ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Multiple endocrine neoplasia type 1: Genetics

Multiple endocrine neoplasia type 1: Genetics
Literature review current through: Jan 2024.
This topic last updated: Sep 27, 2022.

INTRODUCTION — The multiple endocrine neoplasia (MEN) syndromes are rare, but recognition is important both for treatment and for evaluation of family members.

This topic will review the genetics of the MEN type 1 (MEN1) syndrome (OMIM #131100). The clinical manifestations, diagnosis, and therapy of MEN1 and the MEN type 2 (MEN2) syndromes are discussed separately:

(See "Multiple endocrine neoplasia type 1: Clinical manifestations and diagnosis".)

(See "Multiple endocrine neoplasia type 1: Management".)

(See "Classification and genetics of multiple endocrine neoplasia type 2".)

(See "Clinical manifestations and diagnosis of multiple endocrine neoplasia type 2".)

(See "Approach to therapy in multiple endocrine neoplasia type 2".)

DEFINITION — Multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant disorder classically characterized by predisposition to tumors of the parathyroid glands, anterior pituitary, and pancreatic islet cells [1,2]. MEN1 also includes a predisposition to gastrinomas in the duodenum, bronchopulmonary and thymic neuroendocrine tumors (NETs), gastric carcinoids, adrenal adenomas (occasionally carcinomas), angiofibromas, lipomas, and other tumors (figure 1). Other associated tumors include angiofibromas, angiomyolipomas, spinal cord ependymomas, and a two- to threefold increased risk of breast cancer has been reported [3]. (See "Multiple endocrine neoplasia type 1: Clinical manifestations and diagnosis", section on 'Clinical manifestations'.)

The presence of MEN1 is defined clinically as the occurrence of two or more primary MEN1 tumor types (parathyroid, enteropancreatic endocrine, and pituitary tumors), or in family members of a patient with a clinical diagnosis of MEN1, the occurrence of one of the MEN1-associated tumors. Multiple parathyroid tumors causing primary hyperparathyroidism are the most common component of MEN1, occurring in the large majority of patients by age 50 years, and is the initial manifestation of the disorder in most patients (figure 2) [2,4,5]. (See "Multiple endocrine neoplasia type 1: Clinical manifestations and diagnosis", section on 'Diagnosis'.)

The current major morbidity and premature mortality associated with MEN1 is most frequently related to pancreatic NETs (in particular, nonfunctioning pancreatic NETs and, to a lesser extent, gastrinoma), as well as thymic NETs [6]. Subclinical involvement is more common; anatomic or intensive biochemical studies reveal evidence of microscopic pancreatic islet abnormalities in almost all patients. (See "Multiple endocrine neoplasia type 1: Clinical manifestations and diagnosis", section on 'Nonfunctioning pancreatic tumors' and "Multiple endocrine neoplasia type 1: Clinical manifestations and diagnosis", section on 'Thymic and bronchopulmonary neuroendocrine tumors'.)

The prevalence of MEN1 is approximately 2 per 100,000 [2]. The incidence ranges from 1 to 18, 16 to 38, and less than 3 percent in patients with parathyroid adenomas, gastrinomas, and pituitary adenomas, respectively [1].

GENETICS

MEN1 gene — The inheritance of classical MEN1 follows an autosomal dominant pattern, indicating that Mendelian inheritance of a single mutant gene is responsible for transmitting the tumor predisposition within a given family. In 1988, genetic linkage analysis implicated a region on the long arm of chromosome 11 (11q13) as the site of the "MEN1 gene" [7]. A decade later, the critical gene in this region was identified, given the gene symbol designation MEN1, and its protein product termed "menin" [8].

Across multiple studies, pathogenic deoxyribonucleic acid (DNA) sequence variants (ie, "mutations") in the MEN1 gene have been detected in 70 to 90 percent of unrelated MEN1 kindreds [9-12], although this figure is variable across studies and is sensitive to the mode of case selection. More than 600 different germline MEN1 mutations have been identified, with the majority (approximately 70 percent) predicting premature truncation or loss of the Menin protein (eg, as a result of frameshift, nonsense, or splice site DNA variants), although missense variants (eg, single amino acid substitutions) are also reported and are presumed to disrupt key functional domains of the protein or alter protein stability. In addition, larger structural variants (eg, partial or whole gene deletions) may occur, accounting for <5 percent of cases. Furthermore, it has been reasonably hypothesized that most typical MEN1 kindreds without detectable pathogenic variants in the MEN1 gene nonetheless have inactivating germline variants in (or near and in "cis" with) the same gene but outside the coding region that is typically sequenced in diagnostic and research laboratories. Disease-associated pathogenic MEN1 variants have been reported in noncoding untranslated regions (eg, 5' UTR) of the MEN1 gene [13].

