Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Mol Cell Endocrinol ; 408: 138-44, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25449416

ABSTRACT

SF-1 and LRH-1 are transcription factors that belong to the NR5A family of nuclear receptors that both have an essential role during development. Recent studies at the genome-wide scale have enabled the characterization of the cistrome and transcriptome regulated by SF-1 and LRH-1 in different cell lines and tissues. Those studies have allowed us to make a significant leap forward in our understanding of the mechanisms of transcriptional regulation of NR5A nuclear receptors in stem cells and cancer.


Subject(s)
Gene Expression Regulation , Genomics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription, Genetic , Animals , Humans , Models, Genetic , Organ Specificity/genetics
2.
Mol Cell Endocrinol ; 381(1-2): 66-9, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-23906534

ABSTRACT

Mitotane is a widely used drug in the therapy of adrenocortical carcinoma (ACC). It is important to set up preclinical protocols to study the possible synergistic effects of its association with new drugs for ACC therapy. We assessed the efficacy of different routes of administration of mitotane (i.p. and oral) in inhibiting growth of H295R ACC cell xenografts in an adjuvant setting. Both formulations of mitotane could inhibit H295R xenografts growth only at short times after carcinoma cells inoculation, even though plasma mitotane levels approached or fell within the therapeutic range in humans. Our results show that mitotane adjuvant therapy is inadequate to antagonize long-term growth of H295R cancer cells xenografts and that care should then be taken in the design of preclinical protocols to evaluate the performance of new drugs in association with mitotane.


Subject(s)
Adrenal Cortex Neoplasms/drug therapy , Adrenocortical Carcinoma/drug therapy , Antineoplastic Agents, Hormonal/administration & dosage , Mitotane/administration & dosage , Administration, Oral , Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/pathology , Animals , Antineoplastic Agents, Hormonal/pharmacokinetics , Cell Line, Tumor , Cell Proliferation/drug effects , Chemotherapy, Adjuvant , Drug Resistance, Neoplasm , Female , Humans , Inhibitory Concentration 50 , Injections, Intraperitoneal , Mice , Mice, Inbred NOD , Mice, SCID , Mitotane/pharmacokinetics , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
3.
Nucleic Acids Res ; 41(19): 8896-907, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23907384

ABSTRACT

Steroidogenic Factor-1 (SF-1) is a nuclear receptor that has a pivotal role in the development of adrenal glands and gonads and in the control of steroid hormone production, being also implicated in the pathogenesis of adrenocortical tumors. We have analyzed the mechanisms how SF-1 controls gene expression in adrenocortical cells and showed that it regulates different categories of genes according to its dosage. Significant correlations exist between the localization of SF-1-binding sites in chromatin under different dosage conditions and dosage-dependent regulation of gene expression. Our study revealed unexpected functional interactions between SF-1 and Neuron-Restrictive Silencer Factor/RE1-Silencing Transcription Factor (NRSF/REST), which was first characterized as a repressor of neuronal gene expression in non-neuronal tissues, in the regulation of gene expression in steroidogenic cells. When overexpressed, SF-1 reshapes the repertoire of NRSF/REST-regulated genes, relieving repression of key steroidogenic genes. These data show that NRSF/REST has a novel function in regulating gene expression in steroidogenic cells and suggest that it may have a broad role in regulating tissue-specific gene expression programs.


Subject(s)
Gene Expression Regulation, Neoplastic , Steroidogenic Factor 1/metabolism , Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/metabolism , Base Sequence , Binding Sites , Cell Line, Tumor , Chromatin/metabolism , DNA/chemistry , DNA/metabolism , Gene Dosage , Genome , Humans , Repressor Proteins/metabolism , Steroidogenic Factor 1/genetics
4.
Mol Cell Endocrinol ; 371(1-2): 154-9, 2013 May 22.
Article in English | MEDLINE | ID: mdl-23168267

ABSTRACT

Steroidogenic Factor-1 (SF-1) is a nuclear receptor transcription factor that has an essential role in the development of adrenal glands and gonads and in the regulation of steroidogenic gene expression. Recent studies using genomic approaches have revealed that SF-1 also has an important role in regulating proliferation of adrenocortical cells and have revealed its role in the control of a variety of biological processes as diverse as angiogenesis, adhesion to the extracellular matrix, cytoskeleton dynamics, transcriptional and post-transcriptional regulation of gene expression and apoptosis in the adrenal cortex. The identification of the complete set of SF-1 target genes will be of great importance to open new avenues for therapeutic intervention in adrenal diseases.


