Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Hum Genet ; 99(5): 1034-1044, 2016 Nov 03.
Article in English | MEDLINE | ID: mdl-27745835

ABSTRACT

Primary hyperparathyroidism (PHPT) is a common endocrine disease characterized by parathyroid hormone excess and hypercalcemia and caused by hypersecreting parathyroid glands. Familial PHPT occurs in an isolated nonsyndromal form, termed familial isolated hyperparathyroidism (FIHP), or as part of a syndrome, such as multiple endocrine neoplasia type 1 or hyperparathyroidism-jaw tumor syndrome. The specific genetic or other cause(s) of FIHP are unknown. We performed exome sequencing on germline DNA of eight index-case individuals from eight unrelated kindreds with FIHP. Selected rare variants were assessed for co-segregation in affected family members and screened for in an additional 32 kindreds with FIHP. In eight kindreds with FIHP, we identified three rare missense variants in GCM2, a gene encoding a transcription factor required for parathyroid development. Functional characterization of the GCM2 variants and deletion analyses revealed a small C-terminal conserved inhibitory domain (CCID) in GCM2. Two of the three rare variants were recurrent, located in the GCM2 CCID, and found in seven of the 40 (18%) kindreds with FIHP. These two rare variants acted as gain-of-function mutations that increased the transcriptional activity of GCM2, suggesting that GCM2 is a parathyroid proto-oncogene. Our results demonstrate that germline-activating mutations affecting the CCID of GCM2 can cause FIHP.


Subject(s)
Adenoma/genetics , Fibroma/genetics , Germ-Line Mutation , Hyperparathyroidism, Primary/genetics , Hyperparathyroidism/genetics , Jaw Neoplasms/genetics , Multiple Endocrine Neoplasia Type 1/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Adenoma/diagnosis , Adolescent , Adult , Aged , Amino Acid Sequence , Exome , Female , Fibroma/diagnosis , Genetic Variation , Humans , Hyperparathyroidism/diagnosis , Hyperparathyroidism, Primary/diagnosis , Jaw Neoplasms/diagnosis , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/diagnosis , Parathyroid Hormone , Pedigree , Proto-Oncogene Mas , Proto-Oncogenes/genetics , Sequence Analysis, DNA , Young Adult
2.
Gastroenterology ; 154(4): 998-1008.e1, 2018 03.
Article in English | MEDLINE | ID: mdl-29155309

ABSTRACT

BACKGROUND & AIMS: Survival times vary among patients with neuroendocrine tumors (NETs) - even among those with the same site, stage, and grade of primary tumor. This makes it difficult to select treatment for patients with unresectable NETs because some patients can survive decades without treatment. 68Gallium-DOTATATE positron emission tomography with computed tomography (68Ga-DOTATATE PET/CT) is a sensitive imaging technique for detection of NETs. We investigated the prognostic accuracy of 68Ga-DOTATATE PET/CT-based analysis of tumor volume in patients with NETs. METHODS: We performed a prospective study of 184 patients with NETs (128 [69.6%] with metastases and 11 patients [6.0%] with locally advanced disease) at the National Institutes of Health Clinical Center (Bethesda, MD) from 2013 through 2017. All patients underwent 68Ga-DOTATATE PET/CT image analysis and total 68Ga-DOTATATE-Avid tumor volume (68Ga-DOTATATE TV) was determined. We also measured fasting serum chromogranin A, neuron-specific enolase, gastrin, glucagon, vasoactive intestinal peptide, pancreatic polypeptide, and 24-hour urinary 5-hydroxyindoleacetic acid levels in all patients. Disease progression was defined as a new lesion or a growth of a known lesion during the interval between baseline 68Ga-DOTATATE PET/CT scan and follow-up imaging (14.0 ± 6.1 months; range, 1-35 months). The primary outcomes were progression-free survival (PFS) and disease-specific mortality during a median follow-up time of 18 months (range, 4-35 months). RESULTS: We found an inverse correlation between quartiles of 68Ga-DOTATATE TV and PFS (P = .001) and disease-specific survival (P = .002). A 68Ga-DOTATATE TV of 7.0 mL or more was associated with higher odds of disease progression (hazard ratio, 3.0; P = .04). A 68Ga-DOTATATE TV of 35.8 mL or more was associated with increased risk of disease-specific death (hazard ratio, 10.6) in multivariable analysis (P = .01), as well as in subgroup analysis of patients with pancreatic NETs. CONCLUSIONS: In a prospective study, we demonstrated the prognostic utility of 68Ga-DOTATATE TV in a large cohort of patients with NETs, in terms of PFS and disease-specific mortality.


