MIA2 Human

Melanoma Inhibitory Activity 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction

MIA2 Human (Melanoma Inhibitory Activity 2) is a secreted cytokine belonging to the MIA/OTOR protein family, characterized by a Src homology-3 (SH3)-like domain. Predominantly expressed in hepatocytes, MIA2 has emerged as a critical regulator in liver physiology and pathology, with roles in hepatocellular carcinoma (HCC) suppression, fibrosis, and inflammatory responses . This article synthesizes structural, functional, and clinical insights from diverse studies to provide a comprehensive overview of MIA2.

Biological Function and Mechanism

MIA2 exhibits dual roles in health and disease:

  • Tumor Suppression: Inhibits HCC cell proliferation and invasion via HNF1α-dependent pathways .

  • Acute-Phase Response: Upregulated by IL-6, TGF-β, and hepatic stellate cell-derived factors during liver injury .

  • Diagnostic Marker: Elevated serum levels correlate with hepatic fibrosis severity and chronic hepatitis C progression .

Key Findings:

  • Expression Dynamics: MIA2 is downregulated in 85% of HCC tissues compared to non-tumorous liver .

  • Functional Studies:

    • Recombinant MIA2 reduces HCC cell invasion by 60% and proliferation by 45% in vitro .

    • In vivo, MIA2-expressing HCC xenografts show 70% smaller tumor volumes vs. controls .

  • Regulatory Mechanism: Loss of HNF1α in HCC disrupts MIA2 transcription, promoting tumorigenesis .

Clinical Significance as a Biomarker

MIA2 levels are quantifiable via ELISA (detection range: 16–2000 pg/mL) . Clinical correlations include:

ConditionMIA2 ExpressionSource
Chronic hepatitis C↑ 3.5-fold in severe fibrosis
HCC progression↓ in advanced tumor stages
Liver inflammation↑ with IL-6/TGF-β stimulation

Experimental Highlights:

  • HCC Mouse Models: Tumors from MIA2-transfected cells weighed 0.8 ± 0.2 g vs. 2.1 ± 0.5 g in controls .

  • Fibrosis Correlation: MIA2 mRNA levels are 4.2-fold higher in severe vs. mild fibrosis (p < 0.001) .

  • Therapeutic Potential: Exogenous MIA2 reduces Ki67 proliferation index by 35% in HCC tissues .

Future Directions

Current research focuses on:

  • Developing MIA2-based therapies for HCC .

  • Validating MIA2 as a non-invasive biomarker for liver disease staging .

  • Exploring its interplay with other MIA/OTOR family members in tissue repair .

Product Specs

Introduction
MIA2, a novel acute phase protein primarily found in the liver, acts as a tumor suppressor in hepatocellular carcinoma. Its expression levels react to liver damage in chronic liver diseases, showing significant elevation in patients with severe fibrosis. MIA2 expression is induced by IL6, TGF-β, and conditioned medium from activated hepatic stellate cells, with its primary expression occurring in hepatocytes.
Description
Recombinant human MIA2, produced in E. coli, is a single, non-glycosylated polypeptide chain containing 101 amino acids, resulting in a molecular weight of 11.5 kDa. Purification of MIA2 is achieved using proprietary chromatographic methods.
Physical Appearance
Sterile Filtered White lyophilized (freeze-dried) powder.
Formulation
The lyophilized MIA2 protein is prepared in a concentrated solution (1mg/ml) containing 20mM Phosphate buffer at pH 7.4 and 150mM NaCl.
Solubility
For reconstitution, it is recommended to dissolve the lyophilized MIA2 in sterile 18 MΩ-cm H2O at a concentration of at least 100 µg/ml. This solution can be further diluted in other aqueous solutions as needed.
Stability
While lyophilized MIA2 remains stable at room temperature for up to 3 weeks, it is recommended to store it desiccated below -18°C. After reconstitution, MIA2 should be stored at 4°C for 2-7 days. For long-term storage, keep it below -18°C. Avoid repeated freeze-thaw cycles.
Purity
The purity is determined to be greater than 97.0% based on the following analyses: (a) RP-HPLC and (b) SDS-PAGE.
Synonyms
MIA-2, MIA2, Melanoma Inhibitory Activity 2, FLJ22404.
Source
Escherichia Coli.
Amino Acid Sequence
MLESTKLLAD LKKCGDLECE ALINRVSAMR DYRGPDCRYL NFTKGEEISV YVKLAGERED LWAGSKGKEF GYFPRDAVQI EEVFISEEIQ MSTKESDFLC L.