Subject(s)
Adrenal Cortex/metabolism , Gonads/metabolism , Steroidogenic Factor 1/metabolism , Adrenal Cortex Neoplasms/genetics , Apoptosis/genetics , Cell Adhesion , Cell Proliferation , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Gene Dosage , Gene Expression Regulation , Genomics , Humans , MicroRNAs/genetics , Neovascularization, Physiologic/genetics , Steroidogenic Factor 1/genetics , Steroids/biosynthesis
5.
Mol Cell Endocrinol ; 364(1-2): 101-4, 2012 Nov 25.
Article in English | MEDLINE | ID: mdl-22960230

ABSTRACT

Adrenocortical cancer is a rare malignancy for which current pharmacological therapies are still insufficient. We tested the effect of a novel PI3 kinase - mammalian target of rapamycin dual inhibitor (NVP-BEZ235) on proliferation of the H295R adrenocortical cancer cell line in vitro and grown as xenografts in immunodeficient mice. NVP-BEZ235 was able to significantly inhibit phosporylation of Akt kinase and S6 ribosomal protein in H295R cells and to significantly reduce their proliferation in vitro and xenograft growth in vivo. The drug also induced activation of Erk phosphorylation, which could be inhibited by simultaneous treatment with the Erk inhibitor FR180204. This latter drug synergized with NVP-BEZ235 in the inhibition of H295R proliferation in vitro. Our data suggest that dual PI3K/mTOR inhibitors may represent a useful pharmacological tool in the therapy of advanced adrenocortical cancer and that simultaneous inhibition of both Erk and PI3K - mTOR pathways may be required to obtain a higher antiproliferative effect in this type of tumor.


Subject(s)
Adrenal Cortex Neoplasms/drug therapy , Adrenocortical Carcinoma/drug therapy , Antineoplastic Agents/pharmacology , Imidazoles/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Quinolines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adrenal Cortex Neoplasms/metabolism , Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/metabolism , Adrenocortical Carcinoma/pathology , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mice , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation/drug effects , Pyrazoles/pharmacology , Pyridazines/pharmacology , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
6.
J Clin Endocrinol Metab ; 97(7): E1284-93, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22539591

ABSTRACT

CONTEXT: Childhood adrenocortical tumors (ACT) are rare malignancies, except in southern Brazil, where a higher incidence rate is associated to a high frequency of the founder R337H TP53 mutation. To date, copy number alterations in these tumors have only been analyzed by low-resolution comparative genomic hybridization. OBJECTIVE: We analyzed an international series of 25 childhood ACT using high-resolution single nucleotide polymorphism arrays to: 1) detect focal copy number alterations highlighting candidate driver genes; and 2) compare genetic alterations between Brazilian patients carrying the R337H TP53 mutation and non-Brazilian patients. RESULTS: We identified 16 significantly recurrent chromosomal alterations (q-value < 0.05), the most frequent being -4q34, +9q33-q34, +19p, loss of heterozygosity (LOH) of chromosome 17 and 11p15. Focal amplifications and homozygous deletions comprising well-known oncogenes (MYC, MDM2, PDGFRA, KIT, MCL1, BCL2L1) and tumor suppressors (TP53, RB1, RPH3AL) were identified. In addition, eight focal deletions were detected at 4q34, defining a sharp peak region around the noncoding RNA LINC00290 gene. Although non-Brazilian tumors with a mutated TP53 were similar to Brazilian tumors, those with a wild-type TP53 displayed distinct genomic profiles, with significantly fewer rearrangements (P = 0.019). In particular, three alterations (LOH of chromosome 17, +9q33-q34, and -4q34) were significantly more frequent in TP53-mutated samples. Finally, two of four TP53 wild-type tumors displayed as sole rearrangement a copy-neutral LOH of the imprinted region at 11p15, supporting a major role for this region in ACT development. CONCLUSIONS: Our findings highlight potential driver genes and cellular pathways implicated in childhood ACT and demonstrate the existence of different oncogenic routes in this pathology.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenocortical Adenoma/genetics , Adrenocortical Carcinoma/genetics , Cell Transformation, Neoplastic/genetics , Microarray Analysis , Polymorphism, Single Nucleotide , Adolescent , Adrenal Cortex Neoplasms/complications , Adrenal Cortex Neoplasms/epidemiology , Adrenocortical Adenoma/complications , Adrenocortical Adenoma/epidemiology , Adrenocortical Carcinoma/complications , Adrenocortical Carcinoma/epidemiology , Age of Onset , Child , Child, Preschool , Cohort Studies , Female , Gene Expression Profiling , Genetic Association Studies , Humans , Infant , Infant, Newborn , Male , Polymorphism, Single Nucleotide/physiology , Signal Transduction/genetics
7.
Am J Cancer Res ; 1(3): 356-361, 2011.
Article in English | MEDLINE | ID: mdl-21968616

