Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 290
Filtrar
1.
BMC Endocr Disord ; 24(1): 116, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39010034

RESUMO

BACKGROUND: Somatic mutations have been observed to induce aldosterone-producing adenomas (APAs). These may be accelerated during pregnancy. Somatic PRKACA mutations are common in cortisol-producing adenomas (CPAs). However, their role in APAs, particularly aldosterone- and cortisol-producing adenomas (A/CPAs), is not well understood. This study aims to investigate the association between PRKACA mutations and the accelerated development of A/CPAs during pregnancy. CASE PRESENTATION: A patient with primary aldosteronism (PA) associated with severe Cushing's syndrome (CS) underwent surgical resection of an adrenal tumor one year after delivery. Pathologic examination revealed an adrenocortical adenoma characterized primarily by zona glomerulosa hyperplasia. Somatic mutation analysis revealed the presence of the somatic PRKACA mutation, which was validated as a deleterious mutation by various computational databases. Immunohistochemical results showed positive staining for cytochrome P450 family 11 subfamily B member 1 (CYP11B1), cytochrome P450 family 11 subfamily B member 2 (CYP11B2), and luteinizing hormone/chorionic gonadotropin receptor (LHCGR). Our study included a review of 20 previously documented cases of aldosterone- and cortisol-producing adenomas (A/CPAs), two of which were concurrently positive for both CYP11B1 and CYP11B2, consistent with our findings. CONCLUSION: Somatic mutations in PRKACA may correlate with the upregulation of LHCGR, which synergistically drives the accelerated growth of co-secretion tumors during pregnancy, thereby exacerbating disease progression.


Assuntos
Neoplasias do Córtex Suprarrenal , Adenoma Adrenocortical , Aldosterona , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico , Hidrocortisona , Mutação , Complicações Neoplásicas na Gravidez , Humanos , Feminino , Gravidez , Adulto , Hidrocortisona/metabolismo , Adenoma Adrenocortical/genética , Adenoma Adrenocortical/patologia , Adenoma Adrenocortical/metabolismo , Adenoma Adrenocortical/cirurgia , Neoplasias do Córtex Suprarrenal/genética , Neoplasias do Córtex Suprarrenal/patologia , Neoplasias do Córtex Suprarrenal/metabolismo , Aldosterona/metabolismo , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/genética , Complicações Neoplásicas na Gravidez/genética , Complicações Neoplásicas na Gravidez/patologia , Hiperaldosteronismo/genética , Hiperaldosteronismo/patologia , Hiperaldosteronismo/cirurgia , Síndrome de Cushing/genética , Síndrome de Cushing/patologia , Adenoma/genética , Adenoma/patologia , Adenoma/metabolismo
2.
Front Endocrinol (Lausanne) ; 15: 1356870, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39006359

RESUMO

Aims: Primary pigmented nodular adrenocortical disease (PPNAD), as a rare kind of Cushing's syndrome, is frequently misdiagnosed. To get a better understanding of the disease, we analyzed the clinical characteristics and pathogenic variants of PPNAD. Methods: Databases were searched, and the pathogenic variants and clinical manifestations of patients were summarized from the relevant articles. Results: A total of 210 patients in 86 articles were enrolled with a median age of 22 and a female-to-male ratio of 2:1. Sixty-six (31.43%) patients were combined with Carney complex (CNC) and 94.29% were combined with osteoporosis/osteopenia. Among 151 patients who underwent genetic testing, 87.42% (132/151) had pathogenic variants. Six gene mutations (PRKAR1A, PDE11A, PRKACA, CTNNB1, PDE8B, and ARMC5) were detected in the patients. The most common mutation was PKAR1A, accounting for 79.47% (120/151). There was a significant correlation between PRKAR1A pathogenic variant and spotty skin pigmentation in CNC concurrent with PPNAD (p < 0.05). Among pregnant patients with PPNAD, those without surgical treatment and with bilateral adrenalectomy suffered from a high-risk perinatal period. However, patients with unilateral adrenalectomy presented a safe perinatal period. Conclusions: For young patients with Cushing's syndrome, especially female patients with spotty skin pigmentation and osteoporosis/osteopenia, PPNAD should be considered. Unilateral adrenal resection may be considered as an option for women with fertility needs. In view of the difficulty of PPNAD diagnosis, genetic testing before surgery might be a reasonable option. Patients with PPNAD with spotty skin pigmentation should consider the PRKAR1A pathogenic variant and pay attention to CNC. Systematic review registration: https://www.crd.york.ac.uk/prospero, identifier CRD42023416988.


