Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 49
2.
J Clin Invest ; 133(4)2023 02 15.
Article En | MEDLINE | ID: mdl-36538378

Disorders of isolated mineralocorticoid deficiency, which cause potentially life-threatening salt-wasting crisis early in life, have been associated with gene variants of aldosterone biosynthesis or resistance; however, in some patients no such variants are found. WNT/ß-catenin signaling is crucial for differentiation and maintenance of the aldosterone-producing adrenal zona glomerulosa (zG). Herein, we describe a highly consanguineous family with multiple perinatal deaths and infants presenting at birth with failure to thrive, severe salt-wasting crises associated with isolated hypoaldosteronism, nail anomalies, short stature, and deafness. Whole exome sequencing revealed a homozygous splice variant in the R-SPONDIN receptor LGR4 gene (c.618-1G>C) regulating WNT signaling. The resulting transcripts affected protein function and stability and resulted in loss of Wnt/ß-catenin signaling in vitro. The impact of LGR4 inactivation was analyzed by adrenal cortex-specific ablation of Lgr4, using Lgr4fl/fl mice mated with Sf1:Cre mice. Inactivation of Lgr4 within the adrenal cortex in the mouse model caused decreased WNT signaling, aberrant zonation with deficient zG, and reduced aldosterone production. Thus, human LGR4 mutations establish a direct link between LGR4 inactivation and decreased canonical WNT signaling, which results in abnormal zG differentiation and endocrine function. Therefore, variants in WNT signaling and its regulators should systematically be considered in familial hyperreninemic hypoaldosteronism.


Hypoaldosteronism , Receptors, G-Protein-Coupled , Wnt Signaling Pathway , Animals , Humans , Mice , Aldosterone/metabolism , beta Catenin/metabolism , Hypoaldosteronism/complications , Hypoaldosteronism/genetics , Hypoaldosteronism/pathology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
3.
Nat Commun ; 13(1): 7858, 2022 12 21.
Article En | MEDLINE | ID: mdl-36543805

SUMOylation is a dynamic posttranslational modification, that provides fine-tuning of protein function involved in the cellular response to stress, differentiation, and tissue development. In the adrenal cortex, an emblematic endocrine organ that mediates adaptation to physiological demands, the SUMOylation gradient is inversely correlated with the gradient of cellular differentiation raising important questions about its role in functional zonation and the response to stress. Considering that SUMO-specific protease 2 (SENP2), a deSUMOylating enzyme, is upregulated by Adrenocorticotropic Hormone (ACTH)/cAMP-dependent Protein Kinase (PKA) signalling within the zona fasciculata, we generated mice with adrenal-specific Senp2 loss to address these questions. Disruption of SENP2 activity in steroidogenic cells leads to specific hypoplasia of the zona fasciculata, a blunted reponse to ACTH and isolated glucocorticoid deficiency. Mechanistically, overSUMOylation resulting from SENP2 loss shifts the balance between ACTH/PKA and WNT/ß-catenin signalling leading to repression of PKA activity and ectopic activation of ß-catenin. At the cellular level, this blocks transdifferentiation of ß-catenin-positive zona glomerulosa cells into fasciculata cells and sensitises them to premature apoptosis. Our findings indicate that the SUMO pathway is critical for adrenal homeostasis and stress responsiveness.


Cell Transdifferentiation , Cysteine Endopeptidases , Glucocorticoids , Animals , Mice , Adrenal Cortex/metabolism , Adrenal Cortex Hormones/metabolism , Adrenocorticotropic Hormone/metabolism , beta Catenin/metabolism , Cell Transdifferentiation/genetics , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Glucocorticoids/metabolism , Wnt Signaling Pathway
4.
Sci Adv ; 8(41): eadd0422, 2022 10 14.
Article En | MEDLINE | ID: mdl-36240276

Unlike most cancers, adrenocortical carcinomas (ACCs) are more frequent in women than in men, but the underlying mechanisms of this sexual dimorphism remain elusive. Here, we show that inactivation of Znrf3 in the mouse adrenal cortex, recapitulating the most frequent alteration in ACC patients, is associated with sexually dimorphic tumor progression. Although female knockouts develop metastatic carcinomas at 18 months, adrenal hyperplasia regresses in male knockouts. This male-specific phenotype is associated with androgen-dependent induction of senescence, recruitment, and differentiation of highly phagocytic macrophages that clear out senescent cells. In contrast, in females, macrophage recruitment is delayed and dampened, which allows for aggressive tumor progression. Consistently, analysis of TCGA-ACC data shows that phagocytic macrophages are more prominent in men and are associated with better prognosis. Together, these data show that phagocytic macrophages are key players in the sexual dimorphism of ACC that could be previously unidentified allies in the fight against this devastating cancer.


