MEOX2 Human

Mesenchyme Homeobox 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of MEOX2 Human

MEOX2 (Mesenchyme Homeobox 2) is a human gene encoding a homeobox-containing transcription factor critical for mesodermal tissue development, including skeletal muscle, bone, and vascular systems . It regulates cell proliferation, differentiation, and survival, with emerging roles in cancer biology and neurological functions .

Developmental Biology

  • Somitogenesis: Essential for sclerotome development and limb muscle differentiation via regulation of Pax3 and Myf5 .

  • Angiogenesis: Inhibits endothelial cell activation by suppressing NF-κB signaling .

Cancer Biology

  • Pro-Tumorigenic Role in Glioma:

    • Upregulates Cathepsin S (CTSS), promoting cell proliferation and motility .

    • Associated with poor prognosis (HR = 1.8, p < 0.001) .

  • Anti-Tumor Role in Breast Cancer:

    • Low expression correlates with dense vascularization and poor survival (HR = 0.65, p = 0.003) .

    • Induced by chemotherapy agents (cisplatin, epirubicin) .

Cancer TypeMEOX2 ExpressionPrognostic ImpactKey Mechanism
GliomaUpregulatedPoor survivalCTSS activation → EMT promotion
Breast CancerDownregulatedImproved survival if elevatedAngiogenesis suppression

Vascular and Inflammatory Regulation

  • Inhibits NF-κB activity in endothelial cells, reducing responses to proangiogenic factors .

  • Modulates CDKN1A (DNA-dependent) and CDKN2A (DNA-independent) to control cell cycle arrest .

Neurological Functions

  • Expressed in dorsal root ganglia (DRG) nociceptors; haploinsufficiency reduces pain sensitivity in mice .

  • Regulates Scn9a and Scn11a sodium channels, affecting neuronal excitability .

Clinical and Therapeutic Implications

  • Biomarker Potential:

    • Diagnostic marker for breast cancer vascular density .

    • Therapeutic target for anti-angiogenic therapies in tumors .

  • Drug Interactions:

    • Everolimus (mTOR inhibitor) upregulates MEOX2 in breast cancer .

Research Gaps and Future Directions

  • Dual roles in cancer (pro- vs. anti-tumor) require context-specific mechanistic studies.

  • Limited data on MEOX2’s role in non-cancer pathologies (e.g., neurovascular disorders) .

Product Specs

Introduction
Homeobox protein MOX-2, also known as MEOX2, belongs to a family of nonclustered, diverged homeobox genes. These genes exhibit overlapping expression patterns in the paraxial mesoderm and its derivatives. MEOX2 plays a crucial role in regulating vertebrate limb myogenesis. Furthermore, it is involved in mesoderm induction, early regional specification of the mesoderm, somitogenesis, and both myogenic and sclerotomal differentiation. Notably, mutations in the mouse MEOX2 gene have been linked to craniofacial and/or skeletal abnormalities. These mutations are also associated with neurovascular dysfunction, a characteristic feature observed in Alzheimer's disease.
Description
Recombinant human MEOX2, expressed in E. coli, is a single polypeptide chain with a molecular weight of 15.9 kDa. It encompasses amino acids 188-304 and includes a 23 amino acid His-tag fused to its N-terminus. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
The product appears as a clear solution that has undergone sterile filtration.
Formulation
The MEOX2 solution is provided at a concentration of 0.25 mg/ml. It is formulated in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 0.2 M NaCl, 40% glycerol, and 2 mM DTT.
Stability
For short-term storage (2-4 weeks), the product should be kept refrigerated at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure long-term stability during frozen storage, the addition of a carrier protein (0.1% HSA or BSA) is advisable. It is crucial to avoid subjecting the product to repeated freeze-thaw cycles.
Purity
The purity of the product is determined to be greater than 95% based on SDS-PAGE analysis.
Synonyms
GAX, MOX2, Homeobox protein MOX-2, Growth arrest-specific homeobox, Mesenchyme homeobox 2, MEOX2.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSRKERTAF TKEQIRELEA EFAHHNYLTR LRRYEIAVNL DLTERQVKVW FQNRRMKWKR VKGGQQGAAA REKELVNVKK GTLLPSELSG IGAATLQQTG DSIANEDSHD SDHSSEHAHL.

Q&A

What is MEOX2 and what is its primary function in human development?