Q&A

What is MIA2 and what is its functional significance?

MIA2 (Melanoma Inhibitory Activity Protein 2) is a member of the MIA gene family, identified by the UniProt primary accession number Q96PC5 . The protein has been studied extensively in cancer research, with evidence suggesting context-dependent roles in different tissue types.
Methodologically, researchers studying MIA2 should approach its characterization through multiple complementary techniques. The protein can be detected in tissue homogenates, cell lysates, and other biological fluids using standardized assays such as ELISA, with commercially available kits offering detection sensitivity below 0.18 ng/ml and a test range of 0.312-20 ng/ml . The competitive inhibition-based ELISA methodology employs pre-coated antibodies and biotin-conjugated reagents to quantify MIA2 levels with high specificity.
For comprehensive functional studies, researchers should consider multiple experimental models, including cell lines with varying MIA2 expression levels, tissue samples from different origins, and potentially animal models to evaluate its biological significance in vivo.

How does MIA2 relate to other members of the MIA gene family?

MIA2 belongs to a gene family that includes MIA (Melanoma Inhibitory Activity) and TANGO. Research has demonstrated that these family members share certain structural and functional characteristics, though they may display distinct expression patterns across tissues and disease states .
From an experimental perspective, researchers investigating MIA2 should consider comparative analyses with other MIA family members to elucidate shared pathways and distinctive functions. Immunohistochemistry studies have revealed cytoplasmic localization patterns for all three family members across various cancer types, suggesting potential parallels in their cellular functions .
When designing experiments to study MIA2 in relation to other family members, researchers should:

  • Include parallel expression analyses across multiple tissue types

  • Employ consistent detection methodologies for comparative assessments

  • Consider co-expression patterns to identify potential functional relationships

  • Evaluate differential responses to experimental manipulations
    This comparative approach provides a more comprehensive understanding of MIA2's specific role within the broader context of the MIA gene family.

What established methodologies exist for MIA2 detection and quantification?

Researchers have several validated methodologies available for MIA2 detection and quantification:

  • ELISA (Enzyme-Linked Immunosorbent Assay): Commercial kits employ a competitive inhibition reaction between biotin-labeled MIA2 and unlabeled MIA2 on pre-coated antibodies. This methodology offers quantitative measurements with a sensitivity below 0.18 ng/ml .

  • Immunohistochemistry (IHC): This technique allows visualization of MIA2 protein expression in tissue sections, enabling assessment of both expression levels and subcellular localization. Studies have demonstrated cytoplasmic localization of MIA2 in various cancer types including colonic adenocarcinoma, pancreatic adenocarcinoma, and renal cell carcinoma .

  • Western Blotting: For protein expression analysis in cell and tissue lysates, western blotting provides semi-quantitative assessment of MIA2 levels and can confirm antibody specificity.
    When selecting a detection method, researchers should consider:

What is the expression profile of MIA2 across different cancer types?

