Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Eur J Endocrinol ; 190(5): 374-381, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38652802

ABSTRACT

BACKGROUND: Lymph node metastases in papillary thyroid cancer (PTC) increase the risk for persistent and recurrent disease. Data on the predictive value of histopathological features of lymph node metastases, however, are inconsistent. The aim of this study was to evaluate the prognostic significance of known and new histopathological features of lymph node metastases in a well-defined cohort of PTC patients with clinically evident lymph node metastases. METHODS: A total of 1042 lymph node metastases, derived from 129 PTC patients, were reexamined according to a predefined protocol and evaluated for diameter, extranodal extension, cystic changes, necrosis, calcifications, and the proportion of the lymph node taken up by tumor cells. Predictors for a failure to achieve a complete biochemical and structural response to treatment were determined. RESULTS: The presence of more than 5 lymph node metastases was the only independent predictor for a failure to achieve a complete response to treatment (odds ratio [OR] 3.39 [95% CI, 1.57-7.33], P < .05). Diameter nor any of the other evaluated lymph node features were significantly associated with the response to treatment. CONCLUSIONS: Detailed reexamination of lymph nodes revealed that only the presence of more than 5 lymph node metastases was an independent predictor of failure to achieve a complete response to treatment. No predictive value was found for other histopathological features, including the diameter of the lymph node metastases. These findings have the potential to improve risk stratification in patients with PTC and clinically evident lymph node metastases.


Subject(s)
Carcinoma, Papillary , Lymph Nodes , Lymphatic Metastasis , Thyroid Cancer, Papillary , Thyroid Neoplasms , Humans , Thyroid Neoplasms/pathology , Thyroid Neoplasms/therapy , Male , Female , Middle Aged , Lymphatic Metastasis/pathology , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/therapy , Adult , Carcinoma, Papillary/pathology , Aged , Lymph Nodes/pathology , Prognosis , Treatment Outcome , Predictive Value of Tests , Young Adult , Carcinoma/pathology , Carcinoma/secondary , Carcinoma/therapy , Retrospective Studies , Cohort Studies
2.
Head Neck ; 45(9): 2227-2236, 2023 09.
Article in English | MEDLINE | ID: mdl-37490544

ABSTRACT

BACKGROUND: The Gene Expression Classifier (GEC) and Genomic Sequencing Classifier (GSC) were developed to improve risk stratification of indeterminate nodules. Our aim was to assess the clinical utility in a European population with restrictive diagnostic workup. METHODS: Clinical utility of the GEC was assessed in a prospective multicenter cohort of 68 indeterminate nodules. Diagnostic surgical rates for Bethesda III and IV nodules were compared to a historical cohort of 171 indeterminate nodules. Samples were post hoc tested with the GSC. RESULTS: The GEC classified 26% as benign. Surgical rates between the prospective and historical cohort did not differ (72.1% vs. 76.6%). The GSC classified 59% as benign, but misclassified six malignant lesions as benign. CONCLUSION: Implementation of GEC in management of indeterminate nodules in a European country with restrictive diagnostic workup is currently not supported, especially in oncocytic nodules. Prospective studies with the GSC in European countries are needed to determine the clinical utility.


Subject(s)
Thyroid Neoplasms , Thyroid Nodule , Humans , Thyroid Nodule/pathology , Prospective Studies , Netherlands , Gene Expression Profiling , Retrospective Studies , Gene Expression , Thyroid Neoplasms/diagnosis
3.
Endosc Int Open ; 10(4): E549-E557, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35433206

ABSTRACT

Background and study aims In this study, we evaluated the performance of community hospitals involved in the Dutch quality in endosonography team regarding yield of endoscopic ultrasound (EUS)-guided tissue acquisition (TA) of solid pancreatic lesions using cumulative sum (CUSUM) learning curves. The aims were to assess trends in quality over time and explore potential benefits of CUSUM as a feedback-tool. Patients and methods All consecutive EUS-guided TA procedures for solid pancreatic lesions were registered in five community hospitals between 2015 and  2018. CUSUM learning curves were plotted for overall performance and for performance per center. The American Society of Gastrointestinal Endoscopy-defined key performance indicators, rate of adequate sample (RAS), and diagnostic yield of malignancy (DYM) were used for this purpose. Feedback regarding performance was provided on multiple occasions at regional interest group meetings during the study period. Results A total of 431 EUS-guided TA procedures in 403 patients were included in this study. The overall and per center CUSUM curves for RAS improved over time. CUSUM curves for DYM revealed gradual improvement, reaching the predefined performance target (70 %) overall, and in three of five contributing centers in 2018. Analysis of a sudden downslope development in the CUSUM curve of DYM in one center revealed temporary absence of a senior cytopathologist to have had a temporary negative impact on performance. Conclusions CUSUM-derived learning curves allow for assessment of best practices by comparison among peers in a multidisciplinary multicenter quality improvement initiative and proved to be a valuable and easy-to-interpret means to evaluate EUS performance over time.