Adrenal Cortex Neoplasms , Adrenocortical Carcinoma , Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/genetics , Adrenocortical Carcinoma/pathology , Androgens , Animals , Female , Male , Mice , Prognosis
5.
J Invest Dermatol ; 142(11): 2949-2957.e9, 2022 11.
Article En | MEDLINE | ID: mdl-35568059

Carney complex is a rare familial multineoplastic syndrome predisposing to endocrine and nonendocrine tumors due to inactivating mutations of PRKAR1A, leading to perturbations of the cAMP‒protein kinase A signaling pathway. Skin lesions are the most common manifestation of Carney complex, including lentigines, blue nevi, and cutaneous myxomas in unusual locations such as oral and genital mucosa. Unlike endocrine disorders, the pathogenesis of skin lesions remains unexplained. In this study, we show that embryonic invalidation of the Prkar1a gene in steroidogenic factor-1‒expressing cells leads to the development of familial skin pigmentation alterations, reminiscent of those in patients with Carney complex. Immunohistological and molecular analyses, coupled with genetic monitoring of recombinant cell lineages in mouse skin, suggest that familial lentiginosis and myxomas occur in skin areas specifically enriched in dermal melanocytes. In lentigines- and blue nevi‒prone areas from mutant mice and patients, Prkar1a/PRKAR1A invalidation occurs in a subset of dermal fibroblasts capable of inducing, under the influence of protein kinase A signaling, the production of promelanogenic EDN3 and hepatocyte GF signals. Our model strongly suggests that the origin of the typical Carney complex cutaneous lesions is the result of noncell-autonomous promelanogenic activity of a dermal fibroblast population sharing a community of origin with steroidogenic factor-1 lineage.


Carney Complex , Lentigo , Myxoma , Nevus, Blue , Skin Diseases , Animals , Mice , Carney Complex/genetics , Carney Complex/pathology , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Myxoma/genetics , Myxoma/pathology , Syndrome , Lentigo/pathology
6.
Endocr Relat Cancer ; 29(3): 151-162, 2022 02 07.
Article En | MEDLINE | ID: mdl-34935630

Adrenal cortisol-producing tumors can express illicit membrane receptors such as luteinizing hormone (LH), glucose-dependent insulinotropic peptide (GIP) or type 4 and 7 serotonin (5-HT4/7) receptors. Abnormal expression of the LH receptor (LH-R) has been ascribed to the activation of the Wnt/ß-catenin signaling pathway in adrenocortical cells. In the present study, we have investigated whether ß-catenin activation may also trigger the illegitimate expression of GIP and 5-HT receptors. Three models of ß-catenin activation in adrenocortical cells were used: an APC-mutated adrenocortical tumor, human-transfected adrenocortical cells and genetically modified mouse adrenal glands. The methods employed include quantitative reverse transcription PCR, immunohistochemistry and measurement of cortisol secretion by cultured tumor cells. Abnormal expression of the GIP, 5-HT7and LH receptors was observed in the APC-mutated adrenocortical tumor tissue. In addition, GIP, 5-HT and human chorionic gonadotropin stimulated cortisol production from tumor cells in primary culture. Conversely, only the LHCGR was upregulated in human and mouse adrenocortical cells harboring the activation of ß-catenin. Moreover, LH-R immunoreactivity was detected in clusters of zona fasciculata cells in the ß-catenin-activated mouse model. Our data indicate that activation of the ß-catenin signaling pathway can promote the illicit expression of functional LH-Rs in adrenal zona fasciculata cells but does not favor the abnormal expression of GIP and 5-HT receptors.


