MIEN1 Antibody

Shipped with Ice Packs
In Stock

Description

What Is MIEN1 Antibody?

MIEN1 antibodies are immunoreagents designed to detect and quantify MIEN1 protein in experimental settings. These antibodies enable visualization of MIEN1’s spatial expression, interaction partners, and downstream signaling pathways. The E-AB-18767 polyclonal antibody (Elabscience) is a well-characterized example, generated using a human MIEN1 fusion protein and validated for immunohistochemistry (IHC) in breast and thyroid cancers .

Research Applications

MIEN1 antibodies have been instrumental in:

  • Cancer Biomarker Studies: Confirming MIEN1 overexpression in breast , prostate , colorectal , and oral squamous cell carcinomas .

  • Mechanistic Insights:

    • Actin Dynamics: MIEN1 knockdown via CRISPR/Cas9 reduced lamellipodium formation and increased stress fibers in breast cancer cells, detected using MIEN1-specific antibodies .

    • Pathway Inhibition: Anti-MIEN1 peptides (e.g., LA3IK, RP-7) disrupt MIEN1-mediated NF-κB and EMT pathways, validated through antibody-based protein expression assays .

  • Therapeutic Development: Antibodies help evaluate MIEN1-targeted therapies, such as siRNA knockdown reducing tumor growth in prostate xenografts .

Key Research Findings

  • Breast Cancer: MIEN1 antibodies revealed its co-amplification with ERBB2 (Her2) in ~30% of breast cancers, correlating with poor prognosis .

  • Prostate Cancer: MIEN1 overexpression promotes invasion by downregulating E-cadherin and upregulating N-cadherin/Slug, detectable via immunoblotting .

  • Functional Interactions: MIEN1 antibodies identified Annexin A2 as a binding partner, linking MIEN1 to Akt/NF-κB signaling and metastasis .

Limitations and Future Directions

  • Specificity Challenges: No known MIEN1 receptors or normal developmental roles complicate therapeutic targeting .

  • Emerging Tools: MIEN1 antibodies are now paired with CRISPR-edited models (e.g., MDA-MB-231 MIEN1-KO clones) to study off-target effects .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery time may vary depending on the purchasing method and location. Please consult your local distributor for specific delivery timelines.
Synonyms
C17orf37 antibody; C17orf37 homolog antibody; C35 antibody; chromosome 17 open reading frame 37 antibody; HBV X transactivated gene 4 protein antibody; HBV X-transactivated gene 4 protein antibody; HBV XAg transactivated protein 4 antibody; HBV XAg-transactivated protein 4 antibody; MGC14832 antibody; Mien1 antibody; MIEN1_HUMAN antibody; Migration and invasion enhancer 1 antibody; ORB3 antibody; Protein C17orf37 antibody; Protein C35 antibody; RDX12 antibody; Redox protein, 12-KD antibody; XTP4 antibody
Target Names
MIEN1
Uniprot No.

Target Background

Function
MIEN1 antibody promotes cell migration by inducing filopodia formation at the leading edge of migrating cells. It plays a role in the regulation of apoptosis, potentially through the control of CASP3. MIEN1 may also be involved in redox-related processes.
Gene References Into Functions
  1. Research indicates that the MIEN1 promoter contains a short interspersed nuclear Alu element region that is hypermethylated in normal cells, leading to gene repression. In cancer cells, hypomethylation of a portion of this repeat, in addition to the binding of USF, results in MIEN1 expression. PMID: 27589566
  2. Studies demonstrate that migration and invasion enhancer 1 (MIEN1) plays a crucial role in maintaining the plasticity of the dynamic membrane-associated actin cytoskeleton, resulting in increased cell motility. This suggests that targeting MIEN1 may be a promising strategy for preventing breast tumor metastasis. PMID: 27462783
  3. Down-regulation of C35 has been shown to decrease the viability and migration of breast ductal carcinoma cells. PMID: 28859163
  4. Evidence indicates that overexpression of MIEN1 may facilitate migration and invasion in breast cancer. PMID: 28198505
  5. A study concluded that miR-26b suppresses NSCLC metastasis by targeting MIEN1 via NF-kappaB/MMP-9/VEGF pathways, suggesting its potential as a prognostic biomarker and therapeutic target for NSCLC treatment. PMID: 26827826
  6. MIEN1, a novel interactor of Annexin A2, promotes tumor cell migration by enhancing AnxA2 cell surface expression. PMID: 26272794
  7. MIEN1 expression not only positively correlates with oral cancer progression but also appears to be a critical molecular determinant in the migration and invasion of oral cancer cells. PMID: 25996585
  8. C35 might serve as a biomarker or therapeutic target for managing colorectal cancer. PMID: 26173296
  9. While MIEN1 is a direct target of miR-940, miR-940 alters MIEN1 RNA. PMID: 25406943
  10. Findings suggest that DeltaNp73 might cooperate with C35 to promote tumor progression and contribute to cisplatin resistance in ovarian cancer cells. PMID: 23880825
  11. Results indicate that MIEN1 may have a significant regulatory role in the phosphorylation of AKT with its redox potential. PMID: 23284973
  12. Prenylation is necessary for the function of the C17orf37 protein in cancer cells. PMID: 21628459
  13. Inhibition of C35 gene expression using small interfering RNA induces apoptosis in breast cancer cells. PMID: 21068479
  14. Data strongly suggests that C17orf37 overexpression in prostate cancer functionally enhances migration and invasion of tumor cells, making it a critical target for cancer therapy. PMID: 19503095
  15. The MGC14832 gene is located within human chromosome 17q12 amplicon, which includes PPP1R1B, STARD3, TCAP, PNMT, ERBB2, MGC14832, and GRB7 genes. The PPP1R1B ~ ERBB2 ~ GRB7 locus is amplified in human gastric cancer and breast cancer. PMID: 12739007

Show More

Hide All

Database Links

HGNC: 28230

OMIM: 611802

KEGG: hsa:84299

STRING: 9606.ENSP00000377778

UniGene: Hs.333526

Protein Families
SelWTH family
Subcellular Location
Cytoplasm, cytosol. Cell membrane; Lipid-anchor; Cytoplasmic side. Note=Concentrates at the leading edge of migrating cells. Localizes outside membrane raft regions.
Tissue Specificity
Among normal tissues, present only in Leydig cells. Strongly up-regulated in breast cancers and in brain cancer distant metastasis (at protein level). Up-regulated in prostate cancer cells and in the higher grades of prostate adenocarcinoma (at protein le

Q&A

What is MIEN1 and why is it significant in cancer research?

MIEN1 (Migration and Invasion Enhancer 1) is a novel gene abundantly expressed in breast and prostate tumor tissues that functions as a critical regulator of tumor cell migration and invasion. It promotes systemic metastases by facilitating cytoskeletal rearrangements and enhancing cell motility. MIEN1 is particularly significant because it localizes to focal adhesions and stress fibers in the lamellum, a region that plays a major role in actin-rich membrane protrusions during cell migration . Studies have demonstrated that MIEN1 is predominantly overexpressed in Her-2 and luminal B subtypes of breast tumors, and its increased expression correlates with poor disease-free survival . In prostate cancer, MIEN1 expression is significantly higher in carcinoma cells (LNCaP, PC-3, and DU145) compared to normal prostate cells, with immunohistochemical analysis confirming lower expression in normal prostate tissues than in high-grade prostate cancer tissues .

What are the key considerations for selecting MIEN1 antibodies for different experimental applications?

When selecting MIEN1 antibodies for research, several critical factors must be considered. For immunofluorescence studies examining subcellular localization, antibodies with minimal background and high specificity for endogenous MIEN1 are essential. These antibodies should be validated for their ability to detect MIEN1 at focal adhesions and in the lamellum of migrating cells . For Western blotting applications, researchers should select antibodies that can reliably detect both endogenous MIEN1 and overexpressed variants, particularly when studying post-translational modifications like isoprenylation that affect MIEN1 function . For immunohistochemistry on tissue samples, consider antibodies validated for paraformaldehyde-fixed tissues that can distinguish normal from tumor expression levels, as demonstrated in prostate tissue arrays . Additionally, researchers should verify cross-reactivity for their specific model organism, as MIEN1 sequence conservation varies across species.

How should researchers design experiments to study MIEN1's role in cytoskeletal dynamics?

To effectively study MIEN1's role in cytoskeletal dynamics, researchers should implement a multi-faceted experimental approach. Begin with modulating MIEN1 expression through both knockdown (siRNA, shRNA) and overexpression (GFP-tagged MIEN1) strategies in appropriate cancer cell lines such as MDA-MB-231 for breast cancer or PC-3 for prostate cancer studies . Wound healing assays represent an excellent starting point to observe migration effects, followed by detailed immunofluorescence to examine actin filament organization using G-actin/F-actin staining. This allows visualization of how MIEN1 affects the G-actin to F-actin ratio . For functional validation, include actin polymerization assays combined with inhibitors like Cytochalasin D to confirm MIEN1's direct influence on actin dynamics. Additionally, researchers should examine MIEN1's effect on key signaling molecules by measuring phosphorylation of FAK (particularly at Tyr-925), cofilin, Akt (Ser-473), ERK1/2, and NF-κB . Time-lapse microscopy of fluorescently labeled cells provides valuable insights into real-time cytoskeletal rearrangements and membrane protrusion formation in response to MIEN1 modulation.

What are the optimal protocols for detecting MIEN1 protein-protein interactions in cancer cells?

For detecting MIEN1 protein-protein interactions in cancer cells, researchers should employ complementary approaches to ensure robust validation. Co-immunoprecipitation (co-IP) using specific MIEN1 antibodies represents the primary method, ideally performed under both endogenous conditions and with tagged constructs to confirm results . When investigating novel interactions, such as MIEN1's binding to Annexin A2, proximity-based techniques offer superior resolution - Fluorescence Resonance Energy Transfer (FRET) can confirm direct physical interactions as demonstrated in previous studies . For identifying broader interaction networks, mass spectrometry following MIEN1 pulldown provides an unbiased approach. To validate functional relevance of identified interactions, researchers should perform domain mapping experiments using truncated constructs, particularly focusing on the C-terminal isoprenylation site and the immunoreceptor tyrosine-based activation motif (ITAM) which are critical for MIEN1's membrane localization and function . For interactions occurring at specific subcellular locations such as focal adhesions, proximity ligation assays (PLA) offer in situ visualization capabilities with high sensitivity for detecting transient or weak interactions during cell migration events.

What controls are essential when evaluating MIEN1 antibody specificity in immunostaining experiments?

When evaluating MIEN1 antibody specificity in immunostaining experiments, several rigorous controls are essential. The primary negative control should involve MIEN1 knockdown cells (using validated siRNA or shRNA) to demonstrate significant reduction in staining intensity compared to control cells . This knockdown validation is particularly important when examining cellular localization patterns. A complementary approach involves rescue experiments where MIEN1-depleted cells are transfected with GFP-tagged MIEN1 to restore the staining pattern, confirming antibody specificity . For tissue immunohistochemistry, include normal adjacent tissue alongside tumor samples to confirm differential expression patterns . When available, testing multiple MIEN1 antibodies recognizing different epitopes helps validate localization findings. For phospho-specific applications, include phosphatase-treated samples as negative controls. Additionally, peptide competition assays, where the antibody is pre-incubated with excess immunizing peptide, can further confirm binding specificity. For dual-labeling experiments examining MIEN1 colocalization with actin or focal adhesion proteins, appropriate secondary antibody-only controls must be included to rule out cross-reactivity or bleed-through artifacts.

How can MIEN1 antibodies be utilized to investigate the relationship between MIEN1 and the NF-κB pathway in cancer progression?

MIEN1 antibodies can be strategically deployed to dissect the complex relationship between MIEN1 and the NF-κB pathway through several methodological approaches. Chromatin immunoprecipitation (ChIP) assays using MIEN1 antibodies can reveal whether MIEN1 associates with chromatin regions containing NF-κB binding sites, particularly on promoters of target genes such as IL-6 . For investigating regulatory relationships, researchers should perform co-immunoprecipitation studies to examine physical interactions between MIEN1 and NF-κB pathway components like NIK (NF-κB-inducing kinase), which has been shown to upregulate MIEN1 expression . Immunofluorescence microscopy using MIEN1 antibodies in conjunction with NF-κB p65 staining can demonstrate nuclear translocation events and potential colocalization during pathway activation. To examine functional relationships, researchers should conduct dual reporter assays measuring both NF-κB and MIEN1 promoter activities while manipulating expression levels of each component, as previous studies have shown reciprocal regulation . Western blotting with phospho-specific antibodies should be employed to monitor activation status of key NF-κB pathway components in response to MIEN1 modulation, particularly examining IκBα degradation and p65 phosphorylation levels as indicators of pathway activation.

What techniques can be combined with MIEN1 antibodies to study its role in actin polymerization and focal adhesion dynamics?

To comprehensively investigate MIEN1's role in actin polymerization and focal adhesion dynamics, researchers should combine multiple advanced techniques with MIEN1 antibody applications. Super-resolution microscopy (STORM, PALM) with dual-labeled samples using MIEN1 antibodies alongside F-actin stains or focal adhesion markers (paxillin, vinculin) provides nanoscale visualization of spatial relationships during migration . For quantitative assessment of actin dynamics, fluorescence recovery after photobleaching (FRAP) of fluorescently-tagged actin in the presence or absence of MIEN1 can measure polymerization rates in live cells. In vitro F-actin polymerization assays with purified components, including recombinant MIEN1 protein, allow direct assessment of MIEN1's effect on actin assembly kinetics, as previously demonstrated . Focal adhesion turnover analysis using time-lapse microscopy combined with MIEN1 antibody staining at fixed timepoints can correlate adhesion dynamics with MIEN1 localization. Proximity biotinylation (BioID) using MIEN1 as bait can identify novel interaction partners at focal adhesions. For mechanical studies, traction force microscopy coupled with MIEN1 manipulation can determine how MIEN1 affects cell-generated forces, which are critical for migration. Additionally, phosphoproteomic analysis following MIEN1 knockdown helps identify downstream signaling changes affecting cytoskeletal regulators, particularly focusing on FAK and cofilin pathway components .

How do MIEN1 antibodies contribute to research on the MIEN1-Annexin A2 interaction and its role in tumor cell motility?

MIEN1 antibodies serve as essential tools for investigating the functional relationship between MIEN1 and Annexin A2 (AnxA2) in tumor cell motility. Dual immunofluorescence staining with MIEN1 and AnxA2 antibodies enables visualization of their colocalization at the cell membrane during migration, particularly in plasma membrane protrusions . For biochemical validation, co-immunoprecipitation experiments using MIEN1 antibodies followed by AnxA2 detection (and vice versa) confirm their physical interaction, while proximity-based assays like FRET provide more definitive evidence of direct binding . Phosphorylation-specific antibodies detecting Tyr23-phosphorylated AnxA2 are crucial for examining MIEN1's role in promoting this post-translational modification, which facilitates AnxA2 translocation to the cell surface . To establish functional connections, researchers should employ MIEN1 antibodies in combination with phospho-specific antibodies against downstream effectors while performing rescue experiments - for example, examining whether expressing a phosphomimetic AnxA2 variant can restore migration defects in MIEN1-depleted cells. Extracellular antibody blocking experiments targeting cell-surface AnxA2 can determine whether the MIEN1-AnxA2 axis operates through extracellular mechanisms involving plasmin generation. Finally, in vivo metastasis models analyzed with immunohistochemistry using both MIEN1 and AnxA2 antibodies can validate the clinical relevance of this interaction in tumor dissemination.

How should researchers interpret contradictory results between MIEN1 protein levels and functional outcomes in different cancer cell lines?

When encountering contradictory results between MIEN1 protein levels and functional outcomes across different cancer cell lines, researchers should implement a systematic analytical approach. First, assess the baseline expression levels of MIEN1 in each cell line using standardized Western blotting techniques, as threshold effects may explain why additional MIEN1 in already high-expressing cells (like LNCaP) might show diminished functional impact compared to moderately-expressing lines . Examine genetic backgrounds of each cell line, particularly the status of pathways known to interact with MIEN1 function, such as NF-κB, Akt, and FAK signaling components, which may explain differential responses . Evaluate post-translational modifications of MIEN1 between cell lines, especially isoprenylation status which is critical for membrane localization and function . For migration/invasion studies showing discrepancies, analyze the experimental timeframes, as rapid versus sustained MIEN1 effects may differ significantly between cell types with varying doubling times. Consider cell-specific expression of MIEN1 interacting partners, notably Annexin A2 levels, which may create bottlenecks in the functional pathway . Additionally, examine the subcellular localization of MIEN1 through fractionation studies combined with immunofluorescence, as differences in trafficking to focal adhesions or membrane protrusions between cell lines may explain functional variations despite similar total protein levels .

What are the common technical challenges in MIEN1 antibody-based experiments and how can they be addressed?

Researchers frequently encounter several technical challenges when working with MIEN1 antibodies. Background staining issues during immunofluorescence can be addressed by implementing more stringent blocking (5% BSA with 0.1% Triton X-100), using monoclonal antibodies where available, and titrating primary antibody concentrations to optimize signal-to-noise ratios . For inconsistent Western blot detection, optimize lysis conditions to ensure complete MIEN1 extraction, particularly from membrane fractions where isoprenylated MIEN1 localizes . Consider using RIPA buffer supplemented with 0.1% SDS and membrane-disrupting detergents. Cross-reactivity with related proteins can be evaluated by including MIEN1 knockdown samples as negative controls, while examining bands at molecular weights corresponding to MIEN1 splice variants or post-translationally modified forms . For co-immunoprecipitation difficulties, milder lysis conditions that preserve protein interactions should be tested, and chemical crosslinking prior to lysis can stabilize transient interactions. Epitope masking due to MIEN1's interaction with binding partners can be addressed by using multiple antibodies targeting different regions of the protein. When studying phosphorylation-dependent events, ensure samples are processed rapidly with appropriate phosphatase inhibitors. For tissue immunohistochemistry, optimize antigen retrieval methods specifically for MIEN1, as standard protocols may not be sufficient for exposing all MIEN1 epitopes in formalin-fixed tissues .

How can researchers reconcile differences between in vitro and in vivo findings when studying MIEN1 functions using antibody-based techniques?

Reconciling differences between in vitro and in vivo findings in MIEN1 research requires careful consideration of several factors. Tumor microenvironment effects absent in cell culture may significantly modify MIEN1 function; therefore, researchers should complement in vitro studies with tissue microarray analyses using MIEN1 antibodies to examine expression in different tumor regions (invasive front, hypoxic areas) and in relation to stromal components . For xenograft models showing different results than cell line studies, researchers should perform immunohistochemistry on tumor sections to verify whether MIEN1 expression levels or localization patterns changed in vivo compared to the injected cells . Examine the phosphorylation status of MIEN1-regulated pathways (Akt, FAK, cofilin) in both systems using phospho-specific antibodies to determine if downstream signaling is differently modulated . Consider developing three-dimensional culture models (spheroids, organoids) as intermediate systems between traditional cell culture and animal models, using MIEN1 antibodies to track localization in these more physiologically relevant contexts. When migration/invasion phenotypes differ between systems, evaluate matrix composition effects by culturing cells on matrices derived from tumor tissue rather than artificial substrates like Matrigel . For functional validation, combine genetic approaches (CRISPR/Cas9 editing of MIEN1) with antibody-based detection methods across different experimental systems. Finally, patient-derived xenograft models may better recapitulate the clinical situation than cell line xenografts and should be evaluated for MIEN1 expression and function using the same antibody-based techniques applied in simpler systems.

How can machine learning approaches enhance MIEN1 antibody-based research in cancer diagnostics and therapeutics?

Machine learning approaches offer transformative potential for MIEN1 antibody-based research in both cancer diagnostics and therapeutics. For diagnostics, deep learning algorithms can be trained on digital pathology images stained with MIEN1 antibodies to identify subtle expression patterns that correlate with patient outcomes, potentially revealing clinically relevant MIEN1 expression thresholds or localization signatures invisible to conventional analysis . Computer vision techniques applied to immunofluorescence microscopy can automate quantification of MIEN1 colocalization with cytoskeletal elements during migration, enabling high-throughput screening of compounds that disrupt these interactions . In antibody engineering, machine learning models trained on antibody-antigen interaction data can predict optimized MIEN1 antibody sequences with enhanced affinity and specificity, addressing current technical limitations . Computational approaches can design synthetic antibody libraries targeting previously inaccessible MIEN1 epitopes that may have therapeutic potential, particularly regions involved in protein-protein interactions with Annexin A2 or cytoskeletal components . For developing MIEN1-targeted therapies, predictive models integrating MIEN1 expression data with patient genomic profiles could identify patient subgroups most likely to benefit from such interventions. Additionally, machine learning analysis of large-scale phosphoproteomic datasets following MIEN1 inhibition could reveal non-obvious pathway connections and potential combination therapy targets that synergize with MIEN1 blockade in specific cancer subtypes .

How can research on MIEN1's role in cytoskeletal dynamics inform studies of other migration-related cancer proteins?

Research methodologies developed for studying MIEN1's influence on cytoskeletal dynamics provide a valuable framework for investigating other migration-related cancer proteins. The experimental approaches combining live-cell imaging, specific antibody staining, and quantitative analysis of actin polymerization established for MIEN1 can be adapted to study proteins with similar functions . MIEN1's role at the intersection of multiple signaling pathways (Akt, FAK, cofilin) demonstrates how migration regulators can simultaneously influence several aspects of cell motility, suggesting that other metastasis-promoting proteins should be examined for similar multi-pathway effects . The discovery of MIEN1's interaction with Annexin A2 highlights the importance of identifying binding partners that might function as effectors for migration-related proteins, potentially revealing new therapeutic targets . The correlation between MIEN1 overexpression and increased F-actin formation at the leading edge of migrating cells establishes a phenotypic signature that can be used to screen for other proteins with analogous functions . MIEN1's negative regulation of the metastasis suppressor NDRG1 suggests that investigating antagonistic relationships between pro-migratory and anti-migratory factors might be crucial for understanding metastatic progression . Methodologically, the combination of in vitro biochemical assays (actin polymerization), cellular studies (migration, invasion), and animal models (xenografts) used to characterize MIEN1 provides a comprehensive experimental pipeline applicable to other migration regulators . Finally, the identification of post-translational modifications like isoprenylation that are critical for MIEN1 function underscores the importance of studying similar modifications in other cytoskeletal regulators as potential drug targets .

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.