ABSTRACT

Adrenocortical carcinoma (ACC) is a very aggressive tumor with a poor prognosis. Available treatments for this type of cancer are far from being satisfactory. The IGF signalling pathway represents an important mechanism for ACT growth and constitutes a relevant therapeutic target. We investigated the effect of picropodophyllin (PPP), a member of the cyclolignan family and a new inhibitor of IGF-1R, on proliferation of human adrenocortical cell lines H295R and SW-13. PPP inhibits proliferation and induces an important accumulation in G2/M phase and apoptosis of H295R and SW-13 cells. Our data suggest that PPP may be a promising candidate for drug development for adrenocortical carcinoma.

8.
Mol Cell Endocrinol ; 336(1-2): 169-73, 2011 Apr 10.
Article in English | MEDLINE | ID: mdl-21094206

ABSTRACT

Adrenocortical tumors in children are usually diagnosed because of signs of virilization and their prognosis is poor. They possess several distinct pathological features compared to adrenocortical tumors in adults and have an exceptional prevalence in southern Brazil, where they are nearly invariably linked to the presence of a germline specific TP53 (R337H) mutation. Other important factors in childhood adrenocortical tumor pathogenesis are overexpression of the Steroidogenic Factor-1 transcription factor and imprinting defects in the 11p15 genomic region, causing overexpression of Insulin-like Growth Factor-2. Genomic studies have revealed the prognostic relevance of the expression of some Major Histocompatibility Complex genes and the deregulation of the Insulin-like Growth Factor/mammalian Target Of Rapamycin pathway by microRNAs in these tumors. Our hope is that these findings will constitute the basis for the development of novel therapies that will be more active against these tumors and less toxic for the patients.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Genomics , Child , Chromosomes, Human, Pair 11/genetics , Gene Expression Profiling , Genomic Imprinting/genetics , Humans , Signal Transduction/genetics , Somatomedins/genetics , Somatomedins/metabolism , Steroidogenic Factor 1/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/genetics
9.
Methods Enzymol ; 485: 3-23, 2010.
Article in English | MEDLINE | ID: mdl-21050908

ABSTRACT

The transcription factor Steroidogenic Factor-1 (Ad4BP/SF-1; NR5A1 according to the standard nomenclature) has an essential role in adrenogonadal development. Furthermore, SF-1 is amplified and overexpressed in most cases of adrenocortical tumor occurring in children; studies performed in transgenic mice have shown that an increased SF-1 dosage triggers tumor formation in the adrenal cortex. For these reasons, drugs interfering with SF-1 action would represent a promising tool to be added to the current pharmacological protocols in the therapy of adrenocortical cancer. Here, we describe the methods how isoquinolinone compounds inhibiting the constitutive transcriptional activity of SF-1 (SF-1 inverse agonists) were identified and characterized. These compounds have the attributes to inhibit the increase in proliferation triggered by an augmented SF-1 dosage in adrenocortical tumor cells and to reduce their steroid production. This latter property may also reveal beneficial for drugs used in the therapy of adrenocortical tumors to alleviate symptoms of virilization and Cushing often associated with tumor burden.


