoter gene in vivo and it enhances the activity of a human NPM1-luciferase reporter gene providing an opportunity for direct control. Interestingly, rapamycin did not dislodge mTOR from the Npm1 promoter but rather strongly destabilized the Npm1 transcript by increasing its turnover. Using a prostate-specific Pten-deleted mouse model of cancer, Npm1 mRNA levels were found up-regulated and sensitive to rapamycin. Finally, we also showed that Npm1 is required to promote mTOR-dependent cell proliferation. We therefore proposed a model whereby mTOR is closely involved in the transcriptional and posttranscriptional regulation of Npm1 gene expression with implications in development and diseases including cancer.


Subject(s)
Nuclear Proteins/metabolism , PTEN Phosphohydrolase/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , HeLa Cells , Humans , Male , Mice , Mice, Knockout , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nucleophosmin , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Promoter Regions, Genetic , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Messenger/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Transplantation, Heterologous , Up-Regulation/drug effects
11.
Hum Mol Genet ; 23(20): 5418-28, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-24865460

ABSTRACT

Primary pigmented nodular adrenocortical disease (PPNAD) is associated with inactivating mutations of the PRKAR1A tumor suppressor gene that encodes the regulatory subunit R1α of the cAMP-dependent protein kinase (PKA). In human and mouse adrenocortical cells, these mutations lead to increased PKA activity, which results in increased resistance to apoptosis that contributes to the tumorigenic process. We used in vitro and in vivo models to investigate the possibility of a crosstalk between PKA and mammalian target of rapamycin (mTOR) pathways in adrenocortical cells and its possible involvement in apoptosis resistance. Impact of PKA signaling on activation of the mTOR pathway and apoptosis was measured in a mouse model of PPNAD (AdKO mice), in human and mouse adrenocortical cell lines in response to pharmacological inhibitors and in PPNAD tissues by immunohistochemistry. AdKO mice showed increased mTOR complex 1 (mTORC1) pathway activity. Inhibition of mTORC1 by rapamycin restored sensitivity of adrenocortical cells to apoptosis in AdKO but not in wild-type mice. In both cell lines and mouse adrenals, rapid phosphorylation of mTORC1 targets including BAD proapoptotic protein was observed in response to PKA activation. Accordingly, BAD hyperphosphorylation, which inhibits its proapoptotic activity, was increased in both AdKO mouse adrenals and human PPNAD tissues. In conclusion, mTORC1 pathway is activated by PKA signaling in human and mouse adrenocortical cells, leading to increased cell survival, which is correlated with BAD hyperphosphorylation. These alterations could be causative of tumor formation.


Subject(s)
Adrenal Cortex Diseases/metabolism , Adrenal Cortex Diseases/pathology , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , bcl-Associated Death Protein/metabolism , Adrenal Cortex Diseases/genetics , Adrenocorticotropic Hormone/administration & dosage , Adrenocorticotropic Hormone/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Disease Models, Animal , Gene Knockout Techniques , Humans , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/metabolism , Phosphorylation , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism
12.
PLoS One ; 7(8): e44171, 2012.
Article in English | MEDLINE | ID: mdl-22952916

ABSTRACT

Adrenal cortical carcinomas (ACC) are rare but aggressive tumours associated with poor prognosis. The two most frequent alterations in ACC in patients are overexpression of the growth factor IGF2 and constitutive activation of Wnt/ß-catenin signalling. Using a transgenic mouse model, we have previously shown that constitutive active ß-catenin is a bona fide adrenal oncogene. However, although all these mice developed benign adrenal hyperplasia, malignant progression was infrequent, suggesting that secondary genetic events were required for aggressive tumour development. In the present paper, we have tested IGF2 oncogenic properties by developing two distinct transgenic mouse models of Igf2 overexpression in the adrenal cortex. Our analysis shows that despite overexpression levels ranging from 7 (basal) to 87 (ACTH-induced) fold, Igf2 has no tumour initiating potential in the adrenal cortex. However, it induces aberrant accumulation of Gli1 and Pod1-positive progenitor cells, in a hedgehog-independent manner. We have also tested the hypothesis that Igf2 may cooperate with Wnt signalling by mating Igf2 overexpressing lines with mice that express constitutive active ß-catenin in the adrenal cortex. We show that the combination of both alterations has no effect on tumour phenotype at stages when ß-catenin-induced tumours are benign. However, there is a mild promoting effect at later stages, characterised by increased Weiss score and proliferation. Formation of malignant tumours is nonetheless a rare event, even when Igf2 expression is further increased by ACTH treatment. Altogether these experiments suggest that the growth factor IGF2 is a mild contributor to malignant adrenocortical tumourigenesis.