MEOX2 (also known as GAX or MOX-2) is a transcription factor belonging to the homeobox gene family. It plays a critical role in mesoderm differentiation, including the development of bones, muscles, vasculature, and dermatomes . MEOX2 regulates myogenic genes such as Pax3 and Myf5, controlling the development of limb musculature . It is expressed in the paraxial mesoderm as early as embryonic days 8.5-9, making it essential for proper mammalian muscle and bone development as well as vascular differentiation . Methodologically, developmental studies of MEOX2 require carefully timed embryonic assessments combined with gene expression analyses to track its spatial and temporal expression patterns.

In which human tissues is MEOX2 predominantly expressed?

MEOX2 shows variable expression across different human tissues. Based on mouse model studies (which provide insight into likely human expression patterns), MEOX2 is highly expressed in both peripheral and central nervous systems, including dorsal root ganglia (DRG), spinal cord, cerebellum, hippocampus, hypothalamus, and cortex . It is also expressed at significant levels in the liver and heart, with moderate expression in kidney and lung . The table below summarizes MEOX2 expression patterns:

Tissue TypeMEOX2 Expression Level
DRGHigh
Spinal CordHigh
CerebellumHigh
HippocampusHigh
HypothalamusHigh
CortexHigh
LiverNotable
HeartNotable
KidneyVariable
LungVariable
Normal BrainVery low/undetectable

Importantly, in normal brain tissue, MEOX2 expression is typically very low or undetectable, which contrasts with its upregulation in certain brain cancers .

How does MEOX2 influence pain perception pathways?

MEOX2 plays a significant role in nociception (pain perception). Research using Meox2 heterozygous (Meox2+/−) mouse models has revealed impaired acute and inflammatory pain responses, including significant reduction in behavioral responses to noxious heat and capsaicin injections . At the molecular level, this appears to be related to decreased expression of Scn9a and Scn11a mRNA, which encode voltage-gated sodium channels Nav1.7 and Nav1.9, respectively . These channels are crucial for subthreshold amplification and action potential initiation in nociceptors. Electrophysiological studies have shown that MEOX2 deficiency leads to decreased action potentials fired upon ramp current injection, providing a mechanistic explanation for the altered nociceptive behavioral phenotype .

What methodologies are most effective for studying MEOX2 in sensory neurons?

Several complementary methodologies are essential for comprehensive investigation of MEOX2 in sensory neurons:

  • Protein Expression Analysis:

    • Western blotting for quantitative assessment in various tissues

    • Immunohistochemistry with double-labeling (MEOX2 + sensory neuron markers like CGRP, TrkA) for cellular localization

  • Functional Studies:

    • Electrophysiological recordings to measure neuronal excitability and action potential generation

    • Calcium imaging to assess responses to sensory stimuli

    • Behavioral assays (thermal, mechanical, chemical) to evaluate pain responses in vivo

  • Molecular Characterization:

    • Transcriptomic profiling (RNA-seq) to identify downstream targets

    • qPCR for targeted gene expression analysis, particularly of ion channels (e.g., Scn9a, Scn11a)

    • Chromatin immunoprecipitation to identify direct transcriptional targets

These approaches should be integrated to establish clear connections between molecular changes and functional outcomes in sensory processing.

What is the relationship between MEOX2 and neuropathic pain conditions?

MEOX2 has emerging connections to neuropathic pain conditions. Research has shown that MEOX2 mRNA dysregulation occurs in fibroblasts derived from Congenital Insensitivity to Pain (CIP) patients with mutations in the transcription factor PRDM12 . CIP is a rare genetic disorder affecting nociceptor survival or function, rendering patients unable to feel painful stimuli. The decreased expression of sodium channel genes (Scn9a and Scn11a) in MEOX2-deficient models suggests that MEOX2 may regulate pain sensitivity through controlling ion channel expression in sensory neurons . This finding has significant implications for understanding genetic pain disorders. Experimentally, investigating this relationship requires genetic association studies in patient populations, transcriptomic profiling of human sensory neurons, and functional validation in animal models with altered MEOX2 expression.

How does MEOX2 contribute to glioblastoma pathogenesis?

MEOX2 functions as an oncogenic transcription factor in glioblastoma (GBM), particularly in IDH wild-type tumors. MEOX2 expression is significantly elevated in IDH wild-type GBM compared to other brain tumors, with expression restricted to tumor cells rather than surrounding normal tissue . Mechanistically, MEOX2 overexpression increases ERK phosphorylation, activating the MAPK signaling pathway critical for cancer progression .

The oncogenic activities of MEOX2 in GBM include:

  • Promoting tumor growth: MEOX2 overexpression increases tumor growth in vivo and provides a proliferative advantage in human cerebral organoid models of GBM

  • Enhancing tumor initiation: MEOX2 cooperates with p53 and PTEN loss to promote tumor initiation in cerebral organoid models

  • Activating oncogenic pathways: MEOX2 activates pathways involved in MAPK signaling and extracellular matrix organization

  • Regulating gene networks: MEOX2 binds to oncogenic ETS factors and known glioma oncogenes such as FABP7

MEOX2's location on chromosome 7, which frequently shows gain in GBM, suggests it may be one of the key oncogenes co-opted during gliomagenesis .