MIA2 expression varies considerably across different cancer types, with studies showing distinct prevalence patterns. Comprehensive immunohistochemical analyses have revealed the following expression patterns:

Cancer TypeMIA2 Positive CasesPercentage
Head and neck SCCs12/3040.0%
Esophageal SCCs3/1030.0%
Hepatocellular carcinomas7/1643.8%
Pancreatic adenocarcinomas2/540.0%
Lung SCCs5/1827.8%
Cutaneous malignant melanomas2/1020.0%
Cutaneous SCCs1/333.3%
Cervical SCCs6/2128.6%
Endometrial endometrioid adenocarcinomas3/1127.3%
Renal cell carcinomas5/1338.5%
This expression pattern data demonstrates that MIA2 is expressed in significant proportions of multiple cancer types, with the highest prevalence observed in hepatocellular carcinomas (43.8%) and head and neck SCCs (40%) .
Methodologically, researchers investigating MIA2 expression across cancer types should:
  • Use standardized detection protocols to ensure comparability

  • Include appropriate positive and negative controls

  • Employ semi-quantitative scoring systems for consistency

  • Consider cellular heterogeneity within tumor samples

  • Correlate expression with clinicopathological parameters
    This systematic approach allows for meaningful comparisons across tumor types and facilitates the identification of cancer-specific patterns in MIA2 expression.

How does MIA2 expression correlate with cancer prognosis and progression?

Research has demonstrated significant correlations between MIA2 expression and clinical outcomes in multiple cancer types. Disease-free survival analysis revealed that patients positive for MIA2 experienced significantly shorter disease-free survival compared to MIA2-negative patients . This finding suggests potential prognostic utility for MIA2 as a biomarker.
In cervical cancers specifically, MIA2 expression levels in invasive cervical cancer were upregulated relative to those in cervical intraepithelial neoplasia 3, indicating a potential association with disease progression . Furthermore, expression patterns of MIA gene family members, including MIA2, frequently correlated with nodal and/or distant metastasis in esophageal and lung cancers .
For researchers designing prognostic studies involving MIA2:

What experimental approaches best elucidate MIA2's role in tumor biology?

Understanding MIA2's functional role in tumor biology requires systematic experimental approaches that address multiple aspects of cancer hallmarks. Based on established methodologies in cancer research, the following experimental framework is recommended:

  • Expression modulation studies:

    • Gain-of-function experiments through MIA2 overexpression in low-expressing cell lines

    • Loss-of-function studies using siRNA, shRNA, or CRISPR-Cas9 to suppress MIA2 expression

    • Inducible expression systems to study temporal effects of MIA2 modulation

  • Functional assays to assess cancer-related phenotypes:

    • Proliferation assays (MTT, BrdU incorporation, colony formation)

    • Migration and invasion assays (wound healing, transwell, 3D matrix invasion)

    • Apoptosis assessment (Annexin V/PI staining, caspase activation)

    • In vivo tumor growth and metastasis models

  • Mechanistic investigations:

    • Identification of MIA2-interacting proteins through co-immunoprecipitation

    • Pathway analysis using phosphorylation-specific antibodies for key signaling molecules

    • Transcriptomic profiling to identify genes regulated by MIA2

    • Chromatin immunoprecipitation to identify potential transcriptional regulation
      When designing these experiments, researchers should:

  • Include appropriate positive and negative controls

  • Validate key findings with multiple methodological approaches

  • Consider both short-term and long-term effects of MIA2 modulation

  • Correlate in vitro findings with clinical data when possible
    This comprehensive experimental framework facilitates a thorough understanding of MIA2's functional roles in cancer biology beyond mere association studies.

How should researchers optimize experimental design when studying MIA2 in human samples?

Optimal experimental design for MIA2 studies requires careful consideration of multiple methodological factors to ensure valid and reproducible results. When working with human samples, researchers should implement the following design principles:

  • Sample selection and characterization:

    • Define clear inclusion/exclusion criteria for patient samples

    • Collect comprehensive clinicopathological data

    • Consider tissue heterogeneity and implement microdissection when necessary

    • Include appropriate normal tissue controls matched for age, sex, and other relevant variables

  • Technical considerations:

    • Standardize sample collection, processing, and storage protocols

    • Document pre-analytical variables that might affect MIA2 measurements

    • Implement quality control measures throughout the experimental workflow

    • Consider batch effects and implement appropriate randomization strategies

  • Analytical approach:

    • Select detection methods appropriate for the research question and sample type

    • Include methodological controls (antibody specificity, assay validation)

    • Employ quantitative or semi-quantitative scoring systems with defined criteria

    • Consider inter-observer variability for subjective assessments

  • Statistical considerations:

    • Perform power calculations to determine adequate sample sizes

    • Pre-specify primary and secondary endpoints

    • Select appropriate statistical tests based on data distribution

    • Adjust for multiple comparisons when necessary
      For experimental design involving human subjects research, investigators must also adhere to ethical guidelines and obtain appropriate institutional review board approvals . This includes ensuring minimal risk to participants, obtaining informed consent, and maintaining confidentiality of personal information.

