Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Oct 1.
Published in final edited form as: Endocr Relat Cancer. 2017 Oct;24(10):T119–T134. doi: 10.1530/ERC-17-0199

The future: genetics advances in MEN1 therapeutic approaches and management strategies

Sunita K Agarwal 1
PMCID: PMC5679100  NIHMSID: NIHMS899047  PMID: 28899949

Abstract

The identification of the multiple endocrine neoplasia type 1 (MEN1) gene in 1997 has shown that germline heterozygous mutations in the MEN1 gene located on chromosome 11q13, predisposes to the development of tumors in the MEN1 syndrome. Tumor development occurs upon loss of the remaining normal copy of the MEN1 gene in MEN1-target tissues. Therefore, MEN1 is a classic tumor suppressor gene in the context of MEN1. This tumor suppressor role of the protein encoded by the MEN1 gene, menin, holds true in mouse models with germline heterozygous Men1 loss wherein MEN1-associated tumors develop in adult mice after spontaneous loss of the remaining non-targeted copy of the Men1 gene. The availability of genetic testing for mutations in the MEN1 gene has become an essential part of the diagnosis and management of MEN1. Genetic testing is also helping to exclude mutation negative cases in MEN1 families from the burden of lifelong clinical screening. In the past 20 years, efforts of various groups world-wide have been directed at mutation analysis, molecular genetic studies, mouse models, gene expression studies, epigenetic regulation analysis, biochemical studies, and anti-tumor effects of candidate therapies in mouse models. This review will focus on the findings and advances from these studies to identify MEN1 germline and somatic mutations, the genetics of MEN1-related states, several protein partners of menin, the three-dimensional structure of menin, and menin-dependent target genes. The ongoing impact of all these studies on disease prediction, management and outcomes will continue in the years to come.

Keywords: Multiple Endocrine Neoplasia, Neuroendocrine, Parathyroid, Pituitary

Introduction

Multiple endocrine neoplasia type 1 (MEN1) is a rare tumor syndrome with an autosomal dominant pattern of inheritance with >95% penetrance by age 40–50 years (Online Mendelian Inheritance in Man, OMIM number: 131100). The approximate prevalence of MEN1 has been reported as 1 in 30,000 individuals with no apparent gender bias. The main characteristic clinical features of MEN1 include at least two of the following three endocrine tumors in an individual: multi-gland parathyroid adenomas, anterior pituitary adenomas and entero-pancreatic neuroendocrine tumors (gastrinoma/Zollinger-Ellison syndrome and pancreatic neuroendocrine tumors). These characteristics can present in a non-hereditary form with no family history of MEN1 (sporadic MEN1) or in several members of a family (familial MEN1) (Brandi, et al. 2001; Marx, et al. 1998; Thakker, et al. 2012). The penetrance of the main MEN1 tumors by age 50 years is as follows: the parathyroids (90–100%), entero-pancreatic neuroendocrine (30–70%), and anterior pituitary (30–40%) (Brandi et al. 2001; Thakker et al. 2012). In addition to these main endocrine tumors causing associated circulating hormone excess, MEN1 patients present with various hormone-secreting, hormone non-secreting, and non-endocrine tumors. These include adrenal cortical tumor; foregut carcinoids of the lung, thymus, or gastric enterochromaffin-like cells; skin lesions facial angiofibroma, truncal collagenoma, and lipoma; central nervous system tumors meningioma and ependymoma; and smooth muscle tumors leiomyomas (uterine in females, or in the esophagus) (Agarwal 2013; Brandi et al. 2001; Thakker et al. 2012). Malignant gastrinoma or foregut carcinoid tumors are the cause of death in approximately 25% of MEN1 patients (Dean, et al. 2000; Doherty, et al. 1998; Goudet, et al. 2010; Ito, et al. 2013). The various individual tumor types observed in the MEN1 syndrome also occur sporadically. The cloning of the gene causative for the MEN1 syndrome in 1997 has provided a unique opportunity to gain insights into the normal physiology and pathophysiology of the wide range of MEN1-associated cell types affected. This review covers the advances in molecular genetic studies and basic findings about the MEN1 syndrome and related states, the MEN1 gene and its protein product menin.

Positional cloning of the MEN1 gene and identification of inactivating mutations

Genetic mapping, segregation analysis of candidate markers in MEN1 families, and loss of heterozygosity (LOH) studies in MEN1-associated tumors led to the localization of the putative tumor suppressor gene that causes the MEN1 syndrome to a narrow interval on chromosome 11q13 near the PYGM locus (Bale, et al. 1989; Debelenko, et al. 1997; Emmert-Buck, et al. 1997; Friedman, et al. 1989; Larsson, et al. 1988; Nakamura, et al. 1989; Sawicki, et al. 1992; Thakker, et al. 1989). The polymorphic markers in the 11q13 region were useful in genetic tests to identify disease carriers among family members, however, they could not be used to diagnose MEN1 genetically in index cases.

Two different large collaborative groups used a positional cloning approach to sequence a MEN1-linked minimal interval that mapped at 11q13 to identify the MEN1 gene (Chandrasekharappa, et al. 1997; Guru, et al. 1997a; Guru, et al. 1997b; Lemmens, et al. 1997a; Lemmens, et al. 1997b). They showed that the MEN1 gene spans about 9000 bp of genomic DNA containing 10 exons, and transcribed into a 2.8 kb mRNA with the translational start codon (ATG) in exon-2 and the stop codon in exon-10. The protein product of the MEN1 gene consisting of 610 amino acids was named menin (GenBank Accession No.: U93236.1). Germline heterozygous inactivating mutations were found in the coding region of the MEN1 gene in index cases and affected family members, together with LOH for markers at the MEN1 gene locus in their tumors as expected for a causative tumor suppressor gene (Chandrasekharappa et al. 1997; Knudson 1993, 1971; Lemmens et al. 1997b).

It is important to note that there are two versions of menin in the Gene databases with 610 or 615 amino acids. The 615 amino acids version has an alternative splice site at the end of exon-2 that adds five amino acids after codon 148. While the mutations in most publications are written as per the 610 amino acids version of menin, most current databases have used the 615 amino acids version of menin to annotate MEN1 mutations. Therefore, the same version of menin must be used for mutation annotation to compare MEN1 mutations after amino acid 148.

Germline and somatic mutations in the MEN1 gene

Since 1997, germline DNA samples have been screened for mutations in the MEN1 gene that belong to cases suspected to have familial MEN1 or sporadic MEN1. Germline heterozygous mutations in the MEN1 gene are observed in 70–90% of familial MEN1 cases and the frequency of finding a de novo mutation is significantly lower in sporadic MEN1 cases (Agarwal, et al. 1997; Bassett, et al. 1998; Cardinal, et al. 2005; Cebrian, et al. 2003; de Laat, et al. 2016; Giraud, et al. 1998; Giusti, et al. 2017; Klein, et al. 2005; Sakurai, et al. 2012; Tham, et al. 2007). Over 1200 germline mutations in the MEN1 gene have been reported with no obvious genotype-phenotype correlation of specific mutations with the MEN1-associated tumor spectrum even among family members with the exact same mutation. Clustering of mutations is observed in some regions of exon-2 and exon-10 that have been attributed to the nature of the underlying repetitive nucleotides prone to DNA polymerase errors (Agarwal, et al. 1998; Bassett et al. 1998; Thakker 2010). Another observation of clustering of mutations at specific nucleotides in apparently unrelated families has been attributed to founder effects (Agarwal et al. 1998; Agarwal et al. 1997; Burgess, et al. 2000; Cardinal et al. 2005; Kytola, et al. 2001; Lourenco, et al. 2008; Olufemi, et al. 1998; Vierimaa, et al. 2007).

Two comprehensive studies have tabulated and analyzed germline MEN1 mutations published from 1997–2007 and 2007–2015 identifying 459 and 117 (total 576) unique mutations, respectively, in MEN1 patients (Concolino, et al. 2015; Lemos and Thakker 2008). These mutations are scattered over the entire coding region of MEN1 with no hot spots. The largest category of MEN1 germline mutations (69%) predict obvious pathologic consequence due to premature truncation of menin from nonsense mutations (14%) and frame-shift mutations (42%), or exon region deletion due to splicing defects (10.5%), or large deletions (2.5%) (Table 1) (Concolino et al. 2015). Missense mutations (25.5%) and single or few amino acids in-frame deletions or insertions (5.5%) do not predict obvious inactivation of menin, and their classification as benign or pathologic needs further investigation (Table 1) (Concolino et al. 2015; Lemos and Thakker 2008). For example, the pathologic nature of many of these missense and in-frame mutations could be verified based on their tracking with the disease in families. Seven common polymorphisms that constitute a synonymous or non-synonymous amino acid change in the menin coding region have also been reported in the MEN1 gene (Table 2).

Table 1.

MEN1 Germline Mutations

TYPE OF MUTATION PERCENT
Protein Truncation
  Nonsense 14.0
  Frameshift 42.0
  Splicing 10.5
  Large deletion 2.5
Missense 25.5
In-frame deletion/insertion 5.5

Table 2.

MEN1 Common Polymorphisms (ExAC Database Minor Allele Frequency >0.05%)

Genomic Location hg19
(dbSNP)
Codon
Change
Consequence Annotation Minor Allele
Frequency (%)
1 11:64577552 G/T (rs371192390) CTG/CTT p.Leu10Leu synonymous 0.07
2 11:64577147 C/T (rs61736636) AGC/AGT p.Ser145Ser synonymous 2.9
3 11:64575505 G/A (rs607969) CGG/CAG p.Arg171Gln non-synonymous 1.2
4 11:64572602 C/T (rs2071313) GAC/GAT p.Asp418Asp synonymous 39.3
5 11:64572560 G/A(rs138770431) CTG/CTA p.Leu432Leu synonymous 0.1
6 11:64572557 C/T (rs540012) CAC/CAT p.His433His synonymous 0.72
7 11:64572018 G/A (rs2959656) GCA/ACA p.Ala541Thr non-synonymous 6.2

ExAC: The Exome Aggregation Consortium database spans 60,706 unrelated individuals sequenced as part of various disease-specific and population genetic studies. dbSNP: Database of Single Nucleotide Polymorphisms. Minor allele frequency (MAF): Frequency of the less (or least) frequent allele in a population (>0.05% considered as a common variant).

The discovery of susceptibility genes for hereditary tumor syndromes can be informative for the identification of the genetic cause of their sporadic counterpart tumors (Marx and Simonds 2005). Indeed, somatic inactivating mutations in the MEN1 gene have been reported in sporadic tumors of the types observed in the MEN1 syndrome. In contrast to MEN1 where the 1st hit to the MEN1 gene is in the germline and the 2nd hit is somatic, in sporadic tumors the 2 hits to the MEN1 gene for biallelic inactivation are both somatic. Using targeted sequencing of MEN1 exons or whole exome sequencing approaches, the frequency of somatic MEN1 mutations reported in sporadic tumors is as follows: glucagonoma (60%), VIPoma (57%), non-functioning PNETs (44%), gastrinoma (38%), bronchial carcinoid (35%), parathyroid adenoma (35%), lipoma (28%) insulinoma (2–19%), angiofibroma (10%), anterior pituitary tumor (3.5%), and adrenocortical tumor (2%) (Scarpa, et al. 2017) (Cao, et al. 2013; Cromer, et al. 2012; Gortz, et al. 1999; Heppner, et al. 1997; Jiao, et al. 2011; Newey, et al. 2012; Thakker 2014; Zhuang, et al. 1997). Similar to the distribution of germline mutations, the somatic mutations are also spread over the entire coding region of MEN1 with no hot spots. The percentage of the types of somatic MEN1 mutations (nonsense, frame-shift, splicing, missense, and in-frame deletions or insertions) is also similar to the germline MEN1 mutations (Ref: COSMIC database, the "Catalogue of Somatic Mutations in Cancer”, Sanger Institute). Also, no obvious genotype-phenotype correlations have been identified for specific somatic mutations and the associated tumor types.

Genetic testing for mutations in the MEN1 gene

Currently, genetic testing of the MEN1 gene includes PCR-based screening for mutations in the coding region and splice junctions, and if a mutation is not identified then multiplex ligation probe amplification (MLPA) based screening is performed for the detection of large deletions of the MEN1 gene. Clinical practice consensus guidelines for MEN1 and related states developed by a panel of experts including physicians, surgeons and geneticists from international centers, have made recommendations for mutational analysis of the MEN1 gene (Brandi et al. 2001; Thakker et al. 2012). Before and after testing, all individuals should be provided genetic counseling. The tests must be performed in a clinical genetics laboratory that is accredited to perform MEN1 mutation analysis. Individuals who are the relatives of a patient with a known MEN1 mutation should be offered genetic testing prior to biochemical and radiological screening for the detection of MEN1-associated tumors, because the chance of inheriting the mutation is 50%, and the germline genetic testing would help to avoid the screening for tumors in mutation negative cases. Individuals who test positive for the known mutation in their family should be screened on an annual basis for the development of MEN1-associated tumors. For asymptomatic members of a family with a known MEN1 mutation, the guidelines recommend that MEN1 germline testing should be offered at the earliest opportunity, as early as age 5 years, because MEN1 manifestations have been observed in some children at age 5–10 years (Goudet, et al. 2015; Machens, et al. 2007; Stratakis, et al. 2000). MEN1 germline testing may be offered to symptomatic young patients (< 40 years) (with no family history) who present with an atypical phenotype such as an MEN1-associated tumor (for example, multi-gland hyperparathyroidism) and who may have not yet developed the full spectrum of MEN1-associated tumors. Also, the guidelines advise to offer genetic testing to symptomatic family members with a known MEN1 mutation in the family, as well as to index cases with two or more of the MEN1-associated endocrine tumors. This is because of the report of phenocopies wherein symptomatic individuals in MEN1 kindreds have tested negative for their known familial MEN1 mutation (Thakker et al. 2012).

MEN1-like disease from germline mutation in MEN1 or other susceptibility genes

MEN1-like cases and families have been reported who show incomplete clinical manifestations of MEN1 with as few as any one of the three main MEN1-associated endocrine tumors. Germline MEN1 mutations have been found in a few kindreds of the parathyroid-only disorder, familial isolated primary hyperparathyroidism (FIHP) (Concolino et al. 2015; Hannan, et al. 2008; Lemos and Thakker 2008; Simonds, et al. 2002). Among these germline mutations a higher frequency of MEN1 missense mutations have been observed but with no hot spots or genotype-phenotype correlations (Concolino et al. 2015; Lemos and Thakker 2008). Also, germline MEN1 mutations have been reported in five cases of apparently sporadic PNETs (with no family history or other tumors of MEN1) (Scarpa et al. 2017). However, MEN1 germline mutations have not been found in the pituitary-only disorder of familial isolated pituitary adenoma (FIPA).

Because the involvement of the MEN1 gene was excluded in cases with MEN1-like characteristics, efforts could be directed to identify susceptibility genes for such conditions. Identification of the susceptibility gene for the hyperparathyroidism-jaw tumor syndrome by a positional cloning approach has shown that germline mutations in the CDC73 gene are causative for parathyroid cancer (Carpten, et al. 2002; Shattuck, et al. 2003). An exome sequencing approach has shown that germline activating mutations in the GCM2 gene occur in 18% of FIHP kindreds (Guan, et al. 2016). Germline mutations in the AIP tumor suppressor gene identified by a positional cloning approach, which is also located on 11q13, occur in 20% of FIPA patients and in 30–50% of patients with familial acromegaly, and germline GPR101 microduplication or mutation occurs in patients with X-linked acrogigantism (X-LAG) (Daly, et al. 2007; Trivellin, et al. 2014; Vierimaa, et al. 2006). Identification of a homozygous germline mutation in the cyclin-dependent kinase inhibitor (CDKI) gene CDKN1B/p27 in a rat strain with combined MEN1 and MEN2 features (MENX) prompted the germline mutation analysis of this gene in human patients with MEN1-like features, and the identification of the MEN4 disorder (Pellegata, et al. 2006). Heterozygous germline mutations in p27 have been identified in 1–2% of MEN1-like cases (MEN4) who present with incomplete features of MEN1 such as primary hyperparathyroidism only, parathyroid and pituitary tumor, or MEN1 features (Pellegata 2012). No obvious genotype-phenotype correlation has been reported for MEN1 or MEN1-like features with p27 mutations. The CDKI genes belong to two families, the INK4 family (p15, p16, p18, and p19) and the Cip/Kip family (p21, p27, and p57). They are cell cycle regulators that inhibit specific cyclin-CDK complexes. Mutation analysis of the members of the INK4 and Cip/Kip family of CDKI genes in MEN1-like cases has shown that less than 1% carry probable p15, p18 or p21 heterozygous germline mutations (Agarwal, et al. 2009; Thakker 2014).

The MEN1 encoded protein menin and its functional characterization

The amino acid sequence of menin does not show homology to any known proteins. Menin is highly conserved in animal species, and there are no homologs of menin in yeast and nematodes. Menin is ubiquitously expressed, and detected at 67 kDa by western blot analysis. Studies of post-translation modifications of menin have shown that phosphorylation of menin could be detected at amino acid residues Ser394, Thr397, Thr399, Ser487, Ser543, and Ser583 (Francis, et al. 2011). Menin has also been shown to undergo SUMOylation and palmitoylation (Feng, et al. 2013; He, et al. 2016). Sub-cellular localization studies have shown that menin is predominantly nuclear, but a very small amount of menin has also been detected in the cytoplasm and at the cell membrane (Cao, et al. 2009; Guru, et al. 1998; He et al. 2016). The amino acid sequence of menin shows two main nuclear localization signals (NLS), NLS1 (amino acid residues 479–497) and NLS2 (amino acid residues 588–608), and a third accessory NLS, NLSa (amino acid residues 546–572) (Guru et al. 1998; La, et al. 2006). Germline and somatic MEN1 mutations that predict premature protein truncation (frame-shift and nonsense) would cause loss of one or both main NLSs leading to abnormal localization of the truncated menin and functional inactivation. Transfection and protein analysis in mammalian cell lines has shown that menin missense mutant proteins undergo degradation by the proteasome pathway that would prevent any functional activity (Canaff, et al. 2012).

Menin does not possess any intrinsic enzymatic activity, and its amino acid sequence does not predict any obvious functional domains. To determine the mechanisms by which menin acts as a tumor suppressor (as well as the consequence after menin loss), the elucidation of the physiological functions of menin has mainly relied on protein-protein interaction methods. Efforts from many groups have identified more than 50 different proteins that could partner with menin (Table 3). These interactions predict that menin is a multi-functional protein with functional contributions in transcriptional regulation as a co-repressor or co-activator (through interaction with chromatin modifying proteins, transcription factors, and transcription initiation or elongation proteins), DNA-repair associated with response to DNA damage, cell signaling, cytoskeletal structure, cell division, cell adhesion, or cell motility (Balogh, et al. 2010; Hendy, et al. 2009; Matkar, et al. 2013; Thakker 2014). Genetic studies in Drosophila melanogaster have shown that loss of menin may lead to genomic instability (Busygina, et al. 2004).

Table 3.

Menin Interacting Proteins

Protein Function Reference
NUCLEAR
Chromatin modification factors
DAXX Transcriptional regulation (Feng, et al. 2017)
HDACs, mSIN3A Transcriptional regulation (Kim, et al. 2003)
LEDGF Transcriptional regulation (Yokoyama and Cleary 2008)
MLL-complex (H3K4me3) (KMT2A/MLL1 or KMT2B/MLL2; ASH2L, hDPY30, HCF-2, RBBP5, RPB2/RNA-Pol-II, WDR5) Transcriptional regulation (Hughes et al. 2004; Yokoyama et al. 2005)
PRMT5 Transcriptional regulation (Gurung, et al. 2013a)
SuV39H1 (H3K9me3) Transcriptional regulation (Yang, et al. 2013)
Transcription factors
DNMT1 (Hedgehog pathway) Transcriptional regulation (Cheng, et al. 2016)
FBP1 Transcriptional regulation (Zaman, et al. 2014)
FOXA2 Transcriptional regulation (Bonnavion, et al. 2017)
HLXB9/MNX1 Transcriptional regulation (Shi, et al. 2013)
JUND Transcriptional regulation (Agarwal, et al. 1999; Gobl, et al. 1999)
c-MYB Transcriptional regulation (Jin, et al. 2010)
c-MYC Transcriptional regulation (Bres, et al. 2009)
NFκB - p50, p52, p65 Transcriptional regulation (Heppner, et al. 2001)
Nuclear receptors (AR, ERα, LXRα, PPARα, PPARϒ, RXR, VDR) Transcriptional regulation (Cheng, et al. 2015; Cheng, et al. 2011; Dreijerink, et al. 2006; Dreijerink, et al. 2009; Malik et al. 2015)
PEM Transcriptional regulation (Lemmens, et al. 2001)
RUNX2 (BMP2 signaling) Transcriptional regulation (Sowa, et al. 2004)
SMADs (TGFβ signaling) (SMAD1, SMAD3, SMAD5) Transcriptional regulation (Sowa et al. 2004)
SIRT1 Transcriptional regulation (Gang, et al. 2013)
SON Transcriptional regulation (Kim, et al. 2016)
TCF3, TCF4, β-Catenin (Wnt signaling) Transcriptional regulation (Cao et al. 2009)
Transcription initiation or elongation factors
RNA-Pol-II isoforms (pSer5 and pSer2) Transcriptional regulation (Francis et al. 2011)
SKIP (HIV-1 Tat:P-TEFb transcription) Transcriptional regulation (Bres et al. 2009)
Factors not associated with transcription
ASK DNA replication and repair (Schnepp, et al. 2004)
CHES1 DNA replication and repair (Busygina, et al. 2006)
FANCD2 DNA replication and repair (Jin, et al. 2003)
RPA2 DNA replication and repair (Sukhodolets, et al. 2003)
ARS2 MicroRNA biogenesis (Gurung, et al. 2014)
CHIP Protein degradation (Yaguchi, et al. 2004)
CYTOPLASMIC
AKT1 Signaling (Wang, et al. 2011)
FOXO1 Signaling (Wuescher, et al. 2011)
NM23β Signaling (Yaguchi, et al. 2002)
GFAP Cell division/Adhesion/Motility (Lopez-Egido, et al. 2002)
IQGAP1 Cell division/Adhesion/Motility (Yan, et al. 2009)
NMMHC-IIA Cell division/Adhesion/Motility (Obungu, et al. 2003)
VIMENTIN Cell division/Adhesion/Motility (Lopez-Egido et al. 2002)

Due to a prominent role of menin in transcriptional regulation many groups have studied the target genes of menin by analyzing menin-dependent differential mRNA expression, and menin-occupancy or menin-dependent histone modifications at target genes by chromatin immunoprecipitation coupled with next-generation sequencing. However, a consensus signature of genes affected upon menin loss in MEN1 target tissues warrants further investigation. Regarding the regulation of menin, a microRNA (miR24-1 and its mature form miR24) has been shown to regulate the expression of menin with a feedback mechanism (Luzi, et al. 2012; Vijayaraghavan, et al. 2014). Further studies of this miRNA that targets menin may shed light on yet another aspect of the multi-functional roles of menin and its regulation.

The functional characterization of menin has been advanced by the deciphering of the three-dimensional (3D) structure of Nematostella menin (the starlet sea anemone) and human menin (Protein Data Bank No. 3RE2 and 3U84). The deletion of disordered regions facilitated the generation of menin crystals - deletion of an internal unstructured loop (amino acid residues 426–442) and truncation of the C-terminus (amino acid residues 487–539) in Nematostella menin, and deletion of a single internal unstructured loop (amino acid residues 460–519) in human menin (Huang, et al. 2012; Murai, et al. 2011). The 3D structure of menin resembles a ‘curved left hand’, with a deep pocket formed by the ‘thumb’ and the ‘palm’. The structure shows 4 domains: a long β-hairpin N-terminal domain, a transglutaminase-like domain that forms the ‘thumb’, a helical ‘palm’ domain that contains three tetratricopeptide motifs, followed by a C-terminal ‘fingers’ domain. The presence of the deep pocket formed by the thumb and the palm has been shown to act as a domain for protein-protein interaction. Co-crystallization efforts with two known menin interacting proteins has shown that the pocket can bind short peptides of the AP1 transcription factor JUND (amino acid residues 27–47) or the mixed lineage leukemia (MLL) protein MLL1 (amino acid residues 6–25) (Huang et al. 2012). In vitro isothermal titration calorimetry analysis using purified menin protein (normal or with missense mutations) and a peptide consisting of the MLL1 menin-binding motif (MBM), or co-immunoprecipitation of transfected MLL1 and menin mutants has revealed that menin missense mutations at key residues located at the menin-MLL1 interface completely abolishes the menin–MLL1 interaction (Huang et al. 2012; Murai et al. 2011). Such assays can prove to be useful to decipher the benign or pathologic nature of menin missense variants of unknown significance (VUS).

