MEN1 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your orders. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
MEA 1 antibody; MEA1 antibody; MEN 1 antibody; Men1 antibody; MEN1_HUMAN antibody; Menin antibody; Multiple Endocrine Adenomatosis 1 antibody; Multiple Endocrine Neoplasia 1 antibody; SCG 2 antibody; SCG2 antibody; Suppressor Candidate Gene 2 antibody; Wermer syndrome antibody; ZES antibody; Zollinger Ellison Syndrome antibody
Target Names
Uniprot No.

Target Background

Function
Menin is an essential component of the MLL/SET1 histone methyltransferase (HMT) complex. This complex specifically methylates lysine 4 of histone H3 (H3K4), which plays a crucial role in transcriptional regulation. Menin acts as a transcriptional regulator, binding to the TERT promoter and repressing telomerase expression. It also participates in TGFB1-mediated inhibition of cell proliferation, potentially regulating SMAD3 transcriptional activity. Additionally, Menin represses JUND-mediated transcriptional activation on AP1 sites, as well as that mediated by NFKB subunit RELA. It positively regulates HOXC8 and HOXC6 gene expression and may be involved in normal hematopoiesis through the activation of HOXA9 expression. Menin may also be involved in DNA repair.
Gene References Into Functions
  1. Menin functions as an oncogenic regulatory factor that is critical for MYC-mediated gene transcription. PMID: 28474697
  2. This novel study reveals evidence supporting a possible association between altered MEN1 promoter methylation and clinical severity of disease PMID: 30149991
  3. De novo mutation in MEN1 is not associated with parental somatic mosaicism.( PMID: 27799361
  4. Results revealed that the expression level of menin was lower in lung cancer. Its expression is regulated by miR24 which directly targets menin and significantly inhibits its activity, thereby promoting the growth and metastasis of lung cancer cells. PMID: 29565463
  5. A case of dorsal pancreatic hemi-agenesis is reported in a heterozygous carrier of a novel MEN1 variant. PMID: 29174091
  6. A missense variant in aryl hydrocarbon receptor-interacting protein (AIP) gene and a truncating mutation in multiple endocrine neoplasia I protein (MEN1) gene were both detected in the proband and his father, showing limited co-segregation with phenotype. PMID: 29848728
  7. menin is regulated by extracellular signaling factors and has a role in nuclear receptor activation and hepatobiliary pathology in various hepatic cell types [review] PMID: 28485270
  8. Each of these autosomal dominant syndromes results from a specific germline mutation in unique genes: MEN1 is due to pathogenic MEN1 variants (11q13), MEN2A and MEN2B are due to pathogenic RET variants (10q11.21), MEN4 is due to pathogenic CDKN1B variants (12p13.1), and the HPT-JT syndrome is due to pathogenic CDC73 variants (1q25). PMID: 28674121
  9. Menin deficiency is the consequence of a MEN1 mutation in most menin-negative primary hyperparathyroidism tissues PMID: 28597079
  10. Data demonstrate an essential role for MLL1 and menin in mediating tumor maintenance and posterior HOXD gene activation in Ewing sarcoma. PMID: 27888797
  11. Data suggest that a novel germline missense mutation in MEN1 (p.Gly42Val) accounts for type 1 multiple endocrine neoplasia in a family; this mutation was found in the patient and his mother. [CASE REPORT] PMID: 29066490
  12. Our study provides important insights into the role of menin in DNA methylation and its impact on the pathogenesis of Multiple endocrine neoplasia type 1 syndrome tumor development. PMID: 26871472
  13. MEN1 exerts an anti-proliferative function by regulating a distinct expression signature. PMID: 28273452
  14. expression increased in late-stage primary sclerosing cholangitis PMID: 28602220
  15. The results provide novel molecular insights into the tumor suppressor activity of menin, which is partly mediated by proteasomal degradation of beta-catenin and inhibition of Wnt/beta-catenin signaling. PMID: 28782520
  16. knockdown of RPA2 promoted formation of the menin-p65 complex and repressed the expression of NF-kappaB-mediated genes. RPA2 expression was induced via an E2F1-dependent mechanism in MCF7 and MDA-MB-231 cells treated with NF-kappaB activators, TNF-alpha or lipopolysaccharide (LPS). PMID: 28007956
  17. Loss of Menin is an early event in pancreatic neuroendocrine tumorigenesis and that ATRX/DAXX loss and alternative lengthening of telomeres are relatively late events. PMID: 27342911
  18. The lack of somatic CDKN1B mutations in our samples points to a rare involvement in parathyroid adenomas, despite the frequent loss of nuclear p27 expression. MEN1 biallelic inactivation seems to be directly related to down-regulation of p27 expression through the inhibition of CDKN1B gene transcription. PMID: 27038812
  19. This result shows a novel mechanism whereby menin, a RNA-binding protein, facilitates the processing of its specific miRNA by regulating the dynamics of the menin-miR-24 Gene Regulatory Network at the level of pri-miRNA processing. PMID: 27098433
  20. findings reveal a previously unappreciated cross-talk between two crucial tumor suppressor genes, MEN1 and DAXX, thought to work by independent pathways PMID: 27872097