What strategies should be implemented to address data contradictions in MIA2 research?

Data contradictions are common challenges in biomedical research, including studies of MIA2. Addressing these contradictions requires systematic approaches to identify sources of variability and implement resolution strategies. Based on principles of health data quality assessment , researchers should:

  • Identify potential sources of contradictions:

    • Methodological differences (detection methods, antibody specificity)

    • Sample characteristics (tumor type, stage, treatment status)

    • Technical variability (inter-laboratory differences, batch effects)

    • Biological heterogeneity (intra-tumoral, inter-patient)

  • Implement structured evaluation methods:

    • Apply the (α, β, θ) notation proposed for health data quality assessment :

      • α: number of interdependent variables measured

      • β: number of contradictory dependencies identified

      • θ: minimal number of Boolean rules needed to assess contradictions

  • Validate findings through:

    • Replication in independent sample sets

    • Use of complementary methodological approaches

    • Cross-validation between different research groups

    • Meta-analysis of published studies when sufficient data is available

  • Report comprehensively:

    • Document all methodological details to enable replication

    • Report both consistent and contradictory findings

    • Discuss potential sources of discrepancies

    • Present data that both supports and challenges the main hypothesis
      This structured approach to addressing contradictions transforms inconsistencies from obstacles into opportunities for deeper understanding of MIA2 biology and more rigorous experimental design in future studies.

How can researchers effectively control for variables in MIA2 expression studies?

Controlling for variables is essential for generating reliable and interpretable data in MIA2 expression studies. Researchers should implement comprehensive control strategies across multiple experimental dimensions:

What statistical approaches are most appropriate for analyzing MIA2 expression data?

Selecting appropriate statistical methods is crucial for valid interpretation of MIA2 expression data. Based on established biostatistical principles, researchers should implement a structured analytical framework:

  • Descriptive statistics:

    • For continuous MIA2 expression data: mean, median, standard deviation, interquartile range

    • For categorical data (positive/negative): frequencies, proportions, contingency tables

    • Graphical representations: box plots for distribution, scatter plots for correlations

  • Inferential statistics for group comparisons:

    • For normally distributed data: t-tests (two groups) or ANOVA (multiple groups)

    • For non-normally distributed data: Mann-Whitney U (two groups) or Kruskal-Wallis (multiple groups)

    • For categorical data: Chi-square or Fisher's exact test

    • Post-hoc corrections for multiple comparisons (e.g., Bonferroni, False Discovery Rate)

  • Correlation and regression analyses:

    • Pearson (parametric) or Spearman (non-parametric) correlation for continuous variables

    • Logistic regression for binary outcomes (e.g., presence/absence of metastasis)

    • Multiple regression to account for confounding variables

  • Survival analysis:

    • Kaplan-Meier curves for visualizing survival differences

    • Log-rank tests for comparing survival distributions

    • Cox proportional hazards models for multivariate analysis

    • Time-dependent ROC curves for evaluating prognostic performance
      When designing the statistical analysis plan, researchers should:

  • Determine sample size requirements through power calculations

  • Pre-specify primary and secondary endpoints

  • Address potential confounding factors through appropriate adjustments

  • Consider both statistical significance and effect size in interpretation
    This comprehensive statistical approach facilitates robust analysis of MIA2 expression data while minimizing the risk of spurious findings or overinterpretation of results.

How should researchers interpret contradictory findings in MIA2 expression studies?

Interpreting contradictory findings requires a structured approach that considers methodological differences, biological heterogeneity, and context-specific factors. When faced with inconsistent results regarding MIA2 expression, researchers should:

  • Evaluate methodological factors:

    • Detection methods: Compare sensitivity and specificity of different techniques

    • Antibody characteristics: Consider epitope specificity, polyclonal vs. monoclonal

    • Scoring systems: Examine threshold definitions for positivity

    • Sample processing: Assess impact of fixation, antigen retrieval, storage conditions

  • Consider biological variables:

    • Tissue context: MIA2 may function differently in different tissues

    • Disease stage: Expression patterns may evolve during disease progression

    • Molecular subtypes: Consider classification systems relevant to the cancer type

    • Treatment effects: Evaluate potential influence of prior therapies

  • Apply structured contradiction analysis:

    • Implement the (α, β, θ) notation system for health data quality assessment

    • Identify the minimum set of Boolean rules needed to characterize contradictions

    • Map interdependencies between variables that might explain discrepancies

  • Synthesize evidence systematically:

    • Weight findings based on methodological rigor

    • Consider consistency across multiple studies

    • Develop integrative models that incorporate context-specific factors

    • Identify knowledge gaps requiring further investigation
      This approach transforms contradictions from obstacles into opportunities for deeper understanding, ultimately advancing knowledge of MIA2 biology through critical evaluation and synthesis of conflicting evidence.

What approaches best integrate MIA2 data with other molecular and clinical parameters?

Integrating MIA2 expression data with other molecular and clinical parameters requires sophisticated approaches that leverage multiple data dimensions. Researchers should implement the following integration strategies:

  • Multivariate statistical models:

    • Multiple regression incorporating clinical variables and MIA2 expression

    • Logistic regression for binary outcomes (response/non-response, recurrence/non-recurrence)

    • Cox proportional hazards models for survival analysis

    • Machine learning approaches for pattern recognition in complex datasets

  • Pathway and network analysis:

    • Map MIA2 to relevant signaling pathways using databases like KEGG

    • Identify protein-protein interaction networks involving MIA2

    • Perform gene set enrichment analysis to identify functional associations

    • Construct regulatory networks incorporating transcriptional and post-transcriptional mechanisms

  • Multi-omics integration:

    • Correlate MIA2 protein expression with mRNA levels

    • Identify genomic alterations associated with MIA2 expression patterns

    • Examine epigenetic regulation through methylation or histone modification data

    • Consider proteomic profiles to identify co-regulated proteins

  • Visualization techniques:

    • Heatmaps for displaying correlation patterns

    • Network diagrams for illustrating molecular interactions

    • Forest plots for summarizing multivariate analyses

    • Nomograms for developing integrated prognostic models
      When implementing these integration strategies, researchers should:

What are the current hypotheses regarding MIA2's mechanistic role in cancer progression?

Several competing hypotheses exist regarding MIA2's mechanistic role in cancer, based on its expression patterns and associations with clinical outcomes. Current research suggests multiple potential mechanisms:

  • Tumor suppressor function:

    • Research indicates MIA2 may act as a tumor suppressor in hepatocellular carcinoma

    • Potential mechanisms include regulation of cell cycle progression, promotion of apoptosis, or inhibition of invasive properties

    • Loss of expression may contribute to malignant transformation in specific tissues

  • Context-dependent functions:

    • MIA2 appears to have tissue-specific roles, with varied expression patterns across cancer types

    • The protein may interact with different molecular partners in different cellular contexts

    • Signaling pathway interactions may determine whether MIA2 promotes or suppresses tumorigenesis

  • Role in metastatic progression:

    • MIA2 expression has been associated with nodal and distant metastasis in multiple cancer types

    • Potential mechanisms include modulation of cell adhesion, extracellular matrix interactions, or epithelial-mesenchymal transition

    • The protein may influence the tumor microenvironment through paracrine signaling
      To investigate these hypotheses, researchers should design experiments that:

  • Compare MIA2 function across multiple tissue and cancer types

  • Examine both gain-of-function and loss-of-function effects

  • Assess impact on multiple cancer hallmarks (proliferation, invasion, angiogenesis)

  • Identify tissue-specific molecular interaction networks
    This mechanistic understanding is essential for determining whether MIA2 represents a potential therapeutic target and in which cancer contexts it might be most relevant.