An unexpected function of menin as a pro-oncogenic factor in MLL-fusion leukemia

The mixed lineage leukemia (MLL) proteins of the lysine methyltransferase (KMT) family, MLL1/KMT2A and MLL2/KMT2B are part of a protein complex that catalyzes a specific histone modification for gene activation, histone H3 lysine-4 trimethylation (referred to as the H3K4me3 mark in chromatin) (Shilatifard 2012). Loss of menin has been shown to coincide with the loss of H3K4me3 at specific genes, and at the same genes, gain of H3K27me3 that is an epigenetic mark of gene repression (Agarwal and Jothi 2012; Lin, et al. 2015; Scacheri, et al. 2006). MLL1 gene (chromosome 11q23) translocation with several different genes that encode transcription factors leads to the formation of MLL1-fusions that activate the expression of genes such as the HOX genes that cause leukemia (Yokoyama 2017). The interaction of menin in the Trithorax-like MLL protein complex has shown that menin is essential in this complex for the oncogenic activity of MLL1-fusion proteins to cause leukemia (Hughes, et al. 2004; Yokoyama, et al. 2005). Evidence from studies conducted by using bone marrow of mouse models have confirmed this pro-oncogenic action of menin (Chen, et al. 2006). Therefore, menin-MLL interaction inhibitors have been developed which has been facilitated by the deciphering of the 3D structure of menin with the MLL1 MBM (Grembecka, et al. 2012). These inhibitors named MI and further improved versions of these inhibitors (MI-463, MI-503, MI-538 and compound 27) have been shown to block the proliferation of MLL1-fusion leukemia cells from patients in vitro and in mouse xenografts in vivo (Borkin, et al. 2016). Such menin-MLL interaction inhibitors hold promise to conduct future clinical trials as a potential treatment for patients with MLL1-fusion leukemia. Menin-MLL interaction inhibitors have also been studied in mice to block the growth of specific types of Prostate cancer, Ewing sarcoma, and childhood Gliomas (Funato, et al. 2014; Malik, et al. 2015; Svoboda, et al. 2017).

Genetically engineered mouse models

Over the past two decades, many genetically engineered mouse models of Men1 loss have been generated to examine the causality of Men1 loss as a driver of tumorigenesis. These mouse models can serve as valuable surrogates to study the initiation, maintenance and evolution of MEN1-associated tumors, and can be utilized to discover and validate pathways downstream of menin loss, and to evaluate the utility of potential drug treatments.

Mice with germline knockout of the Men1 gene have been generated in four different laboratories by deleting genomic regions encompassing different exons of the mouse Men1 gene (U.S.A. (exons 3–8), France (exon 3), Australia (exon 2), and U.K (exons 1–2)) (Bertolino, et al. 2003a; Bertolino, et al. 2003b; Crabtree, et al. 2001; Harding, et al. 2009; Loffler, et al. 2007a). Collectively, these mouse models have shown that germline homozygous loss of the Men1 gene (Men1−/−) leads to death in utero between embryonic days 10.5 and 14.5 due to delayed development with craniofacial abnormalities, defective neural tube closure, heart hypertrophy, abnormal liver organization, hemorrhages, and edemas. Mice with germline heterozygous loss of the Men1 gene (Men1+/−) gestate normally, but they are predisposed to tumor development in a tissue-restricted manner which resembles the condition in human MEN1 patients (Table 4). Also, these tumors exhibit loss of the non-targeted Men1 allele (LOH), thus supporting a tumor suppressor function of the Men1 gene. Adult Men1+/− mice (age 12–18 months) develop MEN1-associated endocrine tumors of the parathyroid, multiple pancreatic islets (mainly insulinoma; also, glucagonoma, glucagon + insulin mixed-hormone tumors, or gastrinoma), anterior pituitary (mainly prolactinoma in female; also, GH-secreting somatotropinoma), adrenal cortex, thyroid, gonads (Leydig cells in male, or ovarian stroma in female), and mammary glands. The delay in tumor development could be due to the time it takes for the spontaneous LOH of the Men1 locus. Among the spectrum of tumors that develop in Men1+/− mice, some are not observed in human MEN1 patients, e.g., bilateral pheochromocytoma and gonadal tumors; and some human MEN1-associated tumors have not been reported in the mouse models, e.g., foregut carcinoids and skin lesions (3% of Men1+/− mice in one mouse model have shown lipoma (Harding et al. 2009)).

Table 4.

Prevalence of MEN1-associated tumors in Men1+/− mouse models compared to human MEN1

Human
(age 40)
Mouse models (age >12 months)
Crabtree,
2001
Bertolino,
2003
Loffler,
2007
Harding
2009
Parathyroid 90% 24% 47% 9% 85%
Pancreas
Gastrinoma 40% -- 15% -- --
Insulinoma 10% 83% 88% 82% 60%
Pituitary
Prolactinoma 20% 26% 12% 10% --
Prolactin + GH secreting 5% -- -- -- 32%
GH secreting 5% -- 7% -- --
ACTH secreting 2% -- -- -- 3%
Adrenal
Cortex NF 25% 20% 35% 10% 10%
Pheochromocytoma -- 7% -- -- 1%
Gonads
Testis -- 22% 88% 47% 60%
(Leydig cell in male)
Ovary -- 31% 8% -- 40%
(Sex-cord stromal in female)

NF = Non-functioning, GH = growth hormone, ACTH = adrenocorticotropic hormone

Genetically engineered mouse models with homozygous somatic loss of the Men1 gene specifically in MEN1-associated endocrine tissues have been generated by the Cre-Lox system that uses floxed Men1 alleles and Cre-recombinase expression driven by tissue-specific promoters. These mice develop normally and show tumor development at an early age because they do not rely on the spontaneous loss of the non-targeted Men1 allele as in the germline Men1-heterozygous mice. However, despite biallelic Men1 loss during early embryogenesis due to Cre expression from promoters that are active during embryogenesis, tumor development still takes 6–12 months. These observations indicate that loss of menin alone may not be sufficient for tumorigenesis. Parathyroid-specific Men1 knockout mice (using parathryroid hormone promoter/PTH-Cre) develop parathyroid hyperplasia and hypercalcemia (Libutti, et al. 2003). Pancreatic islet β-cell specific Men1 knockout mice (using Rat insulin promoter/RIP-Cre) develop insulinomas (adenoma or carcinoma) (Bertolino, et al. 2003c; Biondi, et al. 2004; Crabtree, et al. 2003). Pancreatic α-cell specific Men1 knockout mice (using glucagon promoter/GLU-Cre) show mostly insulinomas rather than the expected glucagonomas (Lu, et al. 2010; Shen, et al. 2010). The reason for this phenotype has been attributed to the possible involvement of paracrine signals that induce β-cell proliferation or due to trans-differentiation of α-cells into β-cells indicating a role of menin in maintaining the cellular plasticity of islet cells. Men1 knockout in the whole pancreas (using Pancreas/duodenum homeobox protein-1/PDX1-Cre) leads to a single tumor only of the endocrine pancreatic β-cells (insulinomas) while other pancreatic endocrine and non-endocrine cells remain unaffected (Shen, et al. 2009). Also, knockout of Men1 in the liver (using albumin promoter/ALB-Cre), a tissue not targeted in MEN1, does not lead to tumors in the liver despite the complete loss of Men1 (Scacheri, et al. 2004). Other mouse models with knockout of Men1 in pancreatic endocrine cell lineages, bone cells, and intestinal cells have also helped to gain insight into the physiological actions of menin (Bonnavion, et al. 2015; Kanazawa, et al. 2015; Liu, et al. 2017; Sundaresan, et al. 2016; Veniaminova, et al. 2012). These elegant studies in genetically engineered mouse models show that menin may function as a cell-type-specific tumor suppressor, and that its action is only required in MEN1-associated target tissues to prevent tumorigenesis.

A search of mouse models that show MEN1-associated tumors has revealed that mice with knockout of some cell cycle regulators (p18 and p27) develop tumors of the parathyroid, pancreatic islets and anterior pituitary (prolactinoma) (Franklin, et al. 1998). Therefore, mice with knockout of genes that are known to regulate the cell cycle (Rb, p53, p18, p27, Cdk2, or Cdk4) have been generated in the Men1+/− background to study the effect of the combined loss on the development of PNETs (insulinomas). Combined loss of Men1 with Rb or p53 has not shown any significant effect (Loffler, et al. 2007b; Loffler, et al. 2012; Matoso, et al. 2008). Combined loss of Men1 with p18 but not p27 results in the acceleration of insulinoma formation and increased tumor incidence (Bai, et al. 2007). Combined loss of Men1 with Cdk4 but not with Cdk2 blocks the formation of insulinomas (Gillam, et al. 2015). These mouse models show that decreased activity of p18 or increased activity of Cdk4 may contribute to the development of PNETs upon menin loss.

Mouse models have also been generated to look at the effect of pancreatic islet β-cell-specific Men1 loss together with other candidate target genes associated with β-cell proliferation and function. These mouse models have shown that the expression of activated K-RAS(p.G12D) enhances rather than inhibits β-cell proliferation, β-catenin loss can suppress insulinoma tumorigenesis, histone demethylase retinoblastoma binding protein-2 (Rbp2) loss decreases insulinoma formation and prolongs survival, ActivinB loss prolongs survival, and MLL1/KMT2A loss leads to earlier onset of tumor formation (4 months vs. 6 months) and shortened lifespan by promoting tumor progression and angiogenesis (Chamberlain, et al. 2014; Jiang, et al. 2014; Lin, et al. 2011; Lin, et al. 2016; Ripoche, et al. 2015). Such studies have dual benefits as they help to understand tumorigenesis and to reveal β-cell proliferation mechanisms for developing β-cell replacement strategies to help diabetic patients who suffer from functional β-cell loss (Garcia-Ocana and Stewart 2014).

The pre-clinical utility of the mouse models of Men1 loss is highlighted by several studies that have investigated the efficacy of potential treatment options in mouse PNETs (insulinoma) and pituitary tumors (prolactinoma). Some of the potential treatment options were predicted from protein-protein interaction studies where loss of menin in the interaction led to pro-tumorigenic actions of proteins or pathways that could be controlled with drugs (Hedgehog pathway, β-catenin, and epigenetic histone modifications). Other potential treatment options targeted receptors associated with tumor angiogenesis (VEGF receptors) or specific somatostatin receptors. These pre-clinical studies include menin replacement therapy, an angiogenesis inhibitor (anti-VEGF-A monoclonal antibody, mAb G6–31), a small molecular tyrosine kinase inhibitor of all VEGF receptors (Sunitinib), a somatostatin analog (Pasireotide/SOM230), a Hedgehog pathway inhibitor GDC-0449, a β-catenin inhibitor PKF115–584, and an epigenetic drug (BET protein inhibitor JQ1) (Gurung, et al. 2013b; Jiang et al. 2014; Korsisaari, et al. 2008; Lines, et al. 2017; Quinn, et al. 2012; Walls, et al. 2012; Walls, et al. 2016).