  21. Multiple endocrine neoplasia type 1-related primary hyperparathyroidism patients experienced more common kidney complications but less skeletal issues, and a milder biochemical manifestation compared with SHPT patients. MEN1 mutation detection rate was 79.4% and 9 of the identified mutations were novel. PMID: 27846313
  22. miR-24-dependent expression of menin may be important in the regulation of nonmalignant and cholangiocarcinoma proliferation. PMID: 28087162
  23. rs2959656, a nonsynonymous variant in MEN1, is associated with the development of clinically active pituitary adenoma. PMID: 27185868
  24. Study acts as a further supplement of the genetic features of neuroendocrine tumors. Somatic mutations of three potential tumor-related genes (HRAS, PAK1 and MEN1) might contribute to the tumorigenesis of thymic neuroendocrine tumors with EAS. PMID: 27913610
  25. The results and clinical course of disease in this case indicate the potential role of menin in the development of non-endocrine or atypical-endocrine tumors in MEN1 patients. PMID: 26732163
  26. cellular staining for menin also reveals the distribution of menin in the cell membrane and the punctate-like cell organelles PMID: 26560942
  27. study to evaluate frequency of Multiple Endocrine Neoplasia type 1 (MEN1) in patients with pituitary adenoma and to perform genetic analysis and familial screening of those with MEN1; genetic analysis showed MEN1 mutations in 4 index cases: IVS4+1 G>A, IVS3-6 C>T, c.1547insC and a new D180A mutation PMID: 23334809
  28. Study reports the coexistence of a germline intronic heterozygote variation at the MEN1 gene (IVS4+1G>T) and a germline mutation of exon 11 of RET proto-oncogene (K666M) in a large Italian family and describes clinical manifestations in the carriers. PMID: 21678021
  29. novel MEN1 c.8251G>A mutation in a family with multiple endocrine neoplasia type 1 PMID: 26239674
  30. progenitors. Our findings define SON as a fine-tuner of the MLL-menin interaction and reveal short SON overexpression as a marker indicating aberrant transcriptional initiation in leukemia. PMID: 26990989
  31. interaction between menin and Dnmt1 reversibly regulates pancreatic cancer cell growth downstream of Hedgehog pathways with complex mutual modulation networks. PMID: 26454216
  32. Molecular analysis of sporadic insulinoma revealed presence of three novel exonic mutations, two novel intronic variations, three reported exon SNPs, and one intronic SNP (rs669976). PMID: 26307114
  33. Data show that menin, encoded by the MEN1 gene, inhibits the transcriptional activity of nuclear receptor liver X receptor alpha (LXRalpha). PMID: 25962847
  34. While there appears be a modest link between MEN1 and breast cancer, causality has not so far been demonstrated. PMID: 25279812
  35. Conditional knockout of beta-catenin suppresses the tumorigenesis and growth of Men1-deficient pancreatic neuroendocrine tumors. PMID: 25517963
  36. miR-17 could inhibit protein levels of Menin through targeting its 3'-untranslated region PMID: 25753880
  37. hyper-expression of the menin protein correlates closely with the poor prognosis of hepatocellular carcinoma patients PMID: 24845612
  38. Mutations of MEN1 gene in a subset of Hurthle cell tumors point to a potential role for this protein and its associated pathways in thyroid tumorigenesis. PMID: 25625803
  39. Menin expression is higher in castration-resistant prostate cancer than in both hormone-naive prostate cancer and benign prostate tissue, and high menin expression correlates with poor overall survival PMID: 25822367
  40. data raise the question of a potential pathogenicity of the p.Ala541Thr missense variant of menin that commonly occurs within the general population PMID: 24997771
  41. MEN1 patients with MEN1 mutations leading to CHES1-loss of interaction have a higher risk of malignant pancreatic neuroendocrine tumors with an aggressive course of disease and disease-related death. PMID: 25210877
  42. miR-421 may promote neuroblastoma cell growth and motility partially by targeting menin PMID: 25012242
  43. The p27 tumor suppressor gene CDKN1B acts as a disease modifier for the multiple endocrine neoplasia (MEN)1 syndrome associated with MEN1 germline mutations. PMID: 24920291
  44. Our observations indicate that MEN1 mutations are involved in human breast carcinogenesis. PMID: 25099597
  45. Loss of MENIN expression is associated with insulinoma. PMID: 24157940
  46. a novel mechanism whereby menin suppresses cell proliferation, at least partly by promoting the processing of certain miRNAs, including let-7a, leading to suppression of Irs2 expression and insulin signaling. PMID: 24563463
  47. presented a case of MEN 1 from its early stage and followed the progression PMID: 24302194
  48. Whole exome capture and sequencing led to the discovery of a missense mutation in the MEN1 gene in familial hyperparathyroidism. PMID: 24074368
  49. Thirteen percent of pulmonary carcinoids harbor MEN1 mutation associated with reduced mRNA expression and poor prognosis PMID: 24276465
  50. role of MEN1 mutation in pituitary carcinomas PMID: 23905891

Show More

Hide All

Database Links

HGNC: 7010

OMIM: 131100

KEGG: hsa:4221

STRING: 9606.ENSP00000337088

UniGene: Hs.423348

Involvement In Disease
Familial multiple endocrine neoplasia type I (MEN1)
Subcellular Location
Nucleus. Note=Concentrated in nuclear body-like structures. Relocates to the nuclear matrix upon gamma irradiation.
Tissue Specificity
Ubiquitous.