Subject(s)
Gastrointestinal Neoplasms/diagnostic imaging , Neuroendocrine Tumors/diagnostic imaging , Organometallic Compounds/administration & dosage , Pancreatic Neoplasms/diagnostic imaging , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals/administration & dosage , Adult , Aged , Chi-Square Distribution , Disease Progression , Disease-Free Survival , Female , Gastrointestinal Neoplasms/mortality , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/therapy , Humans , Kaplan-Meier Estimate , Male , Maryland , Middle Aged , Multivariate Analysis , Neuroendocrine Tumors/mortality , Neuroendocrine Tumors/secondary , Neuroendocrine Tumors/therapy , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Predictive Value of Tests , Proportional Hazards Models , Prospective Studies , Risk Factors , Time Factors , Tumor Burden
3.
Clin Endocrinol (Oxf) ; 89(4): 437-443, 2018 10.
Article in English | MEDLINE | ID: mdl-29927501

ABSTRACT

OBJECTIVE: Cushing disease (CD) is a rare entity caused by ACTH-secreting pituitary tumours, leading to prolonged hypercortisolism. Most cases are sporadic but can rarely occur in the context of familial predisposition, due to germline mutations in genes such as MEN1, leading to multiple endocrine neoplasia type 1, MEN1. We have reported previously that CD can be the first and only presenting manifestation of MEN1. In this report, we describe a cohort of paediatric patients who presented with CD as the first manifestation of MEN1. MATERIALS AND METHODS: A retrospective analysis of paediatric patients admitted to the National Institutes of Health (NIH) Clinical Center for evaluation of hypercortisolism, between 1997 and 2017. MEN1 was diagnosed on a clinical, familial and/or genetic basis. RESULTS: Of a total of 238 children with CD, six patients were subsequently diagnosed with MEN1, three males and three females with a mean age at diagnosis of CD at 13.4 ± 2.9 years. Five of the six patients had familial MEN1 and one patient was a sporadic case. Additional manifestations of MEN1 included primary hyperparathyroidism in three patients and hyperprolactinemia in two patients. DISCUSSION: This report describes a paediatric patient population with MEN1 in whom CD was the initial manifestation, confirming a previous observation that paediatric patients with MEN1 may present first with an ACTH-producing adenoma. Therefore, germline MEN1 mutations should be sought in paediatric CD and tested for when there is a suggestive family history and/or other manifestations.


Subject(s)
Pituitary ACTH Hypersecretion/genetics , Proto-Oncogene Proteins/genetics , Adolescent , Child , Cushing Syndrome/genetics , Female , Humans , Hyperparathyroidism/genetics , Hyperprolactinemia/genetics , Male , Mutation/genetics , Retrospective Studies
4.
Horm Metab Res ; 49(11): 805-815, 2017 11.
Article in English | MEDLINE | ID: mdl-29136674

ABSTRACT

Six syndromes of familial hyperparathyroidism are compared: 1) Familial hypocalciuric hypercalcemia (FHH) expresses primary hyperparathyroidism (PHPT) beginning at birth with lifelong hypercalcemia. There is nonsuppressed PTH secretion from outwardly normal parathyroid glands. It reflects germline heterozygous mutation in CASR, GNA11, or AP2S1. 2) Neonatal severe primary hyperparathyroidism is severest of the six syndromes. It requires urgent total parathyroidectomy in infancy. It usually reflects biallelic inactivation of the CASR. 3) Multiple endocrine neoplasia type 1 (MEN1) is most frequently expressed as PHPT with asymmetric enlargement of 3-4 parathyroids. Benign or malignant tumors may occur among 30 other tissues. It is predisposed by germline inactivation of MEN1 or rarely by inactivation of a cyclin dependent kinase inhibitor, and then termed MEN4. 4) Multiple endocrine neoplasia type 2A from RET activating mutation rarely presents as familial hyperparathyroidism, because medullary thyroid cancer and pheochromocytoma are more prominent. 5) Hyperparathyroidism-jaw tumor syndrome (HPT-JT) has frequent PHPT and benign jaw tumors. Twenty percent develop parathyroid cancer. It is predisposed by inactivating mutation in CDC73. 6) Familial isolated hyperparathyroidism causes multiple parathyroid tumors. It can be an incomplete expression of FHH, MEN1, HPT-JT or even of relatives without a shared driver mutation. However, in 20% of families it reflects GCM2 activating mutation. Five of the PHPT syndromes reflect overgrowth of parathyroid tissue; in contrast, familial hypocalciuric hypercalcemia reflects dysregulation of PTH secretion with little or no parathyroid overgrowth. These differences underlie major differences in clinical expression.


Subject(s)
Hyperparathyroidism, Primary/pathology , Genetic Predisposition to Disease , Humans , Hyperparathyroidism, Primary/diagnosis , Hyperparathyroidism, Primary/genetics , Mutation/genetics , Parathyroid Glands/pathology , Parathyroid Hormone/metabolism
5.
Gastroenterology ; 149(1): 67-78, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25865046