How can advanced technologies be leveraged to elucidate MIA2's functional significance?

Emerging technologies offer unprecedented opportunities to explore MIA2's functional significance with greater precision and depth. Researchers should consider implementing these advanced approaches:

  • CRISPR-based technologies:

    • Gene editing for precise knockout or mutation of MIA2

    • CRISPRa/CRISPRi for modulating expression without permanent genetic alterations

    • CRISPR screens to identify synthetic lethal interactions with MIA2

    • Base editing for introducing specific mutations to study structure-function relationships

  • Single-cell technologies:

    • Single-cell RNA sequencing to examine cell-specific expression patterns

    • Single-cell proteomics to correlate MIA2 with other protein markers

    • Spatial transcriptomics to map MIA2 expression within tissue architecture

    • Cell lineage tracing to study dynamic expression changes during disease progression

  • Advanced imaging approaches:

    • Super-resolution microscopy for subcellular localization

    • Multiplexed immunofluorescence for co-localization studies

    • Live-cell imaging with fluorescent tags to track MIA2 dynamics

    • Intravital microscopy for in vivo visualization in animal models

  • Structural biology techniques:

    • Cryo-electron microscopy for protein structure determination

    • Hydrogen-deuterium exchange mass spectrometry for mapping interaction surfaces

    • Molecular dynamics simulations to predict functional domains

    • Protein engineering to create reporter constructs for functional studies
      When implementing these technologies, researchers should:

  • Validate findings using complementary methodological approaches

  • Integrate data across multiple technological platforms

  • Consider both hypothesis-driven and discovery-based experimental designs

  • Collaborate across disciplines to leverage specialized expertise
    This technology-forward approach accelerates discovery by providing higher-resolution insights into MIA2 biology than would be possible with conventional methodologies alone.

What methodological considerations are essential for translating MIA2 research into clinical applications?

Translating MIA2 research into clinical applications requires rigorous methodological approaches that bridge the gap between basic science and clinical implementation. Researchers pursuing translational studies should address these essential considerations:

  • Biomarker development framework:

    • Analytical validation: Establish assay precision, accuracy, sensitivity, and specificity

    • Clinical validation: Determine associations with clinical outcomes in retrospective cohorts

    • Clinical utility: Demonstrate impact on clinical decision-making and patient outcomes

    • Standardization: Develop reproducible protocols suitable for clinical laboratories

  • Pre-analytical factors:

    • Sample collection and processing standards

    • Stability assessment under different storage conditions

    • Evaluation of pre-analytical variables that might affect measurements

    • Development of reference materials and calibrators

  • Assay development considerations:

    • Platform selection based on sensitivity, specificity, and clinical applicability

    • Cutoff determination using robust statistical approaches

    • Quality control measures for longitudinal stability

    • Inter-laboratory reproducibility assessment

  • Clinical study design:

    • Prospective validation in well-defined patient cohorts

    • Appropriate control groups and blinding procedures

    • Integration with standard-of-care biomarkers

    • Assessment of incremental value over existing prognostic factors

  • Regulatory considerations:

    • Compliance with relevant guidelines for biomarker development

    • Documentation requirements for clinical validation

    • Quality management systems for assay implementation

    • Ethical considerations for patient testing
      This comprehensive translational framework ensures that promising findings regarding MIA2's clinical relevance can be effectively translated into validated clinical applications that ultimately benefit patients through improved diagnosis, prognosis, or treatment selection.

What are the most significant knowledge gaps in current MIA2 research?

  • Mechanistic understanding: The precise molecular mechanisms through which MIA2 influences cancer progression remain incompletely characterized. While associations with metastasis have been documented , the signaling pathways and molecular interactions mediating these effects need further elucidation.