5.
BMC Cancer ; 19(1): 325, 2019 Apr 05.
Article in English | MEDLINE | ID: mdl-30953466

ABSTRACT

BACKGROUND: For progressive metastatic medullary thyroid carcinoma (MTC), the available treatment options with tyrosine kinase inhibitors result in grade 3-4 adverse events in a large number of patients. Peptide Receptor Radionuclide Therapy (PRRT), which has also been suggested to be a useful treatment for MTC, is usually well tolerated, but evidence on its effectivity is very limited. METHODS: Retrospective evaluation of treatment effects of PRRT in a highly selected group of MTC patients, with progressive disease or refractory symptoms. In addition, a retrospective evaluation of uptake on historical 111In-DTPA-octreotide scans was performed in patients with detectable tumor size > 1 cm. RESULTS: Over the last 17 years, 10 MTC patients were treated with PRRT. Four out of 10 patients showed stable disease at first follow-up (8 months after start of therapy) whereas the other 6 were progressive. Patients with stable disease were characterized by a combination of both a high uptake on 111In-DTPA-octreotide scan (uptake grade ≥ 3) and a positive somatostatin receptor type 2a (SSTR2a) expression of the tumor by immunohistochemistry. Retrospective evaluation of historical 111In-DTPA-octreotide scans of 35 non-treated MTC patients revealed low uptake (uptake grade 1) in the vast majority of patients 31/35 (89%) with intermediate uptake (uptake grade 2) in the remaining 4/35 (11%). CONCLUSIONS: PRRT using 177Lu-octreotate could be considered as a treatment in those patients with high uptake on 111In-DTPA-octreotide scan (uptake grade 3) and positive SSTR2a expression in tumor histology. Since this high uptake was present in a very limited number of patients, this treatment is only suitable in a selected group of MTC patients.


Subject(s)
Carcinoma, Neuroendocrine/radiotherapy , Octreotide/analogs & derivatives , Radioimmunotherapy/methods , Receptors, Somatostatin/metabolism , Thyroid Neoplasms/radiotherapy , Adult , Aged , Carcinoma, Neuroendocrine/diagnostic imaging , Carcinoma, Neuroendocrine/mortality , Carcinoma, Neuroendocrine/pathology , Feasibility Studies , Female , Humans , Male , Middle Aged , Octreotide/administration & dosage , Octreotide/therapeutic use , Patient Selection , Pentetic Acid/administration & dosage , Pentetic Acid/analogs & derivatives , Progression-Free Survival , Radionuclide Imaging/methods , Retrospective Studies , Thyroid Gland/diagnostic imaging , Thyroid Gland/pathology , Thyroid Neoplasms/diagnostic imaging , Thyroid Neoplasms/mortality , Thyroid Neoplasms/pathology , Young Adult
6.
J Clin Endocrinol Metab ; 103(1): 46-55, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28938490

ABSTRACT

Context: Pheochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine, usually benign, tumors. Currently, the only reliable criterion of malignancy is the presence of metastases. Objective: The aim of this study was to identify genes associated with malignancy in PPGLs. Design: Transcriptomic profiling was performed on 40 benign and 11 malignant PPGLs. Genes showing a significantly different expression between benign and malignant PPGLs with a ratio ≥4 were confirmed and tested in an independent series by quantitative real-time polymerase chain reaction (qRT-PCR). Immunohistochemistry was performed for the validated genes on 109 benign and 32 malignant PPGLs. Functional assays were performed with hPheo1 cells. Setting: This study was conducted at the Department of Pathology of the Erasmus MC University Medical Center Rotterdam Human Molecular Genetics laboratory of the de Duve Institute, University of Louvain. Patients: PPGL samples from 179 patients, diagnosed between 1972 and 2015, were included. Main outcome measures: Associations between gene expression and malignancy were tested using supervised clustering approaches. Results: Ten differentially expressed genes were selected based on messenger RNA (mRNA) expression array data. Contactin 4 (CNTN4) was overexpressed in malignant vs benign tumors [4.62-fold; false discovery rate (FDR), 0.001]. Overexpression at the mRNA level was confirmed using qRT-PCR (2.90-fold, P = 0.02; validation set: 4.26-fold, P = 0.005). Consistent findings were obtained in The Cancer Genome Atlas cohort (2.7-fold; FDR, 0.02). CNTN4 protein was more frequently expressed in malignant than in benign PPGLs by immunohistochemistry (58% vs 17%; P = 0.002). Survival after 7 days of culture under starvation conditions was significantly enhanced in hPheo1 cells transfected with CNTN4 complementary DNA. Conclusion: CNTN4 expression is consistently associated with malignant behavior in PPGLs.


Subject(s)
Adrenal Gland Neoplasms/diagnosis , Biomarkers, Tumor/metabolism , Contactins/metabolism , Paraganglioma/diagnosis , Pheochromocytoma/diagnosis , RNA, Messenger/metabolism , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/metabolism , Biomarkers, Tumor/genetics , Contactins/genetics , Gene Expression Profiling , Humans , Paraganglioma/genetics , Paraganglioma/metabolism , Pheochromocytoma/genetics , Pheochromocytoma/metabolism , Prognosis , RNA, Messenger/genetics
7.
Am J Surg Pathol ; 40(4): 569-76, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26685085

ABSTRACT

Despite the established role of Ki67 labeling index in prognostic stratification of adrenocortical carcinomas and its recent integration into treatment flow charts, the reproducibility of the assessment method has not been determined. The aim of this study was to investigate interobserver variability among endocrine pathologists using a web-based virtual microscopy approach. Ki67-stained slides of 76 adrenocortical carcinomas were analyzed independently by 14 observers, each according to their method of preference including eyeballing, formal manual counting, and digital image analysis. The interobserver variation was statistically significant (P<0.001) in the absence of any correlation between the various methods. Subsequently, 61 static images were distributed among 15 observers who were instructed to follow a category-based scoring approach. Low levels of interobserver (F=6.99; Fcrit=1.70; P<0.001) as well as intraobserver concordance (n=11; Cohen κ ranging from -0.057 to 0.361) were detected. To improve harmonization of Ki67 analysis, we tested the utility of an open-source Galaxy virtual machine application, namely Automated Selection of Hotspots, in 61 virtual slides. The software-provided Ki67 values were validated by digital image analysis in identical images, displaying a strong correlation of 0.96 (P<0.0001) and dividing the cases into 3 classes (cutoffs of 0%-15%-30% and/or 0%-10%-20%) with significantly different overall survivals (P<0.05). We conclude that current practices in Ki67 scoring assessment vary greatly, and interobserver variation sets particular limitations to its clinical utility, especially around clinically relevant cutoff values. Novel digital microscopy-enabled methods could provide critical aid in reducing variation, increasing reproducibility, and improving reliability in the clinical setting.


Subject(s)
Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/pathology , Biomarkers, Tumor/analysis , Ki-67 Antigen/analysis , Pathology, Clinical/standards , Adolescent , Adult , Aged , Aged, 80 and over , Child , Female , Humans , Image Interpretation, Computer-Assisted , Male , Middle Aged , Observer Variation , Reproducibility of Results , Tissue Array Analysis , User-Computer Interface , Young Adult
8.
J Clin Endocrinol Metab ; 101(2): 453-60, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26670126

ABSTRACT

CONTEXT: Familial pheochromocytoma (PCC) has been associated with germline mutations in 16 genes. Here we investigated three siblings presenting with bilateral pheochromocytomas. In addition, the index patient also exhibited renal oncocytoma and erythrocytosis, whereas the second sibling presented with a lymph node metastasis. DESIGN: First, single-nucleotide polymorphism array and exome sequencing were performed on germline and PCC-derived DNA to identify genomic alterations in the index patient. Second, alterations were confirmed and validated by Sanger sequencing, analyzed by (multiplexed) PCR to determine the loss of the wild-type allele, and investigated by immunohistochemistry in the tumors of the three siblings. RESULTS: The index patient's germline DNA revealed a large complex genomic alteration encompassing the intragenic and promoter regions of Myc-associated factor X (MAX) and alpha-(1,6)-fucosyltransferase (FUT8). In all three siblings the MAX alteration was confirmed, and the loss of the wild-type MAX and FUT8 alleles was demonstrated in all tumors. Uniparental disomy of chromosome 14q, previously demonstrated as a hallmark for MAX-related PCC, was shown in the index patient's PCC by single-nucleotide polymorphism array. Loss of MAX and FUT8 protein expression was demonstrated by immunohistochemistry in the tumors from the three siblings. CONCLUSIONS: Our results indicate that large genomic deletions of MAX should be considered in familial and bilateral PCC with prior negative testing for gene mutations. In addition, our results confirm that MAX is a tumor suppressor gene for renal oncocytomas.


Subject(s)
Adenoma, Oxyphilic/genetics , Adrenal Gland Neoplasms/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Pheochromocytoma/genetics , Polycythemia/genetics , Adenoma, Oxyphilic/complications , Adenoma, Oxyphilic/metabolism , Adrenal Gland Neoplasms/complications , Adrenal Gland Neoplasms/metabolism , Adult , Chromosomes, Human, Pair 14/genetics , Exome , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Gene Rearrangement , Germ-Line Mutation , Humans , Lymphatic Metastasis , Male , Middle Aged , Pedigree , Pheochromocytoma/complications , Pheochromocytoma/metabolism , Polycythemia/complications , Polycythemia/metabolism , Polymorphism, Single Nucleotide/genetics , Uniparental Disomy
9.
Ned Tijdschr Geneeskd ; 159: A9413, 2015.
Article in Dutch | MEDLINE | ID: mdl-26556492

ABSTRACT

Thyroid cancer is comparatively rare. Thyroid nodules, on the other hand, are frequently diagnosed as a result of increasing use of diagnostic imaging. Cytological investigation of small nodules that have been found by chance often reveals micropapillary carcinoma that is probably not clinically relevant. The new guideline 'Thyroid cancer' advises that cytological investigation of these non-palpable, incidentally discovered thyroid nodules should only be performed on indication. The standard treatment for patients with papillary or follicular thyroid cancer consists of thyroidectomy followed by, if indicated, lymph-node dissection, ablation therapy with radioactive iodine and TSH-suppression. The extent of this treatment is determined on the basis of known prognostic factors and the results of initial treatment. Targeted systemic therapy is available for patients with metastatic progressive disease. There is more focus on the effects of short- and long-term treatment, in order to optimise quality of life.