Assuntos
Doenças do Córtex Suprarrenal , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico , Mutação , Humanos , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Doenças do Córtex Suprarrenal/genética , Doenças do Córtex Suprarrenal/patologia , Feminino , Adulto , Masculino , Síndrome de Cushing/genética , Síndrome de Cushing/cirurgia , Síndrome de Cushing/diagnóstico , Complexo de Carney/genética , Complexo de Carney/diagnóstico , Adulto Jovem , Osteoporose/genética
3.
Endocrinol Diabetes Nutr (Engl Ed) ; 71(3): 119-123, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38555108

RESUMO

INTRODUCTION/OBJECTIVES: Mutations in the ARMC5 (armadillo repeat containing 5, OMIM 615549) gene, a putative tumor suppressor gene, have recently been identified as a common cause of sporadic and familial bilateral macronodular adrenal hyperplasia (BMAH). Familial BMAH is thought to be caused by two mutations, one germline and the other somatic, as suggested by the 2-hit theory. The objective is to describe a new mutation and develop its clinical characteristics and implications. METHODS, RESULTS AND CONCLUSIONS: We present an affected family with 11 members carrying a novel mutation of the ARMC5 gene (NM_001288767.1): c.2162T>C p. (Leu721Pro). Two of the carriers developed clinical Cushing's syndrome (CS), two mild autonomous cortisol secretion (MACS) and one presented with autonomous cortisol secretion (ACS). Four patients developed other tumors, three of whom died from this cause. It is not known whether these tumors could be related to the described mutation.


Assuntos
Hiperplasia Suprarrenal Congênita , Síndrome de Cushing , Neoplasias , Humanos , Hiperplasia Suprarrenal Congênita/complicações , Hiperplasia Suprarrenal Congênita/genética , Síndrome de Cushing/genética , Mutação em Linhagem Germinativa , Hidrocortisona , Hiperplasia , Proteínas Supressoras de Tumor/genética
4.
J Endocrinol Invest ; 47(3): 655-669, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37682493

RESUMO

PURPOSE: Impairment of skeletal muscle mass and strength affects 40-70% of patients with active Cushing's syndrome (CS). Glucocorticoid excess sustains muscle atrophy and weakness, while muscle-specific microRNAs (myomiRs) level changes were associated with muscle organization and function perturbation. The aim of the current study is to explore changes in circulating myomiRs in CS patients compared to healthy controls and their involvement in IGFI/PI3K/Akt/mTOR pathway regulation in skeletal muscle. METHODS: C2C12, mouse myocytes, were exposed to hydrocortisone (HC), and atrophy-related gene expression was investigated by RT-qPCR, WB and IF to assess HC-mediated atrophic signalling. miRNAs were evaluated in HC-treated C2C12 by PCR Arrays. MyomiRs significantly overexpressed in C2C12 were investigated in 37 CS patients and 24 healthy controls serum by RT-qPCR. The anti-anabolic role of circulating miRNAs significantly upregulated in CS patients was explored in C2C12 by investigating the IGFI/PI3K/Akt/mTOR pathway regulation. RESULTS: HC induced higher expression of atrophy-related genes, miR-133a-3p, miR-122-5p and miR-200b-3p in C2C12 compared to untreated cells. Conversely, the anabolic IGFI/PI3K/Akt/mTOR signalling was reduced and this effect was mediated by miR-133a-3p. In CS patients miR-133a-3p and miR-200b-3p revealed higher circulating levels (p < 0.0001, respectively) compared to controls. ROC curves for miR-133a-3p (AUC 0.823, p < 0.0001) and miR-200b-3p (AUC 0.850, p < 0.0001) demonstrated that both myomiRs represent potential biomarkers to discriminate between CS and healthy subjects. Pearson's correlation analysis revealed that circulating levels of miR-133a-3p are directly correlated with 24 h urinary-free cortisol level (r = 0.468, p = 0.004) in CS patients. CONCLUSIONS: HC induces atrophic signals by miR-133a-3p overexpression in mouse myocytes and humans. Circulating miR-133a-3p is promising biomarkers of hypercortisolism.


Assuntos
Síndrome de Cushing , MicroRNAs , Humanos , Animais , Camundongos , Síndrome de Cushing/genética , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , MicroRNAs/genética , Atrofia , Biomarcadores , Hidrocortisona , Serina-Treonina Quinases TOR
5.
Heart Vessels ; 39(1): 65-74, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37695542

RESUMO

The causes of adrenal Cushing's syndrome (CS) encompass a wide spectrum of adrenal cortisol proliferations that exhibit clinical and molecular heterogeneity. The aims of our study were to investigate whether clinical and molecular heterogeneity influences endothelial function and metabolic abnormalities in patients with cortisol-producing adenoma (CPA). We retrospectively enrolled 25 patients with CPA and 45 patients with essential hypertension (EH). All CPAs were studied by direct sequencing of PRKACA. Flow-mediated vasodilation (FMD), an index of vascular endothelial function, was significantly lower in CS and subclinical CS (SCS) groups than in the EH group. FMD impairment did not differ significantly between CS and SCS groups. No differences in FMD were seen between PRKACA mutant and wild-type groups. FMD correlated negatively with hemoglobin A1c (HbA1c) in both PRKACA mutant and wild-type groups, as well as in CS and SCS groups. After adrenalectomy, systolic blood pressure (SBP) and HbA1c decreased significantly from baseline in the CS group, and SBP and low-density lipoprotein cholesterol (LDL-C) decreased significantly from baseline in the SCS group. While SBP and LDL-C decreased significantly from baseline in patients with wild-type PRKACA, only HbA1c decreased from baseline in patients harboring PRKACA mutations. Our data showed that patients with CPA have impaired endothelial function compared with EH patients and suggest the need for strict monitoring of atherosclerosis, even in patients with SCS or without PRKACA mutation.


