Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 888
Filter
Add more filters

Publication year range
1.
Adv Anat Pathol ; 30(1): 69-78, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36136401

ABSTRACT

Familial endocrine tumor syndromes are continuously expanding owing to the growing role of genetic testing in routine clinical practice. Pathologists are usually the first on the clinical team to encounter these syndromes at their initial presentation; thus, recognizing them is becoming more pivotal in routine pathology practice to help in properly planning management and further family testing. Our increasing knowledge about them is reflected in the newer syndromes included in the new World Health Organization classification and in the evolving discovery of new endocrine tumors and new familial associations. In many of these syndromes, the clinical features and co-occurrence of multiple neoplasia are the only clues (multiple endocrine neoplasia syndromes). In other syndromes, specific morphologic findings (pituitary blastoma and DICER1 syndrome, cribriform morular thyroid carcinoma, and AFP syndrome) and available ancillary studies (SDHB in SDH-deficient tumor syndromes) can aid pathologists. The aim of this review is to provide a primer on recent updates on familial endocrine tumor syndromes and related tumors, focusing on recent classification changes or tumor syndromes where a clearer role for pathologists is at play.


Subject(s)
Multiple Endocrine Neoplasia , Neoplastic Syndromes, Hereditary , Humans , Neoplastic Syndromes, Hereditary/diagnosis , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology , Multiple Endocrine Neoplasia/genetics , Ribonuclease III , DEAD-box RNA Helicases
2.
Am J Med Genet A ; 182(5): 1008-1020, 2020 05.
Article in English | MEDLINE | ID: mdl-32077592

ABSTRACT

Williams-Beuren syndrome (WBS) is a multisystem disorder caused by a hemizygous deletion on 7q11.23 encompassing 26-28 genes. An estimated 2-5% of patients have "atypical" deletions, which extend in the centromeric and/or telomeric direction from the WBS critical region. To elucidate clinical differentiators among these deletion types, we evaluated 10 individuals with atypical deletions in our cohort and 17 individuals with similarly classified deletions previously described in the literature. Larger deletions in either direction often led to more severe developmental delays, while deletions containing MAGI2 were associated with infantile spasms and seizures in patients. In addition, head size was notably smaller in those with centromeric deletions including AUTS2. Because children with atypical deletions were noted to be less socially engaged, we additionally sought to determine how atypical deletions relate to social phenotypes. Using the Social Responsiveness Scale-2, raters scored individuals with atypical deletions as having different social characteristics to those with typical WBS deletions (p = .001), with higher (more impaired) scores for social motivation (p = .005) in the atypical deletion group. In recognizing these distinctions, physicians can better identify patients, including those who may already carry a clinical or FISH WBS diagnosis, who may benefit from additional molecular evaluation, screening, and therapy. In addition to the clinical findings, we note mild endocrine findings distinct from those typically seen in WBS in several patients with telomeric deletions that included POR. Further study in additional telomeric deletion cases will be needed to confirm this observation.


Subject(s)
Chromosomes, Human, Pair 7/genetics , Multiple Endocrine Neoplasia/genetics , Neurodevelopmental Disorders/genetics , Williams Syndrome/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Chromosome Deletion , Female , Head/abnormalities , Head/physiopathology , Humans , Infant , Male , Middle Aged , Multiple Endocrine Neoplasia/epidemiology , Multiple Endocrine Neoplasia/physiopathology , Neurodevelopmental Disorders/epidemiology , Neurodevelopmental Disorders/physiopathology , Organ Size/genetics , Phenotype , Williams Syndrome/epidemiology , Williams Syndrome/physiopathology , Young Adult
3.
Ann Pathol ; 40(2): 120-133, 2020 Apr.
Article in French | MEDLINE | ID: mdl-32035641

ABSTRACT

About 5% of gastroenteropancreatic and thoracic neuroendocrine neoplasms (NENs) arise in the context of an inherited tumour syndrome. The two most frequent syndromes are: multiple endocrine neoplasia type 1 (MEN1), associated with a large spectrum of endocrine and non endocrine tumours, including duodenopancreatic, thymic and bronchial NENs, and the von Hippel-Lindau syndrome VHL, associated with pancreatic NENs. Two inherited syndromes have a low incidence of NENs: neurofibromatosis type 1 (NF1), associated with duodenal somatostatinomas, and tuberous sclerosis (TSC), associated with pancreatic NENs. Two rare syndromes have a high incidence of NENs: multiple endocrine neoplasia type 4 (MEN4), with a tumour spectrum similar to that of MEN1, and glucagon cell hyperplasia neoplasia (GCHN), involving only the pancreas. It is likely that other syndromes remain to be characterized, especially in familial small-intestinal NENs. The diagnosis is usually raised because of the suggestive clinical setting: young age at diagnosis, multiple tumours in multiple organs, familial history. Except in VHL and NF1, tumours themselves do not show specific pathological features; they usually are well differentiated and of low histological grade; their prognosis is good, except for MEN1-associated thymic NENs. The most suggestive pathological feature is their combination with various endocrine and/or non endocrine lesions in the adjacent tissue. Pathological examination is important, for a correct diagnosis and for an accurate management of the patients and their families, who must be referred to expert centers.