ABSTRACT

BACKGROUND & AIMS: Small intestinal carcinoids are rare and difficult to diagnose and patients often present with advanced incurable disease. Although the disease occurs sporadically, there have been reports of family clusters. Hereditary small intestinal carcinoid has not been recognized and genetic factors have not been identified. We performed a genetic analysis of families with small intestinal carcinoids to establish a hereditary basis and find genes that might cause this cancer. METHODS: We performed a prospective study of 33 families with at least 2 cases of small intestinal carcinoids. Affected members were characterized clinically and asymptomatic relatives were screened and underwent exploratory laparotomy for suspected tumors. Disease-associated mutations were sought using linkage analysis, whole-exome sequencing, and copy number analyses of germline and tumor DNA collected from members of a single large family. We assessed expression of mutant protein, protein activity, and regulation of apoptosis and senescence in lymphoblasts derived from the cases. RESULTS: Familial and sporadic carcinoids are clinically indistinguishable except for the multiple synchronous primary tumors observed in most familial cases. Nearly 34% of asymptomatic relatives older than age 50 were found to have occult tumors; the tumors were cleared surgically from 87% of these individuals (20 of 23). Linkage analysis and whole-exome sequencing identified a germline 4-bp deletion in the gene inositol polyphosphate multikinase (IPMK), which truncates the protein. This mutation was detected in all 11 individuals with small intestinal carcinoids and in 17 of 35 family members whose carcinoid status was unknown. Mutant IPMK had reduced kinase activity and nuclear localization, compared with the full-length protein. This reduced activation of p53 and increased cell survival. CONCLUSIONS: We found that small intestinal carcinoids can occur as an inherited autosomal-dominant disease. The familial form is characterized by multiple synchronous primary tumors, which might account for 22%-35% of cases previously considered sporadic. Relatives of patients with familial carcinoids should be screened to detect curable early stage disease. IPMK haploinsufficiency promotes carcinoid tumorigenesis.


Subject(s)
Carcinoid Tumor/genetics , Germ-Line Mutation , Intestinal Neoplasms/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoid Tumor/diagnosis , Carcinoid Tumor/pathology , Family , Female , Humans , Intestinal Neoplasms/diagnosis , Intestinal Neoplasms/pathology , Laparotomy , Male , Middle Aged , Pedigree , Prospective Studies , Young Adult
6.
Ann Surg Oncol ; 23(2): 416-23, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26542588

ABSTRACT

BACKGROUND: Recently, some surgeons have suggested that minimally invasive parathyroidectomy guided by preoperative localizing studies of patients with multiple endocrine neoplasia type 1 (MEN1)-associated primary hyperparathyroidism (pHPT) provides an acceptable outcome while minimizing the risk of hypoparathyroidism. This study aimed to evaluate the outcome for MEN1 patients who underwent limited parathyroidectomy compared with subtotal parathyroidectomy. METHODS: The authors performed a retrospective analysis of 99 patients with MEN1-associated pHPT who underwent at least one parathyroid operation at their institution. Preoperative imaging studies, intraoperative findings, and clinical outcomes for patients were compared. RESULTS: A total of 99 patients underwent 146 operations. Persistent pHPT was significantly higher in patients whose initial operations involved removal of 1 or 2 glands (69 %) or 2.5 to 3 glands (20 %) compared with those who had 3.5 or more glands removed (6 %) (P < 0.01). Persistent pHPT occurred in 5 % of all operations that cumulatively removed 3.5 or more parathyroid glands compared with 40 % of operations that removed 3 or fewer glands (P < 0.01). The single largest parathyroid gland was correctly identified preoperatively in 69 % (22/32) of the patients. However, preoperative localizing studies missed enlarged contralateral parathyroid glands in 86 % (19/22) of these patients. Preoperative localizing studies missed the largest contralateral parathyroid gland in 16 % (5/32) of the patients. CONCLUSIONS: Limited parathyroidectomy in MEN1 is associated with a high failure rate and should not be performed. Preoperative identification of a single enlarged parathyroid gland in MEN1 is not reliable enough to justify unilateral neck exploration because additional enlarged contralateral parathyroid glands are frequently missed.


Subject(s)
Hyperparathyroidism, Primary/etiology , Multiple Endocrine Neoplasia Type 1/surgery , Parathyroidectomy/adverse effects , Adolescent , Adult , Aged , Female , Follow-Up Studies , Humans , Hyperparathyroidism, Primary/pathology , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/complications , Multiple Endocrine Neoplasia Type 1/pathology , Neoplasm Staging , Retrospective Studies , Treatment Failure , Young Adult
7.
Nat Rev Cancer ; 5(5): 367-75, 2005 May.
Article in English | MEDLINE | ID: mdl-15864278

ABSTRACT

Six multiple endocrine neoplasia (MEN) syndromes have received a level of attention that might seem disproportionate to their low prevalence. The attention has been given because their hormonal excesses cause striking metabolic expressions and because they might clarify pathways disrupted in more common tumours. The recent discovery of the main gene in each MEN syndrome has furthered our understanding of not only hereditary but also sporadic tumours and has fostered new avenues of research.


Subject(s)
Multiple Endocrine Neoplasia Type 1/genetics , Multiple Endocrine Neoplasia Type 2a/genetics , Proto-Oncogene Proteins/genetics , Animals , Disease Models, Animal , Humans , Mice , Mutation , Oncogene Proteins/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics
8.
Clin Endocrinol (Oxf) ; 76(3): 379-86, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21916912

ABSTRACT

OBJECTIVE: In patients with multiple endocrine neoplasia type 1 (MEN1), Cushing's syndrome (CS) from endogenous hypercortisolism can result from pituitary, adrenal or other endocrine tumours. The purpose of this study was to characterize the range of presentations of CS in a large series of MEN1 patients. DESIGN: Retrospective review of NIH Clinical Center inpatient records over an approximately 40-year period. PATIENTS: Nineteen patients (eight males, 11 females) with CS and MEN1. MEASUREMENTS: Biochemical, imaging, surgical and pathological findings. RESULTS: An aetiology was determined for 14 of the 19 patients with CS and MEN1: 11 (79%) had Cushing's disease (CD) and three (21%) had ACTH-independent CS owing to adrenal tumours, frequencies indistinguishable from sporadic CS. Three of 11 MEN1 patients with CD (27%) had additional non-ACTH-secreting pituitary microadenomas identified at surgery, an incidence 10-fold higher than in sporadic CD. Ninety-one per cent of MEN1 patients with CD were cured after surgery. Two of three MEN1 patients with ACTH-independent CS (67%) had adrenocortical carcinoma. One patient with adrenal cancer and another with adrenal adenoma were cured by unilateral adrenalectomy. No case of ectopic ACTH secretion was identified in our patient cohort. The aetiology of CS could not be defined in five patients; in three of these, hypercortisolism appeared to resolve spontaneously. CONCLUSIONS: The tumour multiplicity of MEN1 can be reflected in the anterior pituitary, MEN1-associated ACTH-independent CS may be associated with aggressive adrenocortical disease and an aetiology for CS in MEN1 may be elusive in a substantial minority of patients.


Subject(s)
Cushing Syndrome/complications , Multiple Endocrine Neoplasia Type 1/complications , Pituitary ACTH Hypersecretion/complications , Adolescent , Adrenal Gland Neoplasms/blood , Adrenal Gland Neoplasms/complications , Adrenal Gland Neoplasms/surgery , Adrenocortical Adenoma/blood , Adrenocortical Adenoma/complications , Adrenocortical Adenoma/surgery , Adrenocorticotropic Hormone/blood , Adult , Child , Cushing Syndrome/blood , Cushing Syndrome/surgery , Female , Follow-Up Studies , Humans , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/blood , Multiple Endocrine Neoplasia Type 1/surgery , Pituitary ACTH Hypersecretion/blood , Pituitary ACTH Hypersecretion/surgery , Pituitary Neoplasms/blood , Pituitary Neoplasms/complications , Pituitary Neoplasms/surgery , Retrospective Studies , Time Factors , Young Adult
10.
J Clin Endocrinol Metab ; 105(4)2020 04 01.
Article in English | MEDLINE | ID: mdl-31778168

ABSTRACT

CONTEXT: Neonatal severe hyperparathyroidism (NSHPT) is rare and potentially lethal. It is usually from homozygous or heterozygous germline-inactivating CASR variant(s). NSHPT shows a puzzling range of serum calcium and parathyroid hormone (PTH) levels. Optimal therapy is unclear. EVIDENCE ACQUISITION: We categorized genotype/phenotype pairings related to CASRs. For the 2 pairings in NSHPT, each of 57 cases of neonatal severe hyperparathyroidism required calcium, PTH, upper normal PTH, and dosage of a germline pathogenic CASR variant. EVIDENCE SYNTHESIS: Homozygous and heterozygous NSHPT are 2 among a spectrum of 9 genotype/phenotype pairings relating to CASRs and NSHPT. For the 2 NSHPT pairings, expressions differ in CASR allelic dosage, CASR variant severity, and sufficiency of maternofetal calcium fluxes. Homozygous dosage of CASR variants was generally more aggressive than heterozygous. Among heterozygotes, high-grade CASR variants in vitro were more pathogenic in vivo than low-grade variants. Fetal calcium insufficiency as from maternal hypoparathyroidism caused fetal secondary hyperparathyroidism, which persisted and was reversible in neonates. Among NSHPT pairings, calcium and PTH were higher in CASR homozygotes than in heterozygotes. Extreme hypercalcemia (above 4.5 mM; normal 2.2-2.6 mM) is a robust biomarker, occurring only in homozygotes (83% of that pairing). It could occur during the first week. CONCLUSIONS: In NSHPT pairings, the homozygotes for pathogenic CASR variants show higher calcium and PTH levels than heterozygotes. Calcium levels above 4.5 mM among NSHPT are frequent and unique only to most homozygotes. This cutoff supports early and robust diagnosis of CASR dosage. Thereby, it promotes definitive total parathyroidectomy in most homozygotes.