What experimental models best capture MEOX2 function in glioblastoma research?

Several experimental models have proven valuable for studying MEOX2 in glioblastoma:

  • Patient-derived tumorsphere lines:

    • Maintain genetic and phenotypic characteristics of original tumors

    • Allow for genetic manipulation (e.g., CRISPR/Cas9-mediated knockdown)

    • Enable molecular and functional studies in a renewable resource

  • Cerebral organoid models:

    • Provide 3D architecture that recapitulates human brain tissue

    • Can incorporate genetic alterations (MEOX2 overexpression, p53/PTEN loss)

    • Excellent for studying tumor initiation in a developmentally relevant context

  • In vivo xenograft models:

    • Patient-derived GBM cells implanted into immunocompromised animals

    • Allow assessment of tumor growth kinetics and invasion patterns

    • Enable testing of therapeutic approaches targeting MEOX2 pathways

  • Primary tumor samples:

    • Fresh frozen GBM specimens for direct analysis of MEOX2 binding and expression

    • Provide clinically relevant data on MEOX2's role in human tumors

The integration of these complementary models provides comprehensive insights into MEOX2's oncogenic functions in GBM.

How do we identify direct transcriptional targets of MEOX2 in cancer cells?

Identifying direct transcriptional targets of MEOX2 in cancer cells requires sophisticated genomic approaches:

  • Genome-wide binding assays:

    • Antibody-guided Chromatin Tagmentation Sequencing (ACT-seq) in patient-derived tumorsphere lines

    • Cleavage Under Targets and Tagmentation Sequencing (CUT&Tag) in fresh frozen GBM tumors

    • These techniques reveal that MEOX2 predominantly binds to distal regulatory elements (intergenic and intronic regions)

  • Motif analysis:

    • Computational identification of enriched sequence motifs within MEOX2 binding peaks

    • Reveals significant enrichment for putative MEOX2/MEOX1 binding sites

  • Integrative genomics:

    • Combining binding data with expression profiling after MEOX2 knockdown or overexpression

    • Allows discrimination between direct and indirect targets

  • Pathway analysis:

    • Subjecting MEOX2-bound regions to pathway enrichment analysis

    • Identifies biological functions including PI3K/AKT signaling, MAPK signaling, and cell-cell communication

Key direct MEOX2 target genes identified across multiple GBM samples include:

GeneFunctionPathway Involvement
ETV1, ETV5, ETS1ETS family transcription factorsCell proliferation, invasion
SPRY2MAPK pathway attenuatorRTK/MAPK signaling regulation
DUSP10Dual-specificity phosphataseMAPK pathway regulation
FABP7Fatty acid binding proteinGlioma cell migration, proliferation

These methodologies collectively provide a comprehensive map of MEOX2's direct transcriptional program in cancer cells .

How do post-translational modifications regulate MEOX2 transcriptional activity?

Post-translational modifications (PTMs) critically regulate MEOX2 activity, with phosphorylation emerging as particularly important. Research has identified that:

  • Phosphorylation sites affect transcriptional activity:

    • Specific phosphorylation sites on MEOX2 regulate its transcriptional activity by altering subnuclear localization

    • This spatial regulation determines which genomic regions MEOX2 can access

  • Signaling pathway integration:

    • MEOX2 overexpression increases ERK phosphorylation

    • MEK inhibition with trametinib decreases both p-ERK levels and MEOX2 target gene expression (p21)

    • This suggests a regulatory feedback loop where MEOX2 activity is modulated by the very pathway it activates

  • Context-dependent effects:

    • Phosphorylation may explain why MEOX2 can function either as a tumor suppressor or oncogene depending on cellular context

    • The specific kinases responsible for MEOX2 phosphorylation likely vary by tissue and disease state

Methodological approaches for studying these modifications include mass spectrometry to identify specific phosphorylation sites, phosphomimetic and phospho-deficient mutants to determine functional consequences, and immunofluorescence to track changes in MEOX2 localization. Understanding these regulatory mechanisms could reveal new therapeutic targets in diseases where MEOX2 contributes to pathogenesis.

What genomic approaches best characterize the MEOX2 regulatory network?