Adrenal Cortex Neoplasms , Adrenal Gland Neoplasms , beta Catenin/metabolism , Adrenal Cortex Neoplasms/pathology , Animals , Humans , Hydrocortisone/metabolism , Mice , Receptors, LH , Receptors, Serotonin/metabolism , Serotonin/metabolism
7.
J Clin Invest ; 131(23)2021 12 01.
Article En | MEDLINE | ID: mdl-34850745

Large-cell calcifying Sertoli cell tumors (LCCSCTs) are among the most frequent lesions occurring in male Carney complex (CNC) patients. Although they constitute a key diagnostic criterion for this rare multiple neoplasia syndrome resulting from inactivating mutations of the tumor suppressor PRKAR1A, leading to unrepressed PKA activity, LCCSCT pathogenesis and origin remain elusive. Mouse models targeting Prkar1a inactivation in all somatic populations or separately in each cell type were generated to decipher the molecular and paracrine networks involved in the induction of CNC testis lesions. We demonstrate that the Prkar1a mutation was required in both stromal and Sertoli cells for the occurrence of LCCSCTs. Integrative analyses comparing transcriptomic, immunohistological data and phenotype of mutant mouse combinations led to the understanding of human LCCSCT pathogenesis and demonstrated PKA-induced paracrine molecular circuits in which the aberrant WNT4 signal production is a limiting step in shaping intratubular lesions and tumor expansion both in a mouse model and in human CNC testes.


Carney Complex/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Sertoli Cells/cytology , Testicular Neoplasms/metabolism , Wnt4 Protein/metabolism , Animals , Apoptosis , Carney Complex/genetics , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Disease Models, Animal , Gene Expression Profiling , Genes, Tumor Suppressor , Humans , Male , Mice , Mice, Knockout , Mutation , Oligonucleotide Array Sequence Analysis , Paracrine Communication , Phenotype , Pigmentation , Seminiferous Tubules/metabolism , Testis/metabolism , Transcriptome
8.
Br J Cancer ; 124(4): 805-816, 2021 02.
Article En | MEDLINE | ID: mdl-33214683

BACKGROUND: Understanding the pathways that drive adrenocortical carcinoma (ACC) is essential to the development of more effective therapies. This study investigates the role of the transcription factor HOXB9 and other HOX factors in ACC and its treatment. METHODS: We used transgenic mouse models to determine the role of Hoxb9 in adrenal tumour development. Patient transcriptomic data was analysed for the expression of HOX genes and their association with disease. Drug response studies on various adrenocortical models were done to establish novel therapeutic options. RESULTS: Our human ACC dataset analyses showed high expression of HOXB9, and other HOX factors, are associated with poorer prognosis. Transgenic overexpression of Hoxb9 in the adrenal cortex of mice with activated Ctnnb1 led to larger adrenal tumours. This phenotype was preferentially observed in male mice and was characterised by more proliferating cells and an increase in the expression of cell cycle genes, including Ccne1. Adrenal tumour cells were found to be dependent on HOX function for survival and were sensitive to a specific peptide inhibitor. CONCLUSIONS: These studies show Hoxb9 can promote adrenal tumour progression in a sex-dependent manner and have identified HOX factors as potential drug targets, leading to novel therapeutic approaches in ACC.


Adrenal Cortex Neoplasms/drug therapy , Adrenal Cortex Neoplasms/genetics , Adrenocortical Carcinoma/drug therapy , Adrenocortical Carcinoma/genetics , Homeodomain Proteins/genetics , Peptides/pharmacology , Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/pathology , Animals , Cell Proliferation/genetics , Female , Gene Expression , Homeodomain Proteins/biosynthesis , Humans , Male , Mice , Mice, Transgenic , Molecular Targeted Therapy , Peptides/genetics
9.
Eur Respir Rev ; 29(155)2020 03 31.
Article En | MEDLINE | ID: mdl-32024721

Options to achieve oral corticosteroid (OCS)-sparing have been triggering increasing interest since the 1970s because of the side-effects of OCSs, and this has now become achievable with biologics. The Société de Pneumologie de Langue Française workshop on OCSs aimed to conduct a comprehensive review of the basics for OCS use in asthma and issue key research questions. Pharmacology and definition of regular use were reviewed by the first working group (WG1). WG2 examined whether regular OCS use is associated with T2 endotype. WG3 reported on the specificities of the paediatric area. Key "research statement proposals" were suggested by WG4. It was found that the benefits of regular OCS use in asthma outside episodes of exacerbations are poorly supported by the existing evidence. However, complete OCS elimination couldn't be achieved in any available studies for all patients and the panel felt that it was too early to conclude that regular OCS use could be declared criminal. Repeated or prolonged need for OCS beyond 1 g·year-1 should indicate the need for referral to secondary/tertiary care. A strategic sequential plan aiming at reducing overall exposure to OCS in severe asthma was then held as a conclusion of the workshop.