Assuntos
Adenoma , Doenças Cardiovasculares , Síndrome de Cushing , Humanos , Hidrocortisona/metabolismo , Síndrome de Cushing/genética , Estudos Retrospectivos , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/genética , LDL-Colesterol , Hemoglobinas Glicadas , Fatores de Risco , Adenoma/genética
6.
Front Endocrinol (Lausanne) ; 14: 1265794, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38098864

RESUMO

Background: We performed a transcriptomic analysis of adrenal signaling pathways in various forms of endogenous Cushing's syndrome (CS) to define areas of dysregulated and druggable targets. Methodology: Next-generation sequencing was performed on adrenal samples of patients with primary bilateral macronodular adrenal hyperplasia (PBMAH, n=10) and control adrenal samples (n=8). The validation groups included cortisol-producing adenoma (CPA, n=9) and samples from patients undergoing bilateral adrenalectomy for Cushing's disease (BADX-CD, n=8). In vivo findings were further characterized using three adrenocortical cell-lines (NCI-H295R, CU-ACC2, MUC1). Results: Pathway mapping based on significant expression patterns identified PPARG (peroxisome proliferator-activated receptor gamma) pathway as the top hit. Quantitative PCR (QPCR) confirmed that PPARG (l2fc<-1.5) and related genes - FABP4 (l2fc<-5.5), PLIN1 (l2fc<-4.1) and ADIPOQ (l2fc<-3.3) - were significantly downregulated (p<0.005) in PBMAH. Significant downregulation of PPARG was also found in BADX-CD (l2fc<-1.9, p<0.0001) and CPA (l2fc<-1.4, p<0.0001). In vitro studies demonstrated that the PPARG activator rosiglitazone resulted in decreased cell viability in MUC1 and NCI-H295R (p<0.0001). There was also a significant reduction in the production of aldosterone, cortisol, and cortisone in NCI-H295R and in Dihydrotestosterone (DHT) in MUC1 (p<0.05), respectively. Outcome: This therapeutic effect was independent of the actions of ACTH, postulating a promising application of PPARG activation in endogenous hypercortisolism.


Assuntos
Síndrome de Cushing , Humanos , Adrenalectomia/métodos , Síndrome de Cushing/genética , Síndrome de Cushing/cirurgia , Síndrome de Cushing/tratamento farmacológico , Hidrocortisona/metabolismo , Hiperplasia , PPAR gama/genética
7.
Arch Endocrinol Metab ; 68: e220491, 2023 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-37988664

RESUMO

Primary pigmented nodular adrenocortical disease (PPNAD) is a rare adrenocorticotropin hormone (ACTH)-independent Cushing's syndrome (CS). Pediatric patients with PPNAD typically have unusual skin lesions and slow growth with unknown causes. We present a case of a female Chinese patient with PPNAD caused by the germline PRKACA gene copy number gain of chromosome 19. The patient initially presented with kidney stones, short stature, and obesity. After further testing, it was discovered that the patient had diabetes, mild hypertension, low bone mass, a low ACTH level, and hypercortisolemia, and neither the low-dose or high-dose dexamethasone suppression test was able to inhibit hematuric cortisol, which paradoxically increased. PPNAD was pathologically diagnosed after unilateral adrenalectomy. Chromosome microarrays and whole exon sequencing analyses of the peripheral blood, as well as testing of sectioned adrenal tissue, showed a rise in the copy number of the duplication-containing PRKACA gene on chromosome 19p13.13p13.12, a de novo but not heritable gene defect that causes disease. The clinical signs and symptoms supported the diagnosis of Carney complex (CNC). One significant mechanism of CNC pathogenesis may be the rise in germline PRKACA copy number of chromosome 19. When assessing PPNAD patients for CNC, the possibility of PRKACA gene amplification should be considered. The effect of PRKACA gene amplification on the clinical manifestations of CNC needs to be confirmed by more cases.


Assuntos
Doenças do Córtex Suprarrenal , Síndrome de Cushing , Humanos , Criança , Feminino , Doenças do Córtex Suprarrenal/genética , Doenças do Córtex Suprarrenal/diagnóstico , Doenças do Córtex Suprarrenal/patologia , Síndrome de Cushing/genética , Adrenalectomia/efeitos adversos , Hidrocortisona , Hormônio Adrenocorticotrópico , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico
8.
Hypertension ; 80(11): 2333-2344, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37646167

RESUMO

BACKGROUND: Cushing syndrome (CS) is a rare disease caused by excess cortisol levels with high cardiovascular morbidity and mortality. Hypertension in CS promotes hypercortisolism-associated cardiovascular events. Adipose tissue is a highly plastic tissue with most cell types strongly affected by the excess cortisol exposure. We hypothesized that the molecular and cellular changes of periadrenal adipose tissue in response to cortisol excess impact systemic blood pressure levels in patients with CS. METHODS: We investigated gene expression signatures in periadrenal adipose tissue from patients with adrenal CS collected during adrenal surgery. RESULTS: During active CS we observed a downregulation of gene programs associated with inflammation in periadrenal adipose tissue. In addition, we observed a clustering of the patients based on tissue gene expression profiles into 2 groups that differed in blood pressure levels (CS low blood pressure and CS high blood pressure). The 2 clusters showed significant differences in gene expression pattens of the renin-angiotensin-aldosterone-system. Renin was the strongest regulated gene compared with control patients and its expression correlated with increased blood pressure observed in our patients with CS. In the CS high blood pressure group, systemic renin plasma levels were suppressed indicative of an abnormal blood pressure associated with periadrenal adipose tissue renin-angiotensin-aldosterone-system activation. CONCLUSIONS: Here, we show for the first time a relevant association of the local renin-angiotensin-aldosterone-system and systemic blood pressure levels in patients with CS. Patients from the CS high blood pressure group still had increased blood pressure levels after 6 months in remission, highlighting the importance of local tissue effects on long-term systemic effects observed in CS.


Assuntos
Síndrome de Cushing , Hipertensão , Humanos , Renina , Síndrome de Cushing/complicações , Síndrome de Cushing/genética , Transcriptoma , Aldosterona , Hidrocortisona , Sistema Renina-Angiotensina/fisiologia , Hipertensão/metabolismo , Pressão Sanguínea/genética , Tecido Adiposo , Angiotensinas/metabolismo
9.
Orv Hetil ; 164(32): 1271-1277, 2023 Aug 13.
Artigo em Húngaro | MEDLINE | ID: mdl-37573559

RESUMO

Our 69-year-old female patient was investigated for a 20 kg weight gain over 2 years. The patient's medical history included hypertension, hyperuricemia, bilateral cataract surgery and musculosceletal complaints. Diabetes mellitus was not found. Physical examination revealed abdominal obesity, proximal myopathy and atrophic, vulnerable skin. The "overnight", low-dose and long, low-dose dexamethasone suppression tests indicated autonomous cortisol overproduction (plasma cortisol level: 172.6 and 153.2 nmol/L, cut-off: 50 nmol/L). The suppressed ACTH (<1.11 pmol/L, normal value: 1.12-10.75 pmol/L) suggested ACTH-independent hypercortisolism. Abdominal CT described macronodular enlargement of both adrenals. The size of the largest nodule was 23 × 20 mm in the right, and 24 × 30 mm on the left side (with -33 ± 37 HU density values on native scans). The 131I-cholesterol adrenal scintigraphy and SPECT/CT showed almost equally intensive radiopharmacon uptake on both sides. Based on the clinical results, bilateral macronodular adrenal hyperplasia associated with ACTH-independent hypercortisolism was diagnosed. Genomic DNA was obtained from the peripheral blood leukocytes. Targeted sequencing of 25 genes potentially involved in adrenal tumorigenesis revealed a new disease-causing armadillo repeat-containing 5 (ARMC5) gene mutation (c.1724del28 bp, g.31,476,067-31,476,094). Because of the autosomal dominant inheritance of this genetic alteration, the patient's two children underwent genetic screening for the ARMC5 mutation. The same mutation was found in the younger child of our patient. To the best of our knowledge, this is the first published Hungarian case of ARMC5 mutation with bilateral macronodular adrenal hyperplasia and ACTH-independent Cushing's syndrome. The genetic alteration is present in two generations of the family of the index patient. Orv Hetil. 2023; 164(32): 1271-1277.


Assuntos
Hiperplasia Suprarrenal Congênita , Síndrome de Cushing , Feminino , Criança , Humanos , Idoso , Síndrome de Cushing/genética , Síndrome de Cushing/diagnóstico , Radioisótopos do Iodo , Hidrocortisona , Hiperplasia/genética , Proteínas Supressoras de Tumor/genética , Mutação , Hormônio Adrenocorticotrópico/genética , Proteínas do Domínio Armadillo/genética
10.
BMJ Case Rep ; 16(7)2023 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-37419498

RESUMO

A woman in her 60s with chronic fatigue, depressed mood and proximal muscle weakness was referred to our endocrinology department. Physical examination revealed facial plethora, atrophic skin and ankle oedema. Adjuvant blood and urine analyses indicated endogenous ACTH-independent Cushing syndrome. Abdominal imaging showed bilateral macronodular adrenals, measuring 58.9 × 29.7 mm on the right and 55.6 × 42.6 mm on the left. Primary bilateral macronodular adrenal hyperplasia was confirmed by pathology after bilateral adrenalectomy. Gradual mental and physical recovery was observed in the months following surgery. Genetic sequencing failed to reveal mutations in the ARMC5 gene.Cushing syndrome is a rare entity that should be suspected when typical clinical signs, including skin atrophy with ecchymosis, muscle weakness or coloured stretch marks, are present. Primary bilateral macronodular adrenal hyperplasia is an uncommon cause of endogenous Cushing syndrome. It is a benign condition characterised by adrenal macronodules exceeding 1 cm and hypercorticism.