Q&A

What is MEN1 and why is it significant in endocrine research?

MEN1 (Multiple Endocrine Neoplasia Type 1) is a rare autosomal dominant inherited tumor syndrome affecting approximately 1 in 30,000 individuals. It is characterized by the development of tumors in multiple endocrine organs, primarily the parathyroid glands, anterior pituitary, and enteropancreatic tissues . The condition results from mutations in the MEN1 tumor suppressor gene, which encodes the protein menin .

The significance of MEN1 in endocrine research extends beyond its clinical manifestations. Menin acts as a tumor suppressor by regulating critical cellular functions including DNA replication and repair, apoptosis, and transcriptional regulation . Understanding menin's molecular functions provides insights into fundamental tumor suppressor mechanisms and potential therapeutic targets for both MEN1-related tumors and other cancers.

What is the molecular structure and function of menin protein?

Menin is a 68-70 kDa nuclear protein encoded by the MEN1 gene . It acts as a tumor suppressor by:

  • Regulating cell proliferation and apoptosis

  • Interacting with transcription factors to control gene expression

  • Playing a role in DNA repair and genome stability

  • Contributing to chromatin remodeling through interactions with histone methyltransferase complexes

Structurally, menin is predominantly localized in the nucleus of cells and is ubiquitously expressed across tissues . The protein interacts with numerous binding partners, most notably components of the MLL histone methyltransferase complex that methylates histone H3 at lysine 4 (H3K4) . This interaction is crucial for its role in transcriptional regulation.

Research has demonstrated that menin influences the expression of key proteins including p53 and retinoblastoma protein (Rb), which are critical cell cycle regulators . Menin knockout studies have revealed its complex roles in different cellular contexts, where it can show both tumor-suppressive and oncogenic functions depending on the tissue microenvironment .

What types of MEN1 antibodies are available for research?

There is a diverse array of MEN1 antibodies available for research purposes, which can be categorized based on several characteristics:

CharacteristicTypesExamples
Host speciesRabbit, MouseRabbit polyclonal (15159-1-AP), Mouse monoclonal (68473-1-Ig)
ClonalityMonoclonal, PolyclonalBosterBio Anti-Menin MEN1 Monoclonal Antibody, Bethyl Rabbit anti-Menin Antibody (Polyclonal)
Target regionFull-length, N-terminal, Middle region, C-terminalAviva Systems Biology MEN1 Antibody - middle region (ARP88468_P050)
ApplicationsWB, IF/ICC, IHC, IP, ELISAMyBioSource.com MEN1 Antibody (WB, ELISA, IHC)
ReactivityHuman, Mouse, RatBiorbyt MEN1 antibody (Hu, Ms, Rt)

Selection of the appropriate antibody depends on the specific research application, target species, and experimental design. For instance, a researcher studying protein-protein interactions might prefer a monoclonal antibody for immunoprecipitation, while someone examining tissue expression patterns might opt for a polyclonal antibody with strong IHC capability .

How do I select the appropriate MEN1 antibody for my specific application?

Selecting the optimal MEN1 antibody requires consideration of multiple factors:

  • Experimental Application: Different applications require different antibody properties:

    • For Western blot: Choose antibodies validated for WB with appropriate dilution ranges (e.g., 1:500-1:2000 for polyclonal or 1:5000-1:50000 for high-affinity monoclonal antibodies)

    • For IHC/IF: Select antibodies specifically validated for these applications with proper epitope exposure (e.g., antibodies requiring TE buffer pH 9.0 for antigen retrieval)

    • For IP: Use antibodies specifically validated for immunoprecipitation with recommended amounts (e.g., 0.5-4.0 μg for 1.0-3.0 mg of total protein lysate)

  • Species Reactivity: Ensure the antibody has been validated in your species of interest:

    • For human samples: Most commercial antibodies react with human menin

    • For mouse/rat models: Verify cross-reactivity with rodent menin (e.g., Bethyl Laboratories antibody reacts with both human and mouse)

  • Epitope Consideration: Depending on your research question, target region matters:

    • C-terminal epitopes may be masked in certain protein complexes

    • If studying a specific MEN1 mutation, ensure the antibody's epitope is not affected by the mutation

  • Validation Evidence: Review published literature and validation data:

    • Citation records (e.g., Bethyl Laboratories antibody with 110 citations)

    • Validation data gallery showing expected banding patterns

    • Published validation in applications similar to yours

Always perform appropriate controls when using a new antibody, such as positive control lysates from cells known to express menin (e.g., HeLa, HepG2, Jurkat cells) and negative controls using MEN1 knockout samples or siRNA-mediated knockdown.

What are the optimal protocols for Western blot detection of menin?

