fbpx
Wikipedia

MEN1

Menin is a protein that in humans is encoded by the MEN1 gene.[5] Menin is a putative tumor suppressor associated with multiple endocrine neoplasia type 1 (MEN-1 syndrome) and has autosomal dominant inheritance.[6] Variations in the MEN1 gene can cause pituitary adenomas, hyperparathyroidism, pancreatic neuroendocrine tumors, gastrinoma, and adrenocortical cancers.

MEN1
Available structures
PDBOrtholog search: PDBe RCSB
Identifiers
AliasesMEN1, MEAI, SCG2, menin 1
External IDsOMIM: 613733 MGI: 1316736 HomoloGene: 7418 GeneCards: MEN1
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_001168488
NM_001168489
NM_001168490
NM_008583

RefSeq (protein)

NP_001161960
NP_001161961
NP_001161962
NP_032609

Location (UCSC)Chr 11: 64.8 – 64.81 MbChr 19: 6.39 – 6.39 Mb
PubMed search[3][4]
Wikidata
View/Edit HumanView/Edit Mouse

In vitro studies have shown that menin is localized to the nucleus, possesses two functional nuclear localization signals, and inhibits transcriptional activation by JunD. However, the function of this protein is not known. Two messages have been detected on northern blots but the larger message has not been characterized. Two variants of the shorter transcript have been identified where alternative splicing affects the coding sequence. Five variants where alternative splicing takes place in the 5' UTR have also been identified.[5]

History edit

In 1988, researchers at Uppsala University Hospital and the Karolinska Institute in Stockholm mapped the MEN1 gene to the long arm of chromosome 11.[7] The gene was finally cloned in 1997.[8]

Genomics edit

The gene is located on long arm of chromosome 11 (11q13) between base pairs 64,570,985 and 64,578,765. It has 10 exons and encodes a 610-amino acid protein.

Over 1300 mutations have been reported to date (2010). The majority (>70%) of these are predicted to lead to truncated forms are scattered throughout the gene. Four - c.249_252delGTCT (deletion at codons 83-84), c.1546_1547insC (insertion at codon 516), c.1378C>T (Arg460Ter) and c.628_631delACAG (deletion at codons 210-211) have been reported to occur in 4.5%, 2.7%, 2.6% and 2.5% of families.[6]

Clinical implications edit

The MEN1 phenotype is inherited via an autosomal-dominant pattern and is associated with neoplasms of the pituitary gland, the parathyroid gland, and the pancreas (the 3 "P"s). While these neoplasias are often benign (in contrast to tumours occurring in MEN2A), they are adenomas and, therefore, produce endocrine phenotypes. Pancreatic presentations of the MEN1 phenotype may manifest as Zollinger-Ellison syndrome.

MEN1 pituitary tumours are adenomas of anterior cells, typically prolactinomas or growth hormone-secreting. Pancreatic tumours involve the islet cells, giving rise to gastrinomas or insulinomas. In rare cases, adrenal cortex tumours are also seen.

Role in cancer edit

Most germline or somatic mutations in the MEN1 gene predict truncation or absence of encoded menin resulting in the inability of MEN1 to act as a tumor suppressor gene.[9] Such mutations in MEN1 have been associated with defective binding of encoded menin to proteins implicated in genetic and epigenetic mechanisms.[10] Menin is a 621 amino acid protein associated with insulinomas[11] which acts as an adapter while also interacting with partner proteins involved in vital cell activities such as transcriptional regulation, cell division, cell proliferation, and genome stability. Insulinomas are neuroendocrine tumors of the pancreas with an incidence of 0.4 %[citation needed] which usually are benign solitary tumors but 5-12 % of cases have distant metastasis at diagnosis.[12] These familial MEN-1 and sporadic tumors may arise either due to loss of heterozygosity or the chromosome region 11q13 where MEN1 is located, or due to presence of mutations in the gene.[13][14]

MEN1 mutations comprise mostly frameshift deletions or insertions, followed by nonsense, missense, splice-site mutations and either part or complete gene deletions resulting in disease pathology.[15] Frameshift and nonsense mutations result in a supposed inactive and truncated menin protein while splice-site mutations result in incorrectly spliced mRNA. Missense mutations of MEN1 are especially important as they result in a change to crucial amino acids needed in order to bind and interact with other proteins and molecules. As menin is located predominantly in the nucleus,[16] these mutations can impact the stability of the cell and may further affect functional activity or expression levels of the protein. Studies have also shown that single amino acid changes in genes involved in oncogenic disorders may result in proteolytic degradation leading to loss of function and reduced stability of the mutant protein; a common mechanism for inactivating tumor suppressor gene products.[17][18] MEN1 gene mutations and deletions also play a role in the development of hereditary and a subgroup of sporadic pituitary adenomas and were detected in approximately 5% of sporadic pituitary adenomas.[19] Consequently, alterations of the gene represent a candidate pathogenetic mechanism of pituitary tumorigenesis especially when considered in terms of interactions with other proteins, growth factors, oncogenes play a rule in tumorigenesis.