Comprehensive characterization of the MEOX2 regulatory network requires integrative genomic approaches:

  • Multi-omics integration:

    • Combining transcriptomics (RNA-seq), genomics (DNA-seq), and epigenomics (ATAC-seq, ChIP-seq)

    • Correlating MEOX2 binding sites with chromatin accessibility and gene expression changes

    • Identifying co-occurring transcription factor binding motifs near MEOX2 binding sites

  • Systems biology methods:

    • Network analysis to identify hub genes within MEOX2-regulated pathways

    • Gene Ontology enrichment to categorize biological processes controlled by MEOX2

    • Pathway analysis to identify signaling networks influenced by MEOX2 activity

  • Perturbation studies:

    • CRISPR/Cas9-mediated knockdown combined with RNA-seq to identify differentially expressed genes

    • Overexpression systems to study gain-of-function effects on gene networks

    • Small molecule inhibitors of downstream pathways to dissect regulatory hierarchies

  • Single-cell approaches:

    • Single-cell RNA-seq to identify cell populations with MEOX2 expression and characterize cell-specific regulatory networks

    • Single-cell ATAC-seq to correlate chromatin accessibility with MEOX2-dependent gene expression

These approaches have revealed that MEOX2 regulates several oncogenic pathways in GBM, including MAPK signaling, extracellular matrix organization, and cell-cell communication pathways , providing a comprehensive view of its regulatory network.

How does MEOX2 chromosomal context influence its role in disease?

MEOX2's genomic location on chromosome 7 and its chromosomal context significantly impact its role in disease:

  • Chromosome 7 gain in glioblastoma:

    • GBM frequently exhibits gain of chromosome 7 as an early driver event

    • MEOX2, located on chromosome 7, shows increased expression in GBM likely as a direct consequence of this chromosomal gain

    • This suggests MEOX2 may be one of the key oncogenes on chromosome 7 that drives gliomagenesis

  • Genomic neighbors and regulatory elements:

    • MEOX2 binding predominantly occurs at distal regulatory elements (intergenic and intronic regions)

    • This binding pattern suggests complex long-range chromatin interactions may regulate MEOX2 target gene expression

    • Studying these interactions requires chromosome conformation capture technologies (Hi-C, 4C, etc.)

  • Epigenetic landscape:

    • The chromatin state surrounding the MEOX2 locus likely influences its expression in different tissues or disease states

    • Aberrant epigenetic modifications could contribute to dysregulated MEOX2 expression in cancer

  • Co-amplified genes:

    • Other genes on chromosome 7 may be co-amplified with MEOX2 in GBM

    • The combined effect of multiple co-amplified genes may synergistically contribute to disease progression

    • Disentangling individual contributions requires careful genetic manipulation experiments

Understanding these genomic and chromosomal contexts provides critical insights into why MEOX2 becomes dysregulated in certain diseases and may suggest novel therapeutic approaches that consider its genomic environment.

What are the critical controls needed when studying MEOX2 in different experimental systems?

Robust MEOX2 research requires careful consideration of appropriate controls across various experimental systems:

  • Genetic manipulation studies:

    • CRISPR/Cas9 knockdown: Include multiple sgRNA targets and appropriate non-targeting controls

    • Overexpression: Use empty vector controls and consider physiologically relevant expression levels

    • Rescue experiments: Re-express MEOX2 in knockout models to verify phenotype specificity

  • Antibody-based techniques:

    • Validate antibody specificity using MEOX2 knockout/knockdown samples as negative controls

    • Include isotype controls for immunoprecipitation experiments

    • Perform epitope mapping to ensure antibodies recognize relevant MEOX2 domains

  • Animal models:

    • Compare heterozygous (Meox2+/−) with wild-type littermates to control for genetic background

    • Consider conditional knockout models to avoid developmental confounds

    • Include sham controls for surgical interventions

  • Genomic studies:

    • For ACT-seq/CUT&Tag: Include IgG controls and knockdown samples as specificity controls

    • For RNA-seq: Include appropriate time-matched and treatment-matched controls

    • For pathway analysis: Use multiple algorithm approaches to confirm enriched pathways

  • Cellular models:

    • Patient-derived cells: Compare multiple patient lines to account for inter-tumor heterogeneity

    • Cell lines: Authenticate lines and control for passage number

    • Organoids: Include non-tumor organoids as baseline controls

These rigorous controls ensure that observed phenotypes and molecular changes are specifically attributable to MEOX2 alteration rather than experimental artifacts.

How can researchers resolve contradictory findings about MEOX2 function in different contexts?