For optimal Western blot detection of menin, follow these methodological considerations:

Sample Preparation:

  • Extract nuclear proteins preferentially, as menin is predominantly a nuclear protein

  • Use a lysis buffer containing protease inhibitors to prevent degradation

  • Standard RIPA or NP-40 buffers supplemented with protease inhibitors work well for menin extraction

SDS-PAGE and Transfer:

  • Use 8-10% polyacrylamide gels to adequately resolve the 68-70 kDa menin protein

  • Transfer to PVDF membrane at 100V for 90 minutes or 30V overnight at 4°C

Blocking and Antibody Incubation:

  • Block with 5% non-fat dry milk or BSA in TBST (based on antibody specifications)

  • Primary antibody dilutions:

    • For polyclonal antibodies: 1:500-1:2000 dilution

    • For monoclonal antibodies: 1:5000-1:50000 dilution

  • Incubate with primary antibody overnight at 4°C

  • Wash 3-5 times with TBST, 5-10 minutes each

  • Use appropriate HRP-conjugated secondary antibody (1:5000-1:10000)

Detection:

  • Enhanced chemiluminescence (ECL) detection systems work well for menin

  • Expected molecular weight: 63-70 kDa

Controls:

  • Positive controls: HepG2, HeLa, Jurkat, or HEK-293 cell lysates

  • Negative controls: MEN1 knockout cell lines or siRNA-mediated knockdown samples

  • Loading control: β-actin, GAPDH, or nuclear proteins like HDAC1 or Lamin B1

Troubleshooting Tips:

  • If detecting multiple bands, optimize primary antibody concentration

  • For weak signals, extend exposure time or increase protein loading

  • For high background, increase washing steps or reduce antibody concentration

How can I use MEN1 antibodies to study protein-protein interactions?

MEN1 antibodies can be valuable tools for studying protein-protein interactions involving menin through several methodological approaches:

Immunoprecipitation (IP):

  • Use 0.5-4.0 μg of anti-MEN1 antibody per 1.0-3.0 mg of total protein lysate

  • Cross-linking antibodies to protein A/G beads can reduce heavy chain interference in subsequent Western blot analysis

  • Include appropriate controls (IgG control, input lysate)

  • After IP, analyze co-precipitated proteins by mass spectrometry or Western blot

Co-immunoprecipitation (Co-IP) Strategy:

  • Forward approach: IP with anti-MEN1 antibody and blot for potential interacting proteins

  • Reverse approach: IP with antibodies against suspected interacting proteins and blot with anti-MEN1 antibody

  • This approach has been successfully used to identify menin interactions with transcription factors and components of histone modification complexes

Proximity Ligation Assay (PLA):

  • Use two antibodies targeting menin and its potential interaction partner

  • Secondary antibodies with oligonucleotide probes generate a signal only when proteins are in close proximity

  • This technique allows visualization of protein interactions in situ

Chromatin Immunoprecipitation (ChIP):
MEN1 antibodies have been effectively used for ChIP to study menin's association with chromatin and interaction with other chromatin-associated proteins:

  • Use formaldehyde to cross-link protein-DNA complexes

  • Sonicate chromatin to appropriate fragment sizes (200-500 bp)

  • Immunoprecipitate with anti-menin antibody (e.g., Bethyl Laboratories BL342)

  • Analyze co-occupancy with other factors such as MLL1 (BL1289) and Rbbp5 (BL766)

  • ChIP-seq analysis has revealed menin's binding across the genome, particularly at H3K4me3-marked regions

These approaches have helped identify menin's interactions with various proteins including MLL histone methyltransferase complexes, which has been critical for understanding menin's role in chromatin regulation and gene expression .

How do I design experiments to study menin's dual function in tumor suppression and oncogenesis?

Recent research has revealed that menin exhibits context-dependent functions that can be either tumor-suppressive or oncogenic . To investigate this duality, consider these experimental approaches:

In Vitro vs. In Vivo Comparative Analysis:

  • Set up parallel in vitro and in vivo experiments with the same cell lines

  • Use CRISPR/Cas9 to knockout MEN1 in cancer cell lines (e.g., A549 lung cancer cells)

  • Compare proliferation rates in vitro (2D and 3D cultures) with tumor growth in mouse models

  • Include both immunodeficient (e.g., NOD/SCID) and immunocompetent mouse models

  • As demonstrated in recent research, MEN1 knockout may show minimal effects in vitro but significant and opposing effects in different in vivo models

Microenvironment Investigation:

  • Co-culture MEN1-knockout and wild-type cells with different stromal or immune cells

  • Use transwell assays to examine paracrine effects

  • Analyze cytokine and chemokine production profiles using multiplex assays

  • Monitor immune cell infiltration in tumors using flow cytometry and immunohistochemistry

  • Research has shown MEN1 knockout can affect cytokine gene expression and immune cell recruitment

Molecular Mechanism Exploration:

  • Perform RNA-seq and ChIP-seq in various conditions to identify context-dependent gene regulation

  • Compare chromatin occupancy patterns of menin and associated factors (e.g., MLL1) in different cell types

  • Investigate H3K4me3 distribution, particularly at repetitive genomic regions

  • Examine double-stranded RNA expression and downstream signaling pathways

Therapeutic Response Testing:

  • Evaluate menin-MLL inhibitors in different tumor models

  • Compare responses in immunodeficient vs. immunocompetent settings

  • Test combinations with immunotherapy agents

  • Assess CD8+ T cell-dependent mechanisms

Data Table: Contrasting Effects of MEN1 Knockout in Different Contexts:

Experimental ContextEffect of MEN1 KnockoutProposed Mechanism
In vitro cell cultureMinimal impact on proliferationLimited dependence on immune factors
Immunodeficient miceEnhanced tumor growthIncreased H3K4me3 at repetitive regions, dsRNA activation, neutrophil infiltration
Immunocompetent miceReduced tumor growthEnhanced CD8+ T cell infiltration and activation

This experimental framework addresses the complex roles of menin in different contexts and provides a systematic approach to understanding its dual functions in cancer .