Assuntos
Síndrome de Cushing , Feminino , Humanos , Síndrome de Cushing/etiologia , Síndrome de Cushing/genética , Hiperplasia/patologia , Proteínas Supressoras de Tumor/genética , Glândulas Suprarrenais/diagnóstico por imagem , Glândulas Suprarrenais/cirurgia , Glândulas Suprarrenais/patologia , Adrenalectomia , Proteínas do Domínio Armadillo
11.
Virchows Arch ; 483(4): 465-476, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37405461

RESUMO

Renal neuroendocrine tumors (RenNETs) are rare malignancies with largely unknown biology, hormone expression, and genetic abnormalities. This study aims to improve our understanding of the RenNETs with emphasis of functional, hormonal, and genetic features. Surgically resected RenNETs (N = 13) were retrieved, and immunohistochemistry and next-generation sequencing (NGS) were performed in all cases. In addition, all published RenNETs were systematically reviewed. Our cohort (4 men and 9 women, mean age 42, mean tumor size 7.6 cm) included 2 patients with Cushing syndrome (CS). WHO grade (23% G1, 54% G2, and 23% G3) and tumor progression did not correlate. CS-associated RenNETs (CS-RenNETs) showed a solid and eosinophilic histology and stained for ACTH, while the remaining non-functioning tumors had a trabecular pattern and expressed variably hormones somatostatin (91%), pancreatic polypeptide (63%), glucagon (54%), and serotonin (18%). The transcription factors ISL1 and SATB2 were expressed in all non-functioning, but not in CS-RenNETs. NGS revealed no pathogenic alterations or gene fusions. In the literature review (N = 194), 15 (8%) of the patients had hormonal syndromes, in which CS being the most frequent (7/15). Large tumor size and presence of metastasis were associated with shorter patients' survival (p < 0.01). RenNETs present as large tumors with metastases. CS-RenNETs differ through ACTH production and solid-eosinophilic histology from the non-functioning trabecular RenNETs that produce pancreas-related hormones and express ISL1 and SATB2. MEN1 or DAXX/ARTX abnormalities and fusion genes are not detected in RenNETs, indicating a distinct yet unknown molecular pathogenesis.


Assuntos
Síndrome de Cushing , Neoplasias Renais , Tumores Neuroendócrinos , Neoplasias Pancreáticas , Masculino , Humanos , Feminino , Adulto , Tumores Neuroendócrinos/metabolismo , Síndrome de Cushing/genética , Patologia Molecular , Neoplasias Pancreáticas/patologia , Fatores de Transcrição , Neoplasias Renais/genética , Hormônio Adrenocorticotrópico/metabolismo
12.
Biochem J ; 480(12): 875-890, 2023 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-37306403

RESUMO

Cushing's syndrome is an endocrine disorder caused by excess production of the stress hormone cortisol. Precision medicine strategies have identified single allele mutations within the PRKACA gene that drive adrenal Cushing's syndrome. These mutations promote perturbations in the catalytic core of protein kinase A (PKAc) that impair autoinhibition by regulatory subunits and compartmentalization via recruitment into AKAP signaling islands. PKAcL205R is found in ∼45% of patients, whereas PKAcE31V, PKAcW196R, and L198insW and C199insV insertion mutants are less prevalent. Mass spectrometry, cellular, and biochemical data indicate that Cushing's PKAc variants fall into two categories: those that interact with the heat-stable protein kinase inhibitor PKI, and those that do not. In vitro activity measurements show that wild-type PKAc and W196R activities are strongly inhibited by PKI (IC50 < 1 nM). In contrast, PKAcL205R activity is not blocked by the inhibitor. Immunofluorescent analyses show that the PKI-binding variants wild-type PKAc, E31V, and W196R are excluded from the nucleus and protected against proteolytic processing. Thermal stability measurements reveal that upon co-incubation with PKI and metal-bound nucleotide, the W196R variant tolerates melting temperatures 10°C higher than PKAcL205. Structural modeling maps PKI-interfering mutations to a ∼20 Šdiameter area at the active site of the catalytic domain that interfaces with the pseudosubstrate of PKI. Thus, Cushing's kinases are individually controlled, compartmentalized, and processed through their differential association with PKI.