The majority of pathogenic MEN1 variants are inherited from an affected parent. However, it is estimated that approximately 10 percent of pathogenic MEN1 variants occur de novo (ie, occurring for the first time in the affected individual). In addition, both germline and somatic MEN1 mosaicism have been observed in patients presenting with MEN1 phenotypes [14]. Genetic variants in the MEN1 gene are not responsible for all individuals, or even kindreds, with an MEN1 phenotype (see 'Other genes' below). Furthermore, rare phenocopies have been reported, ie, individuals within MEN1 kindreds who were initially classified as having the syndrome when they developed a typical tumor (eg, prolactinoma) but were then proven by DNA testing to have not inherited the pathologic mutation [15].

Much has been learned about the biochemical and cellular functions of menin, but the precise way(s) in which these functions relate to tumorigenesis is still not well established. However, it is clear that most of the pathogenic MEN1 gene variants found in MEN1 patients would be expected to inactivate or disrupt menin function. Typical of a classical tumor suppressor gene, the spectrum of reported germline MEN1 pathogenic variants occur throughout the gene and yield no strong genotype/phenotype relationships [12]. The genotype-phenotype correlations are often unclear, even within a family [16]. In addition, biallelic somatic mutations within this gene have been found in 12 to 17 percent of typical nonfamilial parathyroid adenomas [17-19] and some sporadic gastrinomas and insulinomas [20], sporadic neuroendocrine tumors (NETs) of the foregut [21], sporadic carcinoid tumors of the lung [22], and sporadic pituitary tumors [23], further supporting the relationship between inactivation of the MEN1 gene (and Menin function) and tumorigenesis (see "Primary hyperparathyroidism: Pathogenesis and etiology"). However, the large majority of non-MEN1-associated pituitary tumors, whether sporadic or familial, do not have an MEN1 mutation [24,25].

Other genes — Syndromes clinically related to but genetically distinct from MEN1 do exist. At least one family with an unusual expression of MEN1 (eg, a lower than expected incidence of hyperparathyroidism and higher than expected incidence of pituitary tumors) was reported to have a predisposing gene at a location distinct from the chromosome 11q13 site [26,27]. Germline AIP mutation, in the absence of MEN1 gene mutation, has been reported in the setting of pituitary plus parathyroid neoplasia [28]. Furthermore, pathogenic variants in the MEN1 gene are infrequent in kindreds with familial isolated hyperparathyroidism [29,30] and were not found in three kindreds with familial pituitary adenoma [31] and one with isolated familial acromegaly [32]. A small minority of patients presenting with a clinical diagnosis of MEN1 (eg, presence of two or more MEN1-assocated tumors) may harbor pathogenic variants in other monogenic disease genes more typically associated with alternate phenotypes (eg, CDC73 variants associated with the hyperparathyroidism jaw tumour syndrome) [33].

Cyclin-dependent kinase inhibitor genes – An inherited mutation of the p27 cyclin-dependent kinase (CDK) inhibitor gene, CDKN1B, was reported in one kindred whose proband had hyperparathyroidism and acromegaly due to a growth hormone-producing pituitary tumor; the proband's father had acromegaly, and the sister had a renal angiomyolipoma [34]. Germline pathogenic variants in CDKN1B were also reported in a few other cases of MEN1 that collectively exhibited features including hyperparathyroidism, Cushing disease, cervical carcinoid tumor, bilateral nonfunctioning adrenal masses, and Zollinger-Ellison syndrome with duodenal and pancreatic masses [35,36]. MEN1-like disease caused by germline CDKN1B mutation has been termed MEN4 (OMIM #610755). However, MEN4 is rare (<30 kindreds reported in the literature) and is estimated to account for only 1 to 3 percent of unrelated MEN1-like cases without identifiable MEN1 mutations [36-38].

Rare germline variants in three other CDK inhibitor genes, CDKN2B, CDKN2C, and CDKN1A, encoding the p15, p18, and p21 proteins, respectively, have also been identified in this setting and may collectively account for another 1 to 2 percent of MEN1-like cases without detectable MEN1 mutations [36].