Resolving contradictory findings about MEOX2 function requires systematic approaches:

  • Context-specific investigation:

    • MEOX2 exhibits context-dependent functions, acting either as a tumor suppressor or oncogene depending on cellular context

    • Systematically study MEOX2 in identical experimental conditions across different cell types

    • Compare expression levels, binding patterns, and downstream targets across contexts

  • Mechanistic dissection:

    • Identify molecular determinants of context-specific activities (co-factors, PTMs)

    • Characterize cell-type-specific protein-protein interactions that may alter MEOX2 function

    • Analyze chromatin landscape differences that might affect accessibility to target genes

  • Methodological standardization:

    • Employ consistent methodologies across studies (antibodies, genetic manipulation techniques)

    • Establish baseline expression levels in reference tissues for meaningful comparisons

    • Use multiple methodological approaches to verify key findings

  • Integrated data analysis:

    • Perform meta-analysis of published datasets to identify consistent patterns

    • Integrate multi-omics data to build comprehensive models of MEOX2 function

    • Use systems biology approaches to reconcile apparently contradictory observations

  • Collaborative research:

    • Establish multi-laboratory validation studies for key findings

    • Develop shared reagents and protocols to minimize technical variation

    • Create centralized repositories of MEOX2-related data for comprehensive analysis

By implementing these approaches, researchers can better understand the true spectrum of MEOX2 functions and the factors that determine its context-specific activities.

What novel methodologies are emerging for studying MEOX2 in human tissues?

Several cutting-edge methodologies are advancing our understanding of MEOX2 in human tissues:

  • Spatial transcriptomics/proteomics:

    • Technologies like Visium or CODEX allow visualization of MEOX2 expression in intact tissue sections

    • Maintains spatial relationship between MEOX2-expressing cells and their microenvironment

    • Particularly valuable for studying tumor heterogeneity and invasion patterns

  • Single-cell multi-omics:

    • Simultaneous profiling of genome, transcriptome, and epigenome in single cells

    • Reveals cell-type-specific MEOX2 regulatory networks and heterogeneity

    • Captures rare cell populations that might be missed in bulk analyses

  • CRISPR screening in primary tissues:

    • Pooled CRISPR screens targeting MEOX2 pathway components in primary human tissues

    • Identifies synthetic lethal interactions and essential co-factors

    • Can be performed in organoid models for higher throughput

  • In situ genome and transcriptome sequencing:

    • Techniques like SeqFISH or MERFISH enable visualization of MEOX2 mRNA in tissue context

    • Can be combined with protein detection for multi-parameter analysis

    • Preserves tissue architecture for understanding MEOX2 function in complex environments

  • Human tissue-derived experimental models:

    • Cerebral organoids derived from patient iPSCs with genetic manipulation of MEOX2

    • Patient-derived xenografts that maintain tumor heterogeneity

    • Ex vivo culture systems that preserve tissue architecture and microenvironment

  • Advanced computational methods:

    • AI/machine learning approaches to integrate complex MEOX2-related datasets

    • Network inference algorithms to predict MEOX2 interactions from large-scale data

    • Virtual screening for compounds that might modulate MEOX2 activity

These emerging methodologies promise to provide unprecedented insights into MEOX2 function in human tissues, potentially revealing new therapeutic targets for MEOX2-associated diseases.

Product Science Overview

Gene and Protein Structure

The MEOX2 gene is located on chromosome 7p21.2 in humans . It belongs to a family of non-clustered, diverged homeobox genes. The protein encoded by MEOX2 contains a conserved homeodomain, which is a characteristic feature of homeobox proteins. This homeodomain is responsible for binding to specific DNA sequences and regulating the expression of target genes .

Function and Mechanism

MEOX2 functions as a transcription factor, meaning it binds to specific DNA sequences to regulate the transcription of genes. It activates the expression of CDKN1A and CDKN2A in endothelial cells, playing a regulatory role in vascular cell proliferation . Specifically, it activates CDKN1A in a DNA-dependent manner and CDKN2A in a DNA-independent manner .

During embryogenesis, MEOX2 is expressed in a wide range of mesodermal structures, including somites, vertebrae, developing limbs, head muscles, and the developing palate . This expression pattern suggests that MEOX2 is involved in the development of craniofacial and skeletal structures .

Clinical Significance

Mutations or dysregulation of the MEOX2 gene have been associated with various developmental disorders and diseases. For example, alterations in MEOX2 expression have been linked to craniofacial and skeletal abnormalities . Additionally, MEOX2 has been implicated in vascular diseases due to its role in regulating vascular cell proliferation .

Research and Applications

Recombinant MEOX2 protein is used in various research applications to study its function and role in development and disease. Researchers utilize recombinant MEOX2 to investigate its regulatory mechanisms and interactions with other proteins and genes. This research is crucial for understanding the molecular basis of diseases associated with MEOX2 and for developing potential therapeutic strategies.

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.