Adrenal Cortex Hormones/administration & dosage , Asthma/drug therapy , Lung/drug effects , Administration, Oral , Adrenal Cortex Hormones/adverse effects , Asthma/diagnosis , Asthma/physiopathology , Disease Progression , Drug Administration Schedule , Evidence-Based Medicine , Humans , Lung/physiopathology , Patient Safety , Risk Assessment , Severity of Illness Index , Time Factors , Treatment Outcome
10.
Br J Cancer ; 121(5): 384-394, 2019 08.
Article En | MEDLINE | ID: mdl-31363169

BACKGROUND: EZH2 is overexpressed and associated with poor prognosis in adrenocortical carcinoma (ACC) and its inhibition reduces growth and aggressiveness of ACC cells in culture. Although EZH2 was identified as the methyltransferase that deposits the repressive H3K27me3 histone mark, it can cooperate with transcription factors to stimulate gene transcription. METHODS: We used bioinformatics approaches on gene expression data from three cohorts of patients and a mouse model of EZH2 ablation, to identify targets and mode of action of EZH2 in ACC. This was followed by ChIP and functional assays to evaluate contribution of identified targets to ACC pathogenesis. RESULTS: We show that EZH2 mostly works as a transcriptional inducer in ACC, through cooperation with the transcription factor E2F1 and identify three positive targets involved in cell cycle regulation and mitosis i.e., RRM2, PTTG1 and ASE1/PRC1. Overexpression of these genes is associated with poor prognosis, suggesting a potential role in acquisition of aggressive ACC features. Pharmacological and siRNA-mediated inhibition of RRM2 blocks cell proliferation, induces apoptosis and inhibits cell migration, suggesting that it may be an interesting target in ACC. CONCLUSIONS: Altogether, these data show an unexpected role of EZH2 and E2F1 in stimulating expression of genes associated with ACC aggressiveness.


Adrenal Cortex Neoplasms/genetics , Adrenocortical Carcinoma/genetics , E2F1 Transcription Factor/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Expression Regulation, Neoplastic , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Cell Cycle Proteins/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chromatin Immunoprecipitation , Computational Biology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Humans , Indoles/pharmacology , Mice, Knockout , Multivariate Analysis , Proportional Hazards Models , Ribonucleoside Diphosphate Reductase/antagonists & inhibitors , Ribonucleoside Diphosphate Reductase/genetics , Securin/genetics
11.
FASEB J ; 33(9): 10218-10230, 2019 09.
Article En | MEDLINE | ID: mdl-31208233

SUMOylation is a highly conserved and dynamic post-translational mechanism primarily affecting nuclear programs for adapting organisms to stressful challenges. Alteration of SUMOylation cycles leads to severe developmental and homeostatic defects and malignancy, but signals coordinating SUMOylation are still unidentified. The adrenal cortex is a zonated endocrine gland that controls body homeostasis and stress response. Here, we show that in human and in mouse adrenals, SUMOylation follows a decreasing centripetal gradient that mirrors cortical differentiation flow and delimits highly and weakly SUMOylated steroidogenic compartments, overlapping glomerulosa, and fasciculata zones. Activation of PKA signaling by acute hormonal treatment, mouse genetic engineering, or in Carney complex results in repression of small ubiquitin-like modifier (SUMO) conjugation in the inner cortex by coordinating expression of SUMO pathway inducers and repressors. Conversely, genetic activation of canonical wingless-related integration site signaling maintains high SUMOylation potential in the outer neoplastic cortex. Thus, SUMOylation is tightly regulated by signaling pathways that orchestrate adrenal zonation and diseases.-Dumontet, T., Sahut-Barnola, I., Dufour, D., Lefrançois-Martinez, A.-M., Berthon, A., Montanier, N., Ragazzon, B., Djari, C., Pointud, J.-C., Roucher-Boulez, F., Batisse-Lignier, M., Tauveron, I., Bertherat, J., Val, P., Martinez, A. Hormonal and spatial control of SUMOylation in the human and mouse adrenal cortex.


Adrenal Cortex/metabolism , Adrenocorticotropic Hormone/pharmacology , Protein Processing, Post-Translational/physiology , Sumoylation/physiology , Adrenal Cortex/drug effects , Adrenal Cortex/ultrastructure , Adrenal Cortex Neoplasms/pathology , Adrenocorticotropic Hormone/administration & dosage , Animals , Carney Complex/metabolism , Cell Line, Tumor , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/physiology , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Delayed-Action Preparations , Dexamethasone/analogs & derivatives , Dexamethasone/pharmacology , Female , Humans , Mice , Mice, Knockout , Mice, Transgenic , Neoplasm Proteins/metabolism , Protein Processing, Post-Translational/drug effects , Signal Transduction/drug effects , Sumoylation/drug effects , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/physiology , Zona Fasciculata/drug effects , Zona Fasciculata/metabolism , Zona Glomerulosa/drug effects , Zona Glomerulosa/metabolism , beta Catenin/deficiency , beta Catenin/genetics
12.
Genes Dev ; 33(3-4): 209-220, 2019 02 01.
Article En | MEDLINE | ID: mdl-30692207

Spatiotemporal control of Wnt signaling is essential for the development and homeostasis of many tissues. The transmembrane E3 ubiquitin ligases ZNRF3 (zinc and ring finger 3) and RNF43 (ring finger protein 43) antagonize Wnt signaling by promoting degradation of frizzled receptors. ZNRF3 and RNF43 are frequently inactivated in human cancer, but the molecular and therapeutic implications remain unclear. Here, we demonstrate that adrenocortical-specific loss of ZNRF3, but not RNF43, results in adrenal hyperplasia that depends on Porcupine-mediated Wnt ligand secretion. Furthermore, we discovered a Wnt/ß-catenin signaling gradient in the adrenal cortex that is disrupted upon loss of ZNRF3. Unlike ß-catenin gain-of-function models, which induce high Wnt/ß-catenin activation and expansion of the peripheral cortex, ZNRF3 loss triggers activation of moderate-level Wnt/ß-catenin signaling that drives proliferative expansion of only the histologically and functionally distinct inner cortex. Genetically reducing ß-catenin dosage significantly reverses the ZNRF3-deficient phenotype. Thus, homeostatic maintenance of the adrenal cortex is dependent on varying levels of Wnt/ß-catenin activation, which is regulated by ZNRF3.


Adrenal Cortex/metabolism , Homeostasis/genetics , Ubiquitin-Protein Ligases/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Adrenal Cortex/cytology , Adrenal Cortex/growth & development , Adrenal Cortex Diseases/physiopathology , Animals , Cell Proliferation/genetics , Female , Gene Knockout Techniques , Male , Mice , Models, Animal , Transcriptional Activation/genetics , Ubiquitin-Protein Ligases/genetics
13.
Proc Natl Acad Sci U S A ; 115(52): E12265-E12274, 2018 12 26.
Article En | MEDLINE | ID: mdl-30541888

Adrenal cortex steroids are essential for body homeostasis, and adrenal insufficiency is a life-threatening condition. Adrenal endocrine activity is maintained through recruitment of subcapsular progenitor cells that follow a unidirectional differentiation path from zona glomerulosa to zona fasciculata (zF). Here, we show that this unidirectionality is ensured by the histone methyltransferase EZH2. Indeed, we demonstrate that EZH2 maintains adrenal steroidogenic cell differentiation by preventing expression of GATA4 and WT1 that cause abnormal dedifferentiation to a progenitor-like state in Ezh2 KO adrenals. EZH2 further ensures normal cortical differentiation by programming cells for optimal response to adrenocorticotrophic hormone (ACTH)/PKA signaling. This is achieved by repression of phosphodiesterases PDE1B, 3A, and 7A and of PRKAR1B. Consequently, EZH2 ablation results in blunted zF differentiation and primary glucocorticoid insufficiency. These data demonstrate an all-encompassing role for EZH2 in programming steroidogenic cells for optimal response to differentiation signals and in maintaining their differentiated state.