What methods are available for studying MEN1 mutations and their functional consequences?

Studying MEN1 mutations and their functional impacts requires a multi-faceted approach:

Genetic Analysis Techniques:

  • Next-Generation Sequencing (NGS): Use targeted sequencing panels or whole exome sequencing to identify MEN1 mutations

  • Multiplex Ligation-dependent Probe Amplification (MLPA): Detect large deletions or duplications that may be missed by sequencing

  • RNA-seq: Identify potential splicing defects and expression changes associated with mutations

  • Digital droplet PCR: Quantify allelic ratios in samples with suspected mosaicism

Loss of Heterozygosity (LOH) Analysis:

  • Compare germline DNA with tumor DNA to detect second-hit events

  • Use microsatellite markers or SNP arrays for LOH detection

  • Perform laser capture microdissection to isolate tumor cells for precise analysis

  • Studies have shown that LOH at the MEN1 locus (11q13) is a critical event in MEN1-associated tumors

Functional Characterization Systems:

  • CRISPR/Cas9 Gene Editing:

    • Create isogenic cell lines with specific MEN1 mutations

    • A recent study successfully used CRISPR/Cas9 to generate an isogenic iPSC line from a MEN1 patient and correct the mutation, creating a valuable experimental system

    • Develop knock-in mouse models of specific mutations

  • Patient-Derived Models:

    • Generate induced pluripotent stem cells (iPSCs) from patients with MEN1 mutations

    • Differentiate iPSCs into relevant cell types (e.g., endocrine cells)

    • Establish patient-derived xenografts from MEN1 tumor samples

  • Protein Function Assays:

    • Protein stability and half-life measurements

    • Protein-protein interaction analyses (co-IP, mammalian two-hybrid)

    • Subcellular localization studies using fluorescently tagged mutant proteins

    • DNA binding and transcriptional regulation assays

Phenotypic Assessment:

  • Cell proliferation and apoptosis assays

  • Colony formation assays

  • Soft agar growth for anchorage-independent growth

  • In vivo tumorigenicity studies

  • Differentiation capacity analysis, particularly for endocrine lineages

Data Table: Examples of MEN1 Mutations and Their Functional Impacts:

Mutation TypeExampleFunctional ImpactReference
Frameshiftc.1252G>TLoss of protein function, associated with MEN1 syndrome
Missensec.836C>A (exon 6)Associated with aggressive clinical phenotype, including recurrent parathyroid adenomas and neuroendocrine tumors
Deletion5kb deletion (promoter and exons 1-2)Associated with aggressive tumor behavior and multiple malignant pNETs
Mutations affecting CHES1 interactionVariousHigher risk of malignant pancreatic NETs with aggressive disease course

These comprehensive approaches enable researchers to connect specific MEN1 mutations with their molecular and cellular consequences, potentially revealing genotype-phenotype correlations that could inform clinical management .

How can I use MEN1 antibodies in a temporal conditional knockout system to study acute effects of MEN1 deletion?

To study the immediate consequences of MEN1 deletion, researchers can implement temporal conditional knockout systems paired with appropriate antibody-based detection methods:

Experimental System Design:

  • Inducible Cre-loxP System:

    • Use mice with floxed MEN1 alleles (Men1^fl/fl)

    • Employ tamoxifen-inducible CreERT2 under tissue-specific promoters

    • This approach allows temporal control of MEN1 deletion in specific tissues

  • Alternative Temporal Control Systems:

    • Tetracycline-regulated gene expression systems (Tet-On/Tet-Off)

    • Degradation domain fusion proteins with small molecule-induced stabilization

    • CRISPR interference (CRISPRi) with inducible promoters

Time-Course Analysis Protocol:

  • Establish baseline measurements before MEN1 deletion

  • Induce MEN1 deletion via tamoxifen or doxycycline administration

  • Collect samples at multiple time points (e.g., 24h, 48h, 72h, 7 days, 14 days post-induction)

  • Research has shown that effects on cell proliferation can be detected as early as 7 days post-MEN1 excision

Antibody-Based Detection Methods:

  • Western Blot:

    • Track menin protein levels using anti-MEN1 antibodies (1:500-1:2000 dilution)

    • Quantify relative protein abundance at each time point

    • Include appropriate loading controls

  • Immunohistochemistry:

    • Process tissue sections at each time point

    • Use anti-MEN1 antibodies (1:250-1:1000 dilution)

    • Perform antigen retrieval with TE buffer pH 9.0 or citrate buffer pH 6.0

    • Quantify percentage of cells with menin expression

  • Co-staining for Proliferation and Differentiation Markers:

    • Combine anti-MEN1 antibodies with proliferation markers (Ki67, BrdU)

    • Include tissue-specific differentiation markers

    • Particularly useful for pancreatic islet studies, where increased proliferation has been observed within 7 days of Men1 excision

Molecular and Cellular Analysis:

  • ChIP-seq Time Course:

    • Track changes in menin occupancy across the genome

    • Monitor H3K4me3 redistribution following menin loss

    • Examine recruitment of interacting factors

  • Transcriptome Analysis:

    • RNA-seq at multiple time points to identify immediate-early response genes

    • Focus on cell cycle regulators and tissue-specific functional genes

    • Pathway analysis to identify biological processes affected acutely by menin loss

  • Cell Cycle Analysis:

    • Flow cytometry for cell cycle phase distribution

    • EdU incorporation to measure S phase entry

    • Phospho-histone H3 staining for mitotic index

    • Previous research has shown that acute effects of Men1 mutation include accelerated S phase entry and enhanced cell proliferation

This temporal approach allows researchers to distinguish primary effects of menin loss from secondary adaptations or compensatory mechanisms, providing crucial insights into menin's direct functions in cellular processes and tumor suppression.

How do I troubleshoot inconsistent results in MEN1 antibody-based experiments?

Inconsistent results with MEN1 antibodies can arise from multiple sources. Here's a systematic troubleshooting approach:

Antibody-Related Issues:

  • Epitope Masking: Menin interacts with numerous proteins which may obscure antibody recognition sites

    • Solution: Try antibodies targeting different epitopes of menin

    • Compare results from C-terminal vs. N-terminal targeting antibodies

  • Antibody Specificity: Some antibodies may cross-react with similar proteins

    • Solution: Validate specificity using MEN1 knockout samples or siRNA knockdown

    • Compare results across multiple antibodies from different manufacturers

  • Antibody Sensitivity: Detection thresholds vary between antibodies

    • Solution: Test different antibody concentrations and detection methods

    • Consider signal amplification techniques for low-abundance samples

Sample Preparation Issues:

  • Protein Extraction Efficiency: Nuclear proteins like menin require efficient extraction

    • Solution: Compare different lysis buffers optimized for nuclear protein extraction

    • Include phosphatase and protease inhibitors to prevent degradation

  • Post-translational Modifications: These may affect antibody recognition

    • Solution: Test samples with phosphatase treatment before analysis

    • Consider using antibodies that recognize modified forms if available

Experimental Design Considerations:

  • Tissue Heterogeneity: Menin expression varies across cell types

    • Solution: Use laser capture microdissection for specific cell populations

    • Employ single-cell techniques when possible

  • Developmental Timing: Menin function may vary temporally

    • Solution: Carefully control for developmental stage in your experiments

    • Include age-matched controls and time-course analyses

Data Interpretation Framework:
When faced with inconsistent results, consider these potential biological explanations:

ObservationPotential ExplanationValidation Approach
Variable menin levels despite consistent MEN1 expressionPost-translational regulationAnalyze protein stability and turnover rates
Discrepancy between antibody staining and functional outcomesFunctionally distinct isoforms or modificationsUse multiple antibodies targeting different epitopes
Context-dependent results across tissuesTissue-specific binding partnersCo-IP studies in different tissue contexts
Differences between in vitro and in vivo resultsMicroenvironment factorsCompare matched cultured cells and tissue samples

As demonstrated in research, MEN1 can have differential effects in vitro versus in vivo , highlighting the importance of experimental context in data interpretation.

What controls should I include when studying MEN1 in different experimental systems?

Rigorous controls are essential for reliable MEN1 research across various experimental systems:

General Controls for MEN1 Antibody Use:

  • Positive Controls:

    • Cell lines with confirmed menin expression (HepG2, HEK-293, HeLa, Jurkat)

    • Tissues known to express menin (e.g., pancreatic islets, parathyroid)

    • Recombinant menin protein for Western blot standardization

  • Negative Controls:

    • MEN1 knockout cell lines or tissues

    • siRNA/shRNA-mediated MEN1 knockdown samples

    • Secondary antibody-only controls for immunostaining

    • IgG isotype controls for immunoprecipitation

Genetic Modification Experiments:

  • For CRISPR/Cas9 MEN1 Editing:

    • Non-targeting sgRNA controls

    • Verification of on-target editing by sequencing

    • Analysis of potential off-target effects at predicted sites

    • Inclusion of isogenic control lines

  • For Conditional Knockout Models:

    • Cre-negative controls with same floxed alleles

    • Vehicle-treated controls for inducible systems

    • Wild-type tissues processed identically to experimental samples

    • Time-matched sampling for temporal studies

Functional Studies:

  • For Proliferation/Growth Assays:

    • Parallel in vitro and in vivo assessments

    • Multiple cell/tissue types to account for context-dependency

    • Both immunodeficient and immunocompetent models for in vivo studies

  • For Gene Expression Analysis:

    • Multiple reference genes for normalization (e.g., GAPDH, β-actin)

    • Samples across different time points to capture dynamics

    • Independent validation using alternative methods (e.g., qPCR confirming RNA-seq findings)

Disease Model Controls:

  • For MEN1 Syndrome Models:

    • Age-matched controls for each time point

    • Single-organ affected controls to understand tissue interactions

    • Comparison of heterozygous vs. homozygous models

    • Inclusion of clinical samples when available

  • For Tumor Studies:

    • Adjacent normal tissue controls

    • Comparison of familial vs. sporadic cases

    • Analysis of multiple tumor regions to account for heterogeneity

    • MEN1 wildtype tumors of the same histological type

Example Control Strategy for CRISPR/Cas9 MEN1 Correction:
A study creating an isogenic cell system for MEN1 syndrome exemplifies comprehensive controls :

Control TypeSpecific Controls UsedPurpose
GeneticSequencing of target regionConfirm precise correction of mutation
Off-targetAnalysis of predicted off-target sitesVerify absence of unintended mutations
FunctionalPluripotency marker expressionEnsure maintenance of stem cell properties
DifferentiationGerm layer differentiation capacityConfirm proper developmental potential
TechnicalMultiple independent corrected clonesAccount for clone-specific variations

Implementing these control strategies ensures reproducibility and robust interpretation of results across different experimental contexts.

How can MEN1 antibodies be used to explore the dual role of menin in cancer and the tumor microenvironment?

Recent research has revealed that menin plays context-dependent roles in cancer, showing both tumor-suppressive and oncogenic functions depending on the tumor microenvironment . MEN1 antibodies can be instrumental in exploring this duality through several innovative approaches:

Spatial Transcriptomics and Proteomics:

  • Combine anti-MEN1 immunostaining with spatial transcriptomics to correlate menin expression with spatial gene expression patterns

  • Use multiplexed immunofluorescence with anti-MEN1 and immune cell markers to map menin expression relative to the immune microenvironment

  • This approach can reveal associations between menin expression and specific microenvironmental niches

Single-Cell Analysis:

  • Perform single-cell Western blot or CyTOF with anti-MEN1 antibodies to quantify menin levels in individual cells

  • Correlate with other proteins like cytokine receptors or immune checkpoint molecules

  • This can identify cell populations where menin levels correlate with specific functional states

In Situ Interaction Mapping:

  • Use proximity ligation assays with anti-MEN1 and antibodies against potential interacting partners

  • Compare interaction patterns between in vitro cultures and tumor tissues

  • Research has shown that menin's interactions may differ between controlled in vitro environments and complex in vivo settings

Chromatin Occupancy in Different Contexts:

  • Perform ChIP-seq with anti-MEN1 antibodies in:

    • In vitro cultured cancer cells

    • Tumor cells isolated from immunodeficient mouse xenografts

    • Tumor cells from immunocompetent syngeneic models

  • Compare menin genomic occupancy and H3K4me3 patterns

  • Recent work has shown that MEN1 knockout redistributes MLL1 chromatin occupancy and increases H3K4me3 at repetitive genomic regions

Cytokine-Mediated Regulation:

  • Treat cells with various cytokines and assess menin levels and localization using anti-MEN1 antibodies

  • Examine how menin regulates cytokine gene expression in different immune contexts

  • Research has identified "cytokine-cytokine receptor interaction" as a top enriched term in MEN1-low patients, suggesting a key role for menin in immune signaling

Experimental Approaches to Study MEN1's Dual Role:

ApproachMethodExpected Insights
Immune infiltration correlationMultiplex IHC with anti-MEN1 and immune cell markersRelationship between menin levels and specific immune cell populations
Microenvironment signalingPhospho-flow with anti-MEN1 and signaling markersHow menin levels correlate with active signaling pathways
Epigenetic landscapeCUT&Tag with anti-MEN1 and histone modification antibodiesContext-specific epigenetic functions of menin
Tumor-immune interactionsEx vivo co-culture systems with anti-MEN1 immunoblottingHow menin levels change during immune cell interactions

These approaches leverage MEN1 antibodies to understand the molecular basis for menin's context-dependent functions, potentially informing therapeutic strategies that target menin in specific tumor microenvironments.

What are the emerging applications of MEN1 antibodies in therapeutic development and precision medicine?

MEN1 antibodies are becoming increasingly valuable in therapeutic development and precision medicine approaches for MEN1-related disorders and beyond:

Target Validation and Drug Discovery:

  • Use MEN1 antibodies to validate molecular targets downstream of menin

  • Screen compound libraries for molecules that modulate menin protein levels or interactions

  • Evaluate effects of menin-MLL interaction inhibitors using co-immunoprecipitation with MEN1 antibodies

  • Recent research has shown that pharmacological inhibition of menin-MLL interaction reduces tumor growth in a CD8+ T cell-dependent manner, highlighting therapeutic potential

Biomarker Development:

  • Develop immunoassays using MEN1 antibodies to quantify menin levels in liquid biopsies

  • Correlate menin expression patterns with treatment responses

  • Create multiplexed assays combining menin with other markers for patient stratification

  • Research has identified aggressive tumor phenotypes associated with specific MEN1 mutations, suggesting potential for biomarker-guided approaches

Companion Diagnostics:

  • Use immunohistochemistry with MEN1 antibodies to identify patients likely to respond to menin-targeting therapies

  • Develop algorithms combining menin expression with other molecular features

  • Create standardized scoring systems for clinical implementation

Therapeutic Monitoring:

  • Apply MEN1 antibodies in longitudinal studies to track changes in menin expression during treatment

  • Correlate with clinical outcomes and treatment resistance mechanisms

  • Use in combination with functional assays to assess dynamic changes in menin activity

Novel Therapeutic Approaches:

  • Immunotherapy Combinations:

    • Use MEN1 antibodies to identify tumors where menin inhibition might enhance immunotherapy responses

    • Recent findings showing MEN1's role in tumor immune microenvironment suggest potential synergy

  • Targeted Protein Degradation:

    • Develop menin-targeting PROTACs (Proteolysis Targeting Chimeras)

    • Use MEN1 antibodies to monitor degradation efficiency and selectivity

  • Cell-Based Therapies:

    • Engineer CAR-T cells targeting MEN1-mutated cells expressing aberrant menin

    • Use antibodies to validate target specificity

Implementation in Precision Medicine Paradigms:

Clinical ScenarioMEN1 Antibody ApplicationPotential Impact
Early detection in MEN1 familiesSensitive immunoassays for circulating meninEarlier intervention in high-risk individuals
Treatment selectionIHC-based predictive biomarkersRational selection of menin-targeting therapies
Therapy resistanceSerial monitoring of menin and interacting proteinsIdentification of resistance mechanisms
Minimal residual diseaseUltrasensitive detection methodsImproved post-treatment surveillance

These emerging applications of MEN1 antibodies demonstrate their potential value beyond basic research, extending into translational medicine and clinical practice. The dual nature of menin function revealed in recent research suggests that menin-targeted therapies may have applications in both MEN1-related tumors and other cancer types where menin plays a context-dependent role .

How can advanced genetic engineering techniques be combined with MEN1 antibodies to develop better disease models?

Integrating advanced genetic engineering with MEN1 antibody technologies enables the development of sophisticated disease models that more accurately recapitulate MEN1 syndrome and related disorders:

CRISPR/Cas9-Based Precision Models:

  • Knock-in of Patient-Specific Mutations:

    • Generate exact mutations identified in MEN1 patients

    • Use MEN1 antibodies to verify protein expression and localization

    • A recent study successfully used CRISPR/Cas9 to correct a pathogenic MEN1 mutation (c.1252G>T) in patient-derived iPSCs, creating isogenic cell lines for comparative analysis

  • Base Editing and Prime Editing:

    • Introduce precise point mutations without double-strand breaks

    • Use anti-MEN1 antibodies to assess effects on protein expression and function

    • This approach minimizes unwanted on-target indels and off-target effects

  • Inducible Degradation Systems:

    • Integrate degron tags into endogenous MEN1

    • Use anti-MEN1 antibodies to monitor temporal control of protein levels

    • This allows studying acute effects of menin loss, similar to conditional knockout systems

Advanced Organoid and iPSC Models:

  • Patient-Derived Organoids:

    • Develop 3D cultures from MEN1 patient tissues

    • Characterize menin expression patterns using immunostaining

    • Use for drug screening and personalized medicine approaches

  • Directed Differentiation of iPSCs:

    • Generate endocrine cell types affected in MEN1 syndrome

    • Track menin expression during differentiation using anti-MEN1 antibodies

    • Research has shown successful differentiation of MEN1 patient-derived iPSCs into endodermal cells, demonstrating the feasibility of this approach

  • Multi-Lineage Organoids:

    • Create complex organoid systems containing multiple cell types

    • Study cell-cell interactions in MEN1 deficiency

    • Use multiplexed antibody staining to track menin in different cell populations

Humanized Mouse Models:

  • Conditional Tissue-Specific Models:

    • Generate mice with human MEN1 variants in specific tissues

    • Use antibodies to verify human menin expression patterns

    • Compare with existing mouse Men1 models for translational relevance

  • Patient-Derived Xenografts:

    • Implant MEN1 patient tumor samples in immunodeficient mice

    • Characterize menin expression and function using antibodies

    • Test therapeutic approaches targeting menin or downstream pathways

Experimental Matrix for Comprehensive MEN1 Modeling:

Genetic Engineering ApproachCellular SystemAntibody ApplicationDisease Aspect Modeled
CRISPR knock-in of hotspot mutationsiPSCsValidation of expression/localizationGenotype-phenotype correlations
Inducible CRISPR interferenceOrganoidsTemporal expression analysisEarly events in tumorigenesis
Base editing of regulatory regionsPrimary endocrine cellsChIP-seq for altered bindingTranscriptional dysregulation
Conditional allelesMouse modelsTissue-specific expression patternsMulti-organ manifestations
AAV-delivered CRISPRIn vivo somatic editingIn situ detection of editing efficiencySporadic tumor formation

Validation and Characterization Strategies:

  • Combine genetic engineering verification (sequencing, digital PCR) with protein-level confirmation using MEN1 antibodies

  • Use antibodies against modified histone marks (H3K4me3) to assess functional consequences of MEN1 mutations

  • Perform multi-omics characterization (transcriptomics, proteomics, epigenomics) to comprehensively profile model systems

  • Compare engineered models with patient samples using standardized antibody-based assays

These integrated approaches enable the development of more accurate and clinically relevant MEN1 disease models, facilitating both mechanistic studies and therapeutic development efforts.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.