Subject(s)
Biomarkers/analysis , Calcium/blood , Hyperparathyroidism, Primary/diagnosis , Infant, Newborn, Diseases/diagnosis , Mutation , Parathyroid Hormone/blood , Receptors, Calcium-Sensing/genetics , Female , Genotype , Heterozygote , Homozygote , Humans , Hyperparathyroidism, Primary/blood , Hyperparathyroidism, Primary/genetics , Infant, Newborn , Infant, Newborn, Diseases/blood , Infant, Newborn, Diseases/genetics , Male , Prognosis
11.
Endocrine ; 67(1): 204-208, 2020 01.
Article in English | MEDLINE | ID: mdl-31782130

ABSTRACT

PURPOSE: Parathyroid cancer is a rare tumor associated with poor prognosis particularly when disseminated. While chemotherapy and/or radiotherapy are of no clinical value in disseminated disease, immunotherapy should be considered. SUBJECT AND RESULTS: A patient with CDC73-associated metastatic parathyroid carcinoma was treated with combined anti-hPTH immunotherapy and surgery. CONCLUSIONS: Following five courses of anti-hPTH immunotherapy and subsequent surgery, a 12-year long remission of disseminated parathyroid cancer is reported. This case further supports the ever-expanding spectrum of cancers that may benefit from immunotherapy.


Subject(s)
Parathyroid Neoplasms , Humans , Immunotherapy , Parathyroid Glands , Parathyroid Neoplasms/therapy
12.
Endocr Rev ; 26(5): 615-61, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15632315

ABSTRACT

Hereditary origin of a tumor helps toward early discovery of its mutated gene; for example, it supports the compilation of a DNA panel from index cases to identify that gene by finding mutations in it. The gene for a hereditary tumor may contribute also to common tumors. For some syndromes, such as hereditary paraganglioma, several genes can cause a similar syndrome. For other syndromes, such as multiple endocrine neoplasia 2, one gene supports variants of a syndrome. Onset usually begins earlier and in more locations with hereditary than sporadic tumors. Mono- or oligoclonal ("clonal") tumor usually implies a postnatal delay, albeit less delay than for sporadic tumor, to onset and potential for cancer. Hormone excess from a polyclonal tissue shows onset at birth and no benefit from subtotal ablation of the secreting organ. Genes can cause neoplasms through stepwise loss of function, gain of function, or combinations of these. Polyclonal hormonal excess reflects abnormal gene dosage or effect, such as activation or haploinsufficiency. Polyclonal hyperplasia can cause the main endpoint of clinical expression in some syndromes or can be a precursor to clonal progression in others. Gene discovery is usually the first step toward clarifying the molecule and pathway mutated in a syndrome. Most mutated pathways in hormone excess states are only partly understood. The bases for tissue specificity of hormone excess syndromes are usually uncertain. In a few syndromes, tissue selectivity arises from mutation in the open reading frame of a regulatory gene (CASR, TSHR) with selective expression driven by its promoter. Polyclonal excess of a hormone is usually from a defect in the sensor system for an extracellular ligand (e.g., calcium, glucose, TSH). The final connections of any of these polyclonal or clonal pathways to hormone secretion have not been identified. In many cases, monoclonal proliferation causes hormone excess, probably as a secondary consequence of accumulation of cells with coincidental hormone-secretory ability.


Subject(s)
Endocrine Gland Neoplasms/genetics , Neoplastic Syndromes, Hereditary/genetics , Endocrine Gland Neoplasms/metabolism , Endocrine Gland Neoplasms/pathology , Humans , Hyperparathyroidism/genetics , Hyperparathyroidism/metabolism , Hyperparathyroidism/pathology , Neoplastic Syndromes, Hereditary/metabolism , Neoplastic Syndromes, Hereditary/pathology , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/metabolism , von Hippel-Lindau Disease/pathology
13.
Horm Res ; 71 Suppl 2: 131-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19407509

ABSTRACT

Sporadic multiple endocrine neoplasia type 1 (MEN1) is defined as the occurrence of tumours in two of three main endocrine tissue types: parathyroid, pituitary and pancreaticoduodenal. A prolactinoma variant or Burin variant of MEN1 was found to occur in three large kindreds, with more prolactinomas and fewer gastrinomas than typical MEN1. MEN1 tumours differ from common tumours by showing features from the MEN1 gene (e.g. larger pituitary tumours). They also show various expressions of tumour multiplicity; however, pituitary tumour in MEN1 is usually solitary. Diagnosis in MEN1 carriers during childhood is not directed at cancers but at benign morbid tumours. Morbid prolactinoma occurred at the age of 5 years in one MEN1 individual; hence, this is the earliest age at which to recommend tumour surveillance in carriers. The MEN1 gene shows biallelic inactivation in 30% of some types of common variety endocrine tumours (e.g. parathyroid adenoma, gastrinoma, insulinoma and bronchial carcinoid), but in only 1-5% of common pituitary tumours. Heterozygous knockout of MEN1 in mice provides a robust model of MEN1 and has been found to support further research on anti-angiogenesis therapy for pituitary tumours. The rarity of MEN1 mutations in some MEN1-like states aids the identification of other mutated genes, such as AIP, HRPT2 and p27(Kip1). We present recent clinical and basic findings about the MEN1 gene, particularly concerning hereditary vs. common variety pituitary tumours.


Subject(s)
Multiple Endocrine Neoplasia Type 1/metabolism , Prolactinoma/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Gene Knockout Techniques , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Multiple Endocrine Neoplasia Type 1/genetics , Mutation , Prolactinoma/genetics , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
14.
PLoS Genet ; 2(4): e51, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16604156

ABSTRACT

Multiple endocrine neoplasia type I (MEN1) is a familial cancer syndrome characterized primarily by tumors of multiple endocrine glands. The gene for MEN1 encodes a ubiquitously expressed tumor suppressor protein called menin. Menin was recently shown to interact with several components of a trithorax family histone methyltransferase complex including ASH2, Rbbp5, WDR5, and the leukemia proto-oncoprotein MLL. To elucidate menin's role as a tumor suppressor and gain insights into the endocrine-specific tumor phenotype in MEN1, we mapped the genomic binding sites of menin, MLL1, and Rbbp5, to approximately 20,000 promoters in HeLa S3, HepG2, and pancreatic islet cells using the strategy of chromatin-immunoprecipitation coupled with microarray analysis. We found that menin, MLL1, and Rbbp5 localize to the promoters of thousands of human genes but do not always bind together. These data suggest that menin functions as a general regulator of transcription. We also found that factor occupancy generally correlates with high gene expression but that the loss of menin does not result in significant changes in most transcript levels. One exception is the developmentally programmed transcription factor, HLXB9, which is overexpressed in islets in the absence of menin. Our findings expand the realm of menin-targeted genes several hundred-fold beyond that previously described and provide potential insights to the endocrine tumor bias observed in MEN1 patients.


Subject(s)
Genome, Human , Proto-Oncogene Proteins/physiology , Cell Line, Tumor , DNA-Binding Proteins , HeLa Cells , Histone-Lysine N-Methyltransferase , Homeodomain Proteins/biosynthesis , Humans , Models, Genetic , Myeloid-Lymphoid Leukemia Protein/physiology , Nuclear Proteins/physiology , Oligonucleotide Array Sequence Analysis , Phenotype , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins/metabolism , Transcription Factors/biosynthesis
15.
J Clin Endocrinol Metab ; 104(9): 4058-4066, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-30848815

ABSTRACT

CONTEXT: Familial isolated hyperparathyroidism (FIHP) is defined as familial primary hyperparathyroidism (FH) without a characteristic extraparathyroidal feature of a more complex hyperparathyroid syndrome. New concepts of FIHP have been developed within this definition. FIHP has been difficult to study due to small kindreds and mildly symptomatic cases. EVIDENCE ACQUISITION: Searches were through PubMed for FIHP, other FH syndromes, and the gene(s) mutated in each. EVIDENCE SYNTHESIS: Within its definition, the current concept of FIHP has clinical and mutational components that can include incomplete expressions of multiple endocrine neoplasia type 1 (MEN1) familial hypocalciuric hypercalcemia, hyperparathyroidism-jaw tumor syndromes, or their mutations. Newest concepts of FIHP focus on kindreds without mutation of the MEN1, CASR, or CDC73 genes; 17% have germline activating mutation of the gene for the GCM2 transcription factor. The FIHP kindreds with or without GCM2 mutation contain a median of only two cases of primary hyperparathyroidism. The small kindred size in both subgroups of FIHP is probably caused by a low rate of screening among relatives. Persons with FIHP and GCM2 mutation present as adults with mild hypercalcemia and multiple parathyroid tumors. CONCLUSION: The current concept of FIHP led to a focus on small kindreds without mutation of MEN1, CASR, or CDC73. These assisted in identifying germline activating GCM2 mutations in 17% of kindreds. Clinical and mutational characterization in more cases is needed to determine if there are any unique clinical features of FIHP, with or without mutation of GCM2.

16.
J Bone Miner Res ; 34(1): 22-37, 2019 01.
Article in English | MEDLINE | ID: mdl-30536424

ABSTRACT

We review advancing and overlapping stages for our understanding of the expressions of six hyperparathyroid (HPT) syndromes: multiple endocrine neoplasia type 1 (MEN1) or type 4, multiple endocrine neoplasia type 2A (MEN2A), hyperparathyroidism-jaw tumor syndrome, familial hypocalciuric hypercalcemia, neonatal severe primary hyperparathyroidism, and familial isolated hyperparathyroidism. During stage 1 (1903 to 1967), the introduction of robust measurement of serum calcium was a milestone that uncovered hypercalcemia as the first sign of dysfunction in many HPT subjects, and inheritability was reported in each syndrome. The earliest reports of HPT syndromes were biased toward severe or striking manifestations. During stage 2 (1959 to 1985), the early formulations of a syndrome were improved. Radioimmunoassays (parathyroid hormone [PTH], gastrin, insulin, prolactin, calcitonin) were breakthroughs. They could identify a syndrome carrier, indicate an emerging tumor, characterize a tumor, or monitor a tumor. During stage 3 (1981 to 2006), the assembly of many cases enabled recognition of further details. For example, hormone non-secreting skin lesions were discovered in MEN1 and MEN2A. During stage 4 (1985 to the present), new genomic tools were a revolution for gene identification. Four principal genes ("principal" implies mutated or deleted in 50% or more probands for its syndrome) (MEN1, RET, CASR, CDC73) were identified for five syndromes. During stage 5 (1993 to the present), seven syndromal genes other than a principal gene were identified (CDKN1B, CDKN2B, CDKN2C, CDKN1A, GNA11, AP2S1, GCM2). Identification of AP2S1 and GCM2 became possible because of whole-exome sequencing. During stages 4 and 5, the newly identified genes enabled many studies, including robust assignment of the carriers and non-carriers of a mutation. Furthermore, molecular pathways of RET and the calcium-sensing receptor were elaborated, thereby facilitating developments in pharmacotherapy. Current findings hold the promise that more genes for HPT syndromes will be identified and studied in the near future. © 2018 American Society for Bone and Mineral Research.


Subject(s)
Hyperparathyroidism , Multiple Endocrine Neoplasia Type 1 , Multiple Endocrine Neoplasia Type 2a , Neoplasm Proteins , Parathyroid Neoplasms , History, 20th Century , History, 21st Century , Humans , Hyperparathyroidism/classification , Hyperparathyroidism/genetics , Hyperparathyroidism/history , Hyperparathyroidism/metabolism , Multiple Endocrine Neoplasia Type 1/classification , Multiple Endocrine Neoplasia Type 1/genetics , Multiple Endocrine Neoplasia Type 1/history , Multiple Endocrine Neoplasia Type 1/metabolism , Multiple Endocrine Neoplasia Type 2a/classification , Multiple Endocrine Neoplasia Type 2a/genetics , Multiple Endocrine Neoplasia Type 2a/history , Multiple Endocrine Neoplasia Type 2a/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Parathyroid Neoplasms/classification , Parathyroid Neoplasms/genetics , Parathyroid Neoplasms/history , Parathyroid Neoplasms/metabolism , Syndrome
17.
Endocr Relat Cancer ; 15(1): 267-75, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18310293

ABSTRACT

Multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant syndrome caused by mutations in the MEN1 tumor suppressor gene. Loss of the functional second copy of the MEN1 gene causes individuals to develop multiple endocrine tumors, primarily affecting the parathyroid, pituitary, and pancreas. While it is clear that the protein encoded by MEN1, menin, suppresses endocrine tumors, its biochemical functions and direct downstream targets remain unclear. Recent studies have suggested that menin may act as a scaffold protein to coordinate gene transcription, and that menin is an oncogenic cofactor for homeobox (HOX) gene expression in hematopoietic cancer. The role of HOX genes in adult cell differentiation is still obscure, but growing evidence suggests that they may play important roles in the development of cancer. Therefore, we hypothesized that specific HOX genes were regulated by menin in parathyroid tumor development. Utilizing quantitative TaqMan RT-PCR, we compared expression profiles of the 39 HOX genes in human familial MEN1 (fMEN1) parathyroid tumors and sporadic parathyroid adenomas with normal samples. We identified a large set of 23 HOX genes whose deregulation is specific for fMEN1 parathyroid tumors, and only 5 HOX genes whose misexpression are specific for sporadic parathyroid tumor development. These findings provide the first evidence that loss of the MEN1 tumor suppressor gene is associated with deregulation of specific HOX gene expression in the development of familial human parathyroid tumors. Our results strongly reinforce the idea that abnormal expression of developmental HOX genes can be critical in human cancer progression.


Subject(s)
Genes, Homeobox/genetics , Multiple Endocrine Neoplasia Type 1/genetics , Mutation/genetics , Parathyroid Neoplasms/metabolism , Parathyroid Neoplasms/pathology , Proto-Oncogene Proteins/genetics , Adolescent , Adult , Aged , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/metabolism , Multiple Endocrine Neoplasia Type 1/pathology , Parathyroid Neoplasms/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
18.
Mol Cancer ; 7: 65, 2008 Aug 07.
Article in English | MEDLINE | ID: mdl-18687124

ABSTRACT

BACKGROUND: Germline and somatic inactivating mutations in the HRPT2 gene occur in the inherited hyperparathyroidism-jaw tumor syndrome, in some cases of parathyroid cancer and in some cases of familial hyperparathyroidism. HRPT2 encodes parafibromin. To identify parafibromin interacting proteins we used the yeast two-hybrid system for screening a heart cDNA library with parafibromin as the bait. RESULTS: Fourteen parafibromin interaction positive preys representing 10 independent clones encoding actinin-2 were isolated. Parafibromin interacted with muscle alpha-actinins (actinin-2 and actinin-3), but not with non-muscle alpha-actinins (actinin-1 and actinin-4). The parafibromin-actinin interaction was verified by yeast two-hybrid, GST pull-down, and co-immunoprecipitation. Yeast two-hybrid analysis revealed that the N-terminal region of parafibromin interacted with actinins. In actin sedimentation assays parafibromin did not dissociate skeletal muscle actinins from actin filaments, but interestingly, parafibromin could also bundle/cross-link actin filaments. Parafibromin was predominantly nuclear in undifferentiated proliferating myoblasts (C2C12 cells), but in differentiated C2C12 myotubes parafibromin co-localized with actinins in the cytoplasmic compartment. CONCLUSION: These data support a possible contribution of parafibromin outside the nucleus through its interaction with actinins and actin bundling/cross-linking. These data also suggest that actinins (and actin) participate in sequestering parafibromin in the cytoplasmic compartment.


Subject(s)
Actinin/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Nucleus/metabolism , Cells, Cultured , Cytoplasm/metabolism , Fluorescent Antibody Technique , Gene Library , HeLa Cells , Humans , Mice , Parathyroid Neoplasms/metabolism , Transfection , Two-Hybrid System Techniques
19.
Mol Cancer Res ; 5(10): 1041-51, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17951404

ABSTRACT

Multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant familial cancer syndrome characterized primarily by endocrine tumors of the parathyroids, anterior pituitary, and enteropancreatic endocrine tissues. Affected individuals carry a germ-line loss-of-function mutation of the MEN1 gene, and tumors arise after loss of the second allele. Homozygous loss of Men1 in the germ line of mice results in early embryonic lethality, with defective development of neural tube, heart, liver, and craniofacial structures. We generated immortalized wild-type (WT) and menin-null mouse embryo fibroblast (MEF) cell lines and evaluated their characteristics, including global expression patterns. The WT and menin-null cell lines were aneuploid, and the nulls did not display tumorigenic characteristics in soft agar assay. Expression arrays in menin-null MEFs revealed altered expression of several extracellular matrix proteins that are critical in organogenesis. Specifically, transcripts for fibulin 2 (Fbln2), periostin (Postn), and versican [chondroitin sulfate proteoglycan (Cspg2)], genes critical for the developing heart and known to be induced by transforming growth factor-beta (TGF-beta), were decreased in their expression in menin-null MEFs. Fbln2 expression was the most affected, and the reduction in menin-null MEFs for Fbln2, Postn, and Cspg2 was 16.18-, 5.37-, and 2.15-fold, respectively. Menin-null MEFs also showed poor response to TGF-beta-induced Smad3-mediated transcription in a reporter assay, supporting a role for menin in this pathway. Postn and Cspg2 expression in WT, unlike in null MEFs, increased on TGF-beta treatment. The expression changes associated with the loss of the tumor suppressor menin provide insights into the defective organogenesis observed during early embryonic development in Men1-null mouse embryos.


Subject(s)
Embryo, Mammalian/metabolism , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Developmental , Proto-Oncogene Proteins/physiology , Tumor Suppressor Proteins/physiology , Animals , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Mice , Organogenesis/genetics , Proto-Oncogene Proteins/genetics , Transforming Growth Factor beta/pharmacology , Tumor Suppressor Proteins/genetics
20.
J Bone Miner Res ; 33(1): 27-31, 2018 01.
Article in English | MEDLINE | ID: mdl-29115694

ABSTRACT

Familial hypocalciuric hypercalcemia (FHH) causes lifelong hypercalcemia with features that overlap with typical primary hyperparathyroidism (PHPT). The incompleteness of this overlap has led to divergent nomenclatures for FHH. I compare two nomenclatures. One sets FHH as an entity distinct from PHPT. The other groups FHH with PHPT but conditions FHH as atypical PHPT. I analyzed selected articles about calcium-sensing receptors, FHH, PHPT, CASR, GNA11, and AP2S1. FHH usually results from a heterozygous germline inactivating mutation of the CASR, and less frequently from mutation of GNA11 or AP2S1. The CASR encodes the calcium-sensing receptors. These are highly expressed on parathyroid cells, where they sense serum calcium concentration and regulate suppression of PTH secretion by serum calcium. Their mutated expression in the kidney in FHH causes increased renal tubular reabsorption of calcium (hypocalciuria). Many FHH features are shared with PHPT and thus support FHH as a form of PHPT. These include a driver mutation expressed mainly in the parathyroid cells. The mutation causes a parathyroid cell insensitivity to extracellular calcium in vivo and in vitro, a right-shift of the set point for suppression of PTH secretion by calcium. Serum PTH is normal or mildly elevated; ie, it is not appropriately suppressed by hypercalcemia. Total parathyroidectomy causes hypoparathyroidism and durable remission of hypercalcemia. Some other features are not shared with PHPT and could support FHH as a distinct entity. These include onset of hypercalcemia in the first week of life, frequent persistence of hypercalcemia after subtotal parathyroidectomy, and hypocalciuria. The features supporting FHH as a form of PHPT are stronger than those favoring FHH as a distinct entity. Classifying FHH as an atypical form of PHPT represents compact nomenclature and supports current concepts of pathophysiology of FHH and PHPT. Published 2017. This article is a U.S. Government work and is in the public domain in the USA.


Subject(s)
Hypercalcemia/congenital , Hyperparathyroidism, Primary/pathology , Bone and Bones/pathology , Humans , Hypercalcemia/blood , Hypercalcemia/pathology , Hypercalcemia/physiopathology , Hyperparathyroidism, Primary/blood , Hyperparathyroidism, Primary/physiopathology , Parathyroid Hormone/blood , Terminology as Topic
SELECTION OF CITATIONS
SEARCH DETAIL