The impact of molecular genetic studies on disease prediction and management

The discovery of the causative gene for the MEN1 syndrome has changed the clinical management of MEN1 and related states with a direct impact on reducing disease-related morbidity and mortality. Genetic counseling and germline MEN1 genetic testing under recommended guidelines has facilitated the accuracy of disease prediction in index cases and their immediate family members and relatives (Brandi et al. 2001; Thakker et al. 2012). Mutation-negative individuals in families with a known MEN1 mutation are spared the uncertainty and anxiety of knowing whether they have the disease or not, and asymptomatic mutation-positive individuals can undergo early screening and monitoring for MEN1-associated tumors. A positive genetic test also helps to confirm the clinical diagnosis in both familial and sporadic cases of MEN1. Although a positive genetic test for MEN1 mutation does not lead to any immediate medical or surgical treatment decisions, the negative-genetic test eases the burden of lifelong clinical investigations. A few studies have shown that in family members of mutation-positive index cases who opted for genetic testing and who tested positive for an MEN1 mutation, early surveillance has helped to detect tumors at an average of 10 years before clinically evident disease, allowing careful monitoring of tumors that have the potential to become malignant. Reports of a few mutation-negative symptomatic individuals in MEN1 kindreds has suggested the existence of MEN1 phenocopies which may occur in 5–10% of families with MEN1, mainly associated with features of parathyroid and pituitary disease (Newey and Thakker 2011). The identification of other susceptibility genes for disease features similar to MEN1 has prompted several clinical genetic testing laboratories to offer genetic testing of a panel of genes that can be screened by whole exome sequencing (WES) to improve early disease prediction and management.

Future Directions

No mutation in the MEN1 gene is identified in 10–30% of cases diagnosed with MEN1 based on clinical features. Screening of MEN1 regions not included in the current mutation analysis methods may help to find the missing mutations, for example mutations in gene regulatory regions that may be located far away from the gene. The identification of such MEN1 gene regulatory regions may facilitate such studies. It is possible that other causative gene/s may be responsible for the clinical features, such as mutations in genes that encode menin interacting proteins. The identification of these genes can be attempted through collaborative efforts using whole exome sequencing or whole genome sequencing of germline and tumor DNA samples of patients (and family members) who present with the various MEN1-associated tumors. Such efforts may also help to identify disease-associated potential genetic modifiers.

Recent studies have proposed rare phenotypes among the MEN1 characteristics such as breast cancer in the Dutch cohort of MEN1 patients (van Leeuwaarde, et al. 2017). Follow-up studies could be conducted to verify this observation in other large cohorts of MEN1 patients such as in the GTE, Italian, and Japanese database of MEN1 (Giusti et al. 2017; Sakurai et al. 2012; Thevenon, et al. 2015). Also, mouse models of Men1 loss could help to elucidate whether menin loss alone or other factors contribute to this phenotype.

The tissue-restricted pattern of the endocrine tumors of MEN1 is also evident among the different mouse models of Men1 loss. The reason for this tissue-selective anti-tumor function of menin remains to be determined. Exploring tissue-specific actions of menin that may result from interactions with the various protein partners that have been identified may help to reveal therapeutic targets to facilitate translational efforts. Stimulating the outcome of these actions of menin may serve as a potential anti-tumor therapeutic option in MEN1-associated endocrine tumors. The development of cell lines derived from mouse or human MEN1-associated endocrine tumors could facilitate such studies. Another area for future investigation is the reason for the delay in tumorigenesis after the loss of both copies of the Men1 gene (as observed in mouse models).

Future studies directed at the identification of the missing genes that contribute to the development of the tumors characteristic of MEN1, and further elucidation of the molecular pathways that are affected upon menin loss hold promise for enhancing management strategies for MEN1 and related states.

Acknowledgments

Funding

Funding support for this review was from the Intramural Research Program of the NIH, National Institute of Diabetes and Digestive and Kidney Diseases.

Footnotes

Declaration of interest

The author declares that there is no conflict of interest that could be perceived as prejudicing the impartiality of this review.

Reference List

  1. Agarwal SK. MEN1. Frontiers in hormone research. 2013:1–15. doi: 10.1159/000345666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Agarwal SK, Debelenko LV, Kester MB, Guru SC, Manickam P, Olufemi SE, Skarulis MC, Heppner C, Crabtree JS, Lubensky IA, et al. Analysis of recurrent germline mutations in the MEN1 gene encountered in apparently unrelated families. Human mutation. 1998;12:75–82. doi: 10.1002/(SICI)1098-1004(1998)12:2<75::AID-HUMU1>3.0.CO;2-T. [DOI] [PubMed] [Google Scholar]
  3. Agarwal SK, Guru SC, Heppner C, Erdos MR, Collins RM, Park SY, Saggar S, Chandrasekharappa SC, Collins FS, Spiegel AM, et al. Menin interacts with the AP1 transcription factor JunD and represses JunD-activated transcription. Cell. 1999;96:143–152. doi: 10.1016/s0092-8674(00)80967-8. [DOI] [PubMed] [Google Scholar]
  4. Agarwal SK, Jothi R. Genome-Wide Characterization of Menin-Dependent H3K4me3 Reveals a Specific Role for Menin in the Regulation of Genes Implicated in MEN1-Like Tumors. PloS one. 2012;7:e37952. doi: 10.1371/journal.pone.0037952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Agarwal SK, Kester MB, Debelenko LV, Heppner C, Emmert-Buck MR, Skarulis MC, Doppman JL, Kim YS, Lubensky IA, Zhuang Z, et al. Germline mutations of the MEN1 gene in familial multiple endocrine neoplasia type 1 and related states. Human molecular genetics. 1997;6:1169–1175. doi: 10.1093/hmg/6.7.1169. [DOI] [PubMed] [Google Scholar]
  6. Agarwal SK, Mateo CM, Marx SJ. Rare germline mutations in cyclin-dependent kinase inhibitor genes in multiple endocrine neoplasia type 1 and related states. The Journal of clinical endocrinology and metabolism. 2009;94:1826–1834. doi: 10.1210/jc.2008-2083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Bai F, Pei XH, Nishikawa T, Smith MD, Xiong Y. p18Ink4c, but not p27Kip1, collaborates with Men1 to suppress neuroendocrine organ tumors. Molecular and cellular biology. 2007;27:1495–1504. doi: 10.1128/MCB.01764-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bale SJ, Bale AE, Stewart K, Dachowski L, McBride OW, Glaser T, Green JE, 3rd, Mulvihill JJ, Brandi ML, Sakaguchi K, et al. Linkage analysis of multiple endocrine neoplasia type 1 with INT2 and other markers on chromosome 11. Genomics. 1989;4:320–322. doi: 10.1016/0888-7543(89)90336-4. [DOI] [PubMed] [Google Scholar]
  9. Balogh K, Patocs A, Hunyady L, Racz K. Menin dynamics and functional insight: take your partners. Mol Cell Endocrinol. 2010;326:80–84. doi: 10.1016/j.mce.2010.04.011. [DOI] [PubMed] [Google Scholar]
  10. Bassett JH, Forbes SA, Pannett AA, Lloyd SE, Christie PT, Wooding C, Harding B, Besser GM, Edwards CR, Monson JP, et al. Characterization of mutations in patients with multiple endocrine neoplasia type 1. Am J Hum Genet. 1998;62:232–244. doi: 10.1086/301729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Bertolino P, Radovanovic I, Casse H, Aguzzi A, Wang ZQ, Zhang CX. Genetic ablation of the tumor suppressor menin causes lethality at mid-gestation with defects in multiple organs. Mechanisms of development. 2003a;120:549–560. doi: 10.1016/s0925-4773(03)00039-x. [DOI] [PubMed] [Google Scholar]
  12. Bertolino P, Tong WM, Galendo D, Wang ZQ, Zhang CX. Heterozygous Men1 mutant mice develop a range of endocrine tumors mimicking multiple endocrine neoplasia type 1. Molecular endocrinology. 2003b;17:1880–1892. doi: 10.1210/me.2003-0154. [DOI] [PubMed] [Google Scholar]
  13. Bertolino P, Tong WM, Herrera PL, Casse H, Zhang CX, Wang ZQ. Pancreatic beta-cell-specific ablation of the multiple endocrine neoplasia type 1 (MEN1) gene causes full penetrance of insulinoma development in mice. Cancer research. 2003c;63:4836–4841. [PubMed] [Google Scholar]
  14. Biondi CA, Gartside MG, Waring P, Loffler KA, Stark MS, Magnuson MA, Kay GF, Hayward NK. Conditional inactivation of the MEN1 gene leads to pancreatic and pituitary tumorigenesis but does not affect normal development of these tissues. Molecular and cellular biology. 2004;24:3125–3131. doi: 10.1128/MCB.24.8.3125-3131.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Bonnavion R, Teinturier R, Gherardi S, Leteurtre E, Yu R, Cordier-Bussat M, Du R, Pattou F, Vantyghem MC, Bertolino P, et al. Foxa2, a novel protein partner of the tumour suppressor menin, is deregulated in mouse and human MEN1 glucagonomas. J Pathol. 2017;242:90–101. doi: 10.1002/path.4885. [DOI] [PubMed] [Google Scholar]
  16. Bonnavion R, Teinturier R, Jaafar R, Ripoche D, Leteurtre E, Chen YJ, Rehfeld JF, Lepinasse F, Hervieu V, Pattou F, et al. Islet Cells Serve as Cells of Origin of Pancreatic Gastrin-Positive Endocrine Tumors. Molecular and cellular biology. 2015;35:3274–3283. doi: 10.1128/MCB.00302-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Borkin D, Pollock J, Kempinska K, Purohit T, Li X, Wen B, Zhao T, Miao H, Shukla S, He M, et al. Property Focused Structure-Based Optimization of Small Molecule Inhibitors of the Protein-Protein Interaction between Menin and Mixed Lineage Leukemia (MLL) J Med Chem. 2016;59:892–913. doi: 10.1021/acs.jmedchem.5b01305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Brandi ML, Gagel RF, Angeli A, Bilezikian JP, Beck-Peccoz P, Bordi C, Conte-Devolx B, Falchetti A, Gheri RG, Libroia A, et al. Guidelines for diagnosis and therapy of MEN type 1 and type 2. The Journal of clinical endocrinology and metabolism. 2001;86:5658–5671. doi: 10.1210/jcem.86.12.8070. [DOI] [PubMed] [Google Scholar]
  19. Bres V, Yoshida T, Pickle L, Jones KA. SKIP interacts with c-Myc and Menin to promote HIV-1 Tat transactivation. Molecular cell. 2009;36:75–87. doi: 10.1016/j.molcel.2009.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Burgess JR, Nord B, David R, Greenaway TM, Parameswaran V, Larsson C, Shepherd JJ, Teh BT. Phenotype and phenocopy: the relationship between genotype and clinical phenotype in a single large family with multiple endocrine neoplasia type 1 (MEN 1) Clin Endocrinol (Oxf) 2000;53:205–211. doi: 10.1046/j.1365-2265.2000.01032.x. [DOI] [PubMed] [Google Scholar]
  21. Busygina V, Kottemann MC, Scott KL, Plon SE, Bale AE. Multiple endocrine neoplasia type 1 interacts with forkhead transcription factor CHES1 in DNA damage response. Cancer research. 2006;66:8397–8403. doi: 10.1158/0008-5472.CAN-06-0061. [DOI] [PubMed] [Google Scholar]
  22. Busygina V, Suphapeetiporn K, Marek LR, Stowers RS, Xu T, Bale AE. Hypermutability in a Drosophila model for multiple endocrine neoplasia type 1. Hum Mol Genet. 2004;13:2399–2408. doi: 10.1093/hmg/ddh271. [DOI] [PubMed] [Google Scholar]
  23. Canaff L, Vanbellinghen JF, Kanazawa I, Kwak H, Garfield N, Vautour L, Hendy GN. Menin missense mutants encoded by the MEN1 gene that are targeted to the proteasome: restoration of expression and activity by CHIP siRNA. The Journal of clinical endocrinology and metabolism. 2012;97:E282–291. doi: 10.1210/jc.2011-0241. [DOI] [PubMed] [Google Scholar]
  24. Cao Y, Gao Z, Li L, Jiang X, Shan A, Cai J, Peng Y, Li Y, Huang X, Wang J, et al. Whole exome sequencing of insulinoma reveals recurrent T372R mutations in YY1. Nature communications. 2013;4:2810. doi: 10.1038/ncomms3810. [DOI] [PubMed] [Google Scholar]
  25. Cao Y, Liu R, Jiang X, Lu J, Jiang J, Zhang C, Li X, Ning G. Nuclear-cytoplasmic shuttling of menin regulates nuclear translocation of {beta}-catenin. Molecular and cellular biology. 2009;29:5477–5487. doi: 10.1128/MCB.00335-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Cardinal JW, Bergman L, Hayward N, Sweet A, Warner J, Marks L, Learoyd D, Dwight T, Robinson B, Epstein M, et al. A report of a national mutation testing service for the MEN1 gene: clinical presentations and implications for mutation testing. J Med Genet. 2005;42:69–74. doi: 10.1136/jmg.2003.017319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Carpten JD, Robbins CM, Villablanca A, Forsberg L, Presciuttini S, Bailey-Wilson J, Simonds WF, Gillanders EM, Kennedy AM, Chen JD, et al. HRPT2, encoding parafibromin, is mutated in hyperparathyroidism-jaw tumor syndrome. Nat Genet. 2002;32:676–680. doi: 10.1038/ng1048. [DOI] [PubMed] [Google Scholar]
  28. Cebrian A, Ruiz-Llorente S, Cascon A, Pollan M, Diez JJ, Pico A, Telleria D, Benitez J, Robledo M. Mutational and gross deletion study of the MEN1 gene and correlation with clinical features in Spanish patients. J Med Genet. 2003;40:e72. doi: 10.1136/jmg.40.5.e72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Chamberlain CE, Scheel DW, McGlynn K, Kim H, Miyatsuka T, Wang J, Nguyen V, Zhao S, Mavropoulos A, Abraham AG, et al. Menin determines K-RAS proliferative outputs in endocrine cells. The Journal of clinical investigation. 2014;124:4093–4101. doi: 10.1172/JCI69004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Chandrasekharappa SC, Guru SC, Manickam P, Olufemi SE, Collins FS, EmmertBuck MR, Debelenko LV, Zhuang ZP, Lubensky IA, Liotta LA, et al. Positional cloning of the gene for multiple endocrine neoplasia-type 1. Science. 1997;276:404–407. doi: 10.1126/science.276.5311.404. [DOI] [PubMed] [Google Scholar]
  31. Chen YX, Yan J, Keeshan K, Tubbs AT, Wang H, Silva A, Brown EJ, Hess JL, Pear WS, Hua X. The tumor suppressor menin regulates hematopoiesis and myeloid transformation by influencing Hox gene expression. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:1018–1023. doi: 10.1073/pnas.0510347103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Cheng P, Li G, Yang SS, Liu R, Jin G, Zhou XY, Hu XG. Tumor suppressor Menin acts as a corepressor of LXRalpha to inhibit hepatic lipogenesis. FEBS Lett. 2015;589:3079–3084. doi: 10.1016/j.febslet.2015.04.049. [DOI] [PubMed] [Google Scholar]
  33. Cheng P, Wang YF, Li G, Yang SS, Liu C, Hu H, Jin G, Hu XG. Interplay between menin and Dnmt1 reversibly regulates pancreatic cancer cell growth downstream of the Hedgehog signaling pathway. Cancer Lett. 2016;370:136–144. doi: 10.1016/j.canlet.2015.09.019. [DOI] [PubMed] [Google Scholar]
  34. Cheng P, Yang SS, Hu XG, Zhou XY, Zhang YJ, Jin G, Zhou YQ. Menin prevents liver steatosis through co-activation of peroxisome proliferator-activated receptor alpha. FEBS Lett. 2011;585:3403–3408. doi: 10.1016/j.febslet.2011.09.043. [DOI] [PubMed] [Google Scholar]
  35. Concolino P, Costella A, Capoluongo E. Multiple endocrine neoplasia type 1 (MEN1): An update of 208 new germline variants reported in the last nine years. Cancer genetics. 2015 doi: 10.1016/j.cancergen.2015.12.002. [DOI] [PubMed] [Google Scholar]
  36. Crabtree JS, Scacheri PC, Ward JM, Garrett-Beal L, Emmert-Buck MR, Edgemon KA, Lorang D, Libutti SK, Chandrasekharappa SC, Marx SJ, et al. A mouse model of multiple endocrine neoplasia, type 1, develops multiple endocrine tumors. Proceedings of the National Academy of Sciences of the United States of America. 2001;98:1118–1123. doi: 10.1073/pnas.98.3.1118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Crabtree JS, Scacheri PC, Ward JM, McNally SR, Swain GP, Montagna C, Hager JH, Hanahan D, Edlund H, Magnuson MA, et al. Of mice and MEN1: Insulinomas in a conditional mouse knockout. Molecular and cellular biology. 2003;23:6075–6085. doi: 10.1128/MCB.23.17.6075-6085.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Cromer MK, Starker LF, Choi M, Udelsman R, Nelson-Williams C, Lifton RP, Carling T. Identification of somatic mutations in parathyroid tumors using whole-exome sequencing. The Journal of clinical endocrinology and metabolism. 2012;97:E1774–1781. doi: 10.1210/jc.2012-1743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Daly AF, Vanbellinghen JF, Khoo SK, Jaffrain-Rea ML, Naves LA, Guitelman MA, Murat A, Emy P, Gimenez-Roqueplo AP, Tamburrano G, et al. Aryl hydrocarbon receptor-interacting protein gene mutations in familial isolated pituitary adenomas: analysis in 73 families. J Clin Endocrinol Metab. 2007;92:1891–1896. doi: 10.1210/jc.2006-2513. [DOI] [PubMed] [Google Scholar]
  40. de Laat JM, van der Luijt RB, Pieterman CR, Oostveen MP, Hermus AR, Dekkers OM, de Herder WW, van der Horst-Schrivers AN, Drent ML, Bisschop PH, et al. MEN1 redefined, a clinical comparison of mutation-positive and mutation-negative patients. BMC Med. 2016;14:182. doi: 10.1186/s12916-016-0708-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Dean PG, van Heerden JA, Farley DR, Thompson GB, Grant CS, Harmsen WS, Ilstrup DM. Are patients with multiple endocrine neoplasia type I prone to premature death? World journal of surgery. 2000;24:1437–1441. doi: 10.1007/s002680010237. [DOI] [PubMed] [Google Scholar]