Subject(s)
Adrenal Cortex Neoplasms/drug therapy , Antineoplastic Agents/agonists , Antineoplastic Agents/chemistry , Drug Inverse Agonism , Isoquinolines/agonists , Isoquinolines/chemistry , Steroidogenic Factor 1/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Isoquinolines/pharmacology , Steroids/metabolism , Structure-Activity Relationship , Transcriptional Activation/drug effects , Transfection/methods
10.
Cancer Res ; 70(11): 4666-75, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20484036

ABSTRACT

MicroRNAs (miRNAs) act at the posttranscriptional level to control gene expression in virtually every biological process, including oncogenesis. Here, we report the identification of a set of miRNAs that are differentially regulated in childhood adrenocortical tumors (ACT), including miR-99a and miR-100. Functional analysis of these miRNAs in ACT cell lines showed that they coordinately regulate expression of the insulin-like growth factor-mammalian target of rapamycin (mTOR)-raptor signaling pathway through binding sites in their 3'-untranslated regions. In these cells, the active Ser(2448)-phosphorylated form of mTOR is present only in mitotic cells in association with the mitotic spindle and midbody in the G(2)-M phases of the cell cycle. Pharmacologic inhibition of mTOR signaling by everolimus greatly reduces tumor cell growth in vitro and in vivo. Our results reveal a novel mechanism of regulation of mTOR signaling by miRNAs, and they lay the groundwork for clinical evaluation of drugs inhibiting the mTOR pathway for treatment of adrenocortical cancer.


Subject(s)
Adrenal Cortex Neoplasms/metabolism , Adrenocortical Adenoma/metabolism , Adrenocortical Carcinoma/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , MicroRNAs/metabolism , Protein Serine-Threonine Kinases/metabolism , Somatomedins/metabolism , Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/pathology , Adrenocortical Adenoma/genetics , Adrenocortical Adenoma/pathology , Adrenocortical Carcinoma/genetics , Adrenocortical Carcinoma/pathology , Animals , Cell Growth Processes/physiology , Everolimus , Female , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Transplantation, Heterologous
11.
Int J Oncol ; 36(4): 983-90, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20198344

ABSTRACT

The Li-Fraumeni syndrome (LFS) is a rare autosomal dominant hereditary cancer syndrome, characterized by a wide spectrum of neoplasms, occurring in children and young adults. The identification of germline TP53 mutations in LFS has given rise to a number of in vitro studies using cultures of cancer cells and non-tumoral fibroblasts presenting germline TP53 mutations. In the present study, we performed a detailed documentation of the pedigree of an LFS family with a comprehensive analysis of genotype-phenotype correlations. We sequenced the TP53 gene and verified that the proband carries a germline nonsense mutation in codon 146 in one allele, the TP53Arg72Pro polymorphism in the second, and other intronic polymorphisms in the TP53 gene. In order to investigate the disruption of the p53 function in a patient presenting this mutation and the TP53Arg72Pro polymorphism who had so far suffered five malignant tumors and a benign meningioma, we tested her fibroblasts in response to DNA damage by evaluating the proliferation rate, apoptosis, and disruption of the TP53 pathway. The proband's heterozygous fibroblasts were not as efficient as control fibroblasts or those of her mother, who carried only the TP53Arg72Pro polymorphism, in causing cell arrest and cell death after DNA damage, which was correlated with diminished TP21 protein levels.


Subject(s)
Codon, Nonsense , DNA Damage , Fibroblasts/metabolism , Germ-Line Mutation , Li-Fraumeni Syndrome/genetics , Tumor Suppressor Protein p53/genetics , Adult , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Cell Cycle , Cell Proliferation , Cells, Cultured , Codon, Terminator , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Etoposide/pharmacology , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Fibroblasts/radiation effects , Genotype , Heterozygote , Humans , Li-Fraumeni Syndrome/metabolism , Li-Fraumeni Syndrome/pathology , Male , Middle Aged , Pedigree , Phenotype , Polymorphism, Genetic , Tumor Suppressor Protein p53/metabolism
12.
J Clin Endocrinol Metab ; 94(6): 2178-83, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19318454

ABSTRACT

CONTEXT: Transcription factor steroidogenic factor-1 (SF-1) plays a pivotal role in the control of adrenocortical cell steroidogenesis and proliferation. SF-1 amplification and overexpression are found in most cases of childhood adrenocortical tumors (ACTs). OBJECTIVE: Our objective was to investigate the effect of SF-1 inverse agonists of the alkyloxyphenol and isoquinolinone classes on the proliferation of human adrenocortical cell lines expressing SF-1 (H295R), in conditions of basal and increased SF-1 expression, or negative for SF-1 expression (SW-13). MAIN OUTCOME MEASURES: Proliferation assays, immunoblots, flow cytometric analyses, steroid hormone assays, and reverse transcription quantitative PCR were used. RESULTS: SF-1 inhibitors of the alkyloxyphenol class displayed a dose-dependent inhibitory effect on both SF-1-positive and -negative ACT cells, whereas SF-1 inverse agonists of the isoquinolinone class selectively inhibited cell proliferation elicited by SF-1 overexpression. These drugs also inhibited stimulated steroid hormone secretion and CYP21 and CYP17 mRNA expression. CONCLUSION: SF-1 inhibitors may represent a useful tool in the chemotherapy of ACTs.


Subject(s)
Adrenal Cortex Neoplasms/drug therapy , Adrenocortical Carcinoma/drug therapy , Cell Proliferation/drug effects , Phenols/therapeutic use , Steroidogenic Factor 1/agonists , Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/pathology , Drug Evaluation, Preclinical , Drug Inverse Agonism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Phenols/pharmacology , Quinolones/pharmacology , Steroids/metabolism , Tumor Cells, Cultured
13.
Cancer Genet Cytogenet ; 186(1): 19-24, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18786438

ABSTRACT

A germline TP53 R337H mutation is present in childhood adrenocortical tumors (ACT) from southern Brazil. Other genetic alterations are also frequently found in these tumors. This study was designed to assess whether alterations of the 11p15 region exist in childhood ACT, accounting for IGF2 overexpression in these tumors, and how they are related to clinical outcome. Tumor DNA of 12 children with ACT (4 adenomas and 8 carcinomas) and from the blood of their parents was analyzed. All patients showed 11p15 loss of heterozygosity (LOH) in the tumor. In contrast to the single case of paternal LOH, IGF2 was overexpressed in tumors with maternal allele loss. Our data show that 11p15 LOH is a widespread finding in childhood ACT not related with malignancy, contrary to adult ACT. Alterations in the expression of other genes in the same region (e.g., CDKN1C) may contribute to ACT tumorigenesis.


Subject(s)
Adenoma/genetics , Adrenal Cortex Neoplasms/genetics , Amino Acid Substitution , Carcinoma/genetics , Chromosomes, Human, Pair 11/genetics , Genes, p53/genetics , Insulin-Like Growth Factor II/genetics , Loss of Heterozygosity , Mutation, Missense , Neoplasm Proteins/genetics , Neoplastic Syndromes, Hereditary/genetics , Point Mutation , Adenoma/epidemiology , Adenoma/mortality , Adolescent , Adrenal Cortex Neoplasms/epidemiology , Adrenal Cortex Neoplasms/mortality , Age Factors , Brazil/epidemiology , Carcinoma/epidemiology , Carcinoma/mortality , Child , Child, Preschool , Chromosomes, Human, Pair 11/ultrastructure , DNA Methylation , Gene Expression Regulation, Neoplastic , Genomic Imprinting , Germ-Line Mutation , Humans , Infant , Insulin-Like Growth Factor II/biosynthesis , Neoplasm Proteins/biosynthesis , Neoplastic Syndromes, Hereditary/epidemiology , Neoplastic Syndromes, Hereditary/mortality , Treatment Outcome
14.
J Clin Endocrinol Metab ; 93(8): 3222-5, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18544621

ABSTRACT

CONTEXT: Mutations of the beta-catenin (CTNNB1) gene are frequently found in adrenocortical tumors. This has important consequences to deregulate the expression of transcriptional targets of the Wnt pathway, which may contribute to tumorigenesis. OBJECTIVE: The objective of the study was to investigate the effect of the small-molecule inhibitor of the T cell factor (Tcf)/beta-catenin complex PKF115-584 on beta-catenin-dependent transcription and proliferation of H295R adrenocortical tumor cells, which harbor mutations in CTNNB1 as well as the TP53 tumor suppressor gene. MAIN OUTCOME MEASURES: Immunofluorescence, transient transfection, proliferation assays, and flow cytometric analyses were used. RESULTS: Nuclear localization of beta-catenin and constitutive activation of beta-catenin-dependent transcription was observed in H295R cells. PKF115-584 dose-dependently inhibited beta-catenin-dependent transcription and H295R proliferation, even in the presence of increased steroidogenic factor-1 levels, which augment proliferation in this cell line. The drug had no effect on HeLa cells, a cell line in which the beta-catenin pathway is not activated. PKF115-584 decreased the percentage of H295R cells in S-phase and increased the percentage of apoptotic cells. CONCLUSIONS: Inhibitors of the Tcf/beta-catenin complex may prove useful in the treatment of adrenocortical tumors in which multiple genetic alterations have accumulated.