Although the exact function of MEN1 is not known, the Knudson "two-hit" hypothesis provides strong evidence that it is a tumor suppressor gene. Familial loss of one copy of MEN1 is seen in association with MEN-1 syndrome. Tumor suppressor carcinogenesis follows Knudson's "two-hit" model.[20] The first hit is a heterozygous MEN1 germline mutation either developed in an early embryonic stage and consequently present in all cells at birth for the sporadic cases, or inherited from one parent in a familial case. The second hit is a MEN1 somatic mutation, oftentimes a large deletion occurring in the predisposed endocrine cell and providing cells with the survival advantaged needed for tumor development.[21] The MEN-1 syndrome often exhibits tumors of parathyroid glands, anterior pituitary, endocrine pancreas, and endocrine duodenum. Less frequently, neuroendocrine tumors of lung, thymus, and stomach or non-endocrine tumors such as lipomas, angiofibromas, and ependymomas are observed neoplasms.[22]

In a study of 12 sporadic carcinoid tumors of the lung, five cases involved inactivation of both copies of the MEN1 gene. Of the five carcinoids, three were atypical and two were typical. The two typical carcinoids were characterized by a rapid proliferative rate with a higher mitotic index and stronger Ki67 positivity than the other typical carcinoids in the study. Consequently, the carcinoid tumors with MEN1 gene inactivation in the study were considered to be characterized by more aggressive molecular and histopathological features than those without MEN1 gene alterations.[23]

Interactions edit

MEN1 has been shown to interact with:

References edit

  1. ^ a b c GRCh38: Ensembl release 89: ENSG00000133895 - Ensembl, May 2017
  2. ^ a b c GRCm38: Ensembl release 89: ENSMUSG00000024947 - Ensembl, May 2017
  3. ^ "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. ^ "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. ^ a b "Entrez Gene: MEN1 multiple endocrine neoplasia I".
  6. ^ a b Thakker RV (June 2010). "Multiple endocrine neoplasia type 1 (MEN1)". Best Practice & Research. Clinical Endocrinology & Metabolism. 24 (3): 355–70. doi:10.1016/j.beem.2010.07.003. PMID 20833329.
  7. ^ Byström C, Larsson C, Blomberg C, Sandelin K, Falkmer U, Skogseid B, Oberg K, Werner S, Nordenskjöld M (March 1990). "Localization of the MEN1 gene to a small region within chromosome 11q13 by deletion mapping in tumors". Proceedings of the National Academy of Sciences of the United States of America. 87 (5): 1968–72. Bibcode:1990PNAS...87.1968B. doi:10.1073/pnas.87.5.1968. PMC 53606. PMID 1968641.
  8. ^ Chandrasekharappa SC, Guru SC, Manickam P, Olufemi SE, Collins FS, Emmert-Buck MR, Debelenko LV, Zhuang Z, Lubensky IA, Liotta LA, Crabtree JS, Wang Y, Roe BA, Weisemann J, Boguski MS, Agarwal SK, Kester MB, Kim YS, Heppner C, Dong Q, Spiegel AM, Burns AL, Marx SJ (April 1997). "Positional cloning of the gene for multiple endocrine neoplasia-type 1". Science. 276 (5311): 404–7. doi:10.1126/science.276.5311.404. PMID 9103196.
  9. ^ Agarwal SK, Lee Burns A, Sukhodolets KE, Kennedy PA, Obungu VH, Hickman AB, Mullendore ME, Whitten I, Skarulis MC, Simonds WF, Mateo C, Crabtree JS, Scacheri PC, Ji Y, Novotny EA, Garrett-Beal L, Ward JM, Libutti SK, Richard Alexander H, Cerrato A, Parisi MJ, Santa Anna-A S, Oliver B, Chandrasekharappa SC, Collins FS, Spiegel AM, Marx SJ (April 2004). "Molecular pathology of the MEN1 gene". Annals of the New York Academy of Sciences. 1014 (1): 189–98. Bibcode:2004NYASA1014..189A. doi:10.1196/annals.1294.020. PMID 15153434. S2CID 27333205.
  10. ^ Jyotsna VP, Malik E, Birla S, Sharma A (2015-01-01). "Novel MEN 1 gene findings in rare sporadic insulinoma--a case control study". BMC Endocrine Disorders. 15: 44. doi:10.1186/s12902-015-0041-2. PMC 4549893. PMID 26307114.
  11. ^ Chandrasekharappa SC, Guru SC, Manickam P, Olufemi SE, Collins FS, Emmert-Buck MR, Debelenko LV, Zhuang Z, Lubensky IA, Liotta LA, Crabtree JS, Wang Y, Roe BA, Weisemann J, Boguski MS, Agarwal SK, Kester MB, Kim YS, Heppner C, Dong Q, Spiegel AM, Burns AL, Marx SJ (April 1997). "Positional cloning of the gene for multiple endocrine neoplasia-type 1". Science. 276 (5311): 404–7. doi:10.1126/science.276.5311.404. PMID 9103196.
  12. ^ Schussheim DH, Skarulis MC, Agarwal SK, Simonds WF, Burns AL, Spiegel AM, Marx SJ (2001-06-01). "Multiple endocrine neoplasia type 1: new clinical and basic findings". Trends in Endocrinology and Metabolism. 12 (4): 173–8. doi:10.1016/s1043-2760(00)00372-6. PMID 11295574. S2CID 32447772.
  13. ^ Thakker RV (April 2014). "Multiple endocrine neoplasia type 1 (MEN1) and type 4 (MEN4)". Molecular and Cellular Endocrinology. 386 (1–2): 2–15. doi:10.1016/j.mce.2013.08.002. PMC 4082531. PMID 23933118.
  14. ^ Friedman E, Sakaguchi K, Bale AE, Falchetti A, Streeten E, Zimering MB, Weinstein LS, McBride WO, Nakamura Y, Brandi ML (July 1989). "Clonality of parathyroid tumors in familial multiple endocrine neoplasia type 1". The New England Journal of Medicine. 321 (4): 213–8. doi:10.1056/nejm198907273210402. PMID 2568586.
  15. ^ Lemos MC, Thakker RV (January 2008). "Multiple endocrine neoplasia type 1 (MEN1): analysis of 1336 mutations reported in the first decade following identification of the gene". Human Mutation. 29 (1): 22–32. doi:10.1002/humu.20605. PMID 17879353. S2CID 394253.
  16. ^ Guru SC, Manickam P, Crabtree JS, Olufemi SE, Agarwal SK, Debelenko LV (June 1998). "Identification and characterization of the multiple endocrine neoplasia type 1 (MEN1) gene". Journal of Internal Medicine. 243 (6): 433–9. doi:10.1046/j.1365-2796.1998.00346.x. PMID 9681840. S2CID 23149408.
  17. ^ Agarwal SK, Guru SC, Heppner C, Erdos MR, Collins RM, Park SY, Saggar S, Chandrasekharappa SC, Collins FS, Spiegel AM, Marx SJ, Burns AL (January 1999). "Menin interacts with the AP1 transcription factor JunD and represses JunD-activated transcription". Cell. 96 (1): 143–52. doi:10.1016/s0092-8674(00)80967-8. PMID 9989505. S2CID 18116746.
  18. ^ Yaguchi H, Ohkura N, Tsukada T, Yamaguchi K (2002). "Menin, the multiple endocrine neoplasia type 1 gene product, exhibits GTP-hydrolyzing activity in the presence of the tumor metastasis suppressor nm23". The Journal of Biological Chemistry. 277 (41): 38197–204. doi:10.1074/jbc.M204132200. PMID 12145286.
  19. ^ Zhuang Z, Ezzat SZ, Vortmeyer AO, Weil R, Oldfield EH, Park WS, Pack S, Huang S, Agarwal SK, Guru SC, Manickam P, Debelenko LV, Kester MB, Olufemi SE, Heppner C, Crabtree JS, Burns AL, Spiegel AM, Marx SJ, Chandrasekharappa SC, Collins FS, Emmert-Buck MR, Liotta LA, Asa SL, Lubensky IA (December 1997). "Mutations of the MEN1 tumor suppressor gene in pituitary tumors". Cancer Research. 57 (24): 5446–51. PMID 9407947.
  20. ^ Knudson AG (December 1993). "Antioncogenes and human cancer". Proceedings of the National Academy of Sciences of the United States of America. 90 (23): 10914–21. Bibcode:1993PNAS...9010914K. doi:10.1073/pnas.90.23.10914. PMC 47892. PMID 7902574.
  21. ^ Marx SJ, Agarwal SK, Kester MB, Heppner C, Kim YS, Emmert-Buck MR, Debelenko LV, Lubensky IA, Zhuang Z, Guru SC, Manickam P, Olufemi SE, Skarulis MC, Doppman JL, Alexander RH, Liotta LA, Collins FS, Chandrasekharappa SC, Spiegel AM, Burns AL (June 1998). "Germline and somatic mutation of the gene for multiple endocrine neoplasia type 1 (MEN1)". Journal of Internal Medicine. 243 (6): 447–53. doi:10.1046/j.1365-2796.1998.00348.x. PMID 9681842. S2CID 20132064.
  22. ^ Metz DC, Jensen RT, Bale AE, Skarulis MC, Eastman RC, Nieman L, Norton JA, Friedman E, Larsson C, Amorosi A, Brandi ML, Marx SJ (1994). "Multiple endocrine neoplasia type I. Clinical features and management". In Bilezikian JP, Levine MA, Marcus, R (eds.). The Parathyroids. New York: Raven Press Publishing Co. pp. 591–646.
  23. ^ Debelenko LV, Brambilla E, Agarwal SK, Swalwell JI, Kester MB, Lubensky IA, Zhuang Z, Guru SC, Manickam P, Olufemi SE, Chandrasekharappa SC, Crabtree JS, Kim YS, Heppner C, Burns AL, Spiegel AM, Marx SJ, Liotta LA, Collins FS, Travis WD, Emmert-Buck MR (December 1997). "Identification of MEN1 gene mutations in sporadic carcinoid tumors of the lung". Human Molecular Genetics. 6 (13): 2285–90. doi:10.1093/hmg/6.13.2285. PMID 9361035.
  24. ^ Jin S, Mao H, Schnepp RW, Sykes SM, Silva AC, D'Andrea AD, Hua X (July 2003). "Menin associates with FANCD2, a protein involved in repair of DNA damage". Cancer Research. 63 (14): 4204–10. PMID 12874027.
  25. ^ a b Heppner C, Bilimoria KY, Agarwal SK, Kester M, Whitty LJ, Guru SC, Chandrasekharappa SC, Collins FS, Spiegel AM, Marx SJ, Burns AL (August 2001). "The tumor suppressor protein menin interacts with NF-kappaB proteins and inhibits NF-kappaB-mediated transactivation". Oncogene. 20 (36): 4917–25. doi:10.1038/sj.onc.1204529. PMID 11526476.
  26. ^ Agarwal SK, Guru SC, Heppner C, Erdos MR, Collins RM, Park SY, Saggar S, Chandrasekharappa SC, Collins FS, Spiegel AM, Marx SJ, Burns AL (January 1999). "Menin interacts with the AP1 transcription factor JunD and represses JunD-activated transcription". Cell. 96 (1): 143–52. doi:10.1016/S0092-8674(00)80967-8. PMID 9989505. S2CID 18116746.
  27. ^ Yokoyama A, Wang Z, Wysocka J, Sanyal M, Aufiero DJ, Kitabayashi I, Herr W, Cleary ML (July 2004). "Leukemia proto-oncoprotein MLL forms a SET1-like histone methyltransferase complex with menin to regulate Hox gene expression". Molecular and Cellular Biology. 24 (13): 5639–49. doi:10.1128/MCB.24.13.5639-5649.2004. PMC 480881. PMID 15199122.
  28. ^ Sukhodolets KE, Hickman AB, Agarwal SK, Sukhodolets MV, Obungu VH, Novotny EA, Crabtree JS, Chandrasekharappa SC, Collins FS, Spiegel AM, Burns AL, Marx SJ (January 2003). "The 32-kilodalton subunit of replication protein A interacts with menin, the product of the MEN1 tumor suppressor gene". Molecular and Cellular Biology. 23 (2): 493–509. doi:10.1128/MCB.23.2.493-509.2003. PMC 151531. PMID 12509449.
  29. ^ Lopez-Egido J, Cunningham J, Berg M, Oberg K, Bongcam-Rudloff E, Gobl A (August 2002). "Menin's interaction with glial fibrillary acidic protein and vimentin suggests a role for the intermediate filament network in regulating menin activity". Experimental Cell Research. 278 (2): 175–83. doi:10.1006/excr.2002.5575. PMID 12169273.

Further reading edit

  • Tsukada T, Yamaguchi K, Kameya T (2002). "The MEN1 gene and associated diseases: an update". Endocrine Pathology. 12 (3): 259–73. doi:10.1385/EP:12:3:259. PMID 11740047. S2CID 30681290.
  • Kong C, Ellard S, Johnston C, Farid NR (November 2001). "Multiple endocrine neoplasia type 1Burin from Mauritius: a novel MEN1 mutation". Journal of Endocrinological Investigation. 24 (10): 806–10. doi:10.1007/bf03343931. PMID 11765051. S2CID 71097157.
  • Thakker RV (2002). "Multiple endocrine neoplasia". Hormone Research. 56 (Suppl 1): 67–72. doi:10.1159/000048138. PMID 11786689. S2CID 85195319.
  • Stowasser M, Gunasekera TG, Gordon RD (December 2001). "Familial varieties of primary aldosteronism". Clinical and Experimental Pharmacology & Physiology. 28 (12): 1087–90. doi:10.1046/j.1440-1681.2001.03574.x. PMID 11903322. S2CID 23091842.
  • Kameya T, Tsukada T, Yamaguchi K (2004). "Recent Advances in MEN 1 Gene Study for Pituitary Tumor Pathogenesis". Recent advances in MEN1 gene study for pituitary tumor pathogenesis. Frontiers of Hormone Research. Vol. 32. pp. 265–91. doi:10.1159/000079050. ISBN 3-8055-7740-0. PMID 15281352.
  • Balogh K, Rácz K, Patócs A, Hunyady L (November 2006). "Menin and its interacting proteins: elucidation of menin function". Trends in Endocrinology and Metabolism. 17 (9): 357–64. doi:10.1016/j.tem.2006.09.004. PMID 16997566. S2CID 8063335.
  • Lytras A, Tolis G (2006). "Growth hormone-secreting tumors: genetic aspects and data from animal models". Neuroendocrinology. 83 (3–4): 166–78. doi:10.1159/000095525. PMID 17047380. S2CID 45606794.

External links edit

  • GeneReviews/NIH/NCBI/UW entry on Multiple Endocrine Neoplasia Type 1
  • MEN1 gene variant database
  • MEN1+protein,+human at the U.S. National Library of Medicine Medical Subject Headings (MeSH)
  • Overview of all the structural information available in the PDB for UniProt: O00255 (Menin) at the PDBe-KB.

men1, disease, multiple, endocrine, neoplasia, type, menin, protein, that, humans, encoded, gene, menin, putative, tumor, suppressor, associated, with, multiple, endocrine, neoplasia, type, syndrome, autosomal, dominant, inheritance, variations, gene, cause, p. For the disease MEN 1 see Multiple endocrine neoplasia type 1 Menin is a protein that in humans is encoded by the MEN1 gene 5 Menin is a putative tumor suppressor associated with multiple endocrine neoplasia type 1 MEN 1 syndrome and has autosomal dominant inheritance 6 Variations in the MEN1 gene can cause pituitary adenomas hyperparathyroidism pancreatic neuroendocrine tumors gastrinoma and adrenocortical cancers MEN1Available structuresPDBOrtholog search PDBe RCSBList of PDB id codes3U84 3U85 3U86 3U88 4GPQ 4GQ3 4GQ4 4I80 4OG3 4OG4 4OG5 4OG6 4OG7 4OG8 4X5Y 4X5Z 5DDF 5DD9 5DDA 5DDE 5DDB 5DDD 5DDC 5DB0 5DB3 5DB1 5DB2IdentifiersAliasesMEN1 MEAI SCG2 menin 1External IDsOMIM 613733 MGI 1316736 HomoloGene 7418 GeneCards MEN1Gene location Human Chr Chromosome 11 human 1 Band11q13 1Start64 803 510 bp 1 End64 811 294 bp 1 Gene location Mouse Chr Chromosome 19 mouse 2 Band19 19 AStart6 385 009 bp 2 End6 390 921 bp 2 RNA expression patternBgeeHumanMouse ortholog Top expressed inright adrenal glandganglionic eminenceright lobe of liverright lobe of thyroid glandleft adrenal glandstromal cell of endometriumleft lobe of thyroid glandanterior pituitarybody of stomachskin of abdomenTop expressed inganglionic eminenceyolk sacadrenal glandmesencephalonneural tuberhombencephalonthymuscerebellumsecondary oocyteproximal tubuleMore reference expression dataBioGPSMore reference expression dataGene ontologyMolecular functionDNA binding protein macromolecule adaptor activity R SMAD binding protein N terminus binding chromatin binding histone lysine N methyltransferase activity protein binding four way junction DNA binding Y form DNA binding double stranded DNA bindingCellular componentcytoplasm cytosol nuclear matrix nucleoplasm chromatin cleavage furrow nucleus histone methyltransferase complex endoplasmic reticulum lumen protein containing complexBiological processnegative regulation of cell substrate adhesion negative regulation of protein phosphorylation positive regulation of transforming growth factor beta receptor signaling pathway regulation of transcription DNA templated negative regulation of cyclin dependent protein serine threonine kinase activity negative regulation of telomerase activity negative regulation of cell cycle regulation of activin receptor signaling pathway negative regulation of transcription by RNA polymerase II negative regulation of DNA binding transcription factor activity negative regulation of osteoblast differentiation negative regulation of cell cycle G1 S phase transition MAPK cascade cellular response to DNA damage stimulus negative regulation of JNK cascade brain development cellular response to peptide hormone stimulus decidualization negative regulation of epithelial cell proliferation mitotic cell cycle osteoblast development positive regulation of protein binding negative regulation of transcription DNA templated response to gamma radiation response to UV type B pancreatic cell differentiation DNA repair regulation of type B pancreatic cell proliferation cellular response to glucose stimulus positive regulation of transcription by RNA polymerase II negative regulation of cell population proliferation response to transforming growth factor beta histone lysine methylation beta catenin TCF complex assembly transcription DNA templated post translational protein modification chromatin organizationSources Amigo QuickGOOrthologsSpeciesHumanMouseEntrez422117283EnsemblENSG00000133895ENSMUSG00000024947UniProtO00255O88559RefSeq mRNA NM 000244NM 130799NM 130800NM 130801NM 130802NM 130803NM 130804NM 001370251NM 001370259NM 001370260NM 001370261NM 001370262NM 001370263NM 001168488NM 001168489NM 001168490NM 008583RefSeq protein NP 000235NP 570711NP 570712NP 570713NP 570714NP 570715NP 570716NP 001357180NP 001357188NP 001357189NP 001357190NP 001357191NP 001357192NP 001161960NP 001161961NP 001161962NP 032609Location UCSC Chr 11 64 8 64 81 MbChr 19 6 39 6 39 MbPubMed search 3 4 WikidataView Edit HumanView Edit MouseIn vitro studies have shown that menin is localized to the nucleus possesses two functional nuclear localization signals and inhibits transcriptional activation by JunD However the function of this protein is not known Two messages have been detected on northern blots but the larger message has not been characterized Two variants of the shorter transcript have been identified where alternative splicing affects the coding sequence Five variants where alternative splicing takes place in the 5 UTR have also been identified 5 Contents 1 History 2 Genomics 3 Clinical implications 4 Role in cancer 5 Interactions 6 References 7 Further reading 8 External linksHistory editIn 1988 researchers at Uppsala University Hospital and the Karolinska Institute in Stockholm mapped the MEN1 gene to the long arm of chromosome 11 7 The gene was finally cloned in 1997 8 Genomics editThe gene is located on long arm of chromosome 11 11q13 between base pairs 64 570 985 and 64 578 765 It has 10 exons and encodes a 610 amino acid protein Over 1300 mutations have been reported to date 2010 The majority gt 70 of these are predicted to lead to truncated forms are scattered throughout the gene Four c 249 252delGTCT deletion at codons 83 84 c 1546 1547insC insertion at codon 516 c 1378C gt T Arg460Ter and c 628 631delACAG deletion at codons 210 211 have been reported to occur in 4 5 2 7 2 6 and 2 5 of families 6 Clinical implications editThe MEN1 phenotype is inherited via an autosomal dominant pattern and is associated with neoplasms of the pituitary gland the parathyroid gland and the pancreas the 3 P s While these neoplasias are often benign in contrast to tumours occurring in MEN2A they are adenomas and therefore produce endocrine phenotypes Pancreatic presentations of the MEN1 phenotype may manifest as Zollinger Ellison syndrome MEN1 pituitary tumours are adenomas of anterior cells typically prolactinomas or growth hormone secreting Pancreatic tumours involve the islet cells giving rise to gastrinomas or insulinomas In rare cases adrenal cortex tumours are also seen Role in cancer editMost germline or somatic mutations in the MEN1 gene predict truncation or absence of encoded menin resulting in the inability of MEN1 to act as a tumor suppressor gene 9 Such mutations in MEN1 have been associated with defective binding of encoded menin to proteins implicated in genetic and epigenetic mechanisms 10 Menin is a 621 amino acid protein associated with insulinomas 11 which acts as an adapter while also interacting with partner proteins involved in vital cell activities such as transcriptional regulation cell division cell proliferation and genome stability Insulinomas are neuroendocrine tumors of the pancreas with an incidence of 0 4 citation needed which usually are benign solitary tumors but 5 12 of cases have distant metastasis at diagnosis 12 These familial MEN 1 and sporadic tumors may arise either due to loss of heterozygosity or the chromosome region 11q13 where MEN1 is located or due to presence of mutations in the gene 13 14 MEN1 mutations comprise mostly frameshift deletions or insertions followed by nonsense missense splice site mutations and either part or complete gene deletions resulting in disease pathology 15 Frameshift and nonsense mutations result in a supposed inactive and truncated menin protein while splice site mutations result in incorrectly spliced mRNA Missense mutations of MEN1 are especially important as they result in a change to crucial amino acids needed in order to bind and interact with other proteins and molecules As menin is located predominantly in the nucleus 16 these mutations can impact the stability of the cell and may further affect functional activity or expression levels of the protein Studies have also shown that single amino acid changes in genes involved in oncogenic disorders may result in proteolytic degradation leading to loss of function and reduced stability of the mutant protein a common mechanism for inactivating tumor suppressor gene products 17 18 MEN1 gene mutations and deletions also play a role in the development of hereditary and a subgroup of sporadic pituitary adenomas and were detected in approximately 5 of sporadic pituitary adenomas 19 Consequently alterations of the gene represent a candidate pathogenetic mechanism of pituitary tumorigenesis especially when considered in terms of interactions with other proteins growth factors oncogenes play a rule in tumorigenesis Although the exact function of MEN1 is not known the Knudson two hit hypothesis provides strong evidence that it is a tumor suppressor gene Familial loss of one copy of MEN1 is seen in association with MEN 1 syndrome Tumor suppressor carcinogenesis follows Knudson s two hit model 20 The first hit is a heterozygous MEN1 germline mutation either developed in an early embryonic stage and consequently present in all cells at birth for the sporadic cases or inherited from one parent in a familial case The second hit is a MEN1 somatic mutation oftentimes a large deletion occurring in the predisposed endocrine cell and providing cells with the survival advantaged needed for tumor development 21 The MEN 1 syndrome often exhibits tumors of parathyroid glands anterior pituitary endocrine pancreas and endocrine duodenum Less frequently neuroendocrine tumors of lung thymus and stomach or non endocrine tumors such as lipomas angiofibromas and ependymomas are observed neoplasms 22 In a study of 12 sporadic carcinoid tumors of the lung five cases involved inactivation of both copies of the MEN1 gene Of the five carcinoids three were atypical and two were typical The two typical carcinoids were characterized by a rapid proliferative rate with a higher mitotic index and stronger Ki67 positivity than the other typical carcinoids in the study Consequently the carcinoid tumors with MEN1 gene inactivation in the study were considered to be characterized by more aggressive molecular and histopathological features than those without MEN1 gene alterations 23 Interactions editMEN1 has been shown to interact with FANCD2 24 GFAP 25 JunD 26 NFKB1 25 MLL 27 RPA2 28 and VIM 29 References edit a b c GRCh38 Ensembl release 89 ENSG00000133895 Ensembl May 2017 a b c GRCm38 Ensembl release 89 ENSMUSG00000024947 Ensembl May 2017 Human PubMed Reference National Center for Biotechnology Information U S National Library of Medicine Mouse PubMed Reference National Center for Biotechnology Information U S National Library of Medicine a b Entrez Gene MEN1 multiple endocrine neoplasia I a b Thakker RV June 2010 Multiple endocrine neoplasia type 1 MEN1 Best Practice amp Research Clinical Endocrinology amp Metabolism 24 3 355 70 doi 10 1016 j beem 2010 07 003 PMID 20833329 Bystrom C Larsson C Blomberg C Sandelin K Falkmer U Skogseid B Oberg K Werner S Nordenskjold M March 1990 Localization of the MEN1 gene to a small region within chromosome 11q13 by deletion mapping in tumors Proceedings of the National Academy of Sciences of the United States of America 87 5 1968 72 Bibcode 1990PNAS 87 1968B doi 10 1073 pnas 87 5 1968 PMC 53606 PMID 1968641 Chandrasekharappa SC Guru SC Manickam P Olufemi SE Collins FS Emmert Buck MR Debelenko LV Zhuang Z Lubensky IA Liotta LA Crabtree JS Wang Y Roe BA Weisemann J Boguski MS Agarwal SK Kester MB Kim YS Heppner C Dong Q Spiegel AM Burns AL Marx SJ April 1997 Positional cloning of the gene for multiple endocrine neoplasia type 1 Science 276 5311 404 7 doi 10 1126 science 276 5311 404 PMID 9103196 Agarwal SK Lee Burns A Sukhodolets KE Kennedy PA Obungu VH Hickman AB Mullendore ME Whitten I Skarulis MC Simonds WF Mateo C Crabtree JS Scacheri PC Ji Y Novotny EA Garrett Beal L Ward JM Libutti SK Richard Alexander H Cerrato A Parisi MJ Santa Anna A S Oliver B Chandrasekharappa SC Collins FS Spiegel AM Marx SJ April 2004 Molecular pathology of the MEN1 gene Annals of the New York Academy of Sciences 1014 1 189 98 Bibcode 2004NYASA1014 189A doi 10 1196 annals 1294 020 PMID 15153434 S2CID 27333205 Jyotsna VP Malik E Birla S Sharma A 2015 01 01 Novel MEN 1 gene findings in rare sporadic insulinoma a case control study BMC Endocrine Disorders 15 44 doi 10 1186 s12902 015 0041 2 PMC 4549893 PMID 26307114 Chandrasekharappa SC Guru SC Manickam P Olufemi SE Collins FS Emmert Buck MR Debelenko LV Zhuang Z Lubensky IA Liotta LA Crabtree JS Wang Y Roe BA Weisemann J Boguski MS Agarwal SK Kester MB Kim YS Heppner C Dong Q Spiegel AM Burns AL Marx SJ April 1997 Positional cloning of the gene for multiple endocrine neoplasia type 1 Science 276 5311 404 7 doi 10 1126 science 276 5311 404 PMID 9103196 Schussheim DH Skarulis MC Agarwal SK Simonds WF Burns AL Spiegel AM Marx SJ 2001 06 01 Multiple endocrine neoplasia type 1 new clinical and basic findings Trends in Endocrinology and Metabolism 12 4 173 8 doi 10 1016 s1043 2760 00 00372 6 PMID 11295574 S2CID 32447772 Thakker RV April 2014 Multiple endocrine neoplasia type 1 MEN1 and type 4 MEN4 Molecular and Cellular Endocrinology 386 1 2 2 15 doi 10 1016 j mce 2013 08 002 PMC 4082531 PMID 23933118 Friedman E Sakaguchi K Bale AE Falchetti A Streeten E Zimering MB Weinstein LS McBride WO Nakamura Y Brandi ML July 1989 Clonality of parathyroid tumors in familial multiple endocrine neoplasia type 1 The New England Journal of Medicine 321 4 213 8 doi 10 1056 nejm198907273210402 PMID 2568586 Lemos MC Thakker RV January 2008 Multiple endocrine neoplasia type 1 MEN1 analysis of 1336 mutations reported in the first decade following identification of the gene Human Mutation 29 1 22 32 doi 10 1002 humu 20605 PMID 17879353 S2CID 394253 Guru SC Manickam P Crabtree JS Olufemi SE Agarwal SK Debelenko LV June 1998 Identification and characterization of the multiple endocrine neoplasia type 1 MEN1 gene Journal of Internal Medicine 243 6 433 9 doi 10 1046 j 1365 2796 1998 00346 x PMID 9681840 S2CID 23149408 Agarwal SK Guru SC Heppner C Erdos MR Collins RM Park SY Saggar S Chandrasekharappa SC Collins FS Spiegel AM Marx SJ Burns AL January 1999 Menin interacts with the AP1 transcription factor JunD and represses JunD activated transcription Cell 96 1 143 52 doi 10 1016 s0092 8674 00 80967 8 PMID 9989505 S2CID 18116746 Yaguchi H Ohkura N Tsukada T Yamaguchi K 2002 Menin the multiple endocrine neoplasia type 1 gene product exhibits GTP hydrolyzing activity in the presence of the tumor metastasis suppressor nm23 The Journal of Biological Chemistry 277 41 38197 204 doi 10 1074 jbc M204132200 PMID 12145286 Zhuang Z Ezzat SZ Vortmeyer AO Weil R Oldfield EH Park WS Pack S Huang S Agarwal SK Guru SC Manickam P Debelenko LV Kester MB Olufemi SE Heppner C Crabtree JS Burns AL Spiegel AM Marx SJ Chandrasekharappa SC Collins FS Emmert Buck MR Liotta LA Asa SL Lubensky IA December 1997 Mutations of the MEN1 tumor suppressor gene in pituitary tumors Cancer Research 57 24 5446 51 PMID 9407947 Knudson AG December 1993 Antioncogenes and human cancer Proceedings of the National Academy of Sciences of the United States of America 90 23 10914 21 Bibcode 1993PNAS 9010914K doi 10 1073 pnas 90 23 10914 PMC 47892 PMID 7902574 Marx SJ Agarwal SK Kester MB Heppner C Kim YS Emmert Buck MR Debelenko LV Lubensky IA Zhuang Z Guru SC Manickam P Olufemi SE Skarulis MC Doppman JL Alexander RH Liotta LA Collins FS Chandrasekharappa SC Spiegel AM Burns AL June 1998 Germline and somatic mutation of the gene for multiple endocrine neoplasia type 1 MEN1 Journal of Internal Medicine 243 6 447 53 doi 10 1046 j 1365 2796 1998 00348 x PMID 9681842 S2CID 20132064 Metz DC Jensen RT Bale AE Skarulis MC Eastman RC Nieman L Norton JA Friedman E Larsson C Amorosi A Brandi ML Marx SJ 1994 Multiple endocrine neoplasia type I Clinical features and management In Bilezikian JP Levine MA Marcus R eds The Parathyroids New York Raven Press Publishing Co pp 591 646 Debelenko LV Brambilla E Agarwal SK Swalwell JI Kester MB Lubensky IA Zhuang Z Guru SC Manickam P Olufemi SE Chandrasekharappa SC Crabtree JS Kim YS Heppner C Burns AL Spiegel AM Marx SJ Liotta LA Collins FS Travis WD Emmert Buck MR December 1997 Identification of MEN1 gene mutations in sporadic carcinoid tumors of the lung Human Molecular Genetics 6 13 2285 90 doi 10 1093 hmg 6 13 2285 PMID 9361035 Jin S Mao H Schnepp RW Sykes SM Silva AC D Andrea AD Hua X July 2003 Menin associates with FANCD2 a protein involved in repair of DNA damage Cancer Research 63 14 4204 10 PMID 12874027 a b Heppner C Bilimoria KY Agarwal SK Kester M Whitty LJ Guru SC Chandrasekharappa SC Collins FS Spiegel AM Marx SJ Burns AL August 2001 The tumor suppressor protein menin interacts with NF kappaB proteins and inhibits NF kappaB mediated transactivation Oncogene 20 36 4917 25 doi 10 1038 sj onc 1204529 PMID 11526476 Agarwal SK Guru SC Heppner C Erdos MR Collins RM Park SY Saggar S Chandrasekharappa SC Collins FS Spiegel AM Marx SJ Burns AL January 1999 Menin interacts with the AP1 transcription factor JunD and represses JunD activated transcription Cell 96 1 143 52 doi 10 1016 S0092 8674 00 80967 8 PMID 9989505 S2CID 18116746 Yokoyama A Wang Z Wysocka J Sanyal M Aufiero DJ Kitabayashi I Herr W Cleary ML July 2004 Leukemia proto oncoprotein MLL forms a SET1 like histone methyltransferase complex with menin to regulate Hox gene expression Molecular and Cellular Biology 24 13 5639 49 doi 10 1128 MCB 24 13 5639 5649 2004 PMC 480881 PMID 15199122 Sukhodolets KE Hickman AB Agarwal SK Sukhodolets MV Obungu VH Novotny EA Crabtree JS Chandrasekharappa SC Collins FS Spiegel AM Burns AL Marx SJ January 2003 The 32 kilodalton subunit of replication protein A interacts with menin the product of the MEN1 tumor suppressor gene Molecular and Cellular Biology 23 2 493 509 doi 10 1128 MCB 23 2 493 509 2003 PMC 151531 PMID 12509449 Lopez Egido J Cunningham J Berg M Oberg K Bongcam Rudloff E Gobl A August 2002 Menin s interaction with glial fibrillary acidic protein and vimentin suggests a role for the intermediate filament network in regulating menin activity Experimental Cell Research 278 2 175 83 doi 10 1006 excr 2002 5575 PMID 12169273 Further reading editTsukada T Yamaguchi K Kameya T 2002 The MEN1 gene and associated diseases an update Endocrine Pathology 12 3 259 73 doi 10 1385 EP 12 3 259 PMID 11740047 S2CID 30681290 Kong C Ellard S Johnston C Farid NR November 2001 Multiple endocrine neoplasia type 1Burin from Mauritius a novel MEN1 mutation Journal of Endocrinological Investigation 24 10 806 10 doi 10 1007 bf03343931 PMID 11765051 S2CID 71097157 Thakker RV 2002 Multiple endocrine neoplasia Hormone Research 56 Suppl 1 67 72 doi 10 1159 000048138 PMID 11786689 S2CID 85195319 Stowasser M Gunasekera TG Gordon RD December 2001 Familial varieties of primary aldosteronism Clinical and Experimental Pharmacology amp Physiology 28 12 1087 90 doi 10 1046 j 1440 1681 2001 03574 x PMID 11903322 S2CID 23091842 Kameya T Tsukada T Yamaguchi K 2004 Recent Advances in MEN 1 Gene Study for Pituitary Tumor Pathogenesis Recent advances in MEN1 gene study for pituitary tumor pathogenesis Frontiers of Hormone Research Vol 32 pp 265 91 doi 10 1159 000079050 ISBN 3 8055 7740 0 PMID 15281352 Balogh K Racz K Patocs A Hunyady L November 2006 Menin and its interacting proteins elucidation of menin function Trends in Endocrinology and Metabolism 17 9 357 64 doi 10 1016 j tem 2006 09 004 PMID 16997566 S2CID 8063335 Lytras A Tolis G 2006 Growth hormone secreting tumors genetic aspects and data from animal models Neuroendocrinology 83 3 4 166 78 doi 10 1159 000095525 PMID 17047380 S2CID 45606794 External links editGeneReviews NIH NCBI UW entry on Multiple Endocrine Neoplasia Type 1 MEN1 gene variant database MEN1 protein human at the U S National Library of Medicine Medical Subject Headings MeSH Overview of all the structural information available in the PDB for UniProt O00255 Menin at the PDBe KB Retrieved from https en wikipedia org w index php title MEN1 amp oldid 1188076095, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.