  • Tissue specificity: The variable expression patterns across different cancer types suggest context-dependent functions that require systematic investigation across diverse tissue environments and molecular subtypes.

  • Functional validation: Most studies have focused on expression patterns rather than functional manipulation, limiting causal inferences about MIA2's role in cancer biology.

  • Standardized methodologies: Variation in detection methods, scoring systems, and threshold definitions complicates cross-study comparisons and meta-analyses.

  • Integration with emerging biomarkers: The relative value of MIA2 in the context of other molecular markers and multi-parameter signatures remains to be fully established.
    Addressing these knowledge gaps through rigorous experimental design, standardized methodologies, and collaborative research efforts will advance understanding of MIA2 biology and facilitate its potential clinical implementation as a biomarker or therapeutic target.

What future research directions hold the most promise for advancing MIA2 applications?

Future research directions that hold particular promise for advancing MIA2 applications include:

  • Comprehensive functional characterization:

    • Systematic CRISPR-based manipulation across diverse cellular contexts

    • Identification of critical domains through structure-function analysis

    • Characterization of post-translational modifications and their functional significance

    • Development of specific inhibitors or activators as experimental tools

  • Multi-omics integration:

    • Correlation of genomic alterations with MIA2 expression patterns

    • Epigenetic regulation studies to understand tissue-specific expression

    • Proteomic identification of interaction partners across different cancer types

    • Single-cell multi-omics to address cellular heterogeneity

  • Prospective clinical validation:

    • Large-scale, prospective studies with standardized methodology

    • Integration with established biomarker panels

    • Evaluation in the context of specific therapeutic interventions

    • Development of companion diagnostic applications

  • Artificial intelligence applications:

    • Machine learning for pattern recognition in MIA2 expression data

    • Predictive models incorporating MIA2 with other clinical and molecular parameters

    • Automated image analysis for standardized quantification in tissue samples

    • Natural language processing to synthesize findings across the research literature

  • Therapeutic targeting strategies:

    • Evaluation of MIA2 as a direct therapeutic target in appropriate contexts

    • Development of context-specific modulators based on mechanistic understanding

    • Combination approaches targeting MIA2-related pathways

    • Personalized approaches based on molecular profiling These forward-looking research directions leverage emerging technologies and interdisciplinary approaches to address current knowledge gaps and translate MIA2 research into meaningful clinical applications that ultimately improve cancer patient outcomes.

Product Science Overview

Gene and Expression

MIA-2 is mapped to the gene locus of human chromosome 14q13 . Its expression is transcriptionally regulated by the hepatocyte nuclear factor (HNF)-1-binding site . Unlike MIA and OTOR, which are exclusively expressed in the cartilage and cochlea respectively, MIA-2 is expressed exclusively in the liver .

Role in Hepatocellular Carcinoma (HCC)

MIA-2 plays a significant role in hepatocellular carcinoma (HCC). It is expressed in HCC but not in bladder, breast, or prostate cancer . MIA-2 inhibits HCC growth and invasion, acting as a tumor suppressor . In HCC cell lines and tissues, HNF-1 expression is lower than in primary human hepatocytes and corresponding non-tumorous tissue, correlating significantly with the down-regulation of MIA-2 expression . Re-expression of HNF-1 in HCC cells reinduces MIA-2 to similar levels as found in primary human hepatocytes .

Recombinant Human MIA-2

Recombinant Human MIA-2 (rhMIA-2) is produced in E. coli cells as a single non-glycosylated polypeptide chain containing 100 amino acids . It has a molecular mass of 11.6 kDa and is obtained through chromatographic techniques . RhMIA-2 is fully biologically active and has been shown to inhibit proliferation and invasion of HCC cells in vitro and in vivo .

Clinical Implications

The loss of MIA-2 expression in HCC is associated with more advanced tumor stages and a higher Ki67 labeling index, indicating a higher proliferation rate . Therefore, MIA-2 serves as a potential biomarker for HCC progression and a target for therapeutic intervention .

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 2024 Thebiotek. All Rights Reserved.