  42. Debelenko LV, Emmert-Buck MR, Manickam P, Kester M, Guru SC, DiFranco EM, Olufemi SE, Agarwal S, Lubensky IA, Zhuang Z, et al. Haplotype analysis defines a minimal interval for the multiple endocrine neoplasia type 1 (MEN1) gene. Cancer research. 1997;57:1039–1042. [PubMed] [Google Scholar]
  43. Doherty GM, Olson JA, Frisella MM, Lairmore TC, Wells SA, Jr, Norton JA. Lethality of multiple endocrine neoplasia type I. World journal of surgery. 1998;22:581–586. doi: 10.1007/s002689900438. discussion 586–587. [DOI] [PubMed] [Google Scholar]
  44. Dreijerink KM, Mulder KW, Winkler GS, Hoppener JW, Lips CJ, Timmers HT. Menin links estrogen receptor activation to histone H3K4 trimethylation. Cancer research. 2006;66:4929–4935. doi: 10.1158/0008-5472.CAN-05-4461. [DOI] [PubMed] [Google Scholar]
  45. Dreijerink KM, Varier RA, van Nuland R, Broekhuizen R, Valk GD, van der Wal JE, Lips CJ, Kummer JA, Timmers HT. Regulation of vitamin D receptor function in MEN1-related parathyroid adenomas. Mol Cell Endocrinol. 2009;313:1–8. doi: 10.1016/j.mce.2009.08.020. [DOI] [PubMed] [Google Scholar]
  46. Emmert-Buck MR, Lubensky IA, Dong Q, Manickam P, Guru SC, Kester MB, Olufemi SE, Agarwal S, Burns AL, Spiegel AM, et al. Localization of the multiple endocrine neoplasia type I (MEN1) gene based on tumor loss of heterozygosity analysis. Cancer research. 1997;57:1855–1858. [PubMed] [Google Scholar]
  47. Feng Z, Wang L, Sun Y, Jiang Z, Domsic J, An C, Xing B, Tian J, Liu X, Metz DC, et al. Menin and Daxx Interact to Suppress Neuroendocrine Tumors through Epigenetic Control of the Membrane Metallo-Endopeptidase. Cancer research. 2017;77:401–411. doi: 10.1158/0008-5472.CAN-16-1567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Feng ZJ, Gurung B, Jin GH, Yang XL, Hua XX. SUMO modification of menin. Am J Cancer Res. 2013;3:96–106. [PMC free article] [PubMed] [Google Scholar]
  49. Francis J, Lin W, Rozenblatt-Rosen O, Meyerson M. The menin tumor suppressor protein is phosphorylated in response to DNA damage. PloS one. 2011;6:e16119. doi: 10.1371/journal.pone.0016119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Franklin DS, Godfrey VL, Lee H, Kovalev GI, Schoonhoven R, Chen-Kiang S, Su L, Xiong Y. CDK inhibitors p18(INK4c) and p27(Kip1) mediate two separate pathways to collaboratively suppress pituitary tumorigenesis. Genes & development. 1998;12:2899–2911. doi: 10.1101/gad.12.18.2899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Friedman E, Sakaguchi K, Bale AE, Falchetti A, Streeten E, Zimering MB, Weinstein LS, McBride WO, Nakamura Y, Brandi ML, et al. Clonality of parathyroid tumors in familial multiple endocrine neoplasia type 1. N Engl J Med. 1989;321:213–218. doi: 10.1056/NEJM198907273210402. [DOI] [PubMed] [Google Scholar]
  52. Funato K, Major T, Lewis PW, Allis CD, Tabar V. Use of human embryonic stem cells to model pediatric gliomas with H3.3K27M histone mutation. Science. 2014;346:1529–1533. doi: 10.1126/science.1253799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Gang D, Hongwei H, Hedai L, Ming Z, Qian H, Zhijun L. The tumor suppressor protein menin inhibits NF-kappaB-mediated transactivation through recruitment of Sirt1 in hepatocellular carcinoma. Mol Biol Rep. 2013;40:2461–2466. doi: 10.1007/s11033-012-2326-0. [DOI] [PubMed] [Google Scholar]
  54. Garcia-Ocana A, Stewart AF. "RAS"ling beta cells to proliferate for diabetes: why do we need MEN? The Journal of clinical investigation. 2014;124:3698–3700. doi: 10.1172/JCI77764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Gillam MP, Nimbalkar D, Sun L, Christov K, Ray D, Kaldis P, Liu X, Kiyokawa H. MEN1 tumorigenesis in the pituitary and pancreatic islet requires Cdk4 but not Cdk2. Oncogene. 2015;34:932–938. doi: 10.1038/onc.2014.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Giraud S, Zhang CX, Serova-Sinilnikova O, Wautot V, Salandre J, Buisson N, Waterlot C, Bauters C, Porchet N, Aubert JP, et al. Germ-line mutation analysis in patients with multiple endocrine neoplasia type 1 and related disorders. American journal of human genetics. 1998;63:455–467. doi: 10.1086/301953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Giusti F, Cianferotti L, Boaretto F, Cetani F, Cioppi F, Colao A, Davi MV, Faggiano A, Fanciulli G, Ferolla P, et al. Multiple endocrine neoplasia syndrome type 1: institution, management, and data analysis of a nationwide multicenter patient database. Endocrine. 2017 doi: 10.1007/s12020-017-1234-4. [DOI] [PubMed] [Google Scholar]
  58. Gobl AE, Berg M, Lopez-Egido JR, Oberg K, Skogseid B, Westin G. Menin represses JunD-activated transcription by a histone deacetylase-dependent mechanism. Biochim Biophys Acta. 1999;1447:51–56. doi: 10.1016/s0167-4781(99)00132-3. [DOI] [PubMed] [Google Scholar]
  59. Gortz B, Roth J, Krahenmann A, de Krijger RR, Muletta-Feurer S, Rutimann K, Saremaslani P, Speel EJ, Heitz PU, Komminoth P. Mutations and allelic deletions of the MEN1 gene are associated with a subset of sporadic endocrine pancreatic and neuroendocrine tumors and not restricted to foregut neoplasms. Am J Pathol. 1999;154:429–436. doi: 10.1016/S0002-9440(10)65289-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Goudet P, Dalac A, Le Bras M, Cardot-Bauters C, Niccoli P, Levy-Bohbot N, du Boullay H, Bertagna X, Ruszniewski P, Borson-Chazot F, et al. MEN1 disease occurring before 21 years old: a 160-patient cohort study from the Groupe d'etude des Tumeurs Endocrines. J Clin Endocrinol Metab. 2015;100:1568–1577. doi: 10.1210/jc.2014-3659. [DOI] [PubMed] [Google Scholar]
  61. Goudet P, Murat A, Binquet C, Cardot-Bauters C, Costa A, Ruszniewski P, Niccoli P, Menegaux F, Chabrier G, Borson-Chazot F, et al. Risk factors and causes of death in MEN1 disease. A GTE (Groupe d'Etude des Tumeurs Endocrines) cohort study among 758 patients. World journal of surgery. 2010;34:249–255. doi: 10.1007/s00268-009-0290-1. [DOI] [PubMed] [Google Scholar]
  62. Grembecka J, He S, Shi A, Purohit T, Muntean AG, Sorenson RJ, Showalter HD, Murai MJ, Belcher AM, Hartley T, et al. Menin-MLL inhibitors reverse oncogenic activity of MLL fusion proteins in leukemia. Nat Chem Biol. 2012;8:277–284. doi: 10.1038/nchembio.773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Guan B, Welch JM, Sapp JC, Ling H, Li Y, Johnston JJ, Kebebew E, Biesecker LG, Simonds WF, Marx SJ, et al. GCM2-Activating Mutations in Familial Isolated Hyperparathyroidism. Am J Hum Genet. 2016;99:1034–1044. doi: 10.1016/j.ajhg.2016.08.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Guru SC, Agarwal SK, Manickam P, Olufemi SE, Crabtree JS, Weisemann JM, Kester MB, Kim YS, Wang Y, Emmert-Buck MR, et al. A transcript map for the 2.8-Mb region containing the multiple endocrine neoplasia type 1 locus. Genome Res. 1997a;7:725–735. doi: 10.1101/gr.7.7.725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Guru SC, Goldsmith PK, Burns AL, Marx SJ, Spiegel AM, Collins FS, Chandrasekharappa SC. Menin, the product of the MEN1 gene, is a nuclear protein. Proceedings of the National Academy of Sciences of the United States of America. 1998;95:1630–1634. doi: 10.1073/pnas.95.4.1630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Guru SC, Olufemi SE, Manickam P, Cummings C, Gieser LM, Pike BL, Bittner ML, Jiang Y, Chinault AC, Nowak NJ, et al. A 2.8-Mb clone contig of the multiple endocrine neoplasia type 1 (MEN1) region at 11q13. Genomics. 1997b;42:436–445. doi: 10.1006/geno.1997.4783. [DOI] [PubMed] [Google Scholar]
  67. Gurung B, Feng Z, Hua X. Menin directly represses Gli1 expression independent of canonical Hedgehog signaling. Molecular cancer research : MCR. 2013a;11:1215–1222. doi: 10.1158/1541-7786.MCR-13-0170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Gurung B, Feng Z, Iwamoto DV, Thiel A, Jin G, Fan CM, Ng JM, Curran T, Hua X. Menin epigenetically represses Hedgehog signaling in MEN1 tumor syndrome. Cancer research. 2013b;73:2650–2658. doi: 10.1158/0008-5472.CAN-12-3158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Gurung B, Katona BW, Hua X. Menin-mediated regulation of miRNA biogenesis uncovers the IRS2 pathway as a target for regulating pancreatic beta cells. Oncoscience. 2014;1:562–566. doi: 10.18632/oncoscience.79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Hannan FM, Nesbit MA, Christie PT, Fratter C, Dudley NE, Sadler GP, Thakker RV. Familial isolated primary hyperparathyroidism caused by mutations of the MEN1 gene. Nat Clin Pract Endocrinol Metab. 2008;4:53–58. doi: 10.1038/ncpendmet0718. [DOI] [PubMed] [Google Scholar]
  71. Harding B, Lemos MC, Reed AA, Walls GV, Jeyabalan J, Bowl MR, Tateossian H, Sullivan N, Hough T, Fraser WD, et al. Multiple endocrine neoplasia type 1 knockout mice develop parathyroid, pancreatic, pituitary and adrenal tumours with hypercalcaemia, hypophosphataemia and hypercorticosteronaemia. Endocrine-related cancer. 2009;16:1313–1327. doi: 10.1677/ERC-09-0082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. He X, Wang L, Yan J, Yuan C, Witze ES, Hua X. Menin localization in cell membrane compartment. Cancer biology & therapy. 2016;17:114–122. doi: 10.1080/15384047.2015.1108497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Hendy GN, Kaji H, Canaff L. Cellular functions of menin. Advances in experimental medicine and biology. 2009;668:37–50. doi: 10.1007/978-1-4419-1664-8_4. [DOI] [PubMed] [Google Scholar]
  74. Heppner C, Bilimoria KY, Agarwal SK, Kester M, Whitty LJ, Guru SC, Chandrasekharappa SC, Collins FS, Spiegel AM, Marx SJ, et al. The tumor suppressor protein menin interacts with NF-kappaB proteins and inhibits NF-kappaB-mediated transactivation. Oncogene. 2001;20:4917–4925. doi: 10.1038/sj.onc.1204529. [DOI] [PubMed] [Google Scholar]
  75. Heppner C, Kester MB, Agarwal SK, Debelenko LV, Emmert-Buck MR, Guru SC, Manickam P, Olufemi SE, Skarulis MC, Doppman JL, et al. Somatic mutation of the MEN1 gene in parathyroid tumours. Nature genetics. 1997;16:375–378. doi: 10.1038/ng0897-375. [DOI] [PubMed] [Google Scholar]
  76. Huang J, Gurung B, Wan B, Matkar S, Veniaminova NA, Wan K, Merchant JL, Hua X, Lei M. The same pocket in menin binds both MLL and JUND but has opposite effects on transcription. Nature. 2012;482:542–546. doi: 10.1038/nature10806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Hughes CM, Rozenblatt-Rosen O, Milne TA, Copeland TD, Levine SS, Lee JC, Hayes DN, Shanmugam KS, Bhattacharjee A, Biondi CA, et al. Menin associates with a trithorax family histone methyltransferase complex and with the hoxc8 locus. Molecular cell. 2004;13:587–597. doi: 10.1016/s1097-2765(04)00081-4. [DOI] [PubMed] [Google Scholar]
  78. Ito T, Igarashi H, Uehara H, Berna MJ, Jensen RT. Causes of death and prognostic factors in multiple endocrine neoplasia type 1: a prospective study: comparison of 106 MEN1/Zollinger-Ellison syndrome patients with 1613 literature MEN1 patients with or without pancreatic endocrine tumors. Medicine. 2013;92:135–181. doi: 10.1097/MD.0b013e3182954af1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Jiang X, Cao Y, Li F, Su Y, Li Y, Peng Y, Cheng Y, Zhang C, Wang W, Ning G. Targeting beta-catenin signaling for therapeutic intervention in MEN1-deficient pancreatic neuroendocrine tumours. Nature communications. 2014;5:5809. doi: 10.1038/ncomms6809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Jiao Y, Shi C, Edil BH, de Wilde RF, Klimstra DS, Maitra A, Schulick RD, Tang LH, Wolfgang CL, Choti MA, et al. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science. 2011;331:1199–1203. doi: 10.1126/science.1200609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Jin S, Mao H, Schnepp RW, Sykes SM, Silva AC, D'Andrea AD, Hua X. Menin associates with FANCD2, a protein involved in repair of DNA damage. Cancer research. 2003;63:4204–4210. [PubMed] [Google Scholar]
  82. Jin S, Zhao H, Yi Y, Nakata Y, Kalota A, Gewirtz AM. c-Myb binds MLL through menin in human leukemia cells and is an important driver of MLL-associated leukemogenesis. The Journal of clinical investigation. 2010;120:593–606. doi: 10.1172/JCI38030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Kanazawa I, Canaff L, Abi Rafeh J, Angrula A, Li J, Riddle RC, Boraschi-Diaz I, Komarova SV, Clemens TL, Murshed M, et al. Osteoblast menin regulates bone mass in vivo. The Journal of biological chemistry. 2015;290:3910–3924. doi: 10.1074/jbc.M114.629899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Kim H, Lee JE, Cho EJ, Liu JO, Youn HD. Menin, a tumor suppressor, represses JunD-mediated transcriptional activity by association with an mSin3A-histone deacetylase complex. Cancer research. 2003;63:6135–6139. [PubMed] [Google Scholar]
  85. Kim JH, Baddoo MC, Park EY, Stone JK, Park H, Butler TW, Huang G, Yan X, Pauli-Behn F, Myers RM, et al. SON and Its Alternatively Spliced Isoforms Control MLL Complex-Mediated H3K4me3 and Transcription of Leukemia-Associated Genes. Molecular cell. 2016;61:859–873. doi: 10.1016/j.molcel.2016.02.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Klein RD, Salih S, Bessoni J, Bale AE. Clinical testing for multiple endocrine neoplasia type 1 in a DNA diagnostic laboratory. Genet Med. 2005;7:131–138. doi: 10.1097/01.gim.0000153663.62300.f8. [DOI] [PubMed] [Google Scholar]
  87. Knudson AG. Antioncogenes and human cancer. Proceedings of the National Academy of Sciences of the United States of America. 1993;90:10914–10921. doi: 10.1073/pnas.90.23.10914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Knudson AG., Jr Mutation and cancer: statistical study of retinoblastoma. Proceedings of the National Academy of Sciences of the United States of America. 1971;68:820–823. doi: 10.1073/pnas.68.4.820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Korsisaari N, Ross J, Wu X, Kowanetz M, Pal N, Hall L, Eastham-Anderson J, Forrest WF, Van Bruggen N, Peale FV, et al. Blocking vascular endothelial growth factor-A inhibits the growth of pituitary adenomas and lowers serum prolactin level in a mouse model of multiple endocrine neoplasia type 1. Clinical cancer research : an official journal of the American Association for Cancer Research. 2008;14:249–258. doi: 10.1158/1078-0432.CCR-07-1552. [DOI] [PubMed] [Google Scholar]
  90. Kytola S, Villablanca A, Ebeling T, Nord B, Larsson C, Hoog A, Wong FK, Valimaki M, Vierimaa O, Teh BT, et al. Founder effect in multiple endocrine neoplasia type 1 (MEN 1) in Finland. J Med Genet. 2001;38:185–189. doi: 10.1136/jmg.38.3.185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. La P, Desmond A, Hou Z, Silva AC, Schnepp RW, Hua X. Tumor suppressor menin: the essential role of nuclear localization signal domains in coordinating gene expression. Oncogene. 2006;25:3537–3546. doi: 10.1038/sj.onc.1209400. [DOI] [PubMed] [Google Scholar]
  92. Larsson C, Skogseid B, Oberg K, Nakamura Y, Nordenskjold M. Multiple endocrine neoplasia type 1 gene maps to chromosome 11 and is lost in insulinoma. Nature. 1988;332:85–87. doi: 10.1038/332085a0. [DOI] [PubMed] [Google Scholar]
  93. Lemmens I, Merregaert J, Van de Ven WJ, Kas K, Zhang CX, Giraud S, Wautot V, Buisson N, De Witte K, Salandre J, et al. Construction of a 1.2-Mb sequence-ready contig of chromosome 11q13 encompassing the multiple endocrine neoplasia type 1 (MEN1) gene. The European Consortium on MEN1. Genomics. 1997a;44:94–100. doi: 10.1006/geno.1997.4872. [DOI] [PubMed] [Google Scholar]
  94. Lemmens I, Van de Ven WJ, Kas K, Zhang CX, Giraud S, Wautot V, Buisson N, De Witte K, Salandre J, Lenoir G, et al. Identification of the multiple endocrine neoplasia type 1 (MEN1) gene. The European Consortium on MEN1. Human molecular genetics. 1997b;6:1177–1183. doi: 10.1093/hmg/6.7.1177. [DOI] [PubMed] [Google Scholar]
  95. Lemmens IH, Forsberg L, Pannett AA, Meyen E, Piehl F, Turner JJ, Van de Ven WJ, Thakker RV, Larsson C, Kas K. Menin interacts directly with the homeobox-containing protein Pem. Biochem Biophys Res Commun. 2001;286:426–431. doi: 10.1006/bbrc.2001.5405. [DOI] [PubMed] [Google Scholar]
  96. Lemos MC, Thakker RV. Multiple endocrine neoplasia type 1 (MEN1): analysis of 1336 mutations reported in the first decade following identification of the gene. Human mutation. 2008;29:22–32. doi: 10.1002/humu.20605. [DOI] [PubMed] [Google Scholar]
  97. Libutti SK, Crabtree JS, Lorang D, Burns AL, Mazzanti C, Hewitt SM, O'Connor S, Ward JM, Emmert-Buck MR, Remaley A, et al. Parathyroid gland-specific deletion of the mouse Men1 gene results in parathyroid neoplasia and hypercalcemic hyperparathyroidism. Cancer research. 2003;63:8022–8028. [PubMed] [Google Scholar]
  98. Lin W, Cao J, Liu J, Beshiri ML, Fujiwara Y, Francis J, Cherniack AD, Geisen C, Blair LP, Zou MR, et al. Loss of the retinoblastoma binding protein 2 (RBP2) histone demethylase suppresses tumorigenesis in mice lacking Rb1 or Men1. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:13379–13386. doi: 10.1073/pnas.1110104108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Lin W, Francis JM, Li H, Gao X, Pedamallu CS, Ernst P, Meyerson M. Kmt2a cooperates with menin to suppress tumorigenesis in mouse pancreatic islets. Cancer biology & therapy. 2016;17:1274–1281. doi: 10.1080/15384047.2016.1250986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Lin W, Watanabe H, Peng S, Francis JM, Kaplan N, Pedamallu CS, Ramachandran A, Agoston A, Bass AJ, Meyerson M. Dynamic epigenetic regulation by menin during pancreatic islet tumor formation. Molecular cancer research : MCR. 2015;13:689–698. doi: 10.1158/1541-7786.MCR-14-0457. [DOI] [PubMed] [Google Scholar]
  101. Lines KE, Vas Nunes RP, Frost M, Yates CJ, Stevenson M, Thakker R. A MEN1 pancreatic neuroendocrine tumour mouse model, under temporal control. Endocr Connect. 2017 doi: 10.1530/EC-17-0040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Liu P, Lee S, Knoll J, Rauch A, Ostermay S, Luther J, Malkusch N, Lerner UH, Zaiss MM, Neven M, et al. Loss of menin in osteoblast lineage affects osteocyte-osteoclast crosstalk causing osteoporosis. Cell death and differentiation. 2017 doi: 10.1038/cdd.2016.165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Loffler KA, Biondi CA, Gartside M, Waring P, Stark M, Serewko-Auret MM, Muller HK, Hayward NK, Kay GF. Broad tumor spectrum in a mouse model of multiple endocrine neoplasia type 1. International journal of cancer. Journal international du cancer. 2007a;120:259–267. doi: 10.1002/ijc.22288. [DOI] [PubMed] [Google Scholar]
  104. Loffler KA, Biondi CA, Gartside MG, Serewko-Auret MM, Duncan R, Tonks ID, Mould AW, Waring P, Muller HK, Kay GF, et al. Lack of augmentation of tumor spectrum or severity in dual heterozygous Men1 and Rb1 knockout mice. Oncogene. 2007b;26:4009–4017. doi: 10.1038/sj.onc.1210163. [DOI] [PubMed] [Google Scholar]
  105. Loffler KA, Mould AW, Waring PM, Hayward NK, Kay GF. Menin and p53 have non-synergistic effects on tumorigenesis in mice. BMC cancer. 2012;12:252. doi: 10.1186/1471-2407-12-252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Lopez-Egido J, Cunningham J, Berg M, Oberg K, Bongcam-Rudloff E, Gobl A. Menin's interaction with glial fibrillary acidic protein and vimentin suggests a role for the intermediate filament network in regulating menin activity. Exp Cell Res. 2002;278:175–183. doi: 10.1006/excr.2002.5575. [DOI] [PubMed] [Google Scholar]
  107. Lourenco DM, Jr, Toledo RA, Mackowiak II, Coutinho FL, Cavalcanti MG, Correia-Deur JE, Montenegro F, Siqueira SA, Margarido LC, Machado MC, et al. Multiple endocrine neoplasia type 1 in Brazil: MEN1 founding mutation, clinical features, and bone mineral density profile. Eur J Endocrinol. 2008;159:259–274. doi: 10.1530/EJE-08-0153. [DOI] [PubMed] [Google Scholar]
  108. Lu J, Herrera PL, Carreira C, Bonnavion R, Seigne C, Calender A, Bertolino P, Zhang CX. Alpha cell-specific Men1 ablation triggers the transdifferentiation of glucagon-expressing cells and insulinoma development. Gastroenterology. 2010;138:1954–1965. doi: 10.1053/j.gastro.2010.01.046. [DOI] [PubMed] [Google Scholar]
  109. Luzi E, Marini F, Giusti F, Galli G, Cavalli L, Brandi ML. The negative feedback-loop between the oncomir Mir-24-1 and menin modulates the Men1 tumorigenesis by mimicking the "Knudson's second hit". PloS one. 2012;7:e39767. doi: 10.1371/journal.pone.0039767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Machens A, Schaaf L, Karges W, Frank-Raue K, Bartsch DK, Rothmund M, Schneyer U, Goretzki P, Raue F, Dralle H. Age-related penetrance of endocrine tumours in multiple endocrine neoplasia type 1 (MEN1): a multicentre study of 258 gene carriers. Clinical endocrinology. 2007;67:613–622. doi: 10.1111/j.1365-2265.2007.02934.x. [DOI] [PubMed] [Google Scholar]
  111. Malik R, Khan AP, Asangani IA, Cieslik M, Prensner JR, Wang X, Iyer MK, Jiang X, Borkin D, Escara-Wilke J, et al. Targeting the MLL complex in castration-resistant prostate cancer. Nat Med. 2015;21:344–352. doi: 10.1038/nm.3830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Marx S, Spiegel AM, Skarulis MC, Doppman JL, Collins FS, Liotta LA. Multiple endocrine neoplasia type 1: clinical and genetic topics. Annals of internal medicine. 1998;129:484–494. doi: 10.7326/0003-4819-129-6-199809150-00011. [DOI] [PubMed] [Google Scholar]
  113. Marx SJ, Simonds WF. Hereditary hormone excess: genes, molecular pathways, and syndromes. Endocr Rev. 2005;26:615–661. doi: 10.1210/er.2003-0037. [DOI] [PubMed] [Google Scholar]
  114. Matkar S, Thiel A, Hua X. Menin: a scaffold protein that controls gene expression and cell signaling. Trends in biochemical sciences. 2013;38:394–402. doi: 10.1016/j.tibs.2013.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Matoso A, Zhou Z, Hayama R, Flesken-Nikitin A, Nikitin AY. Cell lineage-specific interactions between Men1 and Rb in neuroendocrine neoplasia. Carcinogenesis. 2008;29:620–628. doi: 10.1093/carcin/bgm207. [DOI] [PubMed] [Google Scholar]
  116. Murai MJ, Chruszcz M, Reddy G, Grembecka J, Cierpicki T. Crystal Structure of Menin Reveals Binding Site for Mixed Lineage Leukemia (MLL) Protein. The Journal of biological chemistry. 2011;286:31742–31748. doi: 10.1074/jbc.M111.258186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Nakamura Y, Larsson C, Julier C, Bystrom C, Skogseid B, Wells S, Oberg K, Carlson M, Taggart T, O'Connell P, et al. Localization of the genetic defect in multiple endocrine neoplasia type 1 within a small region of chromosome 11. Am J Hum Genet. 1989;44:751–755. [PMC free article] [PubMed] [Google Scholar]
  118. Newey PJ, Nesbit MA, Rimmer AJ, Attar M, Head RT, Christie PT, Gorvin CM, Stechman M, Gregory L, Mihai R, et al. Whole-exome sequencing studies of nonhereditary (sporadic) parathyroid adenomas. The Journal of clinical endocrinology and metabolism. 2012;97:E1995–2005. doi: 10.1210/jc.2012-2303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Newey PJ, Thakker RV. Role of multiple endocrine neoplasia type 1 mutational analysis in clinical practice. Endocr Pract. 2011;17(Suppl 3):8–17. doi: 10.4158/EP10379.RA. [DOI] [PubMed] [Google Scholar]
  120. Obungu VH, Lee Burns A, Agarwal SK, Chandrasekharapa SC, Adelstein RS, Marx SJ. Menin, a tumor suppressor, associates with nonmuscle myosin II-A heavy chain. Oncogene. 2003;22:6347–6358. doi: 10.1038/sj.onc.1206658. [DOI] [PubMed] [Google Scholar]
  121. Olufemi SE, Green JS, Manickam P, Guru SC, Agarwal SK, Kester MB, Dong Q, Burns AL, Spiegel AM, Marx SJ, et al. Common ancestral mutation in the MEN1 gene is likely responsible for the prolactinoma variant of MEN1 (MEN1Burin) in four kindreds from Newfoundland. Hum Mutat. 1998;11:264–269. doi: 10.1002/(SICI)1098-1004(1998)11:4<264::AID-HUMU2>3.0.CO;2-V. [DOI] [PubMed] [Google Scholar]
  122. Pellegata NS. MENX and MEN4. Clinics. 2012;67(Suppl 1):13–18. doi: 10.6061/clinics/2012(Sup01)04. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Pellegata NS, Quintanilla-Martinez L, Siggelkow H, Samson E, Bink K, Hofler H, Fend F, Graw J, Atkinson MJ. Germ-line mutations in p27Kip1 cause a multiple endocrine neoplasia syndrome in rats and humans. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:15558–15563. doi: 10.1073/pnas.0603877103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Quinn TJ, Yuan Z, Adem A, Geha R, Vrikshajanani C, Koba W, Fine E, Hughes DT, Schmid HA, Libutti SK. Pasireotide (SOM230) is effective for the treatment of pancreatic neuroendocrine tumors (PNETs) in a multiple endocrine neoplasia type 1 (MEN1) conditional knockout mouse model. Surgery. 2012;152:1068–1077. doi: 10.1016/j.surg.2012.08.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Ripoche D, Charbord J, Hennino A, Teinturier R, Bonnavion R, Jaafar R, Goehrig D, Cordier-Bussat M, Ritvos O, Zhang CX, et al. ActivinB is induced in Insulinoma to promote tumor plasticity through a beta-cell induced dedifferentiation. Molecular and cellular biology. 2015 doi: 10.1128/MCB.00930-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Sakurai A, Suzuki S, Kosugi S, Okamoto T, Uchino S, Miya A, Imai T, Kaji H, Komoto I, Miura D, et al. Multiple endocrine neoplasia type 1 in Japan: establishment and analysis of a multicentre database. Clinical endocrinology. 2012;76:533–539. doi: 10.1111/j.1365-2265.2011.04227.x. [DOI] [PubMed] [Google Scholar]
  127. Sawicki MP, Wan YJ, Johnson CL, Berenson J, Gatti R, Passaro E., Jr Loss of heterozygosity on chromosome 11 in sporadic gastrinomas. Hum Genet. 1992;89:445–449. doi: 10.1007/BF00194320. [DOI] [PubMed] [Google Scholar]
  128. Scacheri PC, Crabtree JS, Kennedy AL, Swain GP, Ward JM, Marx SJ, Spiegel AM, Collins FS. Homozygous loss of menin is well tolerated in liver, a tissue not affected in MEN1. Mammalian genome : official journal of the International Mammalian Genome Society. 2004;15:872–877. doi: 10.1007/s00335-004-2395-z. [DOI] [PubMed] [Google Scholar]
  129. Scacheri PC, Davis S, Odom DT, Crawford GE, Perkins S, Halawi MJ, Agarwal SK, Marx SJ, Spiegel AM, Meltzer PS, et al. Genome-wide analysis of menin binding provides insights into MEN1 tumorigenesis. PLoS genetics. 2006;2:e51. doi: 10.1371/journal.pgen.0020051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Scarpa A, Chang DK, Nones K, Corbo V, Patch AM, Bailey P, Lawlor RT, Johns AL, Miller DK, Mafficini A, et al. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature. 543:65–71. doi: 10.1038/nature21063. [DOI] [PubMed] [Google Scholar]
  131. Schnepp RW, Hou Z, Wang H, Petersen C, Silva A, Masai H, Hua X. Functional interaction between tumor suppressor menin and activator of S-phase kinase. Cancer research. 2004;64:6791–6796. doi: 10.1158/0008-5472.CAN-04-0724. [DOI] [PubMed] [Google Scholar]
  132. Shattuck TM, Valimaki S, Obara T, Gaz RD, Clark OH, Shoback D, Wierman ME, Tojo K, Robbins CM, Carpten JD, et al. Somatic and germ-line mutations of the HRPT2 gene in sporadic parathyroid carcinoma. N Engl J Med. 2003;349:1722–1729. doi: 10.1056/NEJMoa031237. [DOI] [PubMed] [Google Scholar]
  133. Shen HC, He M, Powell A, Adem A, Lorang D, Heller C, Grover AC, Ylaya K, Hewitt SM, Marx SJ, et al. Recapitulation of pancreatic neuroendocrine tumors in human multiple endocrine neoplasia type I syndrome via Pdx1-directed inactivation of Men1. Cancer research. 2009;69:1858–1866. doi: 10.1158/0008-5472.CAN-08-3662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Shen HC, Ylaya K, Pechhold K, Wilson A, Adem A, Hewitt SM, Libutti SK. Multiple endocrine neoplasia type 1 deletion in pancreatic alpha-cells leads to development of insulinomas in mice. Endocrinology. 2010;151:4024–4030. doi: 10.1210/en.2009-1251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Shi K, Parekh VI, Roy S, Desai SS, Agarwal SK. The embryonic transcription factor Hlxb9 is a menin interacting partner that controls pancreatic beta-cell proliferation and the expression of insulin regulators. Endocrine-related cancer. 2013;20:111–122. doi: 10.1530/ERC-12-0077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Shilatifard A. The COMPASS family of histone H3K4 methylases: mechanisms of regulation in development and disease pathogenesis. Annu Rev Biochem. 2012;81:65–95. doi: 10.1146/annurev-biochem-051710-134100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Simonds WF, James-Newton LA, Agarwal SK, Yang B, Skarulis MC, Hendy GN, Marx SJ. Familial isolated hyperparathyroidism: clinical and genetic characteristics of 36 kindreds. Medicine (Baltimore) 2002;81:1–26. doi: 10.1097/00005792-200201000-00001. [DOI] [PubMed] [Google Scholar]
  138. Sowa H, Kaji H, Hendy GN, Canaff L, Komori T, Sugimoto T, Chihara K. Menin is required for bone morphogenetic protein 2- and transforming growth factor beta-regulated osteoblastic differentiation through interaction with Smads and Runx2. The Journal of biological chemistry. 2004;279:40267–40275. doi: 10.1074/jbc.M401312200. [DOI] [PubMed] [Google Scholar]
  139. Stratakis CA, Schussheim DH, Freedman SM, Keil MF, Pack SD, Agarwal SK, Skarulis MC, Weil RJ, Lubensky IA, Zhuang Z, et al. Pituitary macroadenoma in a 5-year-old: an early expression of multiple endocrine neoplasia type 1. The Journal of clinical endocrinology and metabolism. 2000;85:4776–4780. doi: 10.1210/jcem.85.12.7064. [DOI] [PubMed] [Google Scholar]
  140. Sukhodolets KE, Hickman AB, Agarwal SK, Sukhodolets MV, Obungu VH, Novotny EA, Crabtree JS, Chandrasekharappa SC, Collins FS, Spiegel AM, et al. The 32-kilodalton subunit of replication protein A interacts with menin, the product of the MEN1 tumor suppressor gene. Molecular and cellular biology. 2003;23:493–509. doi: 10.1128/MCB.23.2.493-509.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Sundaresan S, Kang AJ, Hayes MM, Choi EK, Merchant JL. Deletion of Men1 and somatostatin induces hypergastrinemia and gastric carcinoids. Gut. 2016 doi: 10.1136/gutjnl-2015-310928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Svoboda LK, Bailey N, Van Noord RA, Krook MA, Harris A, Cramer C, Jasman B, Patel RM, Thomas D, Borkin D, et al. Tumorigenicity of Ewing sarcoma is critically dependent on the trithorax proteins MLL1 and menin. Oncotarget. 2017;8:458–471. doi: 10.18632/oncotarget.13444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Thakker RV. Multiple endocrine neoplasia type 1 (MEN1) Best practice & research. Clinical endocrinology & metabolism. 2010;24:355–370. doi: 10.1016/j.beem.2010.07.003. [DOI] [PubMed] [Google Scholar]
  144. Thakker RV. Multiple endocrine neoplasia type 1 (MEN1) and type 4 (MEN4) Molecular and cellular endocrinology. 2014;386:2–15. doi: 10.1016/j.mce.2013.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Thakker RV, Bouloux P, Wooding C, Chotai K, Broad PM, Spurr NK, Besser GM, O'Riordan JL. Association of parathyroid tumors in multiple endocrine neoplasia type 1 with loss of alleles on chromosome 11. N Engl J Med. 1989;321:218–224. doi: 10.1056/NEJM198907273210403. [DOI] [PubMed] [Google Scholar]
  146. Thakker RV, Newey PJ, Walls GV, Bilezikian J, Dralle H, Ebeling PR, Melmed S, Sakurai A, Tonelli F, Brandi ML. Clinical practice guidelines for multiple endocrine neoplasia type 1 (MEN1) The Journal of clinical endocrinology and metabolism. 2012;97:2990–3011. doi: 10.1210/jc.2012-1230. [DOI] [PubMed] [Google Scholar]
  147. Tham E, Grandell U, Lindgren E, Toss G, Skogseid B, Nordenskjold M. Clinical testing for mutations in the MEN1 gene in Sweden: a report on 200 unrelated cases. J Clin Endocrinol Metab. 2007;92:3389–3395. doi: 10.1210/jc.2007-0476. [DOI] [PubMed] [Google Scholar]
  148. Thevenon J, Bourredjem A, Faivre L, Cardot-Bauters C, Calender A, Le Bras M, Giraud S, Niccoli P, Odou MF, Borson-Chazot F, et al. Unraveling the intrafamilial correlations and heritability of tumor types in MEN1: a Groupe d'etude des Tumeurs Endocrines study. Eur J Endocrinol. 2015;173:819–826. doi: 10.1530/EJE-15-0691. [DOI] [PubMed] [Google Scholar]
  149. Trivellin G, Daly AF, Faucz FR, Yuan B, Rostomyan L, Larco DO, Schernthaner-Reiter MH, Szarek E, Leal LF, Caberg JH, et al. Gigantism and acromegaly due to Xq26 microduplications and GPR101 mutation. N Engl J Med. 2014;371:2363–2374. doi: 10.1056/NEJMoa1408028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. van Leeuwaarde RS, Dreijerink K, Ausems MG, Beijers HJ, Dekkers OM, de Herder WW, van der Horst-Schrivers AN, Drent ML, Bisschop PH, Havekes B, et al. MEN1-dependent breast cancer: indication for early screening? Results from the Dutch MEN1 study group. J Clin Endocrinol Metab. 2017 doi: 10.1210/jc.2016-3690. [DOI] [PubMed] [Google Scholar]
  151. Veniaminova NA, Hayes MM, Varney JM, Merchant JL. Conditional deletion of menin results in antral G cell hyperplasia and hypergastrinemia. American journal of physiology. Gastrointestinal and liver physiology. 2012;303:G752–764. doi: 10.1152/ajpgi.00109.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Vierimaa O, Ebeling TM, Kytola S, Bloigu R, Eloranta E, Salmi J, Korpi-Hyovalti E, Niskanen L, Orvola A, Elovaara E, et al. Multiple endocrine neoplasia type 1 in Northern Finland; clinical features and genotype phenotype correlation. Eur J Endocrinol. 2007;157:285–294. doi: 10.1530/EJE-07-0195. [DOI] [PubMed] [Google Scholar]
  153. Vierimaa O, Georgitsi M, Lehtonen R, Vahteristo P, Kokko A, Raitila A, Tuppurainen K, Ebeling TM, Salmela PI, Paschke R, et al. Pituitary adenoma predisposition caused by germline mutations in the AIP gene. Science. 2006;312:1228–1230. doi: 10.1126/science.1126100. [DOI] [PubMed] [Google Scholar]
  154. Vijayaraghavan J, Maggi EC, Crabtree JS. miR-24 regulates menin in the endocrine pancreas. Am J Physiol Endocrinol Metab. 2014;307:E84–92. doi: 10.1152/ajpendo.00542.2013. [DOI] [PubMed] [Google Scholar]
  155. Walls GV, Lemos MC, Javid M, Bazan-Peregrino M, Jeyabalan J, Reed AA, Harding B, Tyler DJ, Stuckey DJ, Piret S, et al. MEN1 gene replacement therapy reduces proliferation rates in a mouse model of pituitary adenomas. Cancer research. 2012;72:5060–5068. doi: 10.1158/0008-5472.CAN-12-1821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Walls GV, Stevenson M, Soukup BS, Lines KE, Grossman AB, Schmid HA, Thakker RV. Pasireotide Therapy of Multiple Endocrine Neoplasia Type 1-Associated Neuroendocrine Tumors in Female Mice Deleted for an Men1 Allele Improves Survival and Reduces Tumor Progression. Endocrinology. 2016;157:1789–1798. doi: 10.1210/en.2015-1965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Wang Y, Ozawa A, Zaman S, Prasad NB, Chandrasekharappa SC, Agarwal SK, Marx SJ. The tumor suppressor protein menin inhibits AKT activation by regulating its cellular localization. Cancer research. 2011;71:371–382. doi: 10.1158/0008-5472.CAN-10-3221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Wuescher L, Angevine K, Hinds T, Ramakrishnan S, Najjar SM, Mensah-Osman EJ. Insulin regulates menin expression, cytoplasmic localization, and interaction with FOXO1. Am J Physiol Endocrinol Metab. 2011;301:E474–483. doi: 10.1152/ajpendo.00022.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Yaguchi H, Ohkura N, Takahashi M, Nagamura Y, Kitabayashi I, Tsukada T. Menin missense mutants associated with multiple endocrine neoplasia type 1 are rapidly degraded via the ubiquitin-proteasome pathway. Molecular and cellular biology. 2004;24:6569–6580. doi: 10.1128/MCB.24.15.6569-6580.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Yaguchi H, Ohkura N, Tsukada T, Yamaguchi K. 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. 2002;277:38197–38204. doi: 10.1074/jbc.M204132200. [DOI] [PubMed] [Google Scholar]
  161. Yan J, Yang Y, Zhang H, King C, Kan HM, Cai Y, Yuan CX, Bloom GS, Hua X. Menin interacts with IQGAP1 to enhance intercellular adhesion of beta-cells. Oncogene. 2009;28:973–982. doi: 10.1038/onc.2008.435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Yang YJ, Song TY, Park J, Lee J, Lim J, Jang H, Kim YN, Yang JH, Song Y, Choi A, et al. Menin mediates epigenetic regulation via histone H3 lysine 9 methylation. Cell Death Dis. 2013;4:e583. doi: 10.1038/cddis.2013.98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Yokoyama A. Transcriptional activation by MLL fusion proteins in leukemogenesis. Exp Hematol. 2017;46:21–30. doi: 10.1016/j.exphem.2016.10.014. [DOI] [PubMed] [Google Scholar]
  164. Yokoyama A, Cleary ML. Menin critically links MLL proteins with LEDGF on cancer-associated target genes. Cancer Cell. 2008;14:36–46. doi: 10.1016/j.ccr.2008.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Yokoyama A, Somervaille TC, Smith KS, Rozenblatt-Rosen O, Meyerson M, Cleary ML. The menin tumor suppressor protein is an essential oncogenic cofactor for MLL-associated leukemogenesis. Cell. 2005;123:207–218. doi: 10.1016/j.cell.2005.09.025. [DOI] [PubMed] [Google Scholar]
  166. Zaman S, Sukhodolets K, Wang P, Qin J, Levens D, Agarwal SK, Marx SJ. FBP1 Is an Interacting Partner of Menin. Int J Endocrinol. 2014;2014:535401. doi: 10.1155/2014/535401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Zhuang Z, Vortmeyer AO, Pack S, Huang S, Pham TA, Wang C, Park WS, Agarwal SK, Debelenko LV, Kester M, et al. Somatic mutations of the MEN1 tumor suppressor gene in sporadic gastrinomas and insulinomas. Cancer research. 1997;57:4682–4686. [PubMed] [Google Scholar]

RESOURCES