Subject(s)
Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/pathology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Insulin-Like Growth Factor II/metabolism , Adrenal Glands/metabolism , Adrenal Glands/pathology , Animals , Disease Models, Animal , Disease Progression , Mice , Mice, Transgenic , beta Catenin/metabolism
13.
Curr Top Dev Biol ; 98: 277-301, 2012.
Article in English | MEDLINE | ID: mdl-22305167

ABSTRACT

Understanding gene regulatory pathways underlying diversification of cell types during development is one of the major challenges in developmental biology. Progressive specification of mesodermal lineages that are at the origin of body wall muscles in Drosophila embryos has been extensively studied during past years, providing an attractive framework for dissecting cell type diversification processes. In particular, it has been found that muscle founder cells that are at the origin of individual muscles display specific expression of transcription factors that control diversification of muscle types. These factors, encoded by genes collectively called muscle identity genes, are activated in discrete subsets of muscle founders. As a result, each founder cell is thought to carry a unique combinatorial code of identity gene expression. Considering this, to define temporally and spatially restricted expression of identity genes, a set of coordinated upstream regulatory inputs is required. But also, to realize the identity program and to form specific muscle types with distinct properties, an efficient battery of downstream identity gene targets needs to be activated. Here we review how the specificity of expression and action of muscle identity genes is acquired.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Muscles/embryology , Animals , Cell Differentiation , Drosophila melanogaster/cytology , Muscles/cytology , Muscles/innervation , Muscles/metabolism , Signal Transduction
14.
Mol Cell Endocrinol ; 351(1): 28-36, 2012 Mar 31.
Article in English | MEDLINE | ID: mdl-22019902

ABSTRACT

In the last 10 years, extensive studies showed that the cAMP pathway is deregulated in patients suffering from adrenocortical tumours, and particularly in primary pigmented nodular adrenocortical disease (PPNAD). Here we describe how evidence arising from the analysis of patients' data, mouse models and in vitro experiments, have shed light on the cAMP pathway as a central player in adrenal physiopathology. We also show how novel data generated from mouse models may point to new targets for potential therapies.


Subject(s)
Adrenal Cortex Neoplasms/metabolism , Adrenal Glands , Cyclic AMP/metabolism , Second Messenger Systems , Adrenal Cortex Neoplasms/pathology , Adrenal Glands/embryology , Adrenal Glands/growth & development , Adrenal Glands/metabolism , Adrenal Glands/pathology , Animals , Disease Models, Animal , Humans , Mice
15.
PLoS Genet ; 6(6): e1000980, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20548949

ABSTRACT

Carney complex (CNC) is an inherited neoplasia syndrome with endocrine overactivity. Its most frequent endocrine manifestation is primary pigmented nodular adrenocortical disease (PPNAD), a bilateral adrenocortical hyperplasia causing pituitary-independent Cushing's syndrome. Inactivating mutations in PRKAR1A, a gene encoding the type 1 alpha-regulatory subunit (R1alpha) of the cAMP-dependent protein kinase (PKA) have been found in 80% of CNC patients with Cushing's syndrome. To demonstrate the implication of R1alpha loss in the initiation and development of PPNAD, we generated mice lacking Prkar1a specifically in the adrenal cortex (AdKO). AdKO mice develop pituitary-independent Cushing's syndrome with increased PKA activity. This leads to autonomous steroidogenic genes expression and deregulated adreno-cortical cells differentiation, increased proliferation and resistance to apoptosis. Unexpectedly, R1alpha loss results in improper maintenance and centrifugal expansion of cortisol-producing fetal adrenocortical cells with concomitant regression of adult cortex. Our data provide the first in vivo evidence that loss of R1alpha is sufficient to induce autonomous adrenal hyper-activity and bilateral hyperplasia, both observed in human PPNAD. Furthermore, this model demonstrates that deregulated PKA activity favors the emergence of a new cell population potentially arising from the fetal adrenal, giving new insight into the mechanisms leading to PPNAD.


Subject(s)
Adrenal Cortex/metabolism , Cushing Syndrome/metabolism , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Adrenocorticotropic Hormone/metabolism , Animals , Cell Proliferation , Cushing Syndrome/embryology , Cushing Syndrome/genetics , Cushing Syndrome/pathology , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/deficiency , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Humans , Male , Mice , Mice, Knockout , Signal Transduction , Steroid 17-alpha-Hydroxylase/metabolism
16.
Hum Mol Genet ; 19(8): 1561-76, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20106872

ABSTRACT

Adrenocortical carcinoma is a rare but aggressive cancer with unknown aetiology. Constitutive activation of beta-catenin is the most frequent alteration in benign and malignant adrenocortical tumours in patients. Here, we show that constitutive activation of beta-catenin in the adrenal cortex of transgenic mice resulted in progressive steroidogenic and undifferentiated spindle-shaped cells hyperplasia as well as dysplasia of the cortex and medulla. Over a 17 months time course, transgenic adrenals developed malignant characteristics such as uncontrolled neovascularization and loco-regional metastatic invasion. These oncogenic events were accompanied by ectopic differentiation of glomerulosa at the expense of fasciculata cells, which caused primary hyperaldosteronism. Altogether these observations demonstrate that constitutively active beta-catenin is an adrenal oncogene which triggers benign aldosterone-secreting tumour development and promotes malignancy.


Subject(s)
Adrenal Cortex/pathology , Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/pathology , beta Catenin/metabolism , Adrenal Cortex/metabolism , Adrenal Gland Neoplasms/physiopathology , Aldosterone/metabolism , Animals , Cell Proliferation , Disease Models, Animal , Humans , Hyperplasia , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Metastasis
17.
J Am Soc Nephrol ; 18(9): 2509-16, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17656470

ABSTRACT

Thiazide diuretics are used worldwide as a first-choice drug for patients with uncomplicated hypertension. In addition to their antihypertensive effect, thiazides increase bone mineral density and reduce the prevalence of fractures. Traditionally, these effects have been attributed to increased renal calcium reabsorption that occurs secondary to the inhibition of the thiazide-sensitive sodium chloride cotransporter (NCC) in the distal tubule. The aim of the current study was to determine whether thiazides exert a direct bone-forming effect independent of their renal action. We found that the osteoblasts of human and rat bone also express NCC, suggesting that these bone-forming cells may be an additional target for thiazides. In vitro, NCC protein was virtually absent in proliferating human and fetal rat osteoblasts, whereas its expression dramatically increased during differentiation. Thiazides did not affect osteoblast proliferation, but directly stimulated the production of the osteoblast differentiation markers runt-related transcription factor 2 (runx2) and osteopontin. Using overexpression/knockdown studies in fetal rat calvarial cells, we show that thiazides increase the formation of mineralized nodules, but loop diuretics do not. Overall, our study demonstrates that thiazides directly stimulate osteoblast differentiation and bone mineral formation independent of their effects in the kidney. Therefore, in addition to their use as antihypertensive drugs, our results suggest that thiazides may find a role in the prevention and treatment of osteoporosis.


Subject(s)
Bone and Bones/metabolism , Calcification, Physiologic , Osteoblasts/pathology , Osteogenesis/drug effects , Sodium Chloride Symporter Inhibitors/pharmacology , Sodium Chloride Symporters/metabolism , Animals , Biomarkers , Cell Differentiation/drug effects , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , Mice , Osteopontin/metabolism , Rats , Rats, Sprague-Dawley , Sodium Chloride Symporters/drug effects
18.
Nature ; 426(6966): 555-9, 2003 Dec 04.
Article in English | MEDLINE | ID: mdl-14654840

ABSTRACT

Drosophila thoracic mechanosensory bristles originate from cells that are singled out from 'proneural' groups of competent epithelial cells. Neural competence is restricted to individual sensory organ precursors (SOPs) by Delta/Notch-mediated 'lateral inhibition', whereas other cells in the proneural field adopt an epidermal fate. The precursors of the large macrochaetes differentiate separately from individual proneural clusters that comprise about 20-30 cells or as heterochronic pairs from groups of more than 100 cells, whereas the precursors of the small regularly spaced microchaetes emerge from even larger proneural fields. This indicates that lateral inhibition might act over several cell diameters; it was difficult to reconcile with the fact that the inhibitory ligand Delta is membrane-bound until the observation that SOPs frequently extend thin processes offered an attractive hypothesis. Here we show that the extension of these planar filopodia--a common attribute of wing imaginal disc cells--is promoted by Delta and that their experimental suppression reduces Notch signalling in distant cells and increases bristle density in large proneural groups, showing that these membrane specializations mediate long-range lateral inhibition.


Subject(s)
Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Membrane Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Pseudopodia/metabolism , Animals , Cell Differentiation , Cytoskeletal Proteins , Drosophila Proteins , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Intracellular Signaling Peptides and Proteins , Ligands , Membrane Proteins/genetics , Phenotype , Phosphoproteins/genetics , Phosphoproteins/metabolism , Receptors, Notch , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...