Assuntos
Síndrome de Cushing , Humanos , Síndrome de Cushing/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Mutação , Domínio Catalítico
13.
Eur J Endocrinol ; 188(3): R56-R72, 2023 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-36857084

RESUMO

Thirty years ago, we identified that cortisol secretion in some patients with unilateral adenoma or primary bilateral macronodular adrenal hyperplasia (PBMAH) was stimulated by food intake; this was secondary to the abnormal adrenocortical responsiveness to physiological post-prandial increase in glucose-dependent insulinotropic peptide (GIP). This resulted from the ectopic expression of non-mutated GIP receptor in the pathological adrenal tissues of those patients. Although ectopic GIP receptor (GIPR) was confirmed in a relatively limited number of cases to date, its elucidation leads to the identification of a wide diversity of aberrant G-protein-coupled receptors regulating steroidogenesis and cell proliferation in a high proportion of patients with PBMAH or cortisol-secreting adenomas. In addition, ectopic GIPR was identified in other endocrine tumors including somatotroph pituitary tumors with paradoxical growth hormone response to oral glucose, medullary thyroid carcinomas, and other neuroendocrine tumors. The first molecular pathogenic mechanism responsible for ectopic GIPR expression was elucidated in unilateral GIP-dependent adenomas in which somatic duplication and rearrangements in chromosome region 19q13.32 containing the GIPR locus lead to increased expression of GIPR which was enhanced by the activity of a glucocorticoid response element. Recently, germline lysine demythylase 1A (KDMIA) mutations combined with somatic chromosome 1p deletions were found to be specifically responsible for ectopic GIPR in sporadic or familial GIP-dependent PBMAH and can be associated with adrenal myelolipoma, monoclonal gammopathy of unknown significance (MGUS), or multiple myeloma. Screening for ectopic GIPR should be conducted in all patients with PBMAH; genetic studies to identify KDM1A mutations should be offered to such patients in order to detect affected members and provide early detection of PBMAH and other potential associated neoplasias. The elucidation of GIP-dependent Cushing's syndrome (CS) illustrates that careful bedside phenotyping of rare conditions can lead to identification of genetically determined diseases requiring personalized approaches to investigation and therapy.


Assuntos
Adenoma , Síndrome de Cushing , Humanos , Síndrome de Cushing/genética , Síndrome de Cushing/metabolismo , Polipeptídeo Inibidor Gástrico/metabolismo , Hidrocortisona/metabolismo , Glândulas Suprarrenais/patologia , Adenoma/patologia , Histona Desmetilases/metabolismo
14.
Surg Oncol Clin N Am ; 32(2): 303-313, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36925187

RESUMO

Surgical diseases of the adrenal gland include pheochromocytoma/paraganglioma, primary hyperaldosteronism, Cushing syndrome, and adrenocortical carcinoma. These conditions may be associated with familial syndromes, and genetic testing is available and recommended in most. For adrenal surgeons to be familiar with these syndromes and know when to consider referral for genetic counseling and genetic testing is important. Identification of patients with familial syndromes allows for the detection and screening of associated syndromic neoplasms, guides surgical planning and operative approach, influences recurrence and malignancy risk assessment, aids in the development of a postoperative surveillance plan, and determines the need for screening family members.


Assuntos
Neoplasias das Glândulas Suprarrenais , Síndrome de Cushing , Cirurgiões , Humanos , Adrenalectomia , Neoplasias das Glândulas Suprarrenais/diagnóstico , Neoplasias das Glândulas Suprarrenais/genética , Neoplasias das Glândulas Suprarrenais/cirurgia , Síndrome de Cushing/genética , Síndrome de Cushing/patologia , Síndrome de Cushing/cirurgia , Testes Genéticos
15.
Mol Genet Genomic Med ; 11(4): e2126, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36727580

RESUMO

BACKGROUND: To perform genetic screening for ARMC5 gene germline pathogenic variants in patients with primary bilateral macronodular adrenal hyperplasia (PBMAH). SUBJECTS AND METHODS: In a group of 10 PBMAH patients, we performed complete sequencing of the coding region of the ARMC5 gene and MLPA analysis for large deletion detection. In subjects with the ARMC5 variant, we searched ARMC5 gene somatic variants on tumor samples. RESULTS: Among 10 PBMAH patients, we identified four ARMC5 germline variants (40%). One variant, c:174dupC p.Glu59Argfs*44, was already known; one variant p.Gly323Asp, was already reported and classified as likely disease-causing VUS (class 3-4); two variants p.Leu596Arg and p.Arg811Pro, were never reported before. For p.Gly323Asp and p.Arg811Pro, we identified second deleterious variants at the somatic level, enforcing the possible pathogenic effect of germline variants. CONCLUSIONS: Our results underscore the importance of performing genetic testing also in sporadic PBMAH patients and broaden the spectrum of molecular variants involved in PBMAH syndrome.


Assuntos
Proteínas do Domínio Armadillo , Síndrome de Cushing , Humanos , Proteínas do Domínio Armadillo/genética , Síndrome de Cushing/diagnóstico , Síndrome de Cushing/genética , Síndrome de Cushing/patologia , Mutação em Linhagem Germinativa , Hiperplasia , Proteínas Supressoras de Tumor/genética
16.
BMC Endocr Disord ; 23(1): 35, 2023 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-36740703

