Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
N Engl J Med ; 370(11): 1019-28, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24571724

ABSTRACT

BACKGROUND: Corticotropin-independent Cushing's syndrome is caused by tumors or hyperplasia of the adrenal cortex. The molecular pathogenesis of cortisol-producing adrenal adenomas is not well understood. METHODS: We performed exome sequencing of tumor-tissue specimens from 10 patients with cortisol-producing adrenal adenomas and evaluated recurrent mutations in candidate genes in an additional 171 patients with adrenocortical tumors. We also performed genomewide copy-number analysis in 35 patients with cortisol-secreting bilateral adrenal hyperplasias. We studied the effects of these genetic defects both clinically and in vitro. RESULTS: Exome sequencing revealed somatic mutations in PRKACA, which encodes the catalytic subunit of cyclic AMP-dependent protein kinase (protein kinase A [PKA]), in 8 of 10 adenomas (c.617A→C in 7 and c.595_596insCAC in 1). Overall, PRKACA somatic mutations were identified in 22 of 59 unilateral adenomas (37%) from patients with overt Cushing's syndrome; these mutations were not detectable in 40 patients with subclinical hypercortisolism or in 82 patients with other adrenal tumors. Among 35 patients with cortisol-producing hyperplasias, 5 (including 2 first-degree relatives) carried a germline copy-number gain (duplication) of the genomic region on chromosome 19 that includes PRKACA. In vitro studies showed impaired inhibition of both PKA catalytic subunit mutants by the PKA regulatory subunit, whereas cells from patients with germline chromosomal gains showed increased protein levels of the PKA catalytic subunit; in both instances, basal PKA activity was increased. CONCLUSIONS: Genetic alterations of the catalytic subunit of PKA were found to be associated with human disease. Germline duplications of this gene resulted in bilateral adrenal hyperplasias, whereas somatic PRKACA mutations resulted in unilateral cortisol-producing adrenal adenomas. (Funded by the European Commission Seventh Framework Program and others.).


Subject(s)
Adenoma/genetics , Adrenal Gland Neoplasms/genetics , Adrenal Hyperplasia, Congenital/genetics , Cushing Syndrome/etiology , Cyclic AMP-Dependent Protein Kinases/genetics , Germ-Line Mutation , Adenoma/complications , Adenoma/enzymology , Adrenal Gland Neoplasms/complications , Adrenal Gland Neoplasms/enzymology , Adult , Catalytic Domain , Cushing Syndrome/enzymology , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Exome , Humans , Hydrocortisone/biosynthesis , Middle Aged , Mutation , Protein Conformation , Sequence Analysis, DNA
2.
Pituitary ; 18(2): 274-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25612787

ABSTRACT

BACKGROUND: Cushing's disease (CD) is caused by adrenocorticotropic hormone (ACTH)-producing pituitary adenomas (ACTHomas). Drug treatment for CD consists of three strategies: pituitary tumor-targeted therapy, steroidogenesis inhibitors, and glucocorticoid receptor antagonists. All of these strategies are under development, and several new drugs have recently been approved for clinical use or are being tested in clinical trials. Pituitary-targeted drugs are a particularly important method in the treatment of CD. Available pituitary tumor-targeted drugs include a dopamine receptor agonist and a somatostatin analog. Since disrupted cell cycle signaling is clearly associated with pathogenesis of ACTHomas which express active forms of epithelial growth factor receptor (EGFR), cyclins, and the catalytic subunit of cyclin-dependent kinases (CDKs), we focused on these molecules as therapeutic targets for ACTHomas. METHODS: In this review, a literature search were performed using PubMed with following terms; Cushing's disease, EGFR, CDKs, cell cycle, and targeted therapy. CONCLUSION: Accumulating evidence demonstrates that EGFR and cyclin E-CDK2 may be promising targets for treating ACTHomas.


Subject(s)
ACTH-Secreting Pituitary Adenoma/drug therapy , Adenoma/drug therapy , Antineoplastic Agents/therapeutic use , Cushing Syndrome/drug therapy , Cyclin-Dependent Kinases/antagonists & inhibitors , ErbB Receptors/antagonists & inhibitors , Pituitary Gland/drug effects , Protein Kinase Inhibitors/therapeutic use , ACTH-Secreting Pituitary Adenoma/complications , ACTH-Secreting Pituitary Adenoma/diagnosis , ACTH-Secreting Pituitary Adenoma/enzymology , Adenoma/complications , Adenoma/diagnosis , Adenoma/enzymology , Animals , Cushing Syndrome/diagnosis , Cushing Syndrome/enzymology , Cushing Syndrome/etiology , Cyclin-Dependent Kinases/metabolism , ErbB Receptors/metabolism , Humans , Molecular Targeted Therapy , Pituitary Gland/enzymology , Pituitary Gland/pathology , Signal Transduction/drug effects , Treatment Outcome
3.
Nat Genet ; 38(7): 794-800, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16767104

ABSTRACT

Phosphodiesterases (PDEs) regulate cyclic nucleotide levels. Increased cyclic AMP (cAMP) signaling has been associated with PRKAR1A or GNAS mutations and leads to adrenocortical tumors and Cushing syndrome. We investigated the genetic source of Cushing syndrome in individuals with adrenocortical hyperplasia that was not caused by known defects. We performed genome-wide SNP genotyping, including the adrenocortical tumor DNA. The region with the highest probability to harbor a susceptibility gene by loss of heterozygosity (LOH) and other analyses was 2q31-2q35. We identified mutations disrupting the expression of the PDE11A isoform-4 gene (PDE11A) in three kindreds. Tumor tissues showed 2q31-2q35 LOH, decreased protein expression and high cyclic nucleotide levels and cAMP-responsive element binding protein (CREB) phosphorylation. PDE11A codes for a dual-specificity PDE that is expressed in adrenal cortex and is partially inhibited by tadalafil and other PDE inhibitors; its germline inactivation is associated with adrenocortical hyperplasia, suggesting another means by which dysregulation of cAMP signaling causes endocrine tumors.


Subject(s)
Adrenal Glands/enzymology , Adrenal Glands/pathology , Mutation , Phosphoric Diester Hydrolases/genetics , 3',5'-Cyclic-GMP Phosphodiesterases , Adult , Child , Chromosomes, Human, Pair 2/genetics , Cushing Syndrome/enzymology , Cushing Syndrome/genetics , Cushing Syndrome/pathology , Female , Humans , Hyperplasia , Loss of Heterozygosity , Male , Phosphoric Diester Hydrolases/metabolism , Polymorphism, Single Nucleotide
4.
Horm Metab Res ; 46(12): 863-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25232906

ABSTRACT

The majority of benign adrenal cortex lesions leading to Cushing syndrome are associated to one or another abnormality of the cAMP/cGMP-phosphodiesterase signaling pathway. Phosphodiesterases (PDEs) are key regulatory enzymes of intracellular cAMP/cGMP levels. These second messengers play important regulatory roles in controlling steroidogenesis in the adrenal. Disruption of PDEs has been associated with a number of adrenal diseases. Specifically, genetic mutations have been associated with benign adrenal lesions, leading to Cushing syndrome and/or related adrenal hyperplasias. A Genome Wide Association study, in 2006, led to the identification of mutations in 2 PDE genes: PDE8B and PDE11A; mutations in these 2 genes modulate steroidogenesis. Further human studies have identified PDE2 as also directly regulating steroidogenesis. PDE2 decreases aldosterone production. This review focuses on the most recent knowledge we have gained on PDEs and their association with adrenal steroidogenesis and altered function, through analysis of patient cohorts and what we have learned from mouse studies.


Subject(s)
Adrenal Glands/enzymology , Adrenal Glands/pathology , Phosphoric Diester Hydrolases/metabolism , Adrenal Gland Neoplasms/enzymology , Adrenal Gland Neoplasms/pathology , Animals , Cushing Syndrome/enzymology , Humans , Hyperplasia , Mice , Phosphoric Diester Hydrolases/genetics , Signal Transduction
5.
Horm Metab Res ; 45(5): 329-31, 2013 May.
Article in English | MEDLINE | ID: mdl-23549672

ABSTRACT

The fact that obesity is a prominent feature of Cushing's syndrome (systemic hypercortisolism of adrenocortical origin) stimulated a 40-year search for evidence of systemic hypercortisolism in human obesity. That search has failed to find such evidence. For the past 15 years, however, studies have been done to evaluate a possible alternative type of hypercortisolism in obesity, namely visceral adipose tissue (VAT) intracellular hypercortisolism. The current review summarizes the evidence published so far about this possibility. There have been three types of evidence studied: direct measurement of the VAT levels of 11ß-hydroxysteroid dehydrogenase type I (11-HSD-1), which converts biologically inactive cortisone to biologically active cortisol; direct measurement of splanchnic cortisol production; and evaluation of the effect of a specific inhibitor of 11-HSD-1 on metabolic abnormalities associated with obesity, particularly diabetes mellitus. The results are complex and difficult to interpret. Our conclusion is that the presence of VAT intracellular hypercortisolism in human obesity is possible but unlikely.


Subject(s)
Cushing Syndrome/complications , Intra-Abdominal Fat/metabolism , Intracellular Space/metabolism , Obesity/complications , 11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Cushing Syndrome/blood , Cushing Syndrome/enzymology , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Humans , Hydrocortisone/blood , Obesity/blood , Obesity/enzymology
6.
Endocr J ; 60(3): 329-36, 2013.
Article in English | MEDLINE | ID: mdl-23257735

ABSTRACT

We analyzed the expression profiles of several steroidogenic enzymes in normal adrenals, aldosterone-producing adenomas (APA), cortisol-producing adenomas combined with Cushing's syndrome (CPA) or with subclinical Cushing's syndrome (SCPA), and nonfunctioning adrenal adenomas (NFA) to clarify the nature and characteristics of steroidogenesis in APA. Clinical data were collected for all subjects. In resected adrenal glands (normal adrenals, APA, CPA, SCPA, and NFA), the mRNA expression levels of the CYP17, HSD3B2, CYP11B1, and CYP11B2 genes were studied using real-time quantitative PCR and immunohistochemistry. The CYP11B2 mRNA level in APA was significantly higher than that in other groups. The CYP17/HSD3B2 ratio for mRNA in APA was significantly lower than those in the other groups. Low ratio of CYP17/HSD3B2 with high expression of CYP11B2 seems to explain steroidogenic characteristics of APA.


Subject(s)
Adenoma/enzymology , Adrenal Gland Neoplasms/enzymology , Aldosterone/biosynthesis , Enzymes/genetics , Gene Expression , Steroids/biosynthesis , Adenoma/metabolism , Adrenal Glands/enzymology , Adult , Aged , Cushing Syndrome/enzymology , Cytochrome P-450 CYP11B2/genetics , Female , Humans , Hydrocortisone/biosynthesis , Immunohistochemistry , Male , Middle Aged , Progesterone Reductase/genetics , RNA, Messenger/analysis , Steroid 11-beta-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/genetics
7.
Diabetes Obes Metab ; 14(10): 869-81, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22321826

ABSTRACT

Recent evidence strongly argues for a pathogenic role of glucocorticoids and 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) in obesity and the metabolic syndrome, a cluster of risk factors for atherosclerotic cardiovascular disease and type 2 diabetes mellitus (T2DM) that includes insulin resistance (IR), dyslipidaemia, hypertension and visceral obesity. This has been partially prompted not only by the striking clinical resemblances between the metabolic syndrome and Cushing's syndrome (a state characterized by hypercortisolism that associates with metabolic syndrome components) but also from monogenic rodent models for the metabolic syndrome (e.g. the leptin-deficient ob/ob mouse or the leptin-resistant Zucker rat) that display overall increased secretion of glucocorticoids. However, systemic circulating glucocorticoids are not elevated in obese patients and/or patients with metabolic syndrome. The study of the role of 11ß-HSD system shed light on this conundrum, showing that local glucocorticoids are finely regulated in a tissue-specific manner at the pre-receptor level. The system comprises two microsomal enzymes that either activate cortisone to cortisol (11ß-HSD1) or inactivate cortisol to cortisone (11ß-HSD2). Transgenic rodent models, knockout (KO) for HSD11B1 or with HSD11B1 or HSD11B2 overexpression, specifically targeted to the liver or adipose tissue, have been developed and helped unravel the currently undisputable role of the enzymes in metabolic syndrome pathophysiology, in each of its isolated components and in their prevention. In the transgenic HSD11B1 overexpressing models, different features of the metabolic syndrome and obesity are replicated. HSD11B1 gene deficiency or HSD11B2 gene overexpression associates with improvements in the metabolic profile. In face of these demonstrations, research efforts are now being turned both into the inhibition of 11ß-HSD1 as a possible pharmacological target and into the role of dietary habits on the establishment or the prevention of the metabolic syndrome, obesity and T2DM through 11ß-HSD1 modulation. We intend to review and discuss 11ß-HSD1 and obesity, the metabolic syndrome and T2DM and to highlight the potential of its inhibition for therapeutic or prophylactic approaches in those metabolic diseases.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/pharmacology , Atherosclerosis/enzymology , Cushing Syndrome/enzymology , Diabetes Mellitus, Type 2/enzymology , Glucocorticoids/blood , Metabolic Syndrome/enzymology , Obesity/enzymology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Adipose Tissue/enzymology , Animals , Anti-Inflammatory Agents , Atherosclerosis/drug therapy , Corticosterone/blood , Cushing Syndrome/diagnosis , Cushing Syndrome/diet therapy , Diabetes Mellitus, Type 2/drug therapy , Female , Humans , Liver/enzymology , Male , Metabolic Syndrome/diagnosis , Metabolic Syndrome/drug therapy , Mice , Mice, Transgenic , Obesity/drug therapy , Rats , Rats, Transgenic
8.
FASEB J ; 22(6): 1672-83, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18198220

ABSTRACT

Chronic exposure to glucocorticoid hormones, resulting from either drug treatment or Cushing's syndrome, results in insulin resistance, central obesity, and symptoms similar to the metabolic syndrome. We hypothesized that the major metabolic effects of corticosteroids are mediated by changes in the key metabolic enzyme adenosine monophosphate-activated protein kinase (AMPK) activity. Activation of AMPK is known to stimulate appetite in the hypothalamus and stimulate catabolic processes in the periphery. We assessed AMPK activity and the expression of several metabolic enzymes in the hypothalamus, liver, adipose tissue, and heart of a rat glucocorticoid-excess model as well as in in vitro studies using primary human adipose and primary rat hypothalamic cell cultures, and a human hepatoma cell line treated with dexamethasone and metformin. Glucocorticoid treatment inhibited AMPK activity in rat adipose tissue and heart, while stimulating it in the liver and hypothalamus. Similar data were observed in vitro in the primary adipose and hypothalamic cells and in the liver cell line. Metformin, a known AMPK regulator, prevented the corticosteroid-induced effects on AMPK in human adipocytes and rat hypothalamic neurons. Our data suggest that glucocorticoid-induced changes in AMPK constitute a novel mechanism that could explain the increase in appetite, the deposition of lipids in visceral adipose and hepatic tissue, as well as the cardiac changes that are all characteristic of glucocorticoid excess. Our data suggest that metformin treatment could be effective in preventing the metabolic complications of chronic glucocorticoid excess.


Subject(s)
Cushing Syndrome/metabolism , Glucocorticoids/pharmacology , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Adipose Tissue/enzymology , Adipose Tissue/metabolism , Animals , Cells, Cultured , Cushing Syndrome/enzymology , Dexamethasone/pharmacology , Humans , Hypothalamus/enzymology , Hypothalamus/metabolism , Liver/enzymology , Liver/metabolism , Metabolism/drug effects , Metformin/pharmacology , Myocardium/enzymology , Myocardium/metabolism , Organ Specificity , Rats
9.
BMJ Case Rep ; 12(1)2019 Jan 18.
Article in English | MEDLINE | ID: mdl-30661045

ABSTRACT

Two years after diagnosis of a metastatic neuroendocrine gastrin-secreting tumour and after several cycles of chemotherapy and peptide receptor radionuclide therapy, a 56-year-old woman presented with hypokalaemic metabolic alkalosis, hypertension, leg oedema and new-onset diabetes mellitus. Further investigations revealed renal potassium loss confirmed by a transtubular potassium gradient of 16, fully suppressed serum aldosterone, but instead highly elevated blood levels of morning cortisol and adrenocorticotropic hormone as well as increased urinary excretion of glucocorticoid and mineralocorticoid metabolites. Ruling out other causes, paraneoplastic hypercortisolism was diagnosed. Pharmacological inhibition of the steroid 11ß-hydroxylase with metyrapone resulted in complete resolution of metabolic alkalosis, hypokalaemia, hypertension, hyperglycaemia and leg oedema within 1 week.


Subject(s)
Cushing Syndrome/diagnosis , Cushing Syndrome/drug therapy , Metyrapone/administration & dosage , Alkalosis , Cushing Syndrome/enzymology , Diabetes Mellitus/enzymology , Diabetes Mellitus/etiology , Female , Humans , Hypertension/enzymology , Hypertension/etiology , Hypokalemia/enzymology , Hypokalemia/etiology , Metyrapone/therapeutic use , Middle Aged , Steroid 11-beta-Hydroxylase/antagonists & inhibitors , Treatment Outcome
10.
J Clin Endocrinol Metab ; 93(12): 4969-73, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18782871

ABSTRACT

OBJECTIVE: Features of the metabolic syndrome such as central obesity with insulin resistance and dyslipidemia are typical signs of Cushing's syndrome and common side effects of prolonged glucocorticoid treatment. AMP-activated protein kinase (AMPK), a key regulatory enzyme of lipid and carbohydrate metabolism as well as appetite, is involved in the development of the deleterious metabolic effects of excess glucocorticoids, but no data are available in humans. In the current study, we demonstrate the effect of high glucocorticoid levels on AMPK activity of human adipose tissue samples from patients with Cushing's syndrome. METHODS: AMPK activity and mRNA expression of genes involved in lipid metabolism were assessed in visceral adipose tissue removed at abdominal surgery of 11 patients with Cushing's syndrome, nine sex-, age-, and weight-matched patients with adrenal incidentalomas, and in visceral adipose tissue from four patients with non-endocrine-related abdominal surgery. RESULTS: The patients with Cushing's syndrome exhibited a 70% lower AMPK activity in visceral adipose tissue as compared with both incidentalomas and control patients (P = 0.007 and P < 0.001, respectively). Downstream targets of AMPK fatty acid synthase and phosphoenol-pyruvate carboxykinase were up-regulated in patients with Cushing's syndrome. AMPK activity was inversely correlated with 0900 h serum cortisol and with urinary free cortisol. CONCLUSIONS: Our data suggest that glucocorticoids inhibit AMPK activity in adipose tissue, suggesting a novel mechanism to explain the deposition of visceral adipose tissue and the consequent central obesity observed in patients with iatrogenic or endogenous Cushing's syndrome.


Subject(s)
Cushing Syndrome/complications , Cushing Syndrome/enzymology , Cyclic AMP-Dependent Protein Kinases/metabolism , Obesity/enzymology , Obesity/etiology , Adipose Tissue/enzymology , Adrenal Gland Neoplasms/genetics , Cushing Syndrome/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Female , Humans , Hydrocortisone/blood , Hydrocortisone/urine , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Male , Middle Aged , Obesity/genetics , Phosphoenolpyruvate Carboxykinase (ATP)/genetics , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/genetics , Up-Regulation/physiology
11.
Eur J Hum Genet ; 16(10): 1245-53, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18431404

ABSTRACT

Bilateral adrenocortical hyperplasia (BAH) is the second most common cause of corticotropin-independent Cushing syndrome (CS). Genetic forms of BAH have been associated with complex syndromes such as Carney Complex and McCune-Albright syndrome or may present as isolated micronodular adrenocortical disease (iMAD) usually in children and young adults with CS. A genome-wide association study identified inactivating phosphodiesterase (PDE) 11A (PDE11A)-sequencing defects as low-penetrance predisposing factors for iMAD and related abnormalities; we also described a mutation (c.914A > C/H305P) in cyclic AMP (cAMP)-specific PDE8B, in a patient with iMAD. In this study we further characterize this mutation; we also found a novel PDE8B isoform that is highly expressed in the adrenal gland. This mutation is shown to significantly affect the ability of the protein to degrade cAMP in vitro. Tumor tissues from patients with iMAD and no mutations in the coding PDE8B sequence or any other related genes (PRKAR1A, PDE11A) showed downregulated PDE8B expression (compared to normal adrenal cortex). Pde8b is detectable in the adrenal gland of newborn mice and is widely expressed in other mouse tissues. We conclude that PDE8B is another PDE gene linked to iMAD; it is a candidate causative gene for other adrenocortical lesions linked to the cAMP signaling pathway and possibly for tumors in other tissues.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/genetics , Adrenal Cortex/enzymology , Adrenal Hyperplasia, Congenital/enzymology , Adrenal Hyperplasia, Congenital/genetics , Mutation/genetics , 3',5'-Cyclic-AMP Phosphodiesterases/chemistry , Adolescent , Adrenal Cortex Diseases/enzymology , Adrenal Cortex Diseases/genetics , Adrenal Cortex Neoplasms/enzymology , Adrenal Cortex Neoplasms/genetics , Amino Acid Sequence , Animals , Base Sequence , Child , Child, Preschool , Conserved Sequence , Cushing Syndrome/enzymology , Cushing Syndrome/genetics , Female , Histidine/genetics , Humans , Infant , Isoenzymes/chemistry , Isoenzymes/genetics , Male , Mice , Molecular Sequence Data , Pedigree , Proline/genetics
12.
Curr Med Chem ; 15(26): 2734-47, 2008.
Article in English | MEDLINE | ID: mdl-18991633

ABSTRACT

Cushing's syndrome is a rare disease with significant morbidity and mortality. Surgical intervention represents the most effective treatment option in both adrenocorticotropin-dependent and -independent forms of hypercortisolism. It is not uncommon, however, that surgery fails to cure or control the disease. Pharmacotherapy with drugs inhibiting steroid biosynthesis can be effectively used in these cases in order to alleviate symptoms or even to induce chemical adrenalectomy. A few drugs inhibiting single or multiple steps in adrenal steroid biosynthesis can be used in clinical practice. Drugs predominantly inhibiting single enzymatic steps include the 11beta-hydroxylase inhibitor metyrapone and the 3beta-hydroxysteroid dehydrogenase inhibitor trilostane, whereas mitotane, aminoglutethimide, ketoconazole and etomidate block multiple enzymatic reactions. Etomidate is the only agent available for parenteral administration that renders it as a treatment of choice in critically ill patients requiring a rapid control of hypercortisolemia. Ketoconazole, metyrapone and aminoglutethimide can be used alone or in combination for the treatment of hypercortisolism caused by benign adrenocorticotropin- or cortisol-secreting tumors. The clinical utility of trilostane is variable. Besides blocking multiple steps in adrenal steroid biosynthesis, the DDT (insecticide) analogue mitotane also has adrenolytic properties by inducing mitochondrial degeneration that renders it superior to other drugs in the treatment of adrenocortical cancer. Severe side effects may develop during therapy with each aforementioned drug that include hepatic, endocrine and neurological toxicity. After summarizing the chemical and biological properties of steroid biosynthetic inhibitors, the authors describe their possible clinical applications and limitations.


Subject(s)
Cushing Syndrome/drug therapy , Cushing Syndrome/metabolism , Enzyme Inhibitors/therapeutic use , Steroids/biosynthesis , Animals , Cushing Syndrome/enzymology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans
13.
Mol Cell Endocrinol ; 285(1-2): 10-8, 2008 Mar 26.
Article in English | MEDLINE | ID: mdl-18313835

ABSTRACT

Although glucocorticoid, as "gluco-" literally implies, plays an important role in maintaining the blood glucose level, excess of glucocorticoid production/action is known to cause impaired glucose tolerance and diabetes. Since 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1), which converts inactive cortisone to active cortisol, is primarily expressed in the liver, an enhanced expression of the enzyme may increase the intracellular glucocorticoid level and thus increase the hepatic glucose production. In this study, we examined the effects of multiple humoral factors related to the metabolic syndrome on the transcriptional activity of 11beta-HSD1 gene in hepatocytes in vitro. We found that, among the factors examined, adipocyte-derived cytokines (adipokines), like TNFalpha and IL-1beta, potently stimulated the transcriptional activity of 11beta-HSD1 gene in human HuH7 cells. In contrast, only minimal effects of other humoral factors were observed when they were used alone. Interestingly, however, when applied in combination, they synergistically enhanced the transcriptional activity of 11beta-HSD1 gene. They also potentiated the effects of cytokines. Glucocorticoid receptor (GR)-dependent transcription was indeed increased even with an inactive glucocorticoid cortisone following TNFalpha pretreatment, indicating the enhanced intracellular conversion. Finally, PPARgamma/PPARalpha agonists, clinically used as anti-diabetic drugs, significantly inhibited the transcriptional activity of 11beta-HSD1. Altogether, our data strongly suggest that combination of the humoral factors related to the metabolic syndrome, including the adipokines, synergistically enhances the hepatic expression of 11beta-HSD1 gene and causes the intracellular Cushing state in the liver by increasing the intracellular glucocorticoid level. We assume that the observed synergistic effects of these factors on 11beta-HSD1 may, at least partly, explain the reason whereby accumulation of the multiple risk factors facilitates the derangement of glucose and lipid metabolism in the metabolic syndrome.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1 , Cushing Syndrome/blood , Cushing Syndrome/enzymology , Gene Expression Regulation, Enzymologic , Metabolic Syndrome/blood , Metabolic Syndrome/enzymology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Animals , Anticholesteremic Agents/metabolism , Base Sequence , Cell Line , Chromans/metabolism , Clofibrate/metabolism , Cortisone/metabolism , Cushing Syndrome/physiopathology , Dexamethasone/metabolism , Glucocorticoids/metabolism , Humans , Hydrocortisone/metabolism , Hypoglycemic Agents/metabolism , Insulin/metabolism , Interleukin-1beta/metabolism , Liver/metabolism , Metabolic Syndrome/physiopathology , Metformin/metabolism , Molecular Sequence Data , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Thiazolidinediones/metabolism , Transcription Factor AP-1/metabolism , Troglitazone , Tumor Necrosis Factor-alpha/metabolism
14.
Cancer Res ; 66(24): 11571-5, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17178847

ABSTRACT

Several types of adrenocortical tumors that lead to Cushing syndrome may be caused by aberrant cyclic AMP (cAMP) signaling. We recently identified patients with micronodular adrenocortical hyperplasia who were carriers of inactivating mutations in the 2q-located phosphodiesterase 11A (PDE11A) gene. We now studied the frequency of two missense substitutions, R804H and R867G, in conserved regions of the enzyme in several sets of normal controls, including 745 individuals enrolled in a longitudinal cohort study, the New York Cancer Project. In the latter, we also screened for the presence of the previously identified PDE11A nonsense mutations. R804H and R867G were frequent among patients with adrenocortical tumors; although statistical significance was not reached, these variants affected significantly enzymatic function in vitro with variable increases in cAMP and/or cyclic guanosine 3',5'-monophosphate levels in HeLa and HEK293 cells. Adrenocortical tissues carrying the R804H mutation showed 2q allelic losses and higher cyclic nucleotide levels and cAMP-responsive element binding protein phosphorylation. We conclude that missense mutations of the PDE11A gene that affect enzymatic activity in vitro are present in the general population; protein-truncating PDE11A mutations may also contribute to a predisposition to other tumors, in addition to their association with adrenocortical hyperplasia. We speculate that PDE11A genetic defects may be associated with adrenal pathology in a wider than previously suspected clinical spectrum that includes asymptomatic individuals.


Subject(s)
Adenoma/genetics , Adrenocortical Hyperfunction/enzymology , Adrenocortical Hyperfunction/genetics , Genetic Variation , Mutation , Phosphoric Diester Hydrolases/genetics , Pituitary Neoplasms/genetics , 3',5'-Cyclic-GMP Phosphodiesterases , Adenoma/enzymology , Base Sequence , Carrier State , Cell Line , Codon, Nonsense , Cushing Syndrome/enzymology , Cushing Syndrome/genetics , DNA/genetics , DNA Primers , DNA, Neoplasm/genetics , Gene Frequency , Genotype , HeLa Cells , Humans , Kidney , Loss of Heterozygosity , Pituitary Neoplasms/enzymology
15.
Eur J Endocrinol ; 179(5): L1-L2, 2018 Oct 12.
Article in English | MEDLINE | ID: mdl-30320504

ABSTRACT

We read with interest the paper of Young et al. in which the authors recommend avoiding ketoconazole in the treatment of Cushing's syndrome when patients display increased liver enzymes (>2-fold the upper limit of normal (ULN)). We found in a small series of patients that We read with interest the paper of Young et al. in which the authors recommend avoiding ketoconazole in the treatment of Cushing's syndrome when patients display increased liver enzymes (>2-fold the upper limit of normal (ULN)). Although limited, our experience suggests that liver function tests may improve during ketoconazole treatment and that, in a life-threatening situation such as severe Cushing's syndrome, increased liver enzymes should not preclude ketoconazole prescription.


Subject(s)
Cushing Syndrome/drug therapy , Ketoconazole/therapeutic use , Liver/enzymology , Adult , Cushing Syndrome/enzymology , Drug Therapy, Combination , Enzyme Inhibitors/therapeutic use , Female , Humans , Ketoconazole/adverse effects , Male , Metyrapone/therapeutic use , Middle Aged , Treatment Outcome
16.
Ann Endocrinol (Paris) ; 68(5): 349-56, 2007 Oct.
Article in French | MEDLINE | ID: mdl-17368420

ABSTRACT

11beta-hydroxysteroide dehydrogenase (11beta-OHSD) enzymes exhibit a regulating action upon cortisol metabolism before access to its receptors. Two types of isoenzymes have been described, type 2 being the most anciently known. Type 2 11beta-OHSD, which changes cortisol into cortisone, is a unidirectional dehydrogenase mainly located in kidney, that protects mineralocorticoid receptors from illicit activation by glucocorticoids. Mutations of the gene coding for this enzyme has been demonstrated in apparent mineralocorticoid excess, which induces hypertension and hypokalemia with low renin and aldosterone levels. Polymorphisms of this gene could modulate essential hypertension and also be responsible for certain forms of acquired apparent mineralocorticoid excess especially after liquorice intoxication, in hypothyroidism, Cushing syndrome, and chronic renal insufficiency. Type 1 11beta-OHSD, which changes cortisone into cortisol, is a reductase, mainly located in liver and adipose tissue. Functional defects of this enzyme have been shown in polycystic ovaries and cortisone reductase deficiency. By contrast, metabolic syndrome, corticoid-induced osteoporosis, and glaucoma are linked to a local over-activity of this enzyme. The understanding of action mechanisms of these two enzymes currently leads to 11beta-OHSD inhibitors development, therefore opening new therapeutic strategies, especially in metabolic syndrome.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenases/metabolism , Kidney/enzymology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/deficiency , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , 11-beta-Hydroxysteroid Dehydrogenases/genetics , Cushing Syndrome/enzymology , Female , Genotype , Humans , Hydrocortisone/metabolism , Phenotype , Polycystic Ovary Syndrome/enzymology , Polycystic Ovary Syndrome/genetics , Receptors, Mineralocorticoid/physiology
17.
Arq Bras Endocrinol Metabol ; 51(8): 1397-403, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18209879

ABSTRACT

Glucocorticoids have a major role in determining adipose tissue metabolism and distribution. 11beta-hydroxysteroid dehydrogenase type 1 (11betaHSD1) is a NADPH-dependent enzyme highly expressed in the liver and adipose tissue. In most intact cells and tissues it functions as a reductase (to convert inactive cortisone to active cortisol). It has been hypothesized that tissue-specific deregulation of cortisol metabolism may be involved in the complex pathophysiology of the metabolic syndrome (MS) and obesity. Transgenic mice overexpressing 11betaHSD1 in adipose tissue develop obesity with all features of the MS, whereas 11betaHSD1-knockout mice are protected from both. The bulk of evidences points to an overexpression and increased activity of 11betaHSD1 also in human adipose tissue. However, 11betaHSD1 seems to adjust local cortisol concentrations independently of its plasma levels. In Cushing's syndrome, 11betaHSD1 is downregulated and may not be responsible for the abdominal fat depots; it also undergoes downregulation in response to weight loss in human obesity. The nonselective 11betaHSD1 inhibitor carbenoxolone improves insulin sensitivity in humans, and selective inhibitors enhance insulin action in diabetic mice liver, thereby lowering blood glucose. Thus, 11betaHSD1 is now emerging as a modulator of energy partitioning and a promising pharmacological target to treat the MS and diabetes.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Adipose Tissue/enzymology , Cushing Syndrome/enzymology , Obesity/enzymology , Adipocytes/enzymology , Adipocytes/metabolism , Animals , Down-Regulation , Glucocorticoids/metabolism , Humans , Hydrocortisone/metabolism , Liver/enzymology , Liver/metabolism , Metabolic Syndrome/enzymology , Mice , Mice, Transgenic/metabolism
18.
FEBS Lett ; 591(3): 459-467, 2017 02.
Article in English | MEDLINE | ID: mdl-28100013

ABSTRACT

The PKAL205R hotspot mutation has been implicated in Cushing's syndrome through hyperactive gain-of-function PKA signaling; however, its influence on substrate specificity has not been investigated. Here, we employ the Proteomic Peptide Library (ProPeL) approach to create high-resolution models for PKAWT and PKAL205R substrate specificity. We reveal that the L205R mutation reduces canonical hydrophobic preference at the substrate P + 1 position, and increases acidic preference in downstream positions. Using these models, we designed peptide substrates that exhibit altered selectivity for specific PKA variants, and demonstrate the feasibility of selective PKAL205R loss-of-function signaling. Through these results, we suggest that substrate rewiring may contribute to Cushing's syndrome disease etiology, and introduce a powerful new paradigm for investigating mutation-induced kinase substrate rewiring in human disease.


Subject(s)
Cushing Syndrome/enzymology , Cushing Syndrome/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Mutation/genetics , Amino Acid Sequence , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Assays , Escherichia coli/metabolism , Humans , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Engineering , Substrate Specificity
19.
J Clin Endocrinol Metab ; 91(9): 3626-32, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16772351

ABSTRACT

CONTEXT: Primary adrenocortical hyperplasias leading to Cushing syndrome include primary pigmented nodular adrenocortical disease and ACTH-independent macronodular adrenal hyperplasia (AIMAH). Inactivating mutations of the 17q22-24-located PRKAR1A gene, coding for the type 1A regulatory subunit of protein kinase A (PKA), cause primary pigmented nodular adrenocortical disease and the multiple endocrine neoplasia syndrome Carney complex. PRKAR1A mutations and 17q22-24 chromosomal losses have been found in sporadic adrenal tumors and are associated with aberrant PKA signaling. OBJECTIVE: The objective of the study was to examine whether somatic 17q22-24 changes, PRKAR1A mutations, and/or PKA abnormalities are present in AIMAH. PATIENTS: We studied fourteen patients with Cushing syndrome due to AIMAH. METHODS: Fluorescent in situ hybridization with a PRKAR1A-specific probe was used for investigating chromosome 17 allelic losses. The PRKAR1A gene was sequenced in all samples, and tissue was studied for PKA activity, cAMP responsiveness, and PKA subunit expression. RESULTS: We found 17q22-24 allelic losses in 73% of the samples. There were no PRKAR1A-coding sequence mutations. The RIIbeta PKA subunit was overexpressed by mRNA, whereas the RIalpha, RIbeta, RIIalpha, and Calpha PKA subunits were underexpressed. These findings were confirmed by immunohistochemistry. Total PKA activity and free PKA activity were higher in AIMAH than normal adrenal glands, consistent with the up-regulation of the RIIbeta PKA subunit. CONCLUSIONS: PRKAR1A mutations are not found in AIMAH. Somatic losses of the 17q22-24 region and PKA subunit and enzymatic activity changes show that PKA signaling is altered in AIMAH in a way that is similar to that of other adrenal tumors with 17q losses or PRKAR1A mutations.


Subject(s)
Chromosomes, Human, Pair 17/genetics , Cushing Syndrome/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Adrenal Glands/metabolism , Adrenal Glands/pathology , Alleles , Cushing Syndrome/enzymology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Hyperplasia , Immunohistochemistry , In Situ Hybridization, Fluorescence , Mutation , RNA/chemistry , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction
20.
Pituitary ; 9(3): 211-9, 2006.
Article in English | MEDLINE | ID: mdl-17036196

ABSTRACT

Primary Pigmented Nodular Adrenocortical Disease (PPNAD) is a rare primary bilateral adrenal defect causing corticotropin-independent Cushing's syndrome. It occurs mainly in children and young adults. Macroscopic appearance of the adrenals is characteristic with small pigmented micronodules observed in the cortex. PPNAD is most often diagnosed in patients with Carney complex (CNC), but it can also be observed in patients without other manifestations or familial history (isolated PPNAD). The CNC is an autosomal dominant multiple neoplasia syndrome characterized by the association of myxoma, spotty skin pigmentation and endocrine overactivity. One of the putative CNC genes has been identified as the gene of the regulatory R1A subunit of protein kinase A (PRKAR1A), located at 17q22-24. Germline heterozygous inactivating mutations of PRKAR1A have been reported in about 45% of patients with CNC, and up to 80% of CNC patients with Cushing's syndrome due to PPNAD. Interestingly, such inactivating germline PRKAR1A mutations have also been found in patients with isolated PPNAD. The hot spot PRKAR1A mutation termed c.709[-7-2]del6 predisposes mostly to isolated PPNAD, and is the first clear genotype/phenotype correlation described for this gene. Somatic inactivating mutations of PRKAR1A have been observed in macronodules of PPNAD and in sporadic cortisol secreting adrenal adenomas. Isolated PPNAD is a genetic heterogenous disease, and recently inactivating mutations of the gene of the phosphodiesterase 11A4 (PDE11A4) located at 2q31-2q35 have been identified in patients without PRKAR1A mutations. Interestingly, both PRKAR1A and PDE11A gene products control the cAMP signaling pathway, which can be altered at various levels in endocrine tumors.


Subject(s)
Adrenal Cortex Diseases/genetics , Cushing Syndrome/genetics , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Mutation , Pigmentation Disorders/genetics , Adrenal Cortex Diseases/complications , Adrenal Cortex Diseases/enzymology , Adrenal Cortex Diseases/pathology , Animals , Cushing Syndrome/enzymology , Cushing Syndrome/pathology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Enzymologic , Genetic Predisposition to Disease , Hormones/metabolism , Humans , Multiple Endocrine Neoplasia/enzymology , Multiple Endocrine Neoplasia/genetics , Pigmentation Disorders/complications , Pigmentation Disorders/enzymology , Pigmentation Disorders/pathology , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL