Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
Am J Surg Pathol ; 46(2): 159-168, 2022 02 01.
Article En | MEDLINE | ID: mdl-34560682

Patients with multiple endocrine neoplasia 1 syndrome (MEN1) often develop multifocal duodenopancreatic neuroendocrine tumors (dpNETs). Nonfunctional pancreatic neuroendocrine tumors (PanNETs) and duodenal gastrinomas are the most frequent origins of metastasis. Current guidelines recommend surgery based on tumor functionality, size ≥2 cm, grade or presence of lymph node metastases. However, in case of multiple primary tumors it is often unknown which specific tumor metastasized. This study aims to unravel the relationship between primary dpNETs and metastases in patients with MEN1 by studying endocrine differentiation. First, it was shown that expression of the endocrine differentiation markers ARX and PDX1 was concordant in 18 unifocal sporadic neuroendocrine tumors (NETs) and matched metastases. Thereafter, ARX, PDX1, Ki67 and gastrin expression, and the presence of alternative lengthening of telomeres were determined in 137 microscopic and macroscopic dpNETs and 36 matched metastases in 10 patients with MEN1. ARX and PDX1 H-score clustering was performed to infer relatedness. For patients with multiple metastases, similar intrametastases transcription factor expression suggests that most metastases (29/32) originated from a single NET of origin, while few patients may have multiple metastatic primary NETs. In 6 patients with MEN1 and hypergastrinemia, periduodenopancreatic lymph node metastases expressed gastrin, and clustered with minute duodenal gastrinomas, not with larger PanNETs. PanNET metastases often clustered with high grade or alternative lengthening of telomeres-positive primary tumors. In conclusion, for patients with MEN1-related hypergastrinemia and PanNETs, a duodenal origin of periduodenopancreatic lymph node metastases should be considered, even when current conventional and functional imaging studies do not reveal duodenal tumors preoperatively.


Carcinoma, Neuroendocrine/secondary , Duodenal Neoplasms/pathology , Multiple Endocrine Neoplasia Type 1/pathology , Pancreatic Neoplasms/pathology , Adult , Aged , Biomarkers, Tumor/analysis , Carcinoma, Neuroendocrine/chemistry , Carcinoma, Neuroendocrine/genetics , Databases, Factual , Duodenal Neoplasms/chemistry , Duodenal Neoplasms/genetics , Female , Gastrins/analysis , Homeodomain Proteins/analysis , Humans , Ki-67 Antigen/analysis , Lymphatic Metastasis , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/chemistry , Multiple Endocrine Neoplasia Type 1/genetics , Neoplasm Grading , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/genetics , Trans-Activators/analysis , Transcription Factors/analysis
2.
Magn Reson Med ; 74(5): 1221-6, 2015 Nov.
Article En | MEDLINE | ID: mdl-25392979

PURPOSE: MR spectroscopy (MRS) can improve diagnosis and follow treatment in cancer. However, no study has yet reported application of in vivo (1)H-MRS in malignant pancreatic lesions. This study quantitatively determined whether in vivo (1)H-MRS on multiple endocrine neoplasia type 1 (Men1) conditional knockout (KO) mice and their wild type (WT) littermates could detect differences in total choline (tCho) levels between tumor and control pancreas. METHODS: Relative tCho levels in pancreatic tumors or pancreata from KO and WT mice were determined using in vivo (1)H-MRS at 9.4 T. The levels of Cho-containing compounds were also quantified using in vitro (1)H-NMR on extracts of pancreatic tissues from KO and WT mice, respectively, and on extracts of pancreatic tissues from patients with pancreatic neuroendocrine tumors (PNETs). RESULTS: tCho levels measured by in vivo (1)H-MRS were significantly higher in PNETs from KO mice compared to the normal pancreas from WT mice. The elevated choline-containing compounds were also identified in pancreatic tumors from KO mice and tissues from patients with PNETs via in vitro (1)H-NMR. CONCLUSION: These results indicate the potential use of tCho levels estimated via in vivo (1)H-MRS in differentiating malignant pancreatic tumors from benign tumors.


Magnetic Resonance Imaging/methods , Multiple Endocrine Neoplasia Type 1/chemistry , Multiple Endocrine Neoplasia Type 1/diagnosis , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/diagnosis , Animals , Choline/analysis , Choline/chemistry , Choline/metabolism , Female , Male , Mice , Mice, Knockout , Multiple Endocrine Neoplasia Type 1/pathology , Pancreatic Neoplasms/pathology , Transcription Factors/genetics
3.
Fam Cancer ; 13(2): 273-80, 2014 Jun.
Article En | MEDLINE | ID: mdl-24522746

Multiple endocrine neoplasia type 1 (MEN1) syndrome is an autosomal dominant disease, characterized by parathyroid adenomas, endocrine gastroenteropancreatic tumors and pituitary adenomas, due to inactivating mutations of the MEN1 gene (chromosome 11q13). MEN1 mutations are mainly represented by nonsense, deletions/insertions, splice site or missense mutations that can be detected by direct sequencing of genomic DNA. However, MEN1 patients with large heterozygous deletions may escape classical genetic screening and may be misidentified as phenocopies, thereby hindering proper clinical surveillance. We employed a real-time polymerase chain reaction application, the TaqMan copy number variation assay, to evaluate a family in which we failed to identify an MEN1 mutation by direct sequencing, despite a clear clinical diagnosis of MEN1 syndrome. Using the TaqMan copy number variation assay we identified a large deletion of the MEN1 gene involving exons 1 and 2, in three affected family members, but not in the other nine family members that were to date clinically unaffected. The same genetic alteration was not found in a group of ten unaffected subjects, without family history of endocrine tumors. The MEN1 deletion was further confirmed by multiplex ligation-dependent probe amplification, which showed the deletion extended from exon 1 to exon 3. This new approach allowed us to correctly genetically diagnose three clinical MEN1 patients that were previously considered as MEN1 phenocopies. More importantly, we excluded the presence of genetic alterations in the unaffected family members. These results underline the importance of using a variety of available biotechnology approaches when pursuing a genetic diagnosis in a clinically suggestive setting of inherited endocrine cancer.


Base Sequence , Genetic Carrier Screening/methods , Multiple Endocrine Neoplasia Type 1/genetics , Proto-Oncogene Proteins/genetics , Sequence Deletion , Adolescent , Adult , Aged , Exons , Female , Humans , Italy , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/chemistry , Multiplex Polymerase Chain Reaction , Pedigree , Proto-Oncogene Proteins/analysis , Real-Time Polymerase Chain Reaction , Young Adult
4.
J Clin Endocrinol Metab ; 94(10): 3640-6, 2009 Oct.
Article En | MEDLINE | ID: mdl-19622622

CONTEXT: Multiple endocrine neoplasia type 1 (MEN1) is characterized by the occurrence of parathyroid, pituitary, and pancreatic tumors. MEN1, an autosomal dominant disorder, has a high degree of penetrance, such that more than 95% of patients develop clinical manifestations by the fifth decade, although this is lower at approximately 50% by age 20 yr. However, the lower penetrance in the younger group, which is based on detecting hormone-secreting tumors, may be an underestimate because patients may have nonfunctioning tumors and be asymptomatic. OBJECTIVE: The aim of the study was to evaluate the occurrence of nonfunctioning pancreatic neuroendocrine tumors in asymptomatic children with MEN1. PATIENTS: Twelve asymptomatic Northern European children, aged 6 to 16 yr, who were known to have MEN1 mutations were studied. RESULTS: Two asymptomatic children, who were aged 12 and 14 yr, had normal plasma fasting gastrointestinal hormones and were found to have nonfunctioning pancreatic neuroendocrine tumors that were more than 2 cm in size. Surgery and immunostaining revealed that the tumors did not have significant expression of gastrointestinal hormones but did contain chromogranin A and synaptophysin, features consistent with those of nonfunctioning pancreatic neuroendocrine tumors. The tumors had a loss of menin expression. The 14 yr old also had primary hyperparathyroidism and a microprolactinoma, and the 12 yr old had a nonfunctioning pituitary microadenoma. Three other children had primary hyperparathyroidism and a microprolactinoma. CONCLUSION: Nonfunctioning pancreatic neuroendocrine tumors may occur in asymptomatic children with MEN1 mutations, and screening for such enteropancreatic tumors in MEN1 children should be considered earlier than the age of 20 yr, as is currently recommended by the international guidelines.


Biomarkers, Tumor/analysis , Multiple Endocrine Neoplasia Type 1/genetics , Mutation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/surgery , Proto-Oncogene Proteins/genetics , Adenoma/genetics , Adolescent , Child , Chromogranin A/analysis , Europe , Female , Gene Deletion , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Hyperparathyroidism/genetics , Immunohistochemistry , Male , Multiple Endocrine Neoplasia Type 1/chemistry , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/pathology , Pedigree , Pituitary Neoplasms/genetics , Prolactinoma/genetics , Synaptophysin/analysis
5.
Am J Surg Pathol ; 33(5): 739-48, 2009 May.
Article En | MEDLINE | ID: mdl-19238077

INTRODUCTION: Von Hippel-Lindau (VHL) disease is an inherited syndrome caused by germline mutation in the VHL tumor suppressor gene predisposing to pancreatic endocrine tumors (PET). Whether these tumors derive from preexisting endocrine microadenomatosis as in multiple endocrine neoplasia type 1 (MEN1) is yet unknown. pVHL regulates hypoxia-inducible factor (HIF) that causes transcription activity of target genes like carbonic anhydrase 9 (CA9), vascular endothelial growth factor (VEGF), and cyclin D1. Our aim was to look for overexpression of these molecules to identify precursor endocrine lesions in the pancreas of VHL patients. METHODS: Nontumoral pancreas of 18 VHL patients operated on for PET, was examined for microadenomatosis (70% of VHL patients operated on for PET. These results demonstrate that the pVHL/HIF pathway is involved very early in pancreatic endocrine tumorigenesis in this disease.


Adenoma/chemistry , Basic Helix-Loop-Helix Transcription Factors/analysis , Biomarkers, Tumor/analysis , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Pancreas/chemistry , Pancreatic Neoplasms/chemistry , Precancerous Conditions/chemistry , Von Hippel-Lindau Tumor Suppressor Protein/genetics , von Hippel-Lindau Disease/metabolism , Adenoma/genetics , Adenoma/surgery , Adult , Antigens, CD34/analysis , Antigens, Neoplasm/analysis , Carbonic Anhydrase IX , Carbonic Anhydrases/analysis , Chromogranin A/analysis , Cyclin D1/analysis , Female , Glucagon/analysis , Humans , Immunohistochemistry , Insulin/analysis , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/chemistry , Multiple Endocrine Neoplasia Type 1/genetics , Mutation , Pancreas/surgery , Pancreatectomy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/surgery , Precancerous Conditions/genetics , Precancerous Conditions/surgery , Vascular Endothelial Growth Factor A/analysis , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/surgery
6.
Am J Surg Pathol ; 32(4): 534-43, 2008 Apr.
Article En | MEDLINE | ID: mdl-18300794

Patients affected by the multiple endocrine neoplasia type I syndrome (MEN1) display a high incidence of pituitary adenomas, though it is still unknown whether these pituitary tumors have specific pathologic features that would distinguish them from sporadic pituitary adenomas. Pituitary tissue specimens of 77 MEN1 patients from the GTE (Groupe d'étude des Tumeurs Endocrines) register were compared with unselected 2509 non-MEN1 sporadic pituitary tumors and also to a control subgroup of 296 cases, where 1 MEN1 tumor was matched with 4 sporadic tumors of the same hormonal immunoprofile. Sex, age, size, and invasiveness of tumors, and menin gene mutations were documented. Histologic analysis took into account 33 items, including immunocytochemical data, the proliferative marker Ki-67, and an examination of the juxtatumoral pituitary. MEN1 tumors were significantly larger and more often invasive by histology. MEN1 patients with large pituitary tumors (grade IV) were younger than non-MEN1 patients. MEN1 tumors had no other characteristic histologic features and no predominance of any one hormone producing subtype. However, plurihormonal adenomas versus monohormonal and nonimmunoreactive adenomas were more frequent in MEN1 tumors (39%) than in the control non-MEN1 group (P = 0.001). Especially, the growth hormone and prolactin plurihormonality with unusual association with follicle-stimulating hormone, luteinizing hormone, or adrenocorticotropic hormone was more frequent in MEN1 tumors. In addition, multiple adenomas were significantly more frequent (4% vs. 0.1%; P < 0.0001), especially prolactin-adrenocorticotropic hormone. Somatotroph hyperplasia, with or without a microadenoma was found in only 3 MEN1 patients, with growth hormone-releasing hormone hypersecretion by a pancreatic tumor in 2 of them. All types of mutation were observed, including frameshifts, nonsenses, missenses, and 1 case of germline MEN1 encompassing large deletion, strongly suggesting the absence of any phenotype-genotype correlation.


Adenoma/pathology , Multiple Endocrine Neoplasia Type 1/pathology , Pituitary Gland/pathology , Pituitary Neoplasms/pathology , Adenoma/chemistry , Adenoma/genetics , Adolescent , Adrenocorticotropic Hormone/analysis , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cell Proliferation , Female , Follicle Stimulating Hormone/analysis , Gene Expression Regulation, Neoplastic , Human Growth Hormone/analysis , Humans , Hyperplasia , Immunohistochemistry , Ki-67 Antigen/analysis , Luteinizing Hormone/analysis , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/chemistry , Multiple Endocrine Neoplasia Type 1/genetics , Mutation , Neoplasm Invasiveness , Neoplasm Staging , Pituitary Gland/chemistry , Pituitary Neoplasms/chemistry , Pituitary Neoplasms/genetics , Prolactin/analysis , Proto-Oncogene Proteins/genetics , Thyrotropin/analysis
7.
Clin Endocrinol (Oxf) ; 66(1): 115-22, 2007 Jan.
Article En | MEDLINE | ID: mdl-17201810

OBJECTIVE: Expression of ghrelin has been reported in pancreatic endocrine tumours, but data on ghrelin receptor protein expression are lacking. The aim of this study was to examine the ghrelin receptor, as well as ghrelin, in a selected series of these tumours, including multiple endocrine neoplasia 1 (MEN1) associated tumours, and to correlate data with clinical features including body mass index. DESIGN: Immunohistochemical detection of ghrelin and its receptor was performed on frozen tissue from 31 tumours: 9 MEN1 and 22 sporadic. Twenty tumours were analysed by quantitative PCR. Plasma ghrelin was assessed in 26 patients. RESULTS: Twenty-one (68%) of 31 tumours showed immunoreactivity for ghrelin (8/9 MEN1) and 19/20 expressed ghrelin mRNA. Ghrelin receptor protein was detected in 21/30 (70%) tumours (4/8 MEN1), and mRNA was detected in all analysed tumours. Insulinomas had significantly higher levels of receptor mRNA than other tumours. Five patients had elevated plasma ghrelin (> 2 SD above the control group mean). No significant difference in mean plasma ghrelin levels was found between patients (908 +/- 569 ng/l) and controls (952 +/- 164 ng/l). Mean BMI was 24.3 kg/m(2). There was no association between ghrelin or receptor expression and survival. CONCLUSIONS: We report the first immunohistochemical data on expression of the ghrelin receptor in pancreatic endocrine tumours: 70% of tumours in our material. Concomitant ghrelin and receptor expression was seen in 50% of tumours, indicating an autocrine loop. Ghrelin was expressed in 68% of tumours (8/9 MEN1). Despite frequent ghrelin expression, elevated circulating ghrelin is rare in these patients.


Adenoma, Islet Cell/metabolism , Pancreatic Neoplasms/metabolism , Peptide Hormones/genetics , Receptors, G-Protein-Coupled/genetics , Adenoma, Islet Cell/blood , Adenoma, Islet Cell/chemistry , Adult , Aged , Aged, 80 and over , Body Mass Index , Chi-Square Distribution , Female , Gene Expression , Ghrelin , Humans , Immunohistochemistry , Insulinoma/chemistry , Insulinoma/metabolism , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/chemistry , Multiple Endocrine Neoplasia Type 1/metabolism , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/chemistry , Peptide Hormones/analysis , Peptide Hormones/blood , Receptors, G-Protein-Coupled/analysis , Receptors, Ghrelin , Survival Rate
8.
Int J Cancer ; 120(2): 259-67, 2007 Jan 15.
Article En | MEDLINE | ID: mdl-17044021

Multiple endocrine neoplasia type 1 (MEN1) is an inherited cancer predisposition syndrome typified by development of tumors in parathyroid, pituitary and endocrine pancreas, as well as less common sites including both endocrine and nonendocrine organs. Deletion or mutation of the tumor suppressor gene MEN1 on chromosome 11 has been identified in many cases of MEN1 as well as in sporadic tumors. The molecular biology of menin, the protein encoded by MEN1, remains poorly understood. Here we describe a mouse model of MEN1 in which tumors were seen in pancreatic islets, pituitary, thyroid and parathyroid, adrenal glands, testes and ovaries. The observed tumor spectrum therefore includes types commonly seen in MEN1 patients and additional types. Pancreatic pathology was most common, evident in over 80% of animals, while other tumor types developed with lower frequency and generally later onset. Tumors of multiple endocrine organs were observed frequently, but progression to carcinoma and metastasis were not evident. Tumors in all sites showed loss of heterozygosity at the Men1 locus, though the frequency in testicular tumors was only 36%, indicating that a different molecular mechanism of tumorigenesis occurs in those Leydig tumors that do not show loss of the normal Men1 allele. Menin expression was below the level of detection in ovary, thyroid and testis, but loss of nuclear menin immunoreactivity was observed uniformly in all pancreatic islet adenomas and in some hyperplastic islet cells, suggesting that complete loss of Men1 is a critical point in islet tumor progression in this model.


Adenoma/pathology , Disease Models, Animal , Endocrine Gland Neoplasms/pathology , Mice/genetics , Multiple Endocrine Neoplasia Type 1/pathology , Proto-Oncogene Proteins/genetics , Adenoma/chemistry , Adenoma/genetics , Animals , DNA, Neoplasm/analysis , Endocrine Gland Neoplasms/chemistry , Endocrine Gland Neoplasms/genetics , Exons/genetics , Female , Genes, Tumor Suppressor , Male , Multiple Endocrine Neoplasia Type 1/chemistry , Multiple Endocrine Neoplasia Type 1/genetics , Peptide Chain Initiation, Translational/genetics , Proto-Oncogene Proteins/analysis
9.
Endocr Pathol ; 17(1): 75-81, 2006.
Article En | MEDLINE | ID: mdl-16760583

The cytoplasm of pancreatic endocrine tumors (PET) can show a diverse range of appearances from clear, to oncocytic, to intracellular mucin accumulation, and the presence of intracytoplasmic inclusions. Intracytoplasmic eosinophilic inclusions can vary morphologically and the spectrum ranges from small, dot-like hyaline inclusions, to deeply acidophilic/eosinophilic ones that occupy almost the whole cytoplasm and displace the nucleus eccentrically: the so-called "rhabdoid" phenotype. The aim of this study was to analyze the frequency, morphology, behavior, and relationship to clinicopathological features of large intracytoplasmic inclusions, including the rhabdoid phenotype, in a large number of PET. The morphological features of 84 cases were assessed for the presence of large, globular intracytoplasmic inclusions. Fourteen of 84 cases contained intracytoplasmic inclusions with 5 cases containing cells conforming to the characteristic rhabdoid morphology. The remaining nine cases showed pale intracytoplasmic inclusions. Four of the five cases with rhabdoid cells had spread to lymph nodes and/or peripancreatic fatty tissue. This study confirms that a spectrum of large intracytoplasmic inclusions is encountered in PET, ranging from lightly eosinophilic intracytoplasmic globules to the more typical rhabdoid phenotype (deeply eosinophilic inclusions). This phenotype, in particular the rhabdoid cells, is worthy of attention as a proportion may show lymphovascular invasion with evidence of metastasis at the time of presentation, irrespective of size, mitotic rates, or necrosis.


Adenoma, Islet Cell/pathology , Carcinoma, Islet Cell/pathology , Inclusion Bodies/pathology , Multiple Endocrine Neoplasia Type 1/pathology , Pancreatic Neoplasms/pathology , Rhabdoid Tumor/pathology , Adenoma, Islet Cell/chemistry , Adult , Aged , Biomarkers, Tumor/analysis , Carcinoma, Islet Cell/chemistry , Female , Humans , Immunohistochemistry , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/chemistry , Pancreatic Neoplasms/chemistry , Retrospective Studies , Rhabdoid Tumor/chemistry
10.
Oncology ; 66(6): 468-75, 2004.
Article En | MEDLINE | ID: mdl-15452376

OBJECTIVE: To determine the prognostic value of valosin-containing protein (VCP) expression and the Ki-67 labeling index (LI) in pancreatic endocrine neoplasms (PENs), the present analysis was employed. METHODS: The Ki-67 LI and VCP expression at the mRNA and protein level were evaluated in 32 patients (12 male and 20 female) with PENs aged from 22 to 73 years (median 49 years). VCP staining intensity in tumor cells was categorized as weaker (level 1) or equal to stronger (level 2) compared to nontumorous islet cells. Ki-67 LI was divided into two categories: level 1, Ki-67 LI < 5%, and level 2, > or = 5%. RESULTS: Five cases (15.6%) showed level 1 and 25 (84.4%) level 2 VCP expression by immunohistochemistry. A significant association was observed between VCP expression and the malignant behavior of PENs (p < 0.01). All level 1 VCP tumors were benign PENs. Quantitative reverse transcription polymerase chain reaction analysis showed higher VCP mRNA expression in malignant PENs (n = 5) than benign cases (n = 5) (p < 0.05). For Ki-67 LI, 28 cases (87.5%) showed level 1 and 4 (12.5%) level 2 expression. All patients with level 2 Ki-67 LI had metastasis. CONCLUSION: VCP expression analysis and Ki-67 LI are useful prognosticators for PENs.


Biomarkers, Tumor/analysis , Cell Cycle Proteins/analysis , Ki-67 Antigen/analysis , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/pathology , Adenosine Triphosphatases , Adult , Aged , Cell Cycle Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Ki-67 Antigen/genetics , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/chemistry , Multiple Endocrine Neoplasia Type 1/pathology , Odds Ratio , Predictive Value of Tests , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Valosin Containing Protein
11.
Clin Endocrinol (Oxf) ; 57(1): 131-4, 2002 Jul.
Article En | MEDLINE | ID: mdl-12100081
12.
Cancer ; 92(6): 1393-401, 2001 Sep 15.
Article En | MEDLINE | ID: mdl-11745215

BACKGROUND: Adrenocortical tumors occur as sporadic tumors, as part of the multiple endocrine neoplasia type 1 (MEN1) syndrome, or as part of other hereditary disorders. MEN1 is a tumor suppressor gene located on chromosome 11q13 that encodes a 610-amino acid protein called menin, and plays an important role in the development of MEN1 syndrome. Recent reports indicate that heterozygous germline mutations of this gene are responsible for the disease onset of MEN1. METHODS: To investigate the role of menin in sporadic adrenocortical tumors, the authors examined a series of adrenocortical adenoma cases and a single case of carcinoma and adrenomedulary tumors with the corresponding adjacent tumor tissues using reverse transcriptase-polymerase chain reaction (RT-PCR) for menin mRNA and Western blot analysis for menin protein. Both RNA and protein from these tumors were applied to RT-PCR and Western blot analysis, respectively, although they are not truly quantitative. Primers for RT-PCR were designed to amplify the sequence between exons 2 and 3 of the MEN1 gene. A specific antibody against menin was generated in guinea pigs immunized with the recombinant peptide from the amino acid residues 443-535 of menin made by using glutathione-S-transferase gene fusion. RESULTS: Based on the results of RT-PCR and Western blot analysis, both MEN1 mRNA and menin protein appeared to be highly expressed in Cushing syndrome resulting from adrenocortical adenomas and carcinoma. However, their expression was found to be greatly decreased in primary aldosteronism compared with their expression in Cushing syndrome. Although weak expression of MEN1 mRNA also was detected in pheochromocytoma on RT-PCR, menin expression was not detected in any case of pheochromocytoma by Western blot analysis, possibly due to the lower sensitivity of this assay compared with RT-PCR. Neither MEN1 mRNA nor menin protein was detected in any of the corresponding adjacent tumor tissues examined. CONCLUSIONS: The findings of the current study indicate that menin expression appears to be up-regulated in Cushing syndrome, suggesting that adrenocortical proliferation might be one of the primary lesions in the MEN1 syndrome in which menin might play a significant role in some specific cellular functions.


Adrenal Gland Neoplasms/chemistry , Neoplasm Proteins/analysis , Neoplasm Proteins/physiology , Proto-Oncogene Proteins , Adenoma/chemistry , Adolescent , Adrenal Cortex Neoplasms/chemistry , Adrenal Medulla , Adult , Aged , Blotting, Western , Carcinoma/chemistry , Cushing Syndrome/etiology , Cushing Syndrome/metabolism , Female , Humans , Hyperaldosteronism/etiology , Hyperaldosteronism/metabolism , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/chemistry , Neoplasm Proteins/genetics , Pheochromocytoma/chemistry , Polymerase Chain Reaction , RNA, Messenger/analysis , Up-Regulation
13.
Ann Pathol ; 16(3): 167-73, 1996.
Article Fr | MEDLINE | ID: mdl-8766173

The Multiple Endocrine Neoplasia (MEN I) or Wermer's syndrome is an uncommon disease which is most often inherited and affects mainly parathyroid glands, pancreatic islets and pituitary gland. The aim of this study concerning 61 pancreatic tumors in 16 patients suffering from MEN I was to define the macroscopic, histological and immunohistochemical characteristics of these tumors. The pancreatic endocrine tumors as part of the MEN I syndrome concern multiple tumors of small size, localized most often to the pancreas's tail. In 79% of cases, these tumors have a different predominating peptidic hormonal secretion in a same patient though most of them have plurihormonal secretions. The pancreatic polyendocrinopathy detection imposes a family investigation to look for a type I polyendocrinopathy.


Multiple Endocrine Neoplasia Type 1/pathology , Pancreatic Neoplasms/pathology , Adolescent , Adult , Child , Female , Humans , Immunohistochemistry , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/chemistry , Pancreatic Neoplasms/chemistry
14.
J Clin Endocrinol Metab ; 79(5): 1457-64, 1994 Nov.
Article En | MEDLINE | ID: mdl-7962343

The functional study of SRIH receptors was performed in ectopic GHRH-secreting tumors from two patients with acromegaly; patient 1 presented with multiple endocrine neoplasia type 1 with GHRH- and insulin-secreting pancreatic tumors, and patient 2 presented with a multihormone-secreting carcinoid tumor (including GHRH and alpha-subunit secretion, as demonstrated by clinical and immunohistochemical studies). In both cases, plasma GH levels were responsive to octreotide. In patient 2, plasma GHRH and alpha-subunit levels were responsive to octreotide. In vitro perifusion studies of a tumor fragment from patient 1 also showed inhibition of GHRH secretion by SRIH. A high density of specific SRIH-binding sites was visualized by autoradiography in GHRH tumors from both patients. SRIH specific binding was much higher in the GHRH tumors (6.6-8.4 fmol/surface unit) than in the insulinoma (1.9 fmol/surface unit). The binding inhibition constant (IC50) was in the nanomolar range (0.9-3 nmol/L) in the GHRH tumors. SRIH-14 inhibited forskolin-stimulated adenylate cyclase in the GHRH tumors from both patients, but not in the insulinoma. The functional SRIH receptors negatively coupled to adenylate cyclase present in ectopic GHRH-secreting tumors mediate the inhibitory effect of octreotide on GHRH secretion and on previously underrecognized ectopic alpha-subunit secretion from carcinoid tumors.


Acromegaly/drug therapy , Acromegaly/metabolism , Adenylyl Cyclases/metabolism , Growth Hormone-Releasing Hormone/metabolism , Multiple Endocrine Neoplasia Type 1/chemistry , Octreotide/therapeutic use , Pancreatic Neoplasms/chemistry , Receptors, Somatostatin/analysis , Receptors, Somatostatin/metabolism , Adult , Carcinoid Tumor/chemistry , Carcinoid Tumor/metabolism , Colforsin/pharmacology , Female , Growth Hormone/blood , Growth Hormone-Releasing Hormone/analysis , Growth Hormone-Releasing Hormone/blood , Humans , Immunohistochemistry , Insulinoma/chemistry , Insulinoma/metabolism , Male , Multiple Endocrine Neoplasia Type 1/metabolism , Pancreatic Neoplasms/metabolism
...