Subject(s)
Adrenal Cortex Neoplasms/drug therapy , TCF Transcription Factors/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/pathology , Cell Proliferation/drug effects , Genes, p53 , Humans , Mutation , Tumor Cells, Cultured , beta Catenin/genetics
15.
Mol Endocrinol ; 21(12): 2968-87, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17761949

ABSTRACT

Steroidogenic factor-1 (SF-1/Ad4BP; NR5A1), a nuclear receptor transcription factor, has a pivotal role in adrenal and gonadal development in humans and mice. A frequent feature of childhood adrenocortical tumors is SF-1 amplification and overexpression. Here we show that an increased SF-1 dosage can by itself augment human adrenocortical cell proliferation through concerted actions on the cell cycle and apoptosis. This effect is dependent on an intact SF-1 transcriptional activity. Gene expression profiling showed that an increased SF-1 dosage regulates transcripts involved in steroid metabolism, the cell cycle, apoptosis, and cell adhesion to the extracellular matrix. Consistent with these results, increased SF-1 levels selectively modulate the steroid secretion profile of adrenocortical cells, reducing cortisol and aldosterone production and maintaining dehydroepiandrosterone sulfate secretion. As a model to understand the mechanisms of transcriptional regulation by increased SF-1 dosage, we studied FATE1, coding for a cancer-testis antigen implicated in the control of cell proliferation. Increased SF-1 levels increase its binding to a consensus site in FATE1 promoter and stimulate its activity through modulation of the recruitment of specific cofactors. On the other hand, sphingosine, which can compete with phospholipids for binding to SF-1, had no effect on the SF-1 dosage-dependent increase of adrenocortical cell proliferation and expression of the FATE1 promoter. In mice, increased Sf-1 dosage produces adrenocortical hyperplasia and formation of tumors expressing gonadal markers (Amh, Gata-4), which originate from the subcapsular region of the adrenal cortex. Gene expression profiling revealed that genes involved in cell adhesion and the immune response and transcription factor signal transducer and activator of transcription-3 (Stat3) are differentially expressed in Sf-1 transgenic mouse adrenals compared with wild-type adrenals. Our studies reveal a critical role for SF-1 dosage in adrenocortical tumorigenesis and constitute a rationale for the development of drugs targeting SF-1 transcriptional activity for adrenocortical tumor therapy.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Neoplasms/pathology , Steroidogenic Factor 1/metabolism , Animals , Base Sequence , Binding Sites , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Extracellular Matrix/metabolism , Histidine/genetics , Histidine/metabolism , Humans , Lipid Metabolism , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation/genetics , Neoplasms/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Steroidogenic Factor 1/genetics , Steroids/metabolism , Transcription, Genetic/genetics , Up-Regulation
16.
J Clin Endocrinol Metab ; 92(8): 3253-60, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17566092

ABSTRACT

CONTEXT: Childhood adrenocortical tumors (ACTs) have a fetal adrenal phenotype and overexpress steroidogenic factor-1 (SF-1). Nephroblastoma overexpressed (NOV)/cysteine-rich protein 61/connective tissue growth factor/nephroblastoma overexpressed gene-3 mRNA is significantly down-regulated in childhood ACTs. OBJECTIVE: The objective of the study was to measure NOV protein levels in childhood ACTs and characterize NOV expression regulation and biological function in human adrenocortical cells. DESIGN AND SETTING: Protein extracts from ACT and normal adrenal cortex samples, human adrenocortical carcinoma H295R, primary adrenocortical tumors and fetal adrenal cultures, tissue culture supernatants, and cell lysates from H295R cells overexpressing SF-1 in an inducible fashion were used. MAIN OUTCOME MEASURES: NOV protein levels were measured by enzyme-linked immunoassay and immunoblot. Transient transfection assays were used to study the activity of NOV promoter. Terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling, caspase assays, and flow cytometry were used to assess the proapoptotic activity of NOV on cells in culture. RESULTS: NOV mRNA and protein expression is lower in childhood ACTs than in normal adrenal cortex. No significant difference was observed between adenomas and carcinomas. SF-1 overexpression down-regulates NOV at the transcriptional level. NOV has a selective proapoptotic activity toward human adrenocortical cells. The C-terminal domain of NOV is responsible for its proapoptotic effect. NOV protein is expressed in DAX-1-positive human fetal adrenal cells. CONCLUSIONS: NOV is a selective proapoptotic factor for human adrenocortical cells. Reduced expression of NOV in ACTs may play an important role in the process of childhood ACT tumorigenesis, accounting at least in part for the defect of apoptotic regression of the fetal adrenal that has been proposed to be responsible for tumor formation.


Subject(s)
Adenoma/metabolism , Adrenal Cortex Neoplasms/metabolism , Adrenal Cortex/cytology , Apoptosis/genetics , Apoptosis/physiology , Carcinoma/metabolism , Gene Expression Regulation, Neoplastic/genetics , Immediate-Early Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Adenoma/genetics , Adenoma/pathology , Adrenal Cortex/physiology , Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/pathology , Carcinoma/genetics , Carcinoma/pathology , Caspases/metabolism , Cell Line, Tumor , Child , Connective Tissue Growth Factor , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Down-Regulation/genetics , Down-Regulation/physiology , Enzyme Activation/physiology , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/physiology , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Immediate-Early Proteins/biosynthesis , Immunoblotting , Intercellular Signaling Peptides and Proteins/biosynthesis , Luciferases/biosynthesis , Luciferases/genetics , Nephroblastoma Overexpressed Protein , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics , Reverse Transcriptase Polymerase Chain Reaction , Steroidogenic Factor 1 , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transfection
17.
Mol Cell Endocrinol ; 265-266: 108-12, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17208361

ABSTRACT

The levels of Agouti-related protein (AgRP) mRNA in the adrenal are second only to those in the hypothalamus, raising questions regarding its target binding sites and its specific role in adrenal steroidogenesis. We and others demonstrated the presence of a population of melanocortin receptor-4 (MC4R) positively coupled to steroidogenesis in adrenal cells. Moreover, AgRP inhibited both the acute and long-term steroidogenic responses of these cells to NDP-alphaMSH through its antagonistic properties towards MC4R. Although AgRP had no antagonistic properties towards the MC2R and did not modify the acute steroidogenic response to ACTH, it exerted a biphasic sustained inhibitory effect on the long-term response to ACTH through an undefined alternate mechanism. Since adrenal cells release a relatively large amount of AgRP, this protein likely exerts a local paracrine/autocrine control on adrenal steroidogenesis.


Subject(s)
Adrenal Glands/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Steroids/biosynthesis , Adrenal Glands/cytology , Agouti-Related Protein , Animals , Humans , Paracrine Communication , Receptors, Melanocortin/metabolism
18.
Eur J Cancer ; 42(8): 1040-3, 2006 May.
Article in English | MEDLINE | ID: mdl-16574405

ABSTRACT

The steroidogenic factor 1 (SF-1) gene encodes a transcription factor playing a pivotal role in the regulation of adrenogenital development. We have recently shown that SF-1 is amplified in childhood adrenocortical tumours (ACT). This study was aimed to assess if an increase in SF-1 gene copy number was associated with increased protein levels and to study the correlation between SF-1 expression and ACT clinical parameters. An increased SF-1 copy number was detected in eight of the 10 ACT cases studied. Conversely, the SF-1 protein was found to be overexpressed in all cases, compared to normal age-matched adrenal glands. No significant correlation was found between SF-1 protein levels and its gene copy number. Furthermore, no significant correlation existed with histological grade or with the clinical manifestation or evolution of disease. This data show that SF-1 overexpression is widespread in childhood ACT and is likely to play a role in its pathogenesis.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Homeodomain Proteins/genetics , Neoplasm Proteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Adrenal Cortex Neoplasms/metabolism , Case-Control Studies , Child , Child, Preschool , Diploidy , Female , Germ-Line Mutation/genetics , Homeodomain Proteins/metabolism , Humans , In Situ Hybridization, Fluorescence , Infant , Loss of Heterozygosity , Male , Neoplasm Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Steroidogenic Factor 1 , Transcription Factors/metabolism
19.
Regul Pept ; 124(1-3): 215-9, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15544862

ABSTRACT

The adrenal gland is the second tissue after hypothalamus exhibiting high expression level of Agouti Related Protein (Agrp) mRNA, which suggests that this peptide may control adrenal cell functions. However, its role in this tissue remained to be determined. In this report, we studied the effects of a long-term treatment (24 h) of cultured bovine adrenal cells by Agrp on the (Nle4, d-Phe7)-alphaMSH (NDP-alphaMSH)- or ACTH-induced cortisol release. We showed that Agrp inhibited, in a dose-dependent manner, the 10(-9) M NDP-alphaMSH-induced cortisol production through its antagonistic properties towards MSH at the level of MC4-R. Surprisingly, we found that Agrp in the same conditions of cell treatment also induced a strong inhibition of the ACTH-induced cortisol release. These effects were stronger using low concentrations of Agrp and disappeared for higher concentrations resulting in U-shaped curve data. There was no effect of SHU9119 in the same conditions of stimulation of the cells. Our data confirmed that Agrp is not an antagonist of ACTH at the level of MC2-R and that its sustained effect on ACTH-induced steroidogenesis did not involve its antagonistic properties at the level of MC4-R. The hypothesis would be that Agrp is acting on adrenal steroidogenesis through an alternate mechanism.


Subject(s)
Adrenal Glands/drug effects , Adrenal Glands/metabolism , Adrenocorticotropic Hormone/pharmacology , Hydrocortisone/biosynthesis , Intercellular Signaling Peptides and Proteins/metabolism , Adrenal Glands/cytology , Adrenocorticotropic Hormone/antagonists & inhibitors , Agouti Signaling Protein , Animals , Cattle , Cells, Cultured , Glucocorticoids/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Leptin/pharmacology , Time Factors , alpha-MSH/analogs & derivatives , alpha-MSH/pharmacology
20.
J Biochem ; 135(4): 541-6, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15115780

ABSTRACT

The melanocortin receptor type 4 (MC4-R) is involved in food intake and represents a potential target for the treatment of some forms of obesity. The fluorescent protein EGFP was fused to the wild-type or mutated coding sequence of the human MC4-R. After transfection in HEK 293, clones stably expressing hMC4-R-EGFP were selected. Wild-type chimeric hMC4-R was well addressed to the cell membrane as demonstrated using confocal microscopy and displayed the same pharmacological characteristics as native hMC4R. NDP-alpha MSH induced a time-dependent internalization of MC4-R that was partially prevented by AgRP. The two mutated chimeric receptors studied here (CTCT-deleted and C271A) showed a high alteration of their response to ligand and were retained inside the cells. In conclusion, we have developed a model of clones stably expressing EGFP-tagged-hMC4-R. This is the only such model available to date and it provides a useful tool to follow the trafficking of MC4-R inside living cells.


Subject(s)
Green Fluorescent Proteins/genetics , Receptor, Melanocortin, Type 4/genetics , alpha-MSH/analogs & derivatives , Agouti-Related Protein , Binding, Competitive , Cell Line , Cell Membrane/metabolism , Cyclic AMP/metabolism , Cytoplasmic Vesicles/metabolism , Dose-Response Relationship, Drug , Genetic Vectors/genetics , Green Fluorescent Proteins/metabolism , Humans , Microscopy, Fluorescence , Mutation , Peptide Fragments/pharmacology , Polymerase Chain Reaction , Protein Binding , Protein Transport/drug effects , Protein Transport/genetics , Protein Transport/physiology , Receptor, Melanocortin, Type 4/metabolism , Receptors, Melanocortin/agonists , Receptors, Melanocortin/antagonists & inhibitors , Receptors, Melanocortin/metabolism , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , Transfection , alpha-MSH/metabolism , alpha-MSH/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...