Subject(s)
Practice Guidelines as Topic , Thyroid Neoplasms/diagnosis , Thyroid Nodule/diagnosis , Thyroidectomy/methods , Adenocarcinoma, Follicular/diagnosis , Adenocarcinoma, Follicular/surgery , Diagnosis, Differential , Humans , Incidental Findings , Iodine Radioisotopes/therapeutic use , Lymph Node Excision , Quality of Life , Thyroid Neoplasms/surgery , Thyroid Nodule/surgery
10.
J Clin Endocrinol Metab ; 99(9): 3060-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24905065

ABSTRACT

CONTEXT: Only a small number of case reports has been published on patients with PTHrP-hypersecreting metastatic gastroenteropancreatic (GEP) neuroendocrine tumors (NETs). OBJECTIVE: The objective of this study was to evaluate the clinical, biochemical, and radiological features, management, and treatment outcome of patients with PTHrP-hypersecreting GEP-NETs. DESIGN: Retrospective case series. SETTING: Tertiary referral hospital. MAIN OUTCOME MEASURES: Clinical, biochemical, and radiological features were measured, as well as response to therapy and survival. PATIENTS: Ten patients with PTHrP-secreting GEP-NETs (nine pancreatic and one unknown primary) with a median age of 50.4 years (range, 38.3-61.1) were studied. Multiple endocrine neoplasia type 1 patients were excluded. RESULTS: The median follow-up was 57.2 months (range, 11.6-204.5 mo). Median overall survival was 86.0 months. In total, 51 different treatment interventions and combinations were applied. In seven of the 10 patients, somatostatin analog (SSA) treatment resulted in a temporary normalization of serum calcium levels with a long-term response observed in two patients (up to 35.2 mo). Peptide receptor radiotherapy (PRRT) with radiolabeled SSAs induced long-term responses ranging from 9.0-49.0 months in four of six patients treated with PRRT. CONCLUSIONS: Hypersecretion of PTHrP by metastatic GEP-NETs is very rare and seems to be exclusively associated with metastatic pancreatic NETs. PTHrP production has major clinical impact because poorly controllable hypercalcemia is associated with increased morbidity and mortality. The most successful treatment options for PTHrP-producing GEP-NETs are SSAs and PRRT using radiolabeled SSAs. Isotonic saline and bisphosphonates can be considered as supportive therapies.


Subject(s)
Gastrointestinal Neoplasms , Neuroendocrine Tumors , Pancreatic Neoplasms , Parathyroid Hormone-Related Protein/metabolism , Adult , Antineoplastic Agents, Hormonal/therapeutic use , Combined Modality Therapy , Digestive System Surgical Procedures , Female , Follow-Up Studies , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/surgery , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasms, Unknown Primary , Neuroendocrine Tumors/diagnosis , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/surgery , Octreotide/therapeutic use , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/surgery , Retrospective Studies
11.
Eur J Endocrinol ; 170(1): 1-12, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24096523

ABSTRACT

OBJECTIVE: Although the succinate dehydrogenase (SDH)-related tumor spectrum has been recently expanded, there are only rare reports of non-pheochromocytoma/paraganglioma tumors in SDHx-mutated patients. Therefore, questions still remain unresolved concerning the aforementioned tumors with regard to their pathogenesis, clinicopathological phenotype, and even causal relatedness to SDHx mutations. Absence of SDHB expression in tumors derived from tissues susceptible to SDH deficiency is not fully elucidated. DESIGN AND METHODS: Three unrelated SDHD patients, two with pituitary adenoma (PA) and one with papillary thyroid carcinoma (PTC), and three SDHB patients affected by renal cell carcinomas (RCCs) were identified from four European centers. SDHA/SDHB immunohistochemistry (IHC), SDHx mutation analysis, and loss of heterozygosity analysis of the involved SDHx gene were performed on all tumors. A cohort of 348 tumors of unknown SDHx mutational status, including renal tumors, PTCs, PAs, neuroblastic tumors, seminomas, and adenomatoid tumors, was investigated by SDHB IHC. RESULTS: Of the six index patients, all RCCs and one PA displayed SDHB immunonegativity in contrast to the other PA and PTC. All immunonegative tumors demonstrated loss of the WT allele, indicating bi-allelic inactivation of the germline mutated gene. Of 348 tumors, one clear cell RCC exhibited partial loss of SDHB expression. CONCLUSIONS: These findings strengthen the etiological association of SDHx genes with pituitary neoplasia and provide evidence against a link between PTC and SDHx mutations. Somatic deletions seem to constitute the second hit in SDHB-related renal neoplasia, while SDHx alterations do not appear to be primary drivers in sporadic tumorigenesis from tissues affected by SDH deficiency.


Subject(s)
Adenoma/genetics , Carcinoma, Renal Cell/genetics , Mutation , Pituitary Neoplasms/genetics , Succinate Dehydrogenase/genetics , Thyroid Neoplasms/genetics , Adenoma/metabolism , Adenoma/pathology , Adult , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Carcinoma, Papillary/genetics , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Exons , Female , Gene Deletion , Germ-Line Mutation , Humans , Loss of Heterozygosity , Male , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Succinate Dehydrogenase/metabolism , Thyroid Cancer, Papillary , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
12.
Head Neck ; 36(6): 907-16, 2014 Jun.
Article in English | MEDLINE | ID: mdl-23913591

ABSTRACT

BACKGROUND: The purpose of this study was to give an overview on hereditary syndromes associated with head and neck paragangliomas (HNPGs). METHODS: Our methods were the review and discussion of the pertinent literature. RESULTS: About one third of all patients with HNPGs are carriers of germline mutations. Hereditary HNPGs have been described in association with mutations of 10 different genes. Mutations of the genes succinate dehydrogenase subunit D (SDHD), succinate dehydrogenase complex assembly factor 2 gene (SDHAF2), succinate dehydrogenase subunit C (SDHC), and succinate dehydrogenase subunit B (SDHB) are the cause of paraganglioma syndromes (PGLs) 1, 2, 3, and 4. Succinate dehydrogenase subunit A (SDHA), von Hippel-Lindau (VHL), and transmembrane protein 127 (TMEM127) gene mutations also harbor the risk for HNPG development. HNPGs in patients with rearranged during transfection (RET), neurofibromatosis type 1 (NF1), and MYC-associated factor X (MAX) gene mutations have been described very infrequently. CONCLUSION: All patients with HNPGs should be offered a molecular genetic screening. This screening may usually be restricted to mutations of the genes SDHD, SDHB, and SDHC. Certain clinical parameters can help to set up the order in which those genes should be tested.


Subject(s)
Germ-Line Mutation , Head and Neck Neoplasms/genetics , Paraganglioma/genetics , Succinate Dehydrogenase/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Carcinoma, Renal Cell/genetics , Diagnosis, Differential , Gastrointestinal Stromal Tumors/genetics , Genetic Testing , Heterozygote , Humans , Kidney Neoplasms/genetics , Membrane Proteins/genetics , Mitochondrial Proteins/genetics , Multiple Endocrine Neoplasia Type 1/genetics , Neoplastic Syndromes, Hereditary/genetics , Neurofibromatosis 1/genetics , von Hippel-Lindau Disease/genetics
13.
Orphanet J Rare Dis ; 8: 142, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-24034279

ABSTRACT

BACKGROUND: Adrenal Cushing's syndrome caused by ACTH-independent macronodular adrenocortical hyperplasia (AIMAH) can be accompanied by aberrant responses to hormonal stimuli. We investigated the prevalence of adrenocortical reactions to these stimuli in a large cohort of AIMAH patients, both in vivo and in vitro. METHODS: In vivo cortisol responses to hormonal stimuli were studied in 35 patients with ACTH-independent bilateral adrenal enlargement and (sub-)clinical hypercortisolism. In vitro, the effects of these stimuli on cortisol secretion and steroidogenic enzyme mRNA expression were evaluated in cultured AIMAH and other adrenocortical cells. Arginine-vasopressin (AVP) receptor mRNA levels were determined in the adrenal tissues. RESULTS: Positive serum cortisol responses to stimuli were detected in 27/35 AIMAH patients tested, with multiple responses within individual patients occurring for up to four stimuli. AVP and metoclopramide were the most prevalent hormonal stimuli triggering positive responses in vivo. Catecholamines induced short-term cortisol production more often in AIMAH cultures compared to other adrenal cells. Short- and long-term incubation with AVP increased cortisol secretion in cultures of AIMAH cells. AVP also increased steroidogenic enzyme mRNA expression, among which an aberrant induction of CYP11B1. AVP type 1a receptor was the only AVPR expressed and levels were high in the AIMAH tissues. AVPR1A expression was related to the AVP-induced stimulation of CYP11B1. CONCLUSIONS: Multiple hormonal signals can simultaneously induce hypercortisolism in AIMAH. AVP is the most prevalent eutopic signal and expression of its type 1a receptor was aberrantly linked to CYP11B1 expression.


Subject(s)
Arginine Vasopressin/metabolism , Cushing Syndrome/drug therapy , Cushing Syndrome/metabolism , Steroid 11-beta-Hydroxylase/metabolism , Adrenocorticotropic Hormone/metabolism , Aged , Catecholamines/pharmacology , Cells, Cultured , Cushing Syndrome/blood , Cushing Syndrome/enzymology , Female , Glucagon/metabolism , Glucagon/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Humans , Hydrocortisone/metabolism , In Vitro Techniques , Male , Metoclopramide/metabolism , Metoclopramide/pharmacology , Middle Aged , Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/pharmacology
14.
Am J Physiol Endocrinol Metab ; 305(6): E736-44, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23900415

ABSTRACT

Functional zonation of the adrenal cortex is a consequence of the zone-specific expression of P450c17 (CYP17A1) and its cofactors. Activin and inhibin peptides are differentially produced within the zones of the adrenal cortex and have been implicated in steroidogenic control. In this study, we investigated whether activin and inhibin can function as intermediates in functional zonation of the human adrenal cortex. Activin A suppressed CYP17A1 expression and P450c17 function in adrenocortical cell lines as well as in primary adrenal cell cultures. Inhibin ßA-subunit mRNA and activin A protein levels were found to be increased up to 1,900-fold and 49-fold, respectively, after protein kinase C (PKC) stimulation through PMA or angiotensin II in H295R adrenocortical carcinoma cells. This was confirmed in HAC15 cells and for PMA in primary adrenal cell cultures. Both PMA and Ang II decreased CYP17A1 expression in the adrenocortical cell lines, whereas PMA concurrently suppressed CYP17A1 levels in the primary cultures. Inhibition of activin signaling during PKC stimulation through silencing of the inhibin ßA-subunit or blocking of the activin type I receptor opposed the PMA-induced downregulation of CYP17A1 expression and P450c17 function. In contrast, PKA stimulation through adrenocorticotrophin or forskolin increased expression of the inhibin α-subunit and betaglycan, both of which are antagonists of activin action. These data indicate that activin A acts as a PKC-induced paracrine factor involved in the suppression of CYP17A1 in the zona glomerulosa and can thereby contribute to functional adrenocortical zonation.


Subject(s)
Activins/pharmacology , Adrenal Cortex/metabolism , Protein Kinase C/metabolism , Signal Transduction/physiology , Steroid 17-alpha-Hydroxylase/genetics , Activins/genetics , Activins/metabolism , Adrenal Cortex/drug effects , Androstenedione/biosynthesis , Angiotensin II/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Hydrocortisone/biosynthesis , Inhibins/genetics , Inhibins/metabolism , Progesterone/biosynthesis , Signal Transduction/drug effects , Steroid 17-alpha-Hydroxylase/metabolism , Zona Glomerulosa/metabolism
15.
Mol Immunol ; 53(3): 302-12, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23000520

ABSTRACT

T-cell receptor (TCR) repertoire diversity, thymic output, clonal size and peripheral T-lymphocyte numbers largely depend on intra-thymic and post-thymic T-lymphocyte proliferation. However, quantitative insight into thymocyte and T-lymphocyte proliferation is still lacking. We developed a new TCRG-based TCR excision circle (TREC) assay, the Vγ-Jγ TREC assay, which we used together with an adjusted δREC-ψJα TREC assay to quantify the proliferative history of human thymocyte and T-lymphocyte subpopulations from children and adults. This revealed that thymocytes undergo ∼6-8 intra-thymic cell divisions from the double negative (DN) 3 developmental stage onwards, which appeared independent of age. Thus thymocyte proliferation after the DN3 developmental stages is stable and therefore not contributing to the reduced thymic output upon ageing. Cord blood naive T lymphocytes had already undergone ∼2-3 post-thymic cell divisions, which increased to ∼6-7 cell divisions in naive T lymphocytes of middle-aged adults, indicating the importance of homeostatic naive T-lymphocyte proliferation from a young age onwards in the maintenance of peripheral T-lymphocyte numbers. In conclusion, our data provide quantitative insight into the proliferative history of thymocyte and T-lymphocyte subpopulations and alterations herein upon ageing. This novel TREC assay approach could prove valuable in immune status monitoring in a variety of conditions.


Subject(s)
Aging/genetics , Aging/immunology , Genes, T-Cell Receptor alpha , Genes, T-Cell Receptor gamma , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Adolescent , Adult , Aged , Aging/pathology , Cell Proliferation , Child , Child, Preschool , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor , Gene Rearrangement, gamma-Chain T-Cell Antigen Receptor , Genetic Techniques , Humans , Infant, Newborn , Middle Aged , Polymerase Chain Reaction , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Thymus Gland/cytology , Thymus Gland/immunology
16.
Eur J Endocrinol ; 168(1): 67-74, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23065993

ABSTRACT

CONTEXT: Primary pigmented nodular adrenocortical disease (PPNAD) can lead to steroid hormone overproduction. Mutations in the cAMP protein kinase A regulatory subunit type 1A (PRKAR1A) are causative of PPNAD. Steroidogenesis in PPNAD can be modified through a local glucocorticoid feed-forward loop. OBJECTIVE: Investigation of regulation of steroidogenesis in a case of PPNAD with virilization. MATERIALS AND METHODS: A 33-year-old woman presented with primary infertility due to hyperandrogenism. Elevated levels of testosterone and subclinical ACTH-independent Cushing's syndrome led to the discovery of an adrenal tumor, which was diagnosed as PPNAD. In vivo evaluation of aberrantly expressed hormone receptors showed no steroid response to known stimuli. Genetic analysis revealed a PRKAR1A protein-truncating Q28X mutation. After adrenalectomy, steroid levels normalized. Tumor cells were cultured and steroidogenic responses to ACTH and dexamethasone were measured and compared with those in normal adrenal and adrenocortical carcinoma cells. Expression levels of 17ß-hydroxysteroid dehydrogenase (17ß-HSD) types 3 and 5 and steroid receptors were quantified in PPNAD, normal adrenal, and adrenal adenoma tissues. RESULTS: Isolated PPNAD cells, analogous to normal adrenal cells, showed both increased steroidogenic enzyme expression and steroid secretion in response to ACTH. Dexamethasone did not affect steroid production in the investigated types of adrenal cells. 17ß-HSD type 5 was expressed at a higher level in the PPNAD-associated adenoma compared with control adrenal tissue. CONCLUSION: PPNAD-associated adenomas can cause virilization and infertility by adrenal androgen overproduction. This may be due to steroidogenic control mechanisms that differ from those described for PPNAD without large adenomas.


Subject(s)
Adrenal Cortex Diseases/physiopathology , Adrenal Glands/metabolism , Adrenocortical Adenoma/complications , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Virilism/etiology , 17-Hydroxysteroid Dehydrogenases/metabolism , Adenoma/complications , Adenoma/physiopathology , Adenoma/surgery , Adrenal Cortex Diseases/complications , Adrenal Cortex Diseases/surgery , Adrenocortical Adenoma/physiopathology , Adrenocortical Adenoma/surgery , Adrenocorticotropic Hormone/physiology , Adult , Cushing Syndrome , Female , Humans , Testosterone/blood
17.
Mod Pathol ; 26(3): 456-63, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23174939

ABSTRACT

Most gastrointestinal stromal tumors (GISTs) harbor oncogenic mutations in KIT or platelet-derived growth factor receptor-α. However, a small subset of GISTs lacks such mutations and is termed 'wild-type GISTs'. Germline mutation in any of the subunits of succinate dehydrogenase (SDH) predisposes individuals to hereditary paragangliomas and pheochromocytomas. However, germline mutations of the genes encoding SDH subunits A, B, C or D (SDHA, SDHB, SDHC or SDHD; collectively SDHx) are also identified in GISTs. SDHA and SDHB immunohistochemistry are reliable techniques to identify pheochromocytomas and paragangliomas with mutations in SDHA, SDHB, SDHC and SDHD. In this study, we investigated if SDHA immunohistochemistry could also identify SDHA-mutated GISTs. Twenty-four adult wild-type GISTs and nine pediatric/adolescent wild-type GISTs were analyzed with SDHB, and where this was negative, then with SDHA immunohistochemistry. If SDHA immunohistochemistry was negative, sequencing analysis of the entire SDHA coding sequence was performed. All nine pediatric/adolescent GISTs and seven adult wild-type GISTs were negative for SDHB immunohistochemistry. One pediatric GIST and three SDHB-immunonegative adult wild-type GISTs were negative for SDHA immunohistochemistry. In all four SDHA-negative GISTs, a germline SDHA c.91C>T transition was found leading to a nonsense p.Arg31X mutation. Our results demonstrate that SDHA immunohistochemistry on GISTs can identify the presence of an SDHA germline mutation. Identifying GISTs with deficient SDH activity warrants additional genetic testing, evaluation and follow-up for inherited disorders and paragangliomas.


Subject(s)
Biomarkers, Tumor , Electron Transport Complex II , Gastrointestinal Stromal Tumors/enzymology , Germ-Line Mutation , Adolescent , Adult , Age Factors , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Child , DNA Mutational Analysis , Electron Transport Complex II/genetics , Electron Transport Complex II/immunology , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/immunology , Gastrointestinal Stromal Tumors/pathology , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Male , Middle Aged , Phenotype , Young Adult
18.
Ned Tijdschr Geneeskd ; 156(41): A5146, 2012.
Article in Dutch | MEDLINE | ID: mdl-23062260

ABSTRACT

BACKGROUND: Thyroid abnormalities are found in 70% of patients with primary hyperparathyroidism. The simultaneous occurrence of a parathyroid adenoma and non-medullary thyroid carcinoma is rare. CASE DESCRIPTION: A parathyroid adenoma was suspected in a 39-year-old man with hypertension and a previous history of nephrolithiasis on the basis of laboratory results and scintigraphic investigations of the thyroid gland. Histopathological investigation of perioperatively obtained frozen sections of 2 nodules, however, revealed lymph node metastases of a papillary thyroid carcinoma; thyroid carcinoma was confirmed by studying a biopsy of the thyroid gland. Immunohistochemical staining of this biopsy for parathyroid hormone was negative; a parathyroid adenoma could not be confirmed. Total thyroidectomy and exploration of the parathyroid glands followed. Parathyroid hormone and calcium values declined following extirpation of the thyroid gland. Histopathological investigation of the thyroid gland revealed an intrathyroidally situated parathyroid adenoma and a multifocal papillary thyroid carcinoma. CONCLUSION: The case history emphasises the importance of thorough evaluation of the thyroid gland in patients with primary hyperparathyroidism to reveal or exclude simultaneous presentation of parathyroid adenoma and thyroid carcinoma.


Subject(s)
Adenoma/pathology , Carcinoma, Papillary/secondary , Hyperparathyroidism/etiology , Parathyroid Neoplasms/pathology , Thyroid Neoplasms/secondary , Adenoma/surgery , Adult , Carcinoma, Papillary/surgery , Humans , Hyperparathyroidism/pathology , Hyperthyroidism/etiology , Male , Neoplasms, Multiple Primary , Parathyroid Glands/pathology , Parathyroid Glands/surgery , Parathyroid Neoplasms/surgery , Parathyroidectomy , Thyroid Gland/pathology , Thyroid Gland/surgery , Thyroid Neoplasms/surgery , Thyroidectomy , Treatment Outcome
19.
J Clin Endocrinol Metab ; 97(5): E747-54, 2012 May.
Article in English | MEDLINE | ID: mdl-22419722

ABSTRACT

CONTEXT: ACTH stimulates adrenocortical steroid production through the melanocortin 2 receptor (MC2R). MC2R trafficking and signaling are dependent on the MC2R accessory protein (MRAP). The MRAP homolog MRAP2 also transports the MC2R to the cell surface but might prevent activation. OBJECTIVE: The objective of the investigation was to study the regulatory pathways of MRAP and MRAP2 and their contributions to ACTH responsiveness in human adrenal tissues. DESIGN AND SETTING: MRAP, MRAP2, and MC2R expression levels were studied in 32 human adrenocortical samples. Regulation of these mRNAs was investigated in 43 primary adrenal cultures, stimulated with ACTH, forskolin, angiotensin II (AngII), phorbol-12-myristate-13-acetate (PMA), or dexamethasone. The induction of cortisol, cAMP, and ACTH-responsive genes after treatment with ACTH was related to MRAP, MRAP2, and MC2R expression levels. RESULTS: MRAP and MRAP2 levels were lower in adrenocortical carcinomas (ACC) than in other adrenal tissues (P < 0.001). Patient ACTH and cortisol levels were associated with adrenal levels of MRAP and MC2R in adrenal hyperplasia samples (P < 0.05) but not in tumors. ACTH induced the expression of MRAP 11 ± 2.1-fold and MC2R 20 ± 3.8-fold in all adrenal tissue types (mean ± SEM, both P < 0.0001), whereas AngII augmented these mRNAs 4.0 ± 1.2-fold and 12.6 ± 3.2-fold (P < 0.0001) in all but the ACC. MRAP2 expression was suppressed by forskolin (-24 ± 15%, P = 0.013) and PMA (-22 ± 7%, P = 0.0007). MRAP, MRAP2, or MC2R levels were not associated with the induction of cortisol, cAMP, or gene expression by ACTH in vitro. CONCLUSION: MRAP and MC2R expression is induced by ACTH and AngII, which would facilitate cell surface receptor availability. Physiological expression levels of MRAP, MRAP2, and MC2R were not limiting for ACTH sensitivity.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenal Cortex/metabolism , Adrenocortical Carcinoma/genetics , Adrenocorticotropic Hormone/pharmacology , Carrier Proteins/genetics , Membrane Proteins/genetics , Adaptor Proteins, Signal Transducing , Adrenal Cortex/drug effects , Adrenal Cortex Neoplasms/metabolism , Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/metabolism , Adrenocortical Carcinoma/pathology , Adrenocorticotropic Hormone/metabolism , Carrier Proteins/metabolism , Cells, Cultured , Gene Expression Regulation , Humans , Hydrocortisone/blood , Membrane Proteins/metabolism
20.
J Clin Endocrinol Metab ; 96(9): E1472-6, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21752896

ABSTRACT

CONTEXT: Pheochromocytoma-paraganglioma syndrome is caused by mutations in SDHB, SDHC, and SDHD, encoding subunits of succinate dehydrogenase (SDH), and in SDHAF2, required for flavination of SDHA. A recent report described a patient with an abdominal paraganglioma, immunohistochemically negative for SDHA, and identified a causal germline mutation in SDHA. OBJECTIVE: In this study, we evaluated the significance of SDHA immunohistochemistry in the identification of new patients with SDHA mutations. SETTING: This study was performed in the Erasmus Medical Center in Rotterdam (The Netherlands) and the Université Paris Descartes in Paris (France). METHODS: We investigated 316 pheochromocytomas and paragangliomas for SDHA expression. Sequence analysis of SDHA was performed on all tumors that were immunohistochemically negative for SDHA and on a subset of tumors immunohistochemically positive for SDHA. RESULTS: Six tumors were immunohistochemically negative for SDHA. Four tumors from Dutch patients showed a germline c.91C → T SDHA gene mutation (p.Arg31X). Another tumor (from France) carried a germline SDHA missense mutation c.1753C → T (p.Arg585Trp). Loss of the wild-type SDHA allele was confirmed by loss of heterozygosity analysis. Sequence analysis of 35 SDHA immunohistochemically positive tumors did not reveal additional SDHA mutations. CONCLUSIONS: Our results demonstrate that SDHA immunohistochemistry on paraffin-embedded tumors can reveal the presence of SDHA germline mutations and allowed the identification of SDHA-related tumors in at least 3% of patients affected by apparently sporadic (para)sympathetic paragangliomas and pheochromocytomas.


Subject(s)
Adrenal Gland Neoplasms/genetics , Electron Transport Complex II/genetics , Germ-Line Mutation , Paraganglioma/genetics , Pheochromocytoma/genetics , Adrenal Gland Neoplasms/metabolism , Alleles , DNA Mutational Analysis , Electron Transport Complex II/metabolism , Humans , Immunohistochemistry , Loss of Heterozygosity , Paraganglioma/metabolism , Pheochromocytoma/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...