Adrenal Cortex/enzymology , Cyclic AMP-Dependent Protein Kinase RIbeta Subunit/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Signal Transduction , Adrenal Cortex/metabolism , Animals , Cell Differentiation , Cyclic AMP-Dependent Protein Kinase RIbeta Subunit/genetics , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 7/genetics , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Steroids/metabolism , Zona Fasciculata/cytology , Zona Fasciculata/enzymology , Zona Fasciculata/metabolism , Zona Glomerulosa/cytology , Zona Glomerulosa/enzymology , Zona Glomerulosa/metabolism
15.
Ann Endocrinol (Paris) ; 79(3): 95-97, 2018 Jun.
Article En | MEDLINE | ID: mdl-29673697

The adrenocortical gland undergoes structural and functional remodelling in the fetal and postnatal periods. After birth, the fetal zone of the gland undergoes rapid involution in favor of the definitive cortex, which reaches maturity with the emergence of the zona reticularis(zR) at the adrenarche. The mechanisms underlying the adrenarche, the process leading to pre-puberty elevation of plasma androgens in higher primates, remain unknown, largely due to lack of any experimental model. By following up fetal and definitive cortex cell lines in mice, we showed that activation of protein kinase A (PKA) signaling mainly impacts the adult cortex by stimulating centripetal regeneration, with differentiation and then conversion of the zona fasciculata into a functional zR. Animals developed Cushing syndrome associated with primary hyperaldosteronism, suggesting possible coexistence of these hypersecretions in certain patients. Remarkably, all of these traits were sex-dependent: testicular androgens promoted WNT signaling antagonism on PKA, slowing cortical renewal and delaying onset of Cushing syndrome and the establishment of the zR in male mice, this being corrected by orchidectomy. In conclusion, zR derives from centripetal conversion of the zona fasciculata under cellular renewal induced by PKA signaling, determining the size of the adult cortex. Finally, we demonstrated that this PKA-dependent mobilization of cortical progenitors is sexually dimorphic and could, if confirmed in humans, account for female preponderance in adrenocortical pathologies.


Adrenal Cortex/embryology , Adrenal Cortex/growth & development , Mice , Models, Animal , Adrenal Glands/embryology , Adrenal Glands/growth & development , Animals , Cell Differentiation , Female , Humans , Male , Mice, Knockout , Sexual Maturation/physiology
16.
JCI Insight ; 3(2)2018 01 25.
Article En | MEDLINE | ID: mdl-29367455

The adrenal cortex undergoes remodeling during fetal and postnatal life. How zona reticularis emerges in the postnatal gland to support adrenarche, a process whereby higher primates increase prepubertal androgen secretion, is unknown. Using cell-fate mapping and gene deletion studies in mice, we show that activation of PKA has no effect on the fetal cortex, while it accelerates regeneration of the adult cortex, triggers zona fasciculata differentiation that is subsequently converted into a functional reticularis-like zone, and drives hypersecretion syndromes. Remarkably, PKA effects are influenced by sex. Indeed, testicular androgens increase WNT signaling that antagonizes PKA, leading to slower adrenocortical cell turnover and delayed phenotype whereas gonadectomy sensitizes males to hypercorticism and reticularis-like formation. Thus, reticularis results from ultimate centripetal conversion of adult cortex under the combined effects of PKA and cell turnover that dictate organ size. We show that PKA-induced progenitor recruitment is sexually dimorphic and may provide a paradigm for overrepresentation of women in adrenal diseases.


Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Sex Characteristics , Signal Transduction/physiology , Zona Fasciculata/metabolism , Zona Reticularis/metabolism , Adrenarche/metabolism , Age Factors , Androgens/metabolism , Animals , Cell Differentiation/physiology , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Embryo, Mammalian , Female , Male , Mice , Mice, Knockout , Models, Animal
17.
Nat Commun ; 8(1): 445, 2017 09 05.
Article En | MEDLINE | ID: mdl-28874658

Advanced prostate cancer (PCa) is a clinical challenge as no curative therapeutic is available. In this context, a better understanding of metastasis and resistance mechanisms in PCa is an important issue. As phosphatase and tensin homolog (PTEN) loss is the most common genetic lesion in such cancer, we investigate human data sets for mechanisms that can constrain cancer evolution in this setting. Here we report a liver X receptor (LXR) signature, which tightly correlates with PTEN loss, in PCa. Accordingly, the LXR pathway is deregulated in prostate carcinomas in Pten-null mice. Genetic ablation of LXRs in Pten-null mice, exacerbates PCa invasiveness and metastatic dissemination, which involves mesenchymal transition and accumulation of matrix metalloproteinases. Mechanistically, PTEN deletion governed LXR transcriptional activity through deregulation of cholesterol de novo synthesis, resulting in accumulation of endogenous LXR ligands. Our study therefore reveals a functional circuit linking PTEN and LXR, and highlights LXRs as metabolic gatekeepers that are able to constrain PCa progression.Treatment of prostate cancer, especially in its advanced stage, is still challenging; therefore, strategies to prevent metastatic dissemination are of great interest. Here the authors reveal a crucial role for liver X receptors in suppressing prostate carcinogenesis and metastatic progression in PTEN-null tumors.


Liver X Receptors/genetics , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Signal Transduction/genetics , Animals , Cell Line, Tumor , Cells, Cultured , Cholesterol/metabolism , Disease Progression , Gene Expression Profiling/methods , Humans , Kaplan-Meier Estimate , Liver X Receptors/deficiency , Male , Mice, Knockout , Neoplasm Metastasis , PTEN Phosphohydrolase/deficiency , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
18.
Nat Commun ; 7: 12751, 2016 09 14.
Article En | MEDLINE | ID: mdl-27624192

Adrenal cortex physiology relies on functional zonation, essential for production of aldosterone by outer zona glomerulosa (ZG) and glucocorticoids by inner zona fasciculata (ZF). The cortex undergoes constant cell renewal, involving recruitment of subcapsular progenitors to ZG fate and subsequent lineage conversion to ZF identity. Here we show that WNT4 is an important driver of WNT pathway activation and subsequent ZG differentiation and demonstrate that PKA activation prevents ZG differentiation through WNT4 repression and WNT pathway inhibition. This suggests that PKA activation in ZF is a key driver of WNT inhibition and lineage conversion. Furthermore, we provide evidence that constitutive PKA activation inhibits, whereas partial inactivation of PKA catalytic activity stimulates ß-catenin-induced tumorigenesis. Together, both lower PKA activity and higher WNT pathway activity lead to poorer prognosis in adrenocortical carcinoma (ACC) patients. These observations suggest that PKA acts as a tumour suppressor in the adrenal cortex, through repression of WNT signalling.


Adrenal Gland Neoplasms/etiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Wnt Signaling Pathway , Zona Fasciculata/metabolism , Zona Glomerulosa/metabolism , Animals , Carcinogenesis , Cell Differentiation , Cell Line, Tumor , Female , Humans , Mice , Phosphorylation , Zona Fasciculata/cytology , Zona Glomerulosa/cytology , beta Catenin/metabolism
19.
Article En | MEDLINE | ID: mdl-27471492

Adrenal cortex tumors are divided into benign forms, such as primary hyperplasias and adrenocortical adenomas (ACAs), and malignant forms or adrenocortical carcinomas (ACCs). Primary hyperplasias are rare causes of adrenocorticotropin hormone-independent hypercortisolism. ACAs are the most common type of adrenal gland tumors and they are rarely "functional," i.e., producing steroids. When functional, adenomas result in endocrine disorders, such as Cushing's syndrome (hypercortisolism) or Conn's syndrome (hyperaldosteronism). By contrast, ACCs are extremely rare but highly aggressive tumors that may also lead to hypersecreting syndromes. Genetic analyses of patients with sporadic or familial forms of adrenocortical tumors (ACTs) led to the identification of potentially causative genes, most of them being involved in protein kinase A (PKA), Wnt/ß-catenin, and P53 signaling pathways. Development of mouse models is a crucial step to firmly establish the functional significance of candidate genes, to dissect mechanisms leading to tumors and endocrine disorders, and in fine to provide in vivo tools for therapeutic screens. In this article, we will provide an overview on the existing mouse models (xenografted and genetically engineered) of ACTs by focusing on the role of PKA and Wnt/ß-catenin pathways in this context. We will discuss the advantages and limitations of models that have been developed heretofore and we will point out necessary improvements in the development of next generation mouse models of adrenal diseases.

...