Subject(s)
Neoplastic Syndromes, Hereditary , Neuroendocrine Tumors/pathology , Duodenal Neoplasms/diagnosis , Genetic Predisposition to Disease , Humans , Intestinal Neoplasms/complications , Intestinal Neoplasms/diagnosis , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Intestines/pathology , Multiple Endocrine Neoplasia/complications , Multiple Endocrine Neoplasia/diagnosis , Multiple Endocrine Neoplasia/genetics , Multiple Endocrine Neoplasia/pathology , Multiple Endocrine Neoplasia Type 1/complications , Multiple Endocrine Neoplasia Type 1/diagnosis , Multiple Endocrine Neoplasia Type 1/genetics , Multiple Endocrine Neoplasia Type 1/pathology , Neoplastic Syndromes, Hereditary/complications , Neoplastic Syndromes, Hereditary/diagnosis , Neoplastic Syndromes, Hereditary/pathology , Neuroendocrine Tumors/complications , Neuroendocrine Tumors/diagnosis , Neuroendocrine Tumors/genetics , Pancreas/pathology , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Stomach Neoplasms/complications , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Thoracic Neoplasms/diagnosis , Thorax/pathology , Tuberous Sclerosis/etiology , Tuberous Sclerosis/pathology , von Hippel-Lindau Disease/complications , von Hippel-Lindau Disease/diagnosis , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/pathology
4.
Intern Med J ; 49(8): 954-961, 2019 08.
Article in English | MEDLINE | ID: mdl-31387156

ABSTRACT

The multiple endocrine neoplasia (MEN) syndromes include MEN1, MEN2 (formerly MEN2A), MEN3 (formerly MEN2B) and the recently identified MEN4. Clinical presentations are varied and often relate to the overproduction of specific hormones. Understanding the genetics of each syndrome assists in determining screening timelines. Treatments for each manifestation are dependent on location, risk of recurrence or malignancy, hormone excess and surgical morbidity. Multidisciplinary management should include geneticists, genetic counsellors, endocrinologists and endocrine surgeons.


Subject(s)
Multiple Endocrine Neoplasia/diagnosis , Multiple Endocrine Neoplasia/genetics , Multiple Endocrine Neoplasia/therapy , Combined Modality Therapy , Endocrine Surgical Procedures , Genetic Markers , Genetic Testing , Humans , Multiple Endocrine Neoplasia/classification , Recurrence
5.
Br J Cancer ; 116(2): 163-168, 2017 Jan 17.
Article in English | MEDLINE | ID: mdl-27959889

ABSTRACT

BACKGROUND: Neuroendocrine tumours (NETs) are rare in children and limited data are available. We aimed to specify tumour and patient characteristics and to investigate the role of genetic predisposition in the aetiology of paediatric NETs. METHODS: Using the Dutch Pathology Registry PALGA, we collected patient- and tumour data of paediatric NETs in the Netherlands between 1991 and 2013 (N=483). RESULTS: The incidence of paediatric NETs in the Netherlands is 5.40 per one million per year. The majority of NETs were appendiceal tumours (N=441;91.3%). Additional surgery in appendiceal NETs was indicated in 89 patients, but performed in only 27 of these patients. Four out of five patients with pancreatic NETs were diagnosed with Von Hippel-Lindau disease (N=2) and Multiple Endocrine Neoplasia type 1 (N=2). In one patient with an appendiceal NET Familial Adenomatous Polyposis was diagnosed. On the basis of second primary tumours or other additional diagnoses, involvement of genetic predisposition was suggestive in several others. CONCLUSIONS: We identified a significant number of patients with a confirmed or suspected tumour predisposition syndrome and show that paediatric pancreatic NETs in particular are associated with genetic syndromes. In addition, we conclude that treatment guidelines for appendiceal paediatric NETs need revision and improved implementation.


Subject(s)
Genetic Predisposition to Disease , Neuroendocrine Tumors/epidemiology , Neuroendocrine Tumors/genetics , Adolescent , Appendiceal Neoplasms/epidemiology , Appendiceal Neoplasms/genetics , Child , Child, Preschool , Female , Humans , Incidence , Infant , Male , Multiple Endocrine Neoplasia/epidemiology , Multiple Endocrine Neoplasia/genetics , Netherlands/epidemiology , Pancreatic Neoplasms/epidemiology , Pancreatic Neoplasms/genetics , Registries , von Hippel-Lindau Disease/genetics
6.
Endocr Pract ; 23(9): 1039-1044, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28613942

ABSTRACT

OBJECTIVE: Inherited endocrinopathies are rare tumor predisposition syndromes associated with significant morbidity and mortality and have implications for both patients and their families. Prior studies suggest that early diagnosis of inherited endocrinopathies may reduce morbidity and mortality. Although genetic counseling and testing can help inform the appropriate management of at-risk relatives, barriers to care still exist. We explored patient perceptions to identify barriers and promote the uptake of genetic counseling. METHODS: An anonymous survey of patients from a multidisciplinary inherited endocrinopathy clinic at a tertiary care, university-based medical center was conducted. Data collected and analyzed included demographics, socioeconomic status, perceived risks, benefits, and both motivating and dissuading factors to genetic counseling and testing. RESULTS: Our study suggests barriers to genetic testing include concerns regarding cost and the potential for discrimination with respect to employers and insurers. CONCLUSION: This highlights the importance of genetic counseling to discuss benefits of genetic testing, while dispelling misperceptions. Knowledge of the common barriers to genetic counseling and testing can guide initiatives and education to foster genetic testing of at-risk relatives in the inherited endocrinopathy population. ABBREVIATIONS: FMTC = familial medullary thyroid carcinoma GINA = Genetic Information Nondiscrimination Act MEN1 = multiple endocrine neoplasia 1 MEN2A = multiple endocrine neoplasia 2A MEN2B = multiple endocrine neoplasia 2B MTC = medullary thyroid cancer PGL-PCC = paraganglioma-pheochromocytoma.


Subject(s)
Attitude , Endocrine System Diseases/genetics , Genetic Counseling/psychology , Genetic Testing , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Medullary/congenital , Carcinoma, Medullary/genetics , Child , Female , Humans , Male , Middle Aged , Multiple Endocrine Neoplasia/genetics , Multiple Endocrine Neoplasia Type 2a/genetics , Thyroid Neoplasms/genetics , Young Adult
7.
Rev Endocr Metab Disord ; 17(3): 381-388, 2016 09.
Article in English | MEDLINE | ID: mdl-27943006

ABSTRACT

We often think of the lentiginoses, phacomatoses and other neurocutaneous syndromes as conditions that affect the skin and also predispose to a variety of tumors. However, we rarely think of Peutz-Jeghers syndrome (PJS), Carney complex (CNC), Cowden disease (CD), neurofibromatosis type-1 (NF-1) or tuberous sclerosis (TSC) as conditions that are multiple endocrine neoplasias (MEN). Indeed, all of these conditions predispose to a variety of endocrine tumors, in addition to many other neoplasms. On the other hand, the classic MENs, type 1 and 2 (MEN-1 and MEN-2, respectively) are almost never thought in terms of their skin manifestations. In this review, we present extensively the MEN-1, MEN-2 and PJS syndromes, and briefly refer to CD, NF-1, and TSC. CNC is discussed in another article in this journal issue.


Subject(s)
Multiple Endocrine Neoplasia/complications , Peutz-Jeghers Syndrome/complications , Skin Diseases/etiology , Humans , Multiple Endocrine Neoplasia/genetics , Peutz-Jeghers Syndrome/genetics
8.
Neuroendocrinology ; 103(1): 18-31, 2016.
Article in English | MEDLINE | ID: mdl-25592387

ABSTRACT

Pituitary adenomas are a common feature of a subset of endocrine neoplasia syndromes, which have otherwise highly variable disease manifestations. We provide here a review of the clinical features and human molecular genetics of multiple endocrine neoplasia (MEN) type 1 and 4 (MEN1 and MEN4, respectively) and Carney complex (CNC). MEN1, MEN4, and CNC are hereditary autosomal dominant syndromes that can present with pituitary adenomas. MEN1 is caused by inactivating mutations in the MEN1 gene, whose product menin is involved in multiple intracellular pathways contributing to transcriptional control and cell proliferation. MEN1 clinical features include primary hyperparathyroidism, pancreatic neuroendocrine tumours and prolactinomas as well as other pituitary adenomas. A subset of patients with pituitary adenomas and other MEN1 features have mutations in the CDKN1B gene; their disease has been called MEN4. Inactivating mutations in the type 1α regulatory subunit of protein kinase A (PKA; the PRKAR1A gene), that lead to dysregulation and activation of the PKA pathway, are the main genetic cause of CNC, which is clinically characterised by primary pigmented nodular adrenocortical disease, spotty skin pigmentation (lentigines), cardiac and other myxomas and acromegaly due to somatotropinomas or somatotrope hyperplasia.


Subject(s)
Carney Complex/genetics , Carney Complex/pathology , Endocrine Gland Neoplasms/genetics , Multiple Endocrine Neoplasia/genetics , Multiple Endocrine Neoplasia/pathology , Pituitary Neoplasms/genetics , Acromegaly/genetics , Animals , Endocrine Gland Neoplasms/pathology , Humans , Multiple Endocrine Neoplasia/diagnosis , Pituitary Neoplasms/pathology , Prolactinoma/genetics , Prolactinoma/pathology
9.
PLoS Genet ; 9(3): e1003350, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23555276

ABSTRACT

The CDKN1B gene encodes the cyclin-dependent kinase inhibitor p27(KIP1), an atypical tumor suppressor playing a key role in cell cycle regulation, cell proliferation, and differentiation. Impaired p27(KIP1) expression and/or localization are often observed in tumor cells, further confirming its central role in regulating the cell cycle. Recently, germline mutations in CDKN1B have been associated with the inherited multiple endocrine neoplasia syndrome type 4, an autosomal dominant syndrome characterized by varying combinations of tumors affecting at least two endocrine organs. In this study we identified a 4-bp deletion in a highly conserved regulatory upstream ORF (uORF) in the 5'UTR of the CDKN1B gene in a patient with a pituitary adenoma and a well-differentiated pancreatic neoplasm. This deletion causes the shift of the uORF termination codon with the consequent lengthening of the uORF-encoded peptide and the drastic shortening of the intercistronic space. Our data on the immunohistochemical analysis of the patient's pancreatic lesion, functional studies based on dual-luciferase assays, site-directed mutagenesis, and on polysome profiling show a negative influence of this deletion on the translation reinitiation at the CDKN1B starting site, with a consequent reduction in p27(KIP1) expression. Our findings demonstrate that, in addition to the previously described mechanisms leading to reduced p27(KIP1) activity, such as degradation via the ubiquitin/proteasome pathway or non-covalent sequestration, p27(KIP1) activity can also be modulated by an uORF and mutations affecting uORF could change p27(KIP1) expression. This study adds the CDKN1B gene to the short list of genes for which mutations that either create, delete, or severely modify their regulatory uORFs have been associated with human diseases.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27 , Genetic Predisposition to Disease , Multiple Endocrine Neoplasia/genetics , Protein Biosynthesis , 5' Untranslated Regions , Cell Cycle , Cell Differentiation , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , HeLa Cells , Humans , Multiple Endocrine Neoplasia/metabolism , Multiple Endocrine Neoplasia/pathology , Mutagenesis, Site-Directed , Mutation , Open Reading Frames/genetics
10.
Curr Opin Oncol ; 26(1): 51-61, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24300902

ABSTRACT

PURPOSE OF REVIEW: Knowledge related to hereditary thyroid cancer syndromes has expanded enormously. This review identifies contributions that have changed approaches to diagnosis and broadened treatment options for patients with hereditary medullary and nonmedullary thyroid cancers related to multiple endocrine neoplasia type 2 (MEN2), Cowden syndrome, and familial adenomatous polyposis (FAP). RECENT FINDINGS: A new risk-stratification scheme based on type of RET gene mutation informs the age at which prophylactic thyroidectomy and diagnostic screening for MEN-associated endocrine diseases should occur. Two new US Food and Drug Administration-approved targeted medical therapies are now available for medullary thyroid cancer. There is better understanding of more aggressive clinical features and increased lifetime cancer risks for patients with well differentiated thyroid cancers as part of families with and without Cowden syndrome or FAP. This has led to a clearer appreciation for the role and timing of thyroid ultrasound screening in these populations. It has also informed the appropriate extent of thyroid surgery and the circumstances in which prophylactic thyroidectomy is reasonable to consider as part of hereditary syndromes other than MEN2. SUMMARY: Recognition and early diagnosis of these syndromes allows for comprehensive medical care and may improve thyroid cancer-related outcomes. Ultrasound-based screening programs to detect thyroid disease are advised for patients and family members with hereditary cancer syndromes.


Subject(s)
Carcinoma, Medullary , Multiple Endocrine Neoplasia , Thyroid Neoplasms , Antineoplastic Agents/therapeutic use , Carcinoma, Medullary/diagnosis , Carcinoma, Medullary/therapy , Early Diagnosis , Humans , Mass Screening/methods , Multiple Endocrine Neoplasia/diagnosis , Multiple Endocrine Neoplasia/genetics , Multiple Endocrine Neoplasia/therapy , Thyroid Neoplasms/diagnosis , Thyroid Neoplasms/therapy
11.
Surg Clin North Am ; 104(4): 909-928, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38944508

ABSTRACT

Multiple endocrine neoplasia (MEN) syndromes are rare autosomal dominant diseases that are associated with a mixture of both endocrine and non-endocrine tumors. Traditionally, there are 2 types of MEN that have unique clinical associations: MEN 1 (parathyroid hyperplasia, pancreatic neuroendocrine tumors, and pituitary tumors) and MEN 2 (medullary thyroid carcinoma and pheochromocytoma), which is further classified into MEN 2A (adds parathyroid adenomas) and 2B (adds ganglioneuromas and marfanoid habitus). Many of the endocrine tumors are resected surgically, and the pre, intra, and postoperative management strategies used must take into account the high recurrence rates asscioated with MEN tumors.


Subject(s)
Multiple Endocrine Neoplasia , Humans , Multiple Endocrine Neoplasia/surgery , Multiple Endocrine Neoplasia/diagnosis , Multiple Endocrine Neoplasia/genetics
12.
J Clin Endocrinol Metab ; 109(7): e1482-e1493, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38288531

ABSTRACT

CONTEXT: Germline CDKN1B variants predispose patients to multiple endocrine neoplasia type 4 (MEN4), a rare MEN1-like syndrome, with <100 reported cases since its discovery in 2006. Although CDKN1B mutations are frequently suggested to explain cases of genetically negative MEN1, the prevalence and phenotype of MEN4 patients is poorly known, and genetic counseling is unclear. OBJECTIVE: To evaluate the prevalence of MEN4 in MEN1-suspected patients and characterize the phenotype of MEN4 patients. DESIGN: Retrospective observational nationwide study. Narrative review of literature and variant class reassessment. PATIENTS: We included all adult patients with class 3/4/5 CDKN1B variants identified by the laboratories from the French Oncogenetic Network on Neuroendocrine Tumors network between 2015 and 2022 through germline genetic testing for MEN1 suspicion. After class reassessment, we compared the phenotype of symptomatic patients with class 4/5 CDKN1B variants (ie, with genetically confirmed MEN4 diagnosis) in our series and in literature with 66 matched MEN1 patients from the UMD-MEN1 database. RESULTS: From 5600 MEN1-suspected patients analyzed, 4 with class 4/5 CDKN1B variant were found (0.07%). They presented with multiple duodenal NET, primary hyperparathyroidism (PHPT) and adrenal nodule, isolated PHPT, PHPT, and pancreatic neuroendocrine tumor. We listed 29 patients with CDKN1B class 4/5 variants from the literature. Compared with matched MEN1 patients, MEN4 patients presented lower NET incidence and older age at PHPT diagnosis. CONCLUSION: The prevalence of MEN4 is low. PHPT and pituitary adenoma represent the main associated lesions, NETs are rare. Our results suggest a milder and later phenotype than in MEN1. Our observations will help to improve genetic counseling and management of MEN4 families.


Subject(s)
Multiple Endocrine Neoplasia Type 1 , Humans , Retrospective Studies , France/epidemiology , Male , Female , Adult , Middle Aged , Multiple Endocrine Neoplasia Type 1/genetics , Multiple Endocrine Neoplasia Type 1/epidemiology , Aged , Germ-Line Mutation , Phenotype , Cyclin-Dependent Kinase Inhibitor p27/genetics , Prevalence , Multiple Endocrine Neoplasia/genetics , Multiple Endocrine Neoplasia/epidemiology , Proto-Oncogene Proteins
13.
Dtsch Med Wochenschr ; 149(6): 283-289, 2024 Mar.
Article in German | MEDLINE | ID: mdl-38412983

ABSTRACT

Understanding genetic predisposition has a significant impact on the management of patients with endocrine tumours, including therapy, early detection and prevention. These tumours, which develop as part of a familial predisposition, often manifest early in life and frequently affect several endocrine organs. In the following article, both common syndromes, such as multiple endocrine neoplasia (MEN) syndromes, and rare syndromes, such as familial isolated pituitary adenoma (FIPA), are presented based on their indicator diseases.


Subject(s)
Adenoma , Growth Hormone-Secreting Pituitary Adenoma , Multiple Endocrine Neoplasia Type 1 , Multiple Endocrine Neoplasia , Pituitary Neoplasms , Humans , Pituitary Neoplasms/diagnosis , Pituitary Neoplasms/genetics , Pituitary Neoplasms/therapy , Adenoma/therapy , Multiple Endocrine Neoplasia Type 1/genetics , Multiple Endocrine Neoplasia Type 1/therapy , Multiple Endocrine Neoplasia Type 1/diagnosis , Multiple Endocrine Neoplasia/diagnosis , Multiple Endocrine Neoplasia/genetics , Multiple Endocrine Neoplasia/therapy , Genetic Predisposition to Disease/genetics
14.
Neuropathol Appl Neurobiol ; 39(3): 256-69, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22524684

ABSTRACT

AIMS: The aim of this study is to evaluate the pathological features, serum hormone levels and ex vivo cultures of pituitary adenomas that occur in rats affected by MENX syndrome. MENX is multiple endocrine neoplasia syndrome caused by a germline mutation in the cell cycle inhibitor p27. Characterization of MENX adenomas is a prerequisite to exploit this animal model for molecular and translational studies of pituitary adenomas. METHODS: We investigated MENX pituitary adenomas with immunohistochemistry, double immunofluorescence, electron microscopy, reverse transcription polymerase chain reaction (RT-PCR), measurement of serum hormone levels and ex vivo cultures. RESULTS: Adenomas in MENX rats belong to the gonadotroph lineage. They start from 4 months of age as multiple neoplastic nodules and progress to become large lesions that efface the gland. Adenomas are composed of chromophobic cells predominantly expressing the glycoprotein alpha-subunit (αGSU). They show mitotic activity and high Ki67 labelling. A few neoplastic cells co-express gonadotropins and the transcription factor steroidogenic factor 1, together with growth hormone or prolactin and Pit-1, suggesting that they are not fully committed to one cell lineage. Ex vivo cultures show features similar to the primary tumour. CONCLUSIONS: Our results suggest that p27 function is critical to regulate gonadotroph cells growth. The MENX syndrome represents a unique model to elucidate the physiological and molecular mechanisms mediating the pathogenesis of gonadotroph adenomas.


Subject(s)
Adenoma/pathology , Cyclin-Dependent Kinase Inhibitor p27/genetics , Multiple Endocrine Neoplasia/pathology , Pituitary Neoplasms/pathology , Adenoma/genetics , Adenoma/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Disease Models, Animal , Fluorescent Antibody Technique , Gonadotropins/genetics , Immunohistochemistry , Multiple Endocrine Neoplasia/genetics , Multiple Endocrine Neoplasia/metabolism , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction
15.
Clin Genet ; 83(4): 384-7, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22734615

ABSTRACT

Mutations in RET proto-oncogene cause multiple endocrine neoplasia 2A (MEN2A). Mutations in codons 609 and 611 are not frequent. We identified two MEN2A families with the Cys609Phe RET mutation, which turned out to be the same family. This mutation has been described a couple of times with no clinical details. We have characterized the clinical phenotype of this large kindred. A 54-year-old woman, with a medullary thyroid carcinoma (MTC), and a 33-year-old woman, who was operated on for an adrenal pheochromocytoma, were the index cases. 35 relatives were studied. Sixteen turned out to be carriers and 12 of them have been operated on. This family showed eight patients with C-cell hyperplasia, six patients affected by MTC and two showing pheochromocytoma. A papillary thyroid carcinoma was also found, together with the MTC, in one of the carriers. The phenotype in this large kindred is clearly of MEN2A. In carriers presenting the Cys609Phe mutation, the timing of the presentation of the syndrome is highly unpredictable. Therefore, a strict follow up of MTC must be carried out because of risk, and pheochromocytoma should not be ignored. These results reinforce the scarce data observed on this particular mutation.


Subject(s)
Multiple Endocrine Neoplasia Type 2a/genetics , Mutation , Proto-Oncogene Proteins c-ret/genetics , Adolescent , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/pathology , Adult , Carcinoma/genetics , Carcinoma/pathology , Carcinoma, Papillary , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Multiple Endocrine Neoplasia/genetics , Multiple Endocrine Neoplasia/pathology , Phenotype , Proto-Oncogene Mas , Thyroid Cancer, Papillary , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Young Adult
16.
J Endocrinol Invest ; 36(9): 781-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23800691

ABSTRACT

Multiple endocrine neoplasias (MEN) are autosomal dominant disorders characterized by the occurrence of tumors in at least two endocrine glands. Until recently, two MEN syndromes were known, i.e. the MEN type 1 (MEN1) and type 2 (MEN2), which are caused by germline mutations in the MEN1 and RET genes, respectively. These two syndromes are characterized by a different tumor spectrum. A few years ago we described a variant of the MEN syndromes, which spontaneously developed in a rat colony and was named MENX. Affected animals consistently develop multiple endocrine tumors, with a spectrum that shares features with both MEN1 and MEN2 human syndromes. Genetic studies identified a germline mutation in the Cdkn1b gene, encoding the p27 cell cycle inhibitor, as the causative mutation for MENX. Capitalizing on these findings, germline mutations in the human homologue, CDKN1B, were searched for and identified in patients with multiple endocrine tumors. As a consequence of this discovery, a novel human MEN syndrome, named MEN4, was recognized, which is caused by heterozygous mutations in p27. These studies identified Cdkn1b/CDKN1B as a novel tumor susceptibility gene for multiple endocrine tumors in both rats and humans. Here we review the characteristics of the MENX and MEN4 syndromes and we briefly address the main function of p27 and how it is affected by MENX- or MEN4-associated mutations.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/genetics , Multiple Endocrine Neoplasia/genetics , Adenoma/genetics , Adult , Animals , Germ-Line Mutation , Humans , Multiple Endocrine Neoplasia Type 1/genetics , Mutation, Missense , Parathyroid Neoplasms/genetics , Pituitary Neoplasms/genetics , Polymorphism, Single Nucleotide , Rats
17.
Proc Natl Acad Sci U S A ; 107(43): 18493-8, 2010 Oct 26.
Article in English | MEDLINE | ID: mdl-20937862

ABSTRACT

Pheochromocytomas are rare neoplasias of neural crest origin arising from chromaffin cells of the adrenal medulla and sympathetic ganglia (extra-adrenal pheochromocytoma). Pheochromocytoma that develop in rats homozygous for a loss-of-function mutation in p27Kip1 (MENX syndrome) show a clear progression from hyperplasia to tumor, offering the possibility to gain insight into tumor pathobiology. We compared the gene-expression signatures of both adrenomedullary hyperplasia and pheochromocytoma with normal rat adrenal medulla. Hyperplasia and tumor show very similar transcriptome profiles, indicating early determination of the tumorigenic signature. Overrepresentation of developmentally regulated neural genes was a feature of the rat lesions. Quantitative RT-PCR validated the up-regulation of 11 genes, including some involved in neural development: Cdkn2a, Cdkn2c, Neurod1, Gal, Bmp7, and Phox2a. Overexpression of these genes precedes histological changes in affected adrenal glands. Their presence at early stages of tumorigenesis indicates they are not acquired during progression and may be a result of the lack of functional p27Kip1. Adrenal and extra-adrenal pheochromocytoma development clearly follows diverged molecular pathways in MENX rats. To correlate these findings to human pheochromocytoma, we studied nine genes overexpressed in the rat lesions in 46 sporadic and familial human pheochromocytomas. The expression of GAL, DGKH, BMP7, PHOX2A, L1CAM, TCTE1, EBF3, SOX4, and HASH1 was up-regulated, although with different frequencies. Immunohistochemical staining detected high L1CAM expression selectively in 27 human pheochromocytomas but not in 140 nonchromaffin neuroendocrine tumors. These studies reveal clues to the molecular pathways involved in rat and human pheochromocytoma and identify previously unexplored biomarkers for clinical use.


Subject(s)
Adrenal Gland Neoplasms/genetics , Multiple Endocrine Neoplasia/genetics , Pheochromocytoma/genetics , Adrenal Gland Neoplasms/pathology , Adrenal Medulla/metabolism , Adrenal Medulla/pathology , Animals , Base Sequence , Biomarkers, Tumor/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , DNA Primers/genetics , Disease Models, Animal , Gene Expression Profiling , Homeodomain Proteins/genetics , Humans , Hyperplasia , Multiple Endocrine Neoplasia/pathology , Neural Cell Adhesion Molecule L1/genetics , PC12 Cells , Paraganglioma/genetics , Pheochromocytoma/pathology , Rats , Rats, Mutant Strains , Species Specificity
18.
Nat Genet ; 6(1): 70-4, 1994 Jan.
Article in English | MEDLINE | ID: mdl-7907913

ABSTRACT

We have analysed 118 families with inherited medullary thyroid carcinoma (MTC) for mutations of the RET proto-oncogene. These included cases of multiple endocrine neoplasia types 2A (MEN 2A) and 2B (MEN 2B) and familial MTC (FMTC). Mutations at one of 5 cysteines in the extracellular domain were found in 97% of patients with MEN 2A and 86% with FMTC but not in MEN 2B patients or normal controls. 84% of the MEN2A mutations affected codon 634. MEN 2A patients with a Cys634 to Arg substitution had a greater risk of developing parathyroid disease than those with other codon 634 mutations. Our data show a strong correlation between disease phenotype and the nature and position of the RET mutation, suggesting that a simple, constitutive activation of the RET tyrosine kinase is unlikely to explain the events leading to MEN 2A and FMTC.


Subject(s)
Carcinoma, Medullary/genetics , Drosophila Proteins , Multiple Endocrine Neoplasia/genetics , Proto-Oncogenes , Thyroid Neoplasms/genetics , Base Sequence , DNA Mutational Analysis , DNA Primers/genetics , Exons , Humans , Molecular Sequence Data , Phenotype , Point Mutation , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/genetics
19.
Rev Med Interne ; 44(1): 12-18, 2023 Jan.
Article in French | MEDLINE | ID: mdl-36307322

ABSTRACT

Multiple endocrine neoplasia (MEN) are genetic predisposition syndromes to endocrine tumors including MEN1, MEN2 and exceptionally MEN4. MEN are transmitted in an autosomal dominant fashion with a high penetrance. Classically, there is no genotype/phenotype correlation for NEM1 whereas this is the case for NEM2. Patients with NEM1, linked to an inactivating mutation of the menin gene, may present with: primary hyperparathyroidism, pituitary adenoma, duodeno-pancreatic neuroendocrine tumors (NETs), bronchial tumors with an increased risk of thymoma, adrenal cortical tumors, an increased risk of breast cancer and characteristic skin involvement such as collagenomas, lentiginomas and an increased risk of skin cancer. These patients require at least annual follow-up. Screening of children is proposed from the age of 5 years. Patients with NEM2, linked to an activating mutation of the RET proto-oncogene, all present with medullary thyroid carcinoma (MTC) at a variable age depending on the genotype. Some patients present a pheochromocytoma (50 %) and hyperparathyroidism (20 %). Pediatric forms with aggressive CMT, ganglioneuromatosis and marfanoid syndrome exist (rare NEM2B). Some mutations are associated with a risk of aggressive CMT, justifying prophylactic thyroidectomy before 6 months of age. The age of genetic testing depends on the mutation subtype in the NEM2 parent. NEM4, related to a mutation in the CDKN1B gene, are rare, with a less well-known pathogenesis and their follow-up is not well codified.


Subject(s)
Adrenal Gland Neoplasms , Multiple Endocrine Neoplasia , Pheochromocytoma , Thyroid Neoplasms , Humans , Multiple Endocrine Neoplasia/diagnosis , Multiple Endocrine Neoplasia/genetics , Pheochromocytoma/diagnosis , Adrenal Gland Neoplasms/diagnosis , Genetic Testing , Mutation , Syndrome
20.
Endocrine ; 82(3): 480-490, 2023 12.
Article in English | MEDLINE | ID: mdl-37632635

ABSTRACT

PURPOSE: Multiple endocrine neoplasia type 4 (MEN4) is a rare multiglandular endocrine neoplasia syndrome, associated with a wide tumor spectrum but hallmarked by primary hyperparathyroidism, which represents the most common clinical feature, followed by pituitary (functional and non-functional) adenomas, and neuroendocrine tumors. MEN4 clinically overlaps MEN type 1 (MEN1) but differs from it for milder clinical features and an older patient's age at onset. The underlying mutated gene, CDKN1B, encodes the cell cycle regulator p27, implicated in cellular proliferation, motility and apoptosis. Given the paucity of MEN4 cases described in the literature, possible genotype-phenotype correlations have not been thoroughly assessed, and specific clinical recommendations are lacking. The present review provides an extensive overview of molecular genetics and clinical features of MEN4, with the aim of contributing to delineate peculiar strategies for clinical management, screening and follow-up of the last and least known MEN syndrome. METHODS: A literature search was performed through online databases like MEDLINE and Scopus. CONCLUSIONS: MEN4 is much less common that MEN1, tend to present later in life with a more indolent course, although involving the same primary organs as MEN1. As a consequence, MEN4 patients might need specific diagnostic and therapeutic approaches and a different strategy for screening and follow-up. Further studies are needed to assess the real oncological risk of MEN4 carriers, and to establish a standardized screening protocol. Furthermore, a deeper understanding of molecular genetics of MEN4 is needed in order to explore p27 as a novel therapeutic target.


Subject(s)
Adenoma , Multiple Endocrine Neoplasia Type 1 , Multiple Endocrine Neoplasia , Neuroendocrine Tumors , Humans , Multiple Endocrine Neoplasia/diagnosis , Multiple Endocrine Neoplasia/genetics , Multiple Endocrine Neoplasia/pathology , Multiple Endocrine Neoplasia Type 1/genetics , Multiple Endocrine Neoplasia Type 1/diagnosis , Neuroendocrine Tumors/genetics , Adenoma/genetics , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL