Fact-checked by Grok 2 weeks ago

MEN1

Multiple endocrine neoplasia type 1 (MEN1) is a rare inherited disorder caused by mutations in the MEN1 gene on chromosome 11q13, leading to the development of multiple tumors in endocrine glands, most commonly the parathyroid glands, the endocrine pancreas, and the gland. This autosomal dominant condition, also known as Wermer's syndrome, has a prevalence of approximately 1 in 30,000 individuals and affects males and females equally, with symptoms often first appearing in the second or third decade of life; approximately 10% of cases result from mutations. The MEN1 gene encodes the protein menin, a tumor suppressor that regulates and division; inactivating mutations disrupt this function, resulting in tumor formation, though most tumors are benign but can become functionally active or, rarely, malignant. The hallmark of MEN1 is , occurring in over 90% of patients by age 50, often presenting as multiglandular parathyroid adenomas or that cause hypercalcemia, leading to symptoms such as fatigue, kidney stones, , and gastrointestinal issues. Enteropancreatic neuroendocrine tumors affect 30-80% of individuals, including gastrinomas (causing Zollinger-Ellison syndrome with peptic ulcers and diarrhea) and insulinomas (resulting in ), while pituitary adenomas occur in about 30-40% of cases, frequently prolactinomas that may cause , , or headaches. Less common manifestations include adrenal cortical tumors, thymic or bronchial carcinoids, and facial angiofibromas, with the syndrome's high —reaching 95% by age 40—necessitating lifelong surveillance. Diagnosis of MEN1 relies on clinical criteria, including the presence of two or more endocrine tumors associated with the syndrome, a family history of MEN1 in a first-degree relative, or identification of a MEN1 mutation through , which is recommended for at-risk individuals starting at age 5-10. Management focuses on early detection via regular biochemical screening (e.g., calcium, , ) and imaging (e.g., MRI, ), with treatments such as surgical resection for symptomatic tumors, medical therapies for excess, and for families, as there is no cure; early detection and appropriate management have substantially improved , though it remains reduced compared to the general population, with recent studies showing that only about 28% of deaths in recognized and treated patients are directly related to MEN1.

Gene Overview

Location and Structure

The MEN1 gene is located on the long arm of chromosome 11 at the q13.1 band. Its genomic coordinates, according to the GRCh38 assembly, span from 64,803,516 to 64,811,294, encompassing approximately 8 kb of DNA. This positioning was confirmed through linkage studies and positional cloning efforts that mapped the gene to this region based on familial inheritance patterns of multiple endocrine neoplasia type 1. The gene consists of 10 s separated by 9 introns, with the overall genomic structure spanning about 9 kb, including regulatory elements. 1 is entirely untranslated, and the coding sequence begins in 2 with the initiation codon, extending through 2 to 10 to encode a 610-amino-acid protein known as menin; the 3' portion of 10 (approximately 832 ) serves as the . The intron- boundaries are precisely defined, with splice sites following the GT-AG consensus rule, facilitating accurate pre-mRNA processing. The promoter region lies upstream of exon 1 and includes Alu-type repetitive elements, which are short interspersed nuclear elements characteristic of primate genomes and contribute to transcriptional regulation. This promoter drives the expression of a major 2.8-kb mRNA transcript, with transcription initiation potentially occurring within exon 1. The Alu elements in the upstream regulatory region represent a human-specific , as they are absent in non-primate orthologs, though the core coding sequence shows high conservation. Evolutionarily, the MEN1 gene is highly conserved across , with orthologs identified in vertebrates such as mice (on ) and , as well as like fruit flies and snails, indicating an ancient role in cellular processes; however, it is absent in simpler organisms like and . The exon-intron organization, particularly exons 2 through 9, is well preserved between and murine genomes, underscoring functional importance.

Transcription and Regulation

The MEN1 undergoes primary transcript processing that includes , generating multiple isoforms with potential functional diversity. The predominant isoform, MENIN isoform 2, consists of 610 and is considered the reference transcript, while events occur across the entire , including a novel in 7 that is highly expressed in various tissues. These events produce isoforms featuring in-frame insertions, deletions, or unique amino termini, with at least four alternative splice variants identified through 5' and RT-PCR analyses. Such isoform diversity may influence menin's interactions with partner proteins, though the functional implications in normal physiology remain under investigation. Regulatory elements controlling MEN1 transcription include a minimal promoter region spanning -135 to -36 base pairs upstream of exon 2, which contains initiator (Inr) elements and is modulated by upstream cis-regulatory sequences such as UR1-UR5 and smaller elements C1-C5 located between -325 and -107. Promoter activity exhibits cell-type specificity, with reduced function in endocrine-derived cells compared to non-endocrine lines, and is subject to negative feedback from menin itself, as overexpression in Men1-null fibroblasts dose-dependently suppresses promoter-driven reporter activity. Epigenetic modifications, including promoter hypermethylation, contribute to downregulated MEN1 expression, as observed in pancreatic ductal adenocarcinomas where methylation correlates with decreased menin levels in 5 of 24 tumors, and in pancreatic neuroendocrine tumors where it occurs frequently. Additionally, histone modifications play a role in regulation; for instance, menin interacts with SUV39H1 to promote H3K9 trimethylation at target loci, and ChIP-seq studies have mapped menin binding to distal enhancers that loop to promoters, influencing chromatin accessibility, though direct application to MEN1 autoregulation requires further validation. MEN1 exhibits ubiquitous basal expression across human tissues but shows elevated levels in endocrine organs, consistent with its role in neuroendocrine . reveals widespread MEN1 transcripts in adult tissues, with enhanced expression in proliferating cells entering , suggesting cell cycle-linked transcriptional control. Quantitative data from the GTEx consortium indicate median transcripts per million (TPM) values of approximately 1.8 in the , 1.3 in the and , and 0.85 in (GTEx Analysis Release V10, dbGaP Accession phs000424.v10.p2), underscoring higher expression in these tissues relative to non-endocrine organs like . This pattern aligns with the gene's association with endocrine tumorigenesis when dysregulated.

Menin Protein

Structure and Domains

Menin is a 610-amino acid nuclear scaffold protein encoded by the MEN1 gene, lacking any transmembrane domains and featuring intrinsically disordered regions (IDRs), including one flanked by two nuclear localization signals (NLSs) in the C-terminal half that facilitate its nuclear import. The protein's folded core adopts a distinctive curved left-hand , consisting of an N-terminal domain formed by a β-hairpin, a central palm domain with a deep binding pocket, and C-terminal finger-like extensions. This overall , resolved in crystal form at 2.1–2.9 resolution (e.g., PDB entry 3U84 for unbound menin), lacks to other known eukaryotic protein domains beyond the NLSs. Key structural features include the C-terminal NLSs, which ensure localization, and interaction domains within the that bind partners like the mixed-lineage leukemia (MLL) complex via conserved motifs such as FPX XP. structures of menin-MLL1 complexes (e.g., PDB 3U85) highlight coordinates for critical residues, including Arg-392 and Gln-359 in the pocket, which stabilize MLL binding through hydrogen bonds and hydrophobic interactions. A cryo-electron microscopy structure at 3.2 Å resolution (PDB 8GPN) captures menin in complex with a nucleosome bearing histone H3 dimethylated at lysine 79 (H3K79me2), revealing multivalent contacts where the palm and finger domains engage the histone H3 tail via a π-cation interaction involving Arg-334 and the methylated lysine. Post-translational modifications of menin include phosphorylation primarily at serine/threonine residues, such as the constitutive site at Ser-543, Ser-583, and dynamic sites like Ser-394 (induced by ionizing radiation or UV) and Ser-487, which occur without altering core stability or localization. Ubiquitination involves polyubiquitin chains that target menin for proteasomal degradation, with patterns enhanced by MEN1 mutations disrupting stabilizing interactions.

Cellular Functions

Menin functions as a tumor suppressor by regulating through the transcriptional activation of inhibitors (CDKIs), particularly p27^{Kip1} and p18^{Ink4c}. These inhibitors enforce arrest at the [G1 phase](/page/G1 phase) by binding and inhibiting complexes, thereby preventing uncontrolled cellular growth. Menin achieves this regulation by recruiting the mixed-lineage leukemia (MLL) complex to the promoter and coding regions of the CDKN1B (encoding p27^{Kip1}) and CDKN2C (encoding p18^{Ink4c}) genes, facilitating their expression in a menin-dependent manner. Experimental evidence from Men1-null embryonic fibroblasts demonstrates that re-expression of wild-type menin increases p27^{Kip1} and p18^{Ink4c} transcription by 4- to 5-fold, as measured by reporter assays, while menin mutants associated with MEN1 syndrome (e.g., L22R, A242V) fail to do so due to impaired DNA binding. In hyperplastic parathyroid glands from MEN1 patients, the percentage of p27^{Kip1}-negative cells is increased (34.4% vs. 20% in normal tissues; p=0.11), underscoring menin's role in maintaining proliferative control. Beyond direct transcriptional effects, menin's tumor suppressive activity is reinforced by its epigenetic modifications that promote the expression of these CDKIs, linking proliferation control to . In mouse models, loss of menin leads to down-regulation of p27^{Kip1} and p18^{Ink4c}, resulting in deregulated , as observed in islet cell hyperplasia. This mechanism highlights menin's essential role in balancing cellular growth, with p18^{Ink4c} showing particularly strong collaboration with menin in suppressing neuroendocrine proliferation, as evidenced by accelerated tumorigenesis in combined Men1 and Cdkn2c knockout mice. Menin plays a critical epigenetic role by interacting with histone methyltransferases MLL1 and MLL2, enabling trimethylation of at 4 (H3K4me3), an active mark that promotes transcription. As a , menin stabilizes the MLL1/2 complexes at target promoters, facilitating H3K4 and subsequent opening for transcriptional machinery access. This process is vital for activating genes involved in and , such as Hox cluster genes, where menin ensures stable long-term expression during embryogenesis. Genome-wide studies in menin-null mouse embryonic stem cells reveal that menin is required for H3K4me3 at nearly all Hox loci, with loss leading to profound reductions in and . The is direct and specific to MLL1/2 among the MLL family, as demonstrated by co-immunoprecipitation assays showing menin's binding to the MLL1 SET domain, which catalyzes the reaction. This epigenetic function extends to broader activation, where menin-MLL complexes maintain H3K4me3 patterns at enhancers and promoters, supporting cellular identity and preventing oncogenic . In addition to transcriptional activation, menin's epigenetic contributions ensure precise control of landscapes, with levels directly correlating with menin occupancy in ChIP-seq analyses across multiple cell types. Disruption of menin-MLL binding, as modeled by small-molecule inhibitors, rapidly reduces at target loci, confirming the complex's necessity for methylation fidelity. These roles position menin as a key of epigenetic , influencing long-range interactions essential for normal cellular function. Menin contributes to and genome stability by stimulating (HR), a high-fidelity pathway for repairing double-strand breaks (DSBs). Menin accumulates at DSB sites in response to DNA damage, where it interacts with checkpoint Chk1 to enhance HR efficiency. In human cells engineered with the DR-GFP/I-SceI reporter system, overexpression of menin increases HR-mediated repair by promoting GFP expression in DSB-induced cells, as quantified by (significant increase in double-positive cells, p<0.05). This stimulation depends on menin's N-terminal domain, as deletion mutants (ΔNH₂) abolish the interaction with Chk1 and repair enhancement. and re-ChIP experiments further show menin and Chk1 co-localize at I-SceI-induced breaks, with menin facilitating the recruitment of HR factors. The process involves the ATM/ATR signaling pathway, as inhibition (an ATM/ATR blocker) reduces menin-stimulated repair by approximately 90%. These findings indicate menin's role in maintaining genomic integrity by prioritizing error-free HR over mutagenic alternatives like . Menin's involvement in HR extends to broader genome stability, where its absence sensitizes cells to DSB-inducing agents, as evidenced by increased γ-H2AX foci in menin-depleted lines. By modulating Chk1 activity at damage sites, menin ensures timely resolution of replication-associated breaks, preventing chromosomal aberrations. This function is conserved across cell types, with menin promoting HR in both cycling and quiescent cells. Menin participates in telomere maintenance by regulating telomerase activity, primarily through transcriptional repression of the human telomerase reverse transcriptase (hTERT) , which encodes the catalytic subunit of telomerase. Menin binds to the hTERT promoter in a sequence-independent manner, inhibiting its activity and thereby limiting telomere elongation in normal cells. reporter assays in various cell lines demonstrate that menin overexpression reduces hTERT promoter activity in a cell-type-specific , while siRNA-mediated knockdown of menin fails to significantly up-regulate hTERT mRNA, suggesting indirect or context-dependent regulation. In experimental models, menin knockdown in HEK293T cells promotes telomerase component (TERC) localization to s, slowing telomere erosion rates as measured by telomere restriction fragment analysis. This indicates that menin normally disrupts telomerase assembly or access, contributing to controlled telomere shortening. Evidence from Men1 models supports this, where homozygous null islets exhibit altered telomerase dynamics and mild telomere instability, though not leading to immediate length aberrations. Additionally, menin localizes to s during , as shown by in spermatocytes, where it associates with telomeric repeats without altering overall telomerase activity in overexpressing cells. These observations highlight menin's role in fine-tuning telomerase to prevent excessive telomere maintenance, which could otherwise promote replicative . Menin also regulates T cell and function, influencing immune responses through transcriptional control.

Role in Disease

Association with MEN1 Syndrome

Multiple endocrine neoplasia type 1 (MEN1) syndrome is a rare autosomal dominant hereditary disorder resulting from germline mutations in the MEN1 gene, which encodes the tumor suppressor protein menin. The syndrome exhibits near-complete , with over 95% of mutation carriers developing clinical manifestations by age 40 and approaching 100% by age 50. Its is estimated at approximately 1 in 30,000 individuals, though reported rates vary from 3 to 20 per 100,000 depending on population and diagnostic criteria. About 10% of cases arise from mutations, meaning affected individuals have no family history of the syndrome, while the remainder are inherited from an affected parent with a 50% transmission risk per offspring. The hallmark features of MEN1 syndrome involve multifocal tumors primarily affecting the parathyroid glands, , and enteropancreatic neuroendocrine tissues, often leading to endocrine dysfunction. Parathyroid involvement manifests as in over 90% of cases by age 50, typically due to multiglandular or adenomas causing hypercalcemia. Pituitary adenomas occur in 30-40% of patients, commonly prolactinomas or non-functioning tumors that may result in hormonal excesses or mass effects. Enteropancreatic tumors, seen in 30-70% of cases, include gastrinomas (leading to Zollinger-Ellison syndrome with peptic ulcers) and insulinomas, among others, which can be functional or malignant. These manifestations usually emerge in adulthood, with parathyroid tumors appearing earliest (average onset in the 20s), followed by enteropancreatic and pituitary lesions. The pathogenesis of MEN1 syndrome adheres to Knudson's for tumor suppressor genes, where the inherited MEN1 represents the first hit, inactivating one in all cells. Tumor formation requires a second somatic hit, typically (LOH) at 11q13 encompassing the wild-type MEN1 , observed in over 90% of affected tumors. This biallelic inactivation disrupts menin function, promoting uncontrolled in endocrine tissues.

Involvement in Sporadic Cancers

Somatic inactivating in the MEN1 gene occur in 20-40% of sporadic parathyroid adenomas, contributing to tumorigenesis independent of hereditary syndromes. These often lead to loss of menin function, promoting uncontrolled in parathyroid chief cells. Similarly, approximately 30% of sporadic gastrinomas harbor MEN1 , frequently accompanied by allelic deletions, which drive Zollinger-Ellison syndrome-like tumor development. In non-endocrine sporadic cancers, MEN1 alterations play emerging roles; for instance, reduced menin expression is observed in sporadic breast cancers, where it interacts with to influence tumor progression, though direct mutations are infrequent. In sporadic cancers, particularly castration-resistant forms, MEN1 mutations occur in about 17% of cases and may exert an oncogenic effect by enhancing signaling. Loss-of-heterozygosity (LOH) at the locus encompassing MEN1 is a common mechanism across various sporadic malignancies, including parathyroid adenomas (26-37% frequency), tumors, and carcinoids, facilitating biallelic inactivation. In sporadic neuroendocrine tumors (NETs), MEN1 somatic alterations provide prognostic insights; The Cancer Genome Atlas (TCGA) analyses of pancreatic NETs reveal MEN1 mutations in 37% of cases, often without direct survival correlation but associated with distinct molecular subtypes influencing therapeutic response. Specifically, in sporadic thymic carcinoids, MEN1 mutations and 11q losses are frequent, correlating with heightened malignant potential, local invasion, and early , as highlighted in 2025 molecular reviews.

Mutations and Variants

Types of Pathogenic Mutations

Pathogenic mutations in the MEN1 gene primarily lead to loss of function of the menin protein and are responsible for the majority of (MEN1) cases. As of 2025, more than 1,800 distinct have been reported in the literature, cataloged in specialized databases such as the Open Variation Database (LOVD) and the Universal Mutation Database (UMD-MEN1). These are evenly distributed across the 10 exons of the gene, with relative hotspots observed in exons 2 and 10. The spectrum of pathogenic mutations includes several major types, with frameshift insertions and deletions being the most prevalent at approximately 40-50%, followed by missense mutations (20-25%), nonsense mutations (10-20%), and splice-site alterations (7-11%). Frameshift and nonsense mutations typically result in premature termination codons and truncated proteins, while missense mutations involve single substitutions that may disrupt protein function. Splice-site mutations affect intron-exon boundaries, leading to aberrant mRNA processing. Large genomic deletions, encompassing one or more exons, represent about 5% of cases and are often identified through (MLPA) rather than standard sequencing. Variant classification follows the American College of Medical Genetics and (ACMG) guidelines, distinguishing pathogenic/likely pathogenic from benign/likely benign ones based on criteria such as population frequency, computational predictions, functional assays, and segregation data. Missense , in particular, pose classification challenges and frequently require additional evidence like tools or studies to confirm pathogenicity. Recent case reports continue to identify novel , including double-substitution that alter multiple adjacent and contribute to the expanding genetic landscape.

Genotype-Phenotype Correlations

Although no definitive genotype-phenotype correlations have been established for overall, certain patterns have emerged from large studies, particularly linking type to disease severity and presentation. Truncating , such as nonsense and frameshift variants, are associated with earlier disease onset and greater tumor multiplicity compared to non-truncating , likely due to complete loss of menin function. In contrast, missense , especially those affecting specific domains like the nuclear localization signal (NLS), have been linked to atypical or less penetrant features, including isolated or unusual tumor types such as macro-prolactinomas. Epidemiological studies from diverse cohorts provide supporting evidence for these associations. For instance, in a multicenter of over 100 Korean patients, truncating mutations correlated with a significantly higher prevalence of pituitary neuroendocrine tumors (59.7% vs. 36.0% in non-truncating cases). Similarly, Italian cohort data indicate that and frameshift mutations are more strongly tied to aggressive gastroenteropancreatic neuroendocrine tumors (GEP-NETs) than missense variants. Disease and expression in MEN1 are also influenced by genetic modifiers, including loss of the wild-type (second hit) via mechanisms like , which is observed in nearly all MEN1-associated tumors and drives tumorigenesis. Environmental factors, such as dietary influences on , may interact with mutations to modulate , though evidence remains limited and indirect. Founder effects in specific populations can further shape phenotypic variability; for example, recurrent mutations in cohorts have been associated with distinct patterns of tumor distribution and earlier onset in affected families, highlighting the role of population-specific .

Molecular Interactions

Protein-Protein Interactions

Menin, the protein encoded by the MEN1 gene, participates in a wide array of protein-protein interactions that underpin its function in cellular processes. High-throughput approaches such as two-hybrid (Y2H) screening and affinity purification-mass (AP-MS) have mapped an extensive interactome, identifying over 50 potential binding partners, which underscore menin's integration into diverse multiprotein complexes. A primary set of interactors involves the mixed-lineage family proteins MLL1 and MLL2, which are methyltransferases central to H3K4 and epigenetic regulation. Menin binds directly to the N-terminal region of MLL1 via two distinct menin-binding motifs (MBM1 spanning residues 4-15 and MBM2 spanning residues 23-40), forming a high-affinity complex with a (Kd) of approximately 10 nM. This interaction has been robustly demonstrated through co-immunoprecipitation assays in and epithelial cell lines, where menin stabilizes MLL1 recruitment to promoters without altering its enzymatic activity. Similar binding occurs with MLL2, sharing the same structural motifs and affinity profile, facilitating cooperative epigenetic modification. Menin also forms a direct interaction with SMAD3, the receptor-regulated in the TGF-β signaling pathway. This association promotes SMAD3-dependent activation of growth-inhibitory genes and has been confirmed by co-immunoprecipitation in cells, specific to SMAD3 without involving SMAD2 or SMAD4. The binding requires the MH2 domain of SMAD3. In the context of , menin interacts with RPA2, the 32-kDa subunit of the heterotrimer, which coats single-stranded DNA during replication and repair processes. This partnership was first identified via Y2H screening reconstituting Ras signaling and subsequently validated by co-immunoprecipitation in human cell lysates, revealing menin's recruitment to DNA damage sites to support . Experimental studies indicate that RPA2 engages menin's N-terminal region (up to residue 286). These interactions display tissue-specific modulation, with enhanced binding affinities and stability observed in endocrine tissues such as the and pituitary, where menin’s associations with MLL1/2 and SMAD3 are amplified to enforce control and suppress .

Signaling Pathways

Menin, the protein encoded by the MEN1 gene, serves as a critical scaffold in multiple cellular signaling pathways, primarily exerting tumor-suppressive effects through and epigenetic modifications. As a component of the mixed-lineage leukemia (MLL) complex, menin facilitates lysine 4 trimethylation (), which activates in a context-dependent manner, while also repressing oncogenic signals in pathways such as Wnt/β-catenin and . Dysregulation of these pathways due to MEN1 loss promotes tumorigenesis, particularly in endocrine tissues, by altering , , and metabolic signaling. In the Wnt/β-catenin pathway, menin functions as a transcriptional repressor by directly interacting with β-catenin, reducing its nuclear accumulation and thereby inhibiting downstream target gene activation. This repression is evident in mouse embryonic fibroblasts and islet β-cells, where menin overexpression suppresses β-catenin-mediated transcription, limiting . Conversely, MEN1 deficiency in murine models leads to β-catenin stabilization and pathway hyperactivation, driving the expression of proliferative genes like Axin2 and contributing to formation. This repressive role underscores menin's integration into Wnt signaling to maintain cellular homeostasis. Menin also antagonizes signaling, a pathway involved in and oncogenesis, by epigenetically repressing key effectors such as Gli1. Through recruitment of the protein arginine methyltransferase PRMT5, menin dimethylates arginine 2 (H3R2me2), which competes with and silences Hedgehog target genes in and MEN1-associated tumors. Ablation of menin in murine models enhances Hedgehog pathway activity, promoting islet cell proliferation and tumor progression, independent of canonical ligand-receptor interactions. Regarding apoptosis regulation, menin interacts with the JunD to repress its transcriptional activity, thereby inhibiting cell growth and promoting . In murine embryonic fibroblasts, menin overexpression induces by blocking JunD-mediated anti-apoptotic , while menin-JunD binding prevents JunD by JNK . Loss of this interaction shifts JunD from a growth suppressor to a promoter, facilitating in MEN1-deficient cells. Pathway dysregulation in MEN1 loss is exemplified by elevated c-Myc expression in neuroendocrine tumors, where menin normally represses c-Myc transcription via modulation at its promoter. In pancreatic models, Men1 upregulates c-Myc mRNA, enhancing and neoplastic . This effect integrates with cross-talk to the PI3K/AKT pathway, where menin inhibits AKT activation by regulating its cellular localization, suppressing AKT1-driven and anti-apoptotic signals in endocrine and non-endocrine cells. Consequently, MEN1 deficiency amplifies PI3K/AKT signaling, promoting tumor progression through enhanced survival and metabolic reprogramming. As of 2025, therapeutic targeting of menin protein-protein interactions, such as the menin-MLL interface with small-molecule inhibitors, has advanced understanding of these complexes in and endocrine tumors.

History and Research

Discovery and Cloning

The (MEN1) syndrome, also known as Wermer syndrome, was first linked to chromosome through genetic studies in affected families during the . In 1988, Larsson et al. demonstrated tight linkage between the MEN1 locus and the muscle gene (PYGM) on in a large pedigree, establishing the chromosomal location for further efforts. Subsequent linkage analyses in additional families refined the candidate to approximately a 400-kilobase interval within , supported by (LOH) observations in MEN1-associated tumors indicating a tumor suppressor mechanism. Positional cloning of the MEN1 was achieved in 1997 by two independent groups using complementary strategies centered on LOH analysis and genomic sequencing within the refined interval. The U.S. (NIH) team, led by Chandrasekharappa et al., identified the by systematically sequencing candidate transcripts in the minimal LOH region, isolating a 2.8-kilobase mRNA expressed ubiquitously and encoding a 610-amino-acid protein named menin; they reported in 15 of 21 MEN1 families and with LOH in 13 of 24 tumors. Concurrently, the MEN1 Consortium, led by Lemmens et al., used trapping and cDNA selection from a chromosome 11-specific library to the same , confirming inactivating such as and frameshift alterations in affected individuals, which truncated the menin protein and supported its role as a tumor suppressor. These initial findings established MEN1 as a classic tumor suppressor gene, with biallelic inactivation via germline mutation plus somatic LOH or second-hit mutation mirroring Knudson's two-hit hypothesis in MEN1 tumorigenesis. The gene's discovery, published in April 1997, enabled the first genetic testing for MEN1 and shifted research toward understanding menin's function beyond its identification.

Recent Advances

Recent advances in MEN1 research since the 2020s have leveraged CRISPR-Cas9 technology to dissect menin's tumor-suppressive roles beyond endocrine tissues. Genome-wide CRISPR screens have identified menin as a key regulator in solid tumors, such as liver cancer, where its knockout promotes metastasis in vivo models, revealing a dual function in suppressing tumor growth and invasion in non-endocrine contexts. Similarly, isogenic cell lines generated via CRISPR editing of patient-derived iPSCs have enabled precise studies of MEN1 loss-of-function, highlighting menin's involvement in chromatin remodeling and gene expression in various cancers. These functional studies underscore menin's broader oncogenic implications, expanding from classical endocrine neoplasia to sporadic solid malignancies. Therapeutic developments targeting menin have progressed notably, with inhibitors like revumenib entering clinical trials primarily for harboring MLL (KMT2A) rearrangements, where menin-MLL disruption drives leukemogenesis. Preclinical extensions to MEN1 models demonstrate that menin inhibition can reverse aberrant in contexts, suggesting potential repurposing for MEN1-associated pancreatic and pituitary lesions, though clinical translation remains exploratory. Concurrently, the 2025 American Association of Clinical Endocrinology (AACE) consensus statement emphasizes expanded for MEN1, recommending it for individuals with suggestive clinical features or family history, alongside biochemical surveillance to enable early intervention. Emerging research highlights overlaps between MEN1 and MEN4, where mutations mimic MEN1 phenotypes, with menin regulating p27 ( product) transcription, leading to shared parathyroid and pituitary tumor risks in up to 3% of MEN1-negative cases. AI-driven tools for mutation prediction are gaining traction, integrating with genomic data to classify MEN1 variants' pathogenicity, improving diagnostic accuracy in ambiguous cases. In imaging, 2024-2025 studies validate 68Ga-DOTATATE as superior for detecting MEN1-associated duodeno-pancreatic neuroendocrine tumors, outperforming conventional modalities with higher sensitivity (up to 87%) and guiding precise surgical management. These multimodal advances address persistent gaps in tumor localization and personalized therapy for MEN1 patients.

References

  1. [1]
    Multiple Endocrine Neoplasia Type 1 - NIDDK
    Overview of multiple endocrine neoplasia type 1 (MEN1), an inherited disorder that causes tumors to develop in two or more endocrine glands.What is multiple endocrine... · What are the complications of...
  2. [2]
    Multiple Endocrine Neoplasia Type 1 (MEN1) - Medscape Reference
    Jan 5, 2022 · MEN1 is an endocrine tumor syndrome caused by inactivating mutations of the MEN1 tumor suppressor gene at the 11q13 locus.Practice Essentials · Pathophysiology · Etiology · Epidemiology
  3. [3]
    Multiple endocrine neoplasia, type 1 (MEN 1) - Symptoms and causes
    Oct 4, 2024 · Multiple endocrine neoplasia, type 1 (MEN 1) is a rare condition. It mainly causes tumors in the glands that make and release hormones.
  4. [4]
    Entry - *613733 - MENIN 1 ; MEN1 - OMIM - (OMIM.ORG)
    The MEN1 gene encodes menin, a nuclear scaffold protein that regulates gene transcription by coordinating chromatin remodeling.
  5. [5]
    MEN1 (multiple endocrine neoplasia I)
    ### Summary of MEN1 Gene (Multiple Endocrine Neoplasia I)
  6. [6]
    MEN1 gene and its mutations: Basic and clinical implications - PMC
    The human MEN1 gene comprises 10 exons distributed over 7 kb in the chromosome region 11q13 and encodes mRNA of approximately 2.8 kb (Fig. 2). Exons 2 through ...
  7. [7]
    Alternative splicing generates isoform diversity in MEN1 - PubMed
    Nov 26, 2024 · These splicing events are predicted to contribute to MENIN diversity by generating isoforms with in-frame insertions, deletions or unique amino termini.
  8. [8]
    Differential expression of multiple alternative spliceforms ... - PubMed
    The present study was undertaken to further approach the function of Men1 and its encoded protein menin. By 5' RACE and RT-PCR four alternative splice variants ...
  9. [9]
    Functional characterization of a promoter region in the human MEN1 ...
    Our data highlight the existence of a complex transcriptional regulation of the MEN1 gene, whose activity is clearly modulated depending not only on the ...
  10. [10]
    Transcription regulation of the multiple endocrine neoplasia type 1 ...
    Our data confirm that the expression of the MEN1 gene is regulated by a feedback from its product menin.
  11. [11]
    Decreased expression and promoter methylation of the menin tumor ...
    These findings represent the first evidence that the MEN1 gene is a target of mutation and methylation in PDAC and that menin influences the cell cycle profile ...
  12. [12]
    DNA methylation profiling in MEN1-related pancreatic ... - PubMed
    Promoter hypermethylation is a frequent event in MEN1-related and sporadic PanNETs. Targeting DNA methylation could be of therapeutic value in MEN1 patients ...
  13. [13]
    Menin mediates epigenetic regulation via histone H3 lysine 9 ...
    Apr 11, 2013 · Menin functions as a transcriptional activator by tethering MLL complex to mediate histone H3 K4 methylation. It also functions as a repressor.
  14. [14]
    Enhancer-Mediated Oncogenic Function of the Menin Tumor ...
    Mar 7, 2017 · The MEN1 gene product menin is involved in H3K4 trimethylation and co-activates transcription. We integrated ChIP-seq and RNA-seq data to ...
  15. [15]
    Proliferation-associated expression of the MEN1 gene as ... - PubMed
    The expression increased as cells entered into S phase, indicating cell cycle-associated transcriptional regulation of the MEN1 gene. Increase or decrease ...
  16. [16]
    GTEx Portal
    ### Summary of MEN1 Tissue-Specific Expression (GTEx Portal)
  17. [17]
    MEN1 - Menin - Homo sapiens (Human) | UniProtKB | UniProt
    Nov 8, 2023 · Essential component of a MLL/SET1 histone methyltransferase (HMT) complex, a complex that specifically methylates 'Lys-4' of histone H3 (H3K4).Missing: crystal PDB
  18. [18]
    Full article: Menin: from molecular insights to clinical impact
    Multiple studies have found that both mutant and wild type Menin are post-translationally degraded in the context of NETs and that stabilizing Menin in these ...
  19. [19]
    Menin: a scaffold protein that controls gene expression and cell ...
    Abstract. The protein menin is encoded by the MEN1 gene, which is mutated in patients with multiple endocrine neoplasia type 1 (MEN1) syndrome.<|control11|><|separator|>
  20. [20]
    3U84: Crystal Structure of Human Menin - RCSB PDB
    Feb 15, 2012 · Here we present the crystal structures of human menin in its free form and in complexes with MLL1 or with JUND, or with an MLL1-LEDGF heterodimer.
  21. [21]
    Menin “reads” H3K79me2 mark in a nucleosomal context - Science
    Feb 16, 2023 · Lin et al. identified the protein menin as a bona fide reader of H3K79me2 using a multifunctional nucleosome-based photoaffinity probe.
  22. [22]
    8GPN: Human menin in complex with H3K79Me2 nucleosome
    Feb 15, 2023 · A cryo-electron microscopy structure of menin bound to an H3K79me2 nucleosome revealed that menin engages with the nucleosome using its fingers ...
  23. [23]
    Phosphorylation of the Menin Tumor Suppressor Protein on Serine ...
    Oct 1, 2006 · To detect specifically which residues (Ser/Thr or Tyr) are phosphorylated, cells were subjected to phosphoamino acid analysis by metabolic ...
  24. [24]
    Menin and MLL cooperatively regulate expression of cyclin ...
    Here, we show that menin directly regulates expression of the cyclin-dependent kinase inhibitors p27Kip1 and p18Ink4c. Menin activates transcription by ...
  25. [25]
    Cellular Functions of Menin - Madame Curie Bioscience Database
    Loss of function of either MLL or menin results in down-regulation of p27 and p18 expression and deregulated cell proliferation. By use of a mouse model with ...
  26. [26]
    p18Ink4c, but Not p27Kip1, Collaborates with Men1 To Suppress ...
    Mutant mice lacking both cyclin-dependent kinase (CDK) inhibitors p18Ink4c and p27Kip1 develop a tumor spectrum reminiscent of human multiple endocrine ...
  27. [27]
    Global Analysis of H3K4 Methylation Defines MLL Family Member ...
    Menin, a common component of Mll1 and Mll2 complexes, is required for the expression of nearly all Hox genes and for H3K4 trimethylation at Hox loci. (A) ...
  28. [28]
    A Molecular Switch between Mammalian MLL Complexes Dictates ...
    Jan 9, 2023 · Because Menin lacks a defined DNA binding domain (57), we tested whether a sequence-specific DNA binding factor may regulate the switch between ...
  29. [29]
    Menin, Histone H3 Methyltransferases, and Regulation of Cell ...
    Hence, this review will focus on the recent progress in identifying menin as a regulator of MLLs and histone H3K4 methylation and gene transcription, menin's ...
  30. [30]
    Epigenetic regulation by the menin pathway - PMC - PubMed Central
    Menin and MLL cooperatively regulate expression of cyclin-dependent kinase inhibitors. ... The tumor suppressor protein menin inhibits AKT activation by ...
  31. [31]
    Menin stimulates homology-directed DNA repair - ScienceDirect.com
    Nov 19, 2010 · We report that in human cells menin stimulates homology-directed (HD) DNA repair induced by the rare endonuclease I-SceI and it accumulates with Chk1 at the ...
  32. [32]
    Menin stimulates homology-directed DNA repair - PubMed
    We report that in human cells menin stimulates homology-directed (HD) DNA repair induced by the rare endonuclease I-SceI and it accumulates with Chk1.Missing: primary source
  33. [33]
    Role of Menin in the Regulation of Telomerase Activity in Normal ...
    Thus, while menin appears to have some inhibitory effects on the hTERT promoter, possibly via the sequence-independent binding to the promoter, the present ...Missing: Sp1 | Show results with:Sp1
  34. [34]
    Fyn-mediated phosphorylation of Menin disrupts telomere ...
    Oct 4, 2023 · We report a novel role for the Src family kinase Fyn, which disrupts telomere maintenance in stem cells by phosphorylating the scaffold protein Menin.
  35. [35]
    MEN1 tumor-suppressor protein localizes to telomeres during meiosis
    Cells overexpressing menin had normal telomerase activity, and tumors with homozygous MEN1 mutations showed no aberrations in telomere length, indicating that ...Missing: knockout | Show results with:knockout
  36. [36]
    Multiple Endocrine Neoplasia Type 1 - GeneReviews - NCBI - NIH
    Aug 31, 2005 · Approximately 90% of individuals diagnosed with MEN1 have an affected parent; approximately 10% of individuals diagnosed with MEN1 have the ...
  37. [37]
    Genotype-phenotype correlation in multiple endocrine neoplasia ...
    Feb 13, 2025 · This report pertains to MEN1, which has a prevalence of 3–20/100,000 and is inherited in an autosomal-dominant pattern with a high degree of ...
  38. [38]
    Multiple Endocrine Neoplasias - MEN | Choose the Right Test
    Jan 28, 2025 · Autosomal dominant inheritance – ~10% of mutations are de novo; Germline mutations in the MEN1 gene on 11q13 are causative. Sequence analysis ...
  39. [39]
    131100 - MULTIPLE ENDOCRINE NEOPLASIA, TYPE I; MEN1 - OMIM
    By FISH, Guru et al. (1999) mapped the mouse Men1 gene to chromosome 19 in a region showing homology of synteny to human chromosome 11q13.
  40. [40]
    Multiple Endocrine Neoplasia Type 1 (MEN1): An Update ... - Frontiers
    Jun 10, 2019 · MEN1 is a rare hereditary tumor syndrome inherited in an autosomal dominant manner and characterized by a predisposition to a multitude of endocrine neoplasms.Missing: GTEx | Show results with:GTEx
  41. [41]
    Multiple Endocrine Neoplasia Type 1 (MEN1) Syndrome - NCBI - NIH
    Jul 18, 2008 · MEN1 follows Knudson's “two-hit” model for tumor suppressor gene carcinogenesis (30). The first hit is a heterozygous MEN1 germline mutation, ...
  42. [42]
    [PDF] Presence of Multiple Endocrine Neoplasia-1 Mutations in Patients ...
    Apr 20, 2023 · 10 More than 90% of patients with MEN-1 have lost heterozygosity, which supports Knudson's 2-hit hypothesis.11. The MEN-1 gene consists of 10 ...
  43. [43]
    Somatic mutation of the MEN1 gene in parathyroid tumours - PubMed
    In more than 95% of cases, the disease is caused by sporadic parathyroid adenoma or sporadic hyperplasia. Some cases are caused by inherited syndromes, such as ...
  44. [44]
    Somatic mutations of the MEN1 tumor suppressor gene in ... - PubMed
    MEN1 gene mutations were identified in 9 of 27 (33%) sporadic gastrinomas and 2 of 12 (17%) insulinomas and were not seen in corresponding germ-line DNA ...
  45. [45]
    Involvement of the MEN1 Gene in Hormone-Related Cancers - MDPI
    Intriguingly, recent evidence has suggested that MEN1 could also be involved in the development of breast and prostate cancers, two major hormone-related ...
  46. [46]
    Sporadic Follicular Thyroid Tumors Show Loss of a 200-kb Region ...
    Loss of heterozygosity (LOH) in 11q13 where the tumor suppressor gene for multiple endocrine neoplasia type 1 (MEN 1) is located has been demonstrated in ...
  47. [47]
    Clinical and molecular determinants of survival in pancreatic ...
    May 28, 2025 · Data from the TCGA database included 98 cases, and mutations in MEN1 (37%), DAXX (17%), ATRX (9%), and PTEN (7%) did not correlate with survival ...
  48. [48]
    Recent Clinical and Molecular Advances in the Management ... - NIH
    Jun 13, 2025 · Recent advances have expanded understanding of the genetic landscape of thymic carcinoids. Frequent mutations in the MEN1 gene, as well as in ...
  49. [49]
    Multiple Endocrine Neoplasia Type 1 (09.08.2024)
    This is because thymic carcinoids metastasize early, are fast growing and therefore, despite their rarity, one of the main causes of MEN1-associated mortality ( ...
  50. [50]
    The UMD- MEN1 mutations database Home
    Oct 8, 2023 · The database is being updated. This database includes 44 references and 1761 mutations ( 426 different mutations and 253 proteic variants) ...Missing: LOVD percentages
  51. [51]
    Phenotypes Associated With MEN1 Syndrome: A Focus ... - Frontiers
    Parathyroid tumors, resulting in primary hyperparathyroidism (PHPT), affect up to 95% of MEN1 patients and represent the first manifestation of the syndrome, ...
  52. [52]
    An Overview of the 370 MEN1 Variants Present in 1676 Patients ...
    This database analysis revealed a low frequency (6.6%) of benign or likely benign missense variants in MEN1. Eight families (1.9%) had members with familial ...
  53. [53]
    Phenotypes Associated With MEN1 Syndrome - PubMed Central - NIH
    Nov 18, 2020 · More than 10% of mutations are nonsense mutations, 40% frameshift insertions and deletions, 25% missense mutations while 11% are splice site ...
  54. [54]
    Multiple endocrine neoplasia type 1 (MEN1): An update of 208 new ...
    Approximately 20% of mutations are nonsense mutations, about 50% are frameshift insertions and deletions, 20% are missense mutations while about 7% are splice ...
  55. [55]
    Multiple Endocrine Neoplasia Type 1 - Endotext - NCBI Bookshelf
    Dec 22, 2021 · MEN1 is considered to act as a tumor suppressor gene which is demonstrated by the identification of inactivating mutations, together with loss ...
  56. [56]
    A new double substitution mutation in the MEN1 gene
    Nov 28, 2012 · We postulate that this new double substitution MEN1 mutation affects the interaction of menin with MLL and JunD, and is clinically relevant ...
  57. [57]
    Genetic Landscape and Clinical Manifestations of Multiple ...
    Nov 18, 2024 · Genetic testing revealed 61 distinct MEN1 variants in 101 patients, with 18 being novel. Four variants were reclassified according to the TENGEN ...
  58. [58]
    Variable clinical expression in patients with a germline MEN1 ...
    MEN1 is caused by germline mutations of the MEN1 tumor suppressor gene. It appears that in the MEN1 syndrome, clinical expression differs between families. This ...
  59. [59]
    Multiple endocrine neoplasia type 1: analysis of germline MEN1 ...
    Jul 21, 2018 · Currently, no direct genotype-phenotype correlation is identified. We aim to analyze MEN1 mutation site and features, and possible correlations ...<|separator|>
  60. [60]
    Multi-omics analyses of MEN1 missense mutations identify ...
    Aug 9, 2022 · The protein interactomes of these mutants were assessed by quantitative mass spectrometry, which indicated that seven of the nine mutants ...
  61. [61]
  62. [62]
    Inactivation of menin, a Smad3-interacting protein, blocks ... - PNAS
    Furthermore, we show that menin interacts with the receptor-regulated Smad, Smad3, and antisense menin sup- presses TGF-ß-induced and Smad3-induced ...
  63. [63]
    Menin Is Required for Bone Morphogenetic Protein 2- and ...
    Menin was co-immunoprecipitated with Smad3 (Fig. 8A). Moreover, the co-immunoprecipitation was enhanced with TGF-β in the nucleus (Fig. 8B). However, the co ...
  64. [64]
    The 32-Kilodalton Subunit of Replication Protein A Interacts with ...
    In a yeast two-hybrid system based on reconstitution of Ras signaling, menin was found to interact with the 32-kDa subunit (RPA2) of replication protein A (RPA) ...
  65. [65]
    Investigating the novel-binding site of RPA2 on Menin and ... - Nature
    Jun 8, 2023 · Recently, novel DNA damage-induced phosphorylation site at Thr-98 site of RPA2 has been identified. Alternations at this site lead to ...Missing: Ser | Show results with:Ser
  66. [66]
    Epigenetic regulation by the menin pathway in
    Menin was shown to be crucial for regulating canonical Wnt/β-catenin signaling ... Our previous study indicated that menin antagonizes Hedgehog signaling ...
  67. [67]
    Targeting β-catenin signaling for therapeutic intervention in MEN1 ...
    Dec 17, 2014 · The suppression of β-catenin could repress the growth of menin-null tumours by rescuing the upregulation of proliferative genes. Figure 6: ...
  68. [68]
    Menin Epigenetically Represses Hedgehog signaling in MEN1 ... - NIH
    These findings uncover a novel link between menin and Hh signaling whereby menin/PRMT5 epigenetically suppress Hh signaling, revealing it as a target for ...
  69. [69]
    Menin epigenetically represses Hedgehog signaling in MEN1 tumor ...
    Apr 15, 2013 · Menin recruits PRMT5 to the promoter of the Gas1 gene, a crucial factor for binding of Sonic Hedgehog (Shh) ligand to its receptor PTCH1 and ...
  70. [70]
    Menin Induces Apoptosis in Murine Embryonic Fibroblasts - PMC - NIH
    Here we show that ectopic overexpression of menin via adenoviruses induces apoptosis in murine embryonic fibroblasts (MEFs).
  71. [71]
    Transcription factor JunD, deprived of menin, switches from growth ...
    JunD changed from growth suppressor to growth promoter when its binding to menin was prevented by a JunD mutant unable to bind menin or by Men1-null genetic ...Missing: apoptosis | Show results with:apoptosis
  72. [72]
    Twenty years of menin; emerging opportunities for restoration of ...
    We propose that menin regulates cell type-specific transcriptional programs by linking chromatin regulatory complexes to specific transcription factors.Menin Binds To Transcription... · Mll1/mll2 · Menin-Dependent Gene...
  73. [73]
    The tumor suppressor protein menin inhibits AKT activation by ...
    Menin is a tumor suppressor, its molecular mechanism of action has not been defined. Here we report that menin interacts with AKT1 in vitro and in vivo.
  74. [74]
    Mathematical modeling of histone modifications reveals the ... - NIH
    Jul 23, 2021 · We develop a mathematical model that involves antagonistic histone modifications H3K4me3 and H3K27me3 to capture the key features of bivalent chromatin.Missing: menin equations
  75. [75]
    Multiple endocrine neoplasia type 1 gene maps to chromosome 11 ...
    Mar 3, 1988 · Here we map the MEN-1 locus to chromosome 11 by family studies, and demon-strate tight linkage with the human muscle phosphorylase gene.
  76. [76]
    Linkage Analysis of Multiple Endocrine Neoplasia Type 1 With INT2 ...
    Pairwise and multipoint analyses with INT2 confirm the recent finding by C. Larsson et al. (1988, Nature (London) 332: 85-87) of MEN1 linkage to another marker, ...
  77. [77]
    Identification of the Multiple Endocrine Neoplasia Type 1 (MEN1 ...
    Two novel genes, referred to as SCG1 and SCG2 (i.e. suppressor candidate gene 1 and 2), were isolated (Fig. 1). Use of these bovine clones revealed that SCG1 ...
  78. [78]
    In vivo CRISPR screens identify a dual function of MEN1 in ... - Nature
    Sep 3, 2024 · We identified MEN1 as the top hit that confers differential dropout effects in vitro and in vivo. MEN1 knockout in multiple solid cancer types does not impact ...
  79. [79]
    Generation of isogenic and homozygous MEN1 mutant cell lines ...
    These cell lines will be useful for elucidating subcellular MEN1 pathophysiology and for screening to identify potential MEN1 therapeutic targets.
  80. [80]
    Menin in Cancer - PMC - PubMed Central
    The MEN1 gene was first discovered in the context of multiple endocrine neoplasia type 1 (MEN1) syndrome [1,2], where it functions as a potent tumor suppressor ...
  81. [81]
    The menin inhibitor revumenib in KMT2A-rearranged or ... - Nature
    Mar 15, 2023 · Here, we describe the results of the first-in-human phase 1 clinical trial investigating revumenib (SNDX-5613), a potent and selective oral ...
  82. [82]
    Preclinical efficacy of the potent, selective menin-KMT2A inhibitor ...
    Sep 12, 2024 · Recombinant mutant MEN1 binding with revumenib and other menin-KMT2A inhibitors showed a significant reduction in binding affinity. KMT2A-r ...Key Points · Introduction · Results · Discussion
  83. [83]
    Multiple endocrine neoplasia type 4: a new member of the MEN ...
    Approximately 3% of patients displaying a MEN1-like phenotype but testing negative for MEN1 mutations are reported to have mutations in the causative CDKN1B ...
  84. [84]
    Annals of Reviews & Research (ARR) - Juniper Publishers
    Aug 29, 2025 · The article concludes by highlighting future directions, including the potential for CRISPR gene editing, artificial intelligence in diagnostics ...
  85. [85]
    68Ga-DOTA-TATE PET/CT improves accuracy and guides ...
    Sep 17, 2025 · Ga-DOTA-TATE PET/CT improves accuracy and guides management in multiple endocrine neoplasia type 1 (MEN-1) patients with suspected duodeno- ...