Overall, because patients presenting with the combination of sporadic parathyroid plus pituitary tumor have a much lower yield of detectable MEN1 mutation than in typical MEN1 kindreds or in sporadic cases of parathyroid plus pancreatic tumor [39], it seems likely that this phenotype may often be due to mutation in a gene other than MEN1, or possibly the coincidental presence of sporadic tumors absent of any major genetic predisposition. However, as noted above, the extent to which mutation in CDKN1B or other CDK inhibitor genes is responsible for MEN1-like phenotypes, sporadic or familial, in the absence of MEN1 mutation appears to be small [36,38,40].

MAX gene – Two kindreds have been reported with a spectrum of endocrine tumors partially overlapping with MEN1 including pheochromocytoma, NETs (eg, paravertebral ganglioneuroma, abdominal neuroblastoma, pulmonary adenocarcinoma), pituitary adenomas, and primary hyperparathyroidism. These kindreds were observed to harbor pathogenic germline variants in the MAX gene, and the possibility was raised that this could be considered an additional MEN syndrome (ie, MEN5) [41].

DNA testing — Direct DNA testing for MEN1 mutation is available for clinical use, has a useful role in certain settings, and should be considered on an individual basis [1]. However, in contrast with testing for RET gene mutations in MEN2, presymptomatic DNA diagnosis has not been shown to yield equally clear benefits in preventing morbidity and mortality in individuals at risk for MEN1. DNA testing is reviewed in detail separately. (See "Multiple endocrine neoplasia type 1: Clinical manifestations and diagnosis", section on 'Genetic testing'.)

PATHOGENESIS — Patients with classical MEN1 have often inherited one inactivated copy of the MEN1 gene from an affected parent [8,10]. The actual outgrowth of a tumor is thought to require the subsequent somatic inactivation, often by gross deletion, of the remaining normal copy of the gene in one cell (so-called "two-hit" effect described by Knudson). Such a parathyroid cell, as an example, would then be devoid of the MEN1 gene's normal tumor suppressor function and could gain a selective advantage over its neighbors, resulting in a clonal proliferation (figure 3). The high incidence of endocrine tumors in MEN1 (which has over 90 percent penetrance) and the common multiplicity of these tumors implies that somatic inactivation of the remaining normal copy of the gene occurs at an appreciable frequency and contributes importantly to tumorigenesis in the clinically affected tissues. This model also appears to apply to some of the nonendocrine tumors that occur in patients with MEN1.

Although, a range of in vitro and in vivo studies have provided extensive insights into the function of the Menin protein, the mechanisms leading to tumorigenesis in MEN1-associated tumors remain poorly defined. Notably, Menin is a ubiquitously expressed predominantly nuclear protein implicated in a broad range of cellular activities including transcription and epigenetic regulation through direct or indirect interactions with a large number of protein partners and complexes. However, understanding how tissue-specific functions are established remains to be defined. The identification and analysis of other genes whose somatic alteration is also important in the emergence of clonal tumors in this syndrome should further clarify the relationship between genotype and phenotype in MEN1.

SUMMARY

Definition – Multiple endocrine neoplasia type 1 (MEN1) is a rare autosomal dominant disorder with a prevalence of approximately 2 per 100,000. MEN1 is defined clinically as the presence of two of the three main MEN1 tumor types (parathyroid, enteropancreatic endocrine, and pituitary tumors), or in family members of a patient with a clinical diagnosis of MEN1, the occurrence of one of the MEN1-associated tumors. Patients with MEN1 may have tumors other than those in the parathyroid, pituitary glands, and in the pancreatic islet cells, including duodenal gastrinomas, thymic or bronchial carcinoid tumors, enterochromaffin cell-like gastric tumors, adrenocortical adenomas, lipomas, angiofibromas, angiomyolipomas, and spinal cord ependymomas (figure 2 and figure 1). (See 'Definition' above.)

MEN1 gene – The MEN1 tumor suppressor gene is located on the long arm of chromosome 11 (11q13). Its protein product is termed "menin." Over 1000 MEN1 gene mutations have been detected that inactivate or disrupt menin function. Inactivation of menin results in loss of tumor suppression. Families with the same types of mutations do not necessarily have the same clinical phenotype. (See 'MEN1 gene' above.)

Other genes – Syndromes clinically related to but genetically distinct from MEN1 do exist, and mutations in the MEN1 gene are not responsible for all individuals, or even kindreds, with an MEN1 phenotype. (See 'Other genes' above.)

  1. Thakker RV, Newey PJ, Walls GV, et al. Clinical practice guidelines for multiple endocrine neoplasia type 1 (MEN1). J Clin Endocrinol Metab 2012; 97:2990.
  2. Al-Salameh A, Cadiot G, Calender A, et al. Clinical aspects of multiple endocrine neoplasia type 1. Nat Rev Endocrinol 2021; 17:207.
  3. van Leeuwaarde RS, Dreijerink KM, Ausems MG, et al. MEN1-Dependent Breast Cancer: Indication for Early Screening? Results From the Dutch MEN1 Study Group. J Clin Endocrinol Metab 2017; 102:2083.
  4. Trump D, Farren B, Wooding C, et al. Clinical studies of multiple endocrine neoplasia type 1 (MEN1). QJM 1996; 89:653.
  5. Romanet P, Mohamed A, Giraud S, et al. UMD-MEN1 Database: An Overview of the 370 MEN1 Variants Present in 1676 Patients From the French Population. J Clin Endocrinol Metab 2019; 104:753.
  6. Newey PJ, Newell-Price J. MEN1 Surveillance Guidelines: Time to (Re)Think? J Endocr Soc 2022; 6:bvac001.
  7. Larsson C, Skogseid B, Oberg K, et al. Multiple endocrine neoplasia type 1 gene maps to chromosome 11 and is lost in insulinoma. Nature 1988; 332:85.
  8. Chandrasekharappa SC, Guru SC, Manickam P, et al. Positional cloning of the gene for multiple endocrine neoplasia-type 1. Science 1997; 276:404.
  9. Bassett JH, Forbes SA, Pannett AA, et al. Characterization of mutations in patients with multiple endocrine neoplasia type 1. Am J Hum Genet 1998; 62:232.
  10. Agarwal SK, Lee Burns A, Sukhodolets KE, et al. Molecular pathology of the MEN1 gene. Ann N Y Acad Sci 2004; 1014:189.
  11. Lemos MC, Thakker RV. Multiple endocrine neoplasia type 1 (MEN1): analysis of 1336 mutations reported in the first decade following identification of the gene. Hum Mutat 2008; 29:22.
  12. Brandi ML, Agarwal SK, Perrier ND, et al. Multiple Endocrine Neoplasia Type 1: Latest Insights. Endocr Rev 2021; 42:133.
  13. Kooblall KG, Boon H, Cranston T, et al. Multiple Endocrine Neoplasia Type 1 (MEN1) 5'UTR Deletion, in MEN1 Family, Decreases Menin Expression. J Bone Miner Res 2021; 36:100.
  14. Coppin L, Giraud S, Pasmant E, et al. Multiple endocrine neoplasia type 1 caused by mosaic mutation: clinical follow-up and genetic counseling? Eur J Endocrinol 2022; 187:K1.
  15. Turner JJ, Christie PT, Pearce SH, et al. Diagnostic challenges due to phenocopies: lessons from Multiple Endocrine Neoplasia type1 (MEN1). Hum Mutat 2010; 31:E1089.
  16. Concolino P, Rossodivita A, Carrozza C, et al. A novel MEN1 frameshift germline mutation in two Italian monozygotic twins. Clin Chem Lab Med 2008; 46:824.
  17. Heppner C, Kester MB, Agarwal SK, et al. Somatic mutation of the MEN1 gene in parathyroid tumours. Nat Genet 1997; 16:375.
  18. Carling T, Correa P, Hessman O, et al. Parathyroid MEN1 gene mutations in relation to clinical characteristics of nonfamilial primary hyperparathyroidism. J Clin Endocrinol Metab 1998; 83:2960.
  19. Farnebo F, Teh BT, Kytölä S, et al. Alterations of the MEN1 gene in sporadic parathyroid tumors. J Clin Endocrinol Metab 1998; 83:2627.
  20. Zhuang Z, Vortmeyer AO, Pack S, et al. Somatic mutations of the MEN1 tumor suppressor gene in sporadic gastrinomas and insulinomas. Cancer Res 1997; 57:4682.
  21. Toliat MR, Berger W, Ropers HH, et al. Mutations in the MEN I gene in sporadic neuroendocrine tumours of gastroenteropancreatic system. Lancet 1997; 350:1223.
  22. Debelenko LV, Brambilla E, Agarwal SK, et al. Identification of MEN1 gene mutations in sporadic carcinoid tumors of the lung. Hum Mol Genet 1997; 6:2285.
  23. Zhuang Z, Ezzat SZ, Vortmeyer AO, et al. Mutations of the MEN1 tumor suppressor gene in pituitary tumors. Cancer Res 1997; 57:5446.
  24. Asa SL, Somers K, Ezzat S. The MEN-1 gene is rarely down-regulated in pituitary adenomas. J Clin Endocrinol Metab 1998; 83:3210.
  25. Teh BT, Kytölä S, Farnebo F, et al. Mutation analysis of the MEN1 gene in multiple endocrine neoplasia type 1, familial acromegaly and familial isolated hyperparathyroidism. J Clin Endocrinol Metab 1998; 83:2621.
  26. Stock JL, Warth MR, Teh BT, et al. A kindred with a variant of multiple endocrine neoplasia type 1 demonstrating frequent expression of pituitary tumors but not linked to the multiple endocrine neoplasia type 1 locus at chromosome region 11q13. J Clin Endocrinol Metab 1997; 82:486.
  27. Agarwal SK, Ozawa A, Mateo CM, Marx SJ. The MEN1 gene and pituitary tumours. Horm Res 2009; 71 Suppl 2:131.
  28. Belar O, De La Hoz C, Pérez-Nanclares G, et al. Novel mutations in MEN1, CDKN1B and AIP genes in patients with multiple endocrine neoplasia type 1 syndrome in Spain. Clin Endocrinol (Oxf) 2012; 76:719.
  29. Agarwal SK, Kester MB, Debelenko LV, et al. Germline mutations of the MEN1 gene in familial multiple endocrine neoplasia type 1 and related states. Hum Mol Genet 1997; 6:1169.
  30. Simonds WF, Robbins CM, Agarwal SK, et al. Familial isolated hyperparathyroidism is rarely caused by germline mutation in HRPT2, the gene for the hyperparathyroidism-jaw tumor syndrome. J Clin Endocrinol Metab 2004; 89:96.
  31. Tanaka C, Yoshimoto K, Yamada S, et al. Absence of germ-line mutations of the multiple endocrine neoplasia type 1 (MEN1) gene in familial pituitary adenoma in contrast to MEN1 in Japanese. J Clin Endocrinol Metab 1998; 83:960.
  32. Jorge BH, Agarwal SK, Lando VS, et al. Study of the multiple endocrine neoplasia type 1, growth hormone-releasing hormone receptor, Gs alpha, and Gi2 alpha genes in isolated familial acromegaly. J Clin Endocrinol Metab 2001; 86:542.
  33. Lines KE, Nachtigall LB, Dichtel LE, et al. Multiple Endocrine Neoplasia Type 1 (MEN1) Phenocopy Due to a Cell Cycle Division 73 (CDC73) Variant. J Endocr Soc 2020; 4:bvaa142.
  34. Pellegata NS, Quintanilla-Martinez L, Siggelkow H, et al. Germ-line mutations in p27Kip1 cause a multiple endocrine neoplasia syndrome in rats and humans. Proc Natl Acad Sci U S A 2006; 103:15558.
  35. Georgitsi M, Raitila A, Karhu A, et al. Germline CDKN1B/p27Kip1 mutation in multiple endocrine neoplasia. J Clin Endocrinol Metab 2007; 92:3321.
  36. Agarwal SK, Mateo CM, Marx SJ. Rare germline mutations in cyclin-dependent kinase inhibitor genes in multiple endocrine neoplasia type 1 and related states. J Clin Endocrinol Metab 2009; 94:1826.
  37. Pellegata NS. MENX and MEN4. Clinics (Sao Paulo) 2012; 67 Suppl 1:13.
  38. Frederiksen A, Rossing M, Hermann P, et al. Clinical Features of Multiple Endocrine Neoplasia Type 4: Novel Pathogenic Variant and Review of Published Cases. J Clin Endocrinol Metab 2019; 104:3637.
  39. Klein RD, Salih S, Bessoni J, Bale AE. Clinical testing for multiple endocrine neoplasia type 1 in a DNA diagnostic laboratory. Genet Med 2005; 7:131.
  40. Ozawa A, Agarwal SK, Mateo CM, et al. The parathyroid/pituitary variant of multiple endocrine neoplasia type 1 usually has causes other than p27Kip1 mutations. J Clin Endocrinol Metab 2007; 92:1948.
  41. Seabrook AJ, Harris JE, Velosa SB, et al. Multiple Endocrine Tumors Associated with Germline MAX Mutations: Multiple Endocrine Neoplasia Type 5? J Clin Endocrinol Metab 2021; 106:1163.
Topic 2037 Version 17.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