RESUMO

BACKGROUND: Carney complex (CNC) is a rare multiple endocrine neoplasia syndrome characterized by mucocutaneous lentigines/ blue nevi, cardiac myxoma and endocrine overactivity. Here, we report a CNC case with PRKAR1A gene mutation characterized by left atrial adenomyxoma to explore the diagnosis and treatment of CNC. CASE PRESENTATION: A 42-year-old woman with a history of cardiac tumour surgery presented with typical features of Cushing syndrome, including central obesity, buffalo hump, mild facial plethora, purple striae on the lower abdomen, and spotty skin pigmentation. Left atrial adenomyxoma and thyroid papillary carcinoma were identified by postoperative histologic assays. Genetic screening revealed a pathogenic germline heterozygous mutation of c.682C > T (p.R228X) in exon 7 of the PRKAR1A gene. The clinical features and normal ACTH levels suggest this patient suffered the ACTH-independent primary pigmented nodular adrenocortical disease (PPNAD) with cyclic hypercortisolism or ACTH-dependent Cushing syndrome. CONCLUSION: CNC is uncommon, however, if a patient develops clinical features involving multiple endocrine and non-endocrine tumors, especially Cushing syndrome and cardiac myxoma, CNC should be considered. Genetic analysis is recommended in patients with suspected CNC.


Assuntos
Fibrilação Atrial , Complexo de Carney , Síndrome de Cushing , Mixoma , Humanos , Complexo de Carney/complicações , Complexo de Carney/diagnóstico , Complexo de Carney/genética , Síndrome de Cushing/etiologia , Síndrome de Cushing/genética , Mixoma/complicações , Mixoma/genética , Mixoma/cirurgia , Hormônio Adrenocorticotrópico , Mutação
17.
Am J Case Rep ; 23: e937404, 2022 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-36573035

RESUMO

BACKGROUND Carney complex (CNC) is a rare multiple neoplasia syndrome with autosomal dominant inheritance. CNC is frequently misdiagnosed owing to its diverse clinical characteristics. We reported the case of a 14-year-old Saudi boy with a history of gynecomastia, Cushing syndrome, large-cell calcifying Sertoli cell tumor of the testis, and CNC. CASE REPORT The patient was referred to the pediatric endocrine clinic for evaluation of bilateral slow progressing gynecomastia for 1-year duration. His clinical examination revealed lentigenes, bilateral diffuse breast enlargement (consistent with Tanner stage III), and asymmetrical testicular enlargement, more on the left side. Other systemic examinations were unremarkable. The initial blood workup showed elevated estradiol level with unsuppressed cortisol after an overnight 1-mg dexamethasone suppression test. Breast ultrasound (US) confirmed true gynecomastia. Testicular US revealed microcalcification and the testicular biopsy confirmed diagnoses of large-cell calcifying Sertoli cell tumor (LCCSCT). A 2-step dexamethasone suppression test showed a paradoxical rise in serum and urine cortisol levels, which are characteristic for PPNAD. LCCSCT and PPNAD are 2 major criteria fulfilling a diagnosis of CNC. The gene test showed heterozygous mutation in the PRKAR1A gene, which is diagnostic for CNC. The patient underwent bilateral mastoplasty and was planned for radical left orchiectomy. CONCLUSIONS Gynecomastia and LCCSCT can be presenting features of CNC, which mandates careful, thorough clinical examination and tailored investigation to reach a diagnosis.


Assuntos
Complexo de Carney , Síndrome de Cushing , Ginecomastia , Tumor de Células de Sertoli , Neoplasias Testiculares , Masculino , Criança , Humanos , Adolescente , Tumor de Células de Sertoli/diagnóstico , Tumor de Células de Sertoli/genética , Tumor de Células de Sertoli/patologia , Complexo de Carney/diagnóstico , Complexo de Carney/genética , Síndrome de Cushing/genética , Ginecomastia/diagnóstico , Ginecomastia/etiologia , Hidrocortisona , Arábia Saudita , Células de Sertoli/patologia , Neoplasias Testiculares/diagnóstico , Neoplasias Testiculares/patologia , Dexametasona
18.
Medicine (Baltimore) ; 101(43): e31283, 2022 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-36316863

RESUMO

BACKGROUND: Many clinical studies have reported a relatively high incidence of osteoporosis and fragility fractures in patients with Cushing syndrome (CS). However, few papers have investigated osteoporosis and CS in terms of pathogenesis, so this study explores the association between the 2 and predicts upstream micro-ribonucleic acids (miRNAs) through bioinformatics, which provides potential targets for simultaneous pharmacological interventions in both diseases and also provides a basis for pathological screening. METHODS: We used Genecards, Online Mendelian Inheritance in Man and Therapeutic Target Database databases to screen the targets of osteoporosis and Cushing syndrome; import target genes to Database for Annotation, Visualization and Integrated Discovery for Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis; the intersecting genes were uploaded to Search Tool for the Retrieval of Genes and Genomes database to construct protein-protein interaction network; Cytoscape software was used to screen core genes, and Molecular Complex Detection module was used to analyze cluster modules; finally, the NetworkAnalyst data platform was used to predict the miRNAs that interact with core genes. RESULTS: The core genes of osteoporosis and Cushing syndrome were insulin, tumor necrosis factor, signal transducer and activator of transcription 3 (STAT3), interleukin-6, insulin-like growth factor 1, etc. A total of 340 upstream miRNAs including hsa-let-7a-5p, hsa-mir-30a-5p and hsa-mir-125b-5p were predicted. The biological processes involved include regulating the transcription of ribonucleic acid polymerase II promoter and participating in the transduction of cytokine signaling pathways, which focus on the binding of nerve system ligand, JAK-STAT signaling pathway, Rap1 signaling pathway, PI3K-Akt signaling pathway, etc. CONCLUSION: Osteoporosis and Cushing syndrome are closely related in terms of targets and molecular mechanisms. In this study, bioinformatics methods were used to identify their targets and mechanisms, providing potential targets for drug simultaneous regulation of the 2 diseases, and providing a new direction for exploring the relationship between diseases.


Assuntos
Síndrome de Cushing , MicroRNAs , Osteoporose , Humanos , Biologia Computacional/métodos , Síndrome de Cushing/complicações , Síndrome de Cushing/genética , Fosfatidilinositol 3-Quinases , MicroRNAs/genética , Osteoporose/genética
19.
Front Endocrinol (Lausanne) ; 13: 934748, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35966069

RESUMO

In a small number of cases, the development of ectopic residual adrenal lesions during embryogenesis causing Cushing's syndrome due to the production of excess cortisol has been reported. A 29-year-old woman was admitted to our hospital for fatigue and recent amenorrhea. Her plasma ACTH was <1.5 pg/mL, and her serum cortisol was 21.4 pg/mL after the 8 mg dexamethasone suppression test, revealing the presence of ACTH-independent Cushing's syndrome; however, her bilateral adrenal glands were atrophied. Abdominal CT revealed a 40-mm round tumor on the right renal hilum and remarkably accumulated 131I-labelled adosterol. CT and bone scintigraphy showed that 99mTc-methylene diphosphonate had accumulated in her dissymmetric skull at the right-frontoparietal region. The tumor on the right renal hilum was laparoscopically removed. Her cortisol levels rapidly decreased to below the normal range, and glucocorticoids were administered to rescue adrenal insufficiency. The resected tumor was yellowish in appearance and 4.5×3.0×2.8 cm in size. Immunohistochemical staining for SF-1, P450scc, CYP17A, CYP21A, and CYP11B1 indicated that this tumor produced cortisol. Exome sequencing analysis revealed that the GNAS heterozygous mutation (c.601C>T, p. Arg201Cys; accession number, NM_000516.5) was found in approximately 20% of the adrenal tumor sample. A mutation of GNAS, encoding the Gsα subunit that mediates GPCR signaling, causes the constitutive activation of adenylyl cyclase, resulting in hypersecretion of hormones regulated by the GPCR. GNAS mutation is one of the major genetic causes of cortisol-producing adrenal tumors independent of ACTH secretion. Considering the combination of GNAS mutation with one of the typical clinical triad characteristics, fibrous dysplasia of bone, we diagnosed this patient with McCune-Albright syndrome accompanied by ACTH-independent Cushing's syndrome caused by an ectopic residual adrenal tumor due to GNAS mutation. This case highlights that GNAS involves a previously unknown pathological mechanism in which inhibition of the natural elimination of remnant tissue leads to ectopic endocrine hypersecretion.


Assuntos
Neoplasias das Glândulas Suprarrenais , Síndrome de Cushing , Displasia Fibrosa Poliostótica , Hipotireoidismo , Neoplasias das Glândulas Suprarrenais/complicações , Hormônio Adrenocorticotrópico , Adulto , Amenorreia/complicações , Amenorreia/genética , Cromograninas/genética , Síndrome de Cushing/complicações , Síndrome de Cushing/genética , Feminino , Displasia Fibrosa Poliostótica/complicações , Displasia Fibrosa Poliostótica/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Humanos , Hidrocortisona , Hipotireoidismo/complicações , Mutação
20.
Front Endocrinol (Lausanne) ; 13: 937793, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35992119

RESUMO

Pediatric adrenocortical hyperplasias are rare; they usually present with Cushing syndrome (CS); of them, isolated micronodular adrenal disease and its variant, primary pigmented adrenocortical disease are the most commonly encountered. Most cases are due to defects in the cyclic AMP/protein kinase A (cAMP/PKA) pathway, although a few cases remain without an identified genetic defect. Another cause of adrenal hyperplasia in childhood is congenital adrenal hyperplasia, a group of autosomal recessive disorders that affect steroidogenic enzymes in the adrenal cortex. Clinical presentation varies and depends on the extent of the underlying enzymatic defect. The most common form is due to 21-hydroxylase deficiency; it accounts for more than 90% of the cases. In this article, we discuss the genetic etiology of adrenal hyperplasias in childhood.


Assuntos
Doenças do Córtex Suprarrenal , Córtex Suprarrenal , Hiperplasia Suprarrenal Congênita , Síndrome de Cushing , Córtex Suprarrenal/metabolismo , Doenças do Córtex Suprarrenal/complicações , Doenças do Córtex Suprarrenal/genética , Hiperplasia Suprarrenal Congênita/patologia , Criança , Síndrome de Cushing/genética , Humanos , Hiperplasia/complicações
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA