MAGEA6 Human

Melanoma Antigen Family A, 6 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biological Functions and Mechanisms

MAGEA6 acts as an oncoprotein with dual roles in cancer progression and autophagy regulation:

  • Autophagy Suppression: MAGEA6 inhibits basal and starvation-induced autophagy by stabilizing E3 ubiquitin ligases (e.g., TRIM28), promoting degradation of AMPKα1, and reducing autophagosome formation .

  • Tumorigenesis: Overexpression transforms nonmalignant cells (e.g., pancreatic ductal epithelial cells) into tumorigenic phenotypes in vivo .

  • Chemoresistance: In triple-negative breast cancer (TNBC), MAGEA6 silencing enhances doxorubicin sensitivity by reactivating autophagy and ferroptosis .

Key Research Findings:

Study FocusFindingsImplications
Esophageal Cancer (2022)MAGEA6 upregulates MSMO1, driving cell migration and proliferation via cholesterol biosynthesis pathways .Identifies MAGEA6-MSMO1 axis as a therapeutic target.
Pancreatic Cancer (2020)MAGEA6 degradation under nutrient stress reactivates autophagy, enabling tumor progression .Highlights MAGEA6 as a metabolic switch in cancer.
Lung Cancer Survival (2021)High MAGEA6 correlates with poor survival in stage I patients (HR = 1.96, p < 0.0001) but not in advanced stages .Suggests stage-specific therapeutic targeting.
TNBC Therapy (2024)MAGEA6 knockdown reduces IC₅₀ of doxorubicin by 60% and increases apoptosis in xenografts .Supports MAGEA6 inhibition for chemo-sensitization.

Clinical Relevance:

  • Biomarker Potential: MAGEA6 is overexpressed in 93.9% of esophageal squamous cell carcinoma (ESCC) tissues, with 100% specificity compared to paracancerous tissues .

  • Prognostic Value: High MAGEA6 mRNA levels correlate with advanced glioma, melanoma, and pancreatic cancer .

Recombinant MAGEA6 in Experimental Use

  • Applications: Used in in vitro assays to study ubiquitination, autophagy, and cancer cell viability .

  • Storage: Stable at -20°C in 10% glycerol; avoid freeze-thaw cycles .

Pathways and Interactions

  • Core Pathways:

    • AMPK signaling (degradation of AMPKα1) .

    • Cholesterol biosynthesis (via MSMO1 upregulation) .

    • Ubiquitin-proteasome system (E3 ligase recruitment) .

  • Protein Partners: TRIM28, MAGEA3, MAGEA12, and CTAG1B .

Limitations and Future Directions

  • Therapeutic Challenges: MAGEA6’s role in early vs. late cancer stages complicates targeted strategies .

  • Research Gaps: Mechanisms linking MAGEA6 to immune evasion remain underexplored despite its classification as a cancer/testis antigen .

Product Specs

Introduction
MAGEA6, a member of the MAGEA family, plays a crucial role in the expression of tumor antigens recognized by autologous cytolytic T lymphocytes (CTL) on human melanoma cells. The MAGEA family proteins share 50-80% sequence identity and are encoded by genes clustered at chromosomal location Xq28. Initially isolated from various tumors, MAGE genes are considered potential targets for cancer immunotherapy due to their nearly exclusive tumor-specific expression in adult tissues. The significant variability observed in the promoters and first exons of MAGEA genes suggests that this gene family allows for the expression of a common function under diverse transcriptional control. Notably, MAGEA genes have been implicated in several hereditary disorders, including dyskeratosis congenita.
Description
Recombinant MAGEA6, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 322 amino acids (1-314 a.a.) with a molecular mass of 35.9 kDa. It includes an 8 amino acid His-tag at the C-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The MAGEA6 protein solution (1 mg/ml) is supplied in a buffer containing 20 mM Tris-HCl (pH 8.0), 0.1 M NaCl, 10% glycerol, and 1 mM DTT.
Stability
For short-term storage (up to 4 weeks), keep refrigerated at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity is determined to be greater than 90% using SDS-PAGE analysis.
Synonyms
Melanoma-associated antigen 6, Cancer/testis antigen 1.6, CT1.6, MAGE-6 antigen, MAGE3B antigen, MAGEA6, MAGE6, MAGE3B, MAGE-3b.
Source
E.coli.
Amino Acid Sequence
MPLEQRSQHC KPEEGLEARG EALGLVGAQA PATEEQEAAS SSSTLVEVTL GEVPAAESPD PPQSPQGASS LPTTMNYPLW SQSYEDSSNQ EEEGPSTFPD LESEFQAALS RKVAKLVHFL LLKYRAREPV TKAEMLGSVV GNWQYFFPVI FSKASDSLQL VFGIELMEVD PIGHVYIFAT
CLGLSYDGLL GDNQIMPKTG FLIIILAIIA KEGDCAPEEK IWEELSVLEV FEGREDSIFG DPKKLLTQYF VQENYLEYRQ VPGSDPACYE FLWGPRALIE TSYVKVLHHM VKISGGPRIS YPLLHEWALR EGEELEHHHH HH.

Q&A

What is MAGEA6 and what are its expression patterns in normal versus cancerous tissues?

MAGEA6 is a member of the melanoma-associated antigen (MAGE) family A, which belongs to the cancer/testis antigens (CTAs). These antigens exhibit a highly restricted expression pattern in normal tissues, primarily limited to male germ cells, but are aberrantly expressed in various malignancies .

The MAGEA gene family is located on the X chromosome and consists of thirteen protein-encoding genes (MAGEA1 to A6, A8 to A12, A2B, and A9B) and one pseudogene, MAGEA7P . The aberrant expression of MAGEA proteins in cancer results from promoter hypomethylation due to genome-wide epigenetic reprogramming .

In bladder cancer studies, MAGEA6 expression has been observed in 78.4% male and 21.6% female patients, with a male/female ratio of 3.6, which aligns with the general prevalence of bladder cancer between sexes (estimated at 2:1 to 4:1) . Importantly, MAGEA antigens are expressed in a wide variety of malignant tumors but not in adult somatic cells, making them potentially valuable targets for cancer immunotherapy .

How does MAGEA6 influence autophagy in cancer cells?

MAGEA6 functions as a suppressor of macroautophagy (hereafter referred to as autophagy) in pancreatic ductal adenocarcinoma (PDAC) cell models . This suppression occurs through several mechanisms:

  • MAGEA6 inhibits AMPK Thr172 phosphorylation, which is a key activator of autophagy .

  • It reduces the induction of LC3B expression, an essential component of autophagosomes .

  • It impairs autophagic flux, as demonstrated by the slow accumulation of SQSTM1/p62 and LC3B in the presence of bafilomycin A1 (BafA1) .

When MAGEA6 is downregulated, either through degradation-prone mutations or by long-term starvation-induced protein degradation, its autophagy-suppressing effect is revoked, allowing autophagy re-initiation . This is evidenced by low S6K Thr389 and high AMPK Thr172 phosphorylation under these conditions .

What techniques are most effective for detecting and quantifying MAGEA6 expression?

Several complementary techniques have proven effective for detecting and quantifying MAGEA6 expression:

  • Western blotting: Used to detect MAGEA6 protein levels and phosphorylation status of related signaling molecules such as p70S6K (S6K) and AMPK .

  • Immunohistochemistry: Applied to visualize MAGEA6 expression in tumor tissues, as demonstrated in the BxPC-3 tumor xenograft models .

  • Co-immunoprecipitation assays: Utilized to detect protein-protein interactions and post-translational modifications of MAGEA6, such as polyubiquitination .

  • Bioinformatics approaches: Analysis of gene expression datasets (e.g., TCGA, COSMIC, ICGC) has been used to identify MAGEA6 mutations and expression patterns across cancer types .

  • Stable cell lines expressing MAGEA6 variants: Created using viral transduction to study the functional consequences of wild-type and mutant MAGEA6 expression .

How is MAGEA6 expression regulated in response to nutrient availability?

MAGEA6 expression is highly sensitive to nutrient availability, with significant implications for its function in cancer cells:

  • Nutrient deprivation: Culturing MAGEA6-expressing cells in medium without fetal bovine serum (FBS), glucose, or glutamine robustly downregulates MAGEA6 expression within 5 hours .

  • Carbon source dependence: Supplementation with glucose or glutamine partially rescues MAGEA6 expression under nutrient-deficient conditions, while FBS or combination of carbon sources fully recovers MAGEA6 levels in both short-term (5 hr) and long-term (24 hr) culture conditions .

  • mTORC1 signaling: Nutrient-dependent changes in MAGEA6 expression correlate with changes in p70S6K Thr389 phosphorylation, a target of mTORC1 kinase whose activity is suppressed under nutrient deprivation .

  • E3 ubiquitin ligase regulation: In certain cell lineages, FBS-dependent (but not glucose-dependent) MAGEA3/A6 protein stability is regulated by the CRL4-DCAF12 E3 ubiquitin ligase .

How do cancer-associated mutations affect MAGEA6 protein stability and function?

Cancer-associated mutations frequently lead to reduced MAGEA6 protein stability and altered function:

  • Proteasome-dependent degradation: Many MAGEA6 variants identified in cancer undergo proteasome-mediated degradation. Addition of the proteasome inhibitor MG132 to cells expressing MAGEA6 variants results in dramatic increases in mutant protein levels, reaching those similar to wild-type MAGEA6 .

  • Polyubiquitination: Cancer-associated mutants such as MAGEA6 H305fs* and MAGEA6 N254I show strong polyubiquitination signals, which are further enhanced by MG132 treatment, indicating that they are targeted for degradation via the ubiquitin-proteasome pathway .

  • Functional consequences: Degradation-prone mutations of MAGEA6 (such as H305fs* and N254I) show minimal changes in autophagy signaling compared to wild-type MAGEA6, suggesting that these mutations negate MAGEA6's normal function in autophagy suppression .

  • Distribution of mutations: Bioinformatic analyses have identified approximately 1000 unique non-silent MAGEA gene aberrations across various cancer datasets, with most being missense mutations clustered on specific MAGEA family members, including MAGEA6 .

What is the biphasic role of MAGEA6 in cancer progression?

MAGEA6 exhibits a fascinating biphasic role in cancer progression, particularly in pancreatic ductal adenocarcinoma:

  • Tumor initiation: MAGEA6 functions as a bona fide oncogene in early-stage PDAC by suppressing autophagy, which promotes tumor initiation .

  • Tumor progression: As tumors grow, MAGEA6 degradation (through mutations or nutrient stress) releases autophagy inhibition, which then paradoxically promotes further tumor development in late-stage disease .

  • Experimental evidence:

    • Overexpression of wild-type MAGEA6 in HPDE-iKRAS cells significantly increases tumor volume in xenograft models, while MAGEA6 variants that cannot suppress autophagy (H305fs*, N254I) show reduced tumor formation .

    • Conversely, knockdown of MAGEA6 in established BxPC-3 PDAC cells (with high endogenous wild-type MAGEA6) dramatically increases tumor volume compared to control xenografts .

    • Knockdown of autophagy regulators ATG7 and VPS34 suppresses tumor growth induced by MAGEA6 downregulation, confirming that autophagy activity plays a key role in promoting tumor growth in response to MAGEA6 depletion in established tumors .

This dual nature suggests that MAGEA6-dependent tumorigenicity is most critical at an early disease stage when low autophagy activity is beneficial, while in later stages, MAGEA6 degradation and subsequent autophagy reactivation become advantageous for tumor progression .

What are the implications of MAGEA6 mutation status for cancer immunotherapy?

The mutation status of MAGEA6 has significant implications for cancer immunotherapy approaches:

  • Target availability: Since many cancer-associated MAGEA6 mutations lead to protein degradation, these mutations may reduce the availability of the target antigen for immunotherapy, potentially compromising therapeutic efficacy .

  • Immunogenicity considerations: Changes in MAGEA6 expression during different stages of cancer progression may affect the immunogenicity of MAGEA6-based therapeutic approaches .

  • Clinical trial outcomes: Despite the high tumor specificity of MAGEA expression leading to multiple clinical trials targeting MAGEA genes with immunotherapy agents, responses have been modest . The discovery that cancer-associated mutations can affect MAGEA6 protein levels may partially explain these limited responses.

  • Strategic considerations:

    • Immunotherapeutic strategies may need to account for MAGEA6 expression heterogeneity and mutation-induced expression changes .

    • Combined approaches targeting multiple MAGEA family members might be more effective due to functional redundancy among family members .

    • Timing of MAGEA-targeted immunotherapy may be critical given the changing role of MAGEA6 during cancer progression .

What experimental models are most suitable for studying MAGEA6 in pancreatic cancer?

Several experimental models have proven valuable for investigating MAGEA6 in pancreatic cancer:

  • Cell line models:

    • HPDE-iKRAS cells: Human pancreatic ductal epithelial cells with inducible KRAS expression provide a model for early-stage pancreatic cancer initiation .

    • Established PDAC cell lines: AsPC-1, BxPC-3, and MIA PaCa-2 cells with endogenous MAGEA6 expression are useful for studying advanced disease .

  • Genetic manipulation approaches:

    • Lentiviral transduction: Used to create stable cell lines expressing wild-type or mutant MAGEA6 variants .

    • shRNA knockdown: Applied to reduce endogenous MAGEA6 expression in cell lines with high baseline expression .

    • CRISPR-Cas9: Although not explicitly mentioned in the search results, this technique could be used for precise genetic manipulation of MAGEA6.

  • In vivo models:

    • Xenograft mouse models: Subcutaneous implantation of MAGEA6-modified cells in immunocompromised mice allows assessment of MAGEA6's impact on tumor growth and progression .

    • Tissue analysis: Immunohistochemical staining of xenograft tumors for markers such as LC3B provides insights into autophagy activity in vivo .

  • Nutrient manipulation studies:

    • Culture in nutrient-deficient media: Experiments with media lacking FBS, glucose, or glutamine help elucidate how MAGEA6 responds to metabolic stress .

    • Pharmacological manipulation: Use of compounds like bafilomycin A1 (BafA1) to assess autophagic flux .

How can researchers differentiate between the effects of different MAGEA family members?

Differentiating between the effects of various MAGEA family members presents a significant challenge due to their high sequence homology and potential functional redundancy. Researchers can employ these strategies:

  • Specific antibodies: Use of highly specific antibodies that can distinguish between different MAGEA proteins, particularly between closely related members like MAGEA3 and MAGEA6 .

  • Gene-specific knockdown/knockout: Carefully designed siRNA, shRNA, or CRISPR-Cas9 approaches targeting unique regions of specific MAGEA genes .

  • Overexpression studies: Expression of individual MAGEA family members in appropriate cellular contexts to assess their specific functions .

  • Co-expression analysis: Evaluation of which MAGEA genes are co-expressed and potentially function complementarily, as suggested for MAGEA3 and MAGEA6 in regulating TRIM28 E3 activity and autophagy suppression .

  • Bioinformatic approaches: Analysis of cancer genomic databases to identify tumor types or contexts where specific MAGEA genes are predominantly expressed or mutated .

What are the key signaling pathways that mediate MAGEA6 function in cancer?

MAGEA6 interacts with several key signaling pathways in cancer:

  • Autophagy pathway: MAGEA6 suppresses autophagy by inhibiting:

    • AMPK Thr172 phosphorylation

    • LC3B expression

    • Autophagic flux (measured by SQSTM1/p62 and LC3B accumulation)

  • mTORC1 signaling: MAGEA6 expression correlates with p70S6K Thr389 phosphorylation, a target of mTORC1 kinase .

  • Ubiquitin-proteasome system: MAGEA6 interacts with E3 ubiquitin ligases:

    • MAGEA6 itself can be targeted for ubiquitination and proteasomal degradation

    • MAGEA family members (including MAGEA6) can bind to the TRIM28 E3 ligase and boost its ubiquitin ligase activity against targets like p53

  • Nutrient sensing pathways: MAGEA6 expression is regulated by nutrient availability and is linked to cellular metabolic status .

Understanding these pathway interactions is crucial for comprehending MAGEA6's complex role in cancer and developing targeted interventions.

How can the dynamic expression of MAGEA6 throughout cancer progression be effectively monitored?

Monitoring the dynamic expression of MAGEA6 throughout cancer progression requires a multi-faceted approach:

  • Temporal sampling: Collection of tumor samples at different stages of disease progression, either through longitudinal sampling when possible or through cross-sectional studies of tumors at different stages .

  • Spatial heterogeneity assessment: Analysis of MAGEA6 expression across different regions of the same tumor to account for intratumoral heterogeneity .

  • Multi-omics approach:

    • Transcriptomics: RNA sequencing or qPCR to measure MAGEA6 mRNA levels

    • Proteomics: Mass spectrometry or Western blotting to quantify MAGEA6 protein levels

    • Epigenomics: Analysis of promoter methylation status to understand regulatory mechanisms

  • Functional readouts: Measurement of autophagy markers (LC3B, p62) as functional indicators of MAGEA6 activity .

  • In vivo imaging: For animal models, techniques such as bioluminescence imaging of reporter-tagged MAGEA6 could provide real-time monitoring of expression changes.

  • Correlation with nutrient status: Assessment of MAGEA6 expression in relation to tumor microenvironment factors such as nutrient availability and metabolic stress .

What are the most promising approaches for targeting MAGEA6 in cancer therapy?

Several promising approaches for targeting MAGEA6 in cancer therapy emerge from current research:

  • Stage-specific targeting strategies:

    • Early-stage disease: Mimicking MAGEA6 function or enhancing its stability to maintain autophagy suppression

    • Late-stage disease: Promoting MAGEA6 degradation to inhibit tumor progression

  • Combination therapies:

    • MAGEA6-targeted immunotherapy combined with autophagy modulators

    • Targeting both MAGEA6 and other redundant MAGEA family members simultaneously

  • Synthetic lethality approaches:

    • Identifying cellular dependencies created by MAGEA6 expression

    • Developing drugs that selectively kill cells expressing MAGEA6

  • Nutrient sensitization:

    • Exploiting MAGEA6's nutrient-sensitive stability to enhance tumor cell killing under metabolic stress conditions

  • Ubiquitin-proteasome system modulation:

    • Developing compounds that selectively promote MAGEA6 degradation through the ubiquitin-proteasome pathway

    • Targeting the crosstalk between autophagy and the ubiquitin-proteasome system

What is the relationship between MAGEA6 and therapy resistance in cancer?

While the search results don't directly address therapy resistance, we can infer potential relationships based on MAGEA6's functions:

  • Autophagy modulation: Since MAGEA6 suppresses autophagy, and autophagy is known to contribute to therapy resistance in many cancers, MAGEA6 expression might influence response to therapies that depend on or are affected by autophagy status .

  • Metabolic adaptation: MAGEA6's nutrient-sensitive expression suggests it may play a role in metabolic adaptation to stress, which is a known mechanism of therapy resistance .

  • Cancer stem cell phenotypes: If MAGEA6 contributes to cancer stem cell-like properties (which isn't directly addressed in the provided search results), this could influence therapy resistance.

  • Tumor microenvironment interactions: MAGEA6's role in adaptation to nutrient stress suggests it might influence how cancer cells respond to microenvironmental challenges, including those imposed by therapy .

Future research should directly investigate how MAGEA6 expression and mutational status correlate with response to various cancer therapies, particularly those that modulate autophagy or target metabolic vulnerabilities.

What are the main challenges in studying MAGEA6 and how can they be overcome?

Researchers face several key challenges when studying MAGEA6:

  • Protein sequence homology within the MAGEA family:

    • Challenge: High sequence similarity between MAGEA family members makes specific detection difficult.

    • Solution: Develop highly specific antibodies targeting unique regions; use genetic approaches like CRISPR to specifically modify MAGEA6 .

  • Dynamic expression patterns:

    • Challenge: MAGEA6 expression changes with nutrient availability and disease progression.

    • Solution: Employ time-course experiments with carefully controlled nutrient conditions; use models that capture different disease stages .

  • Functional redundancy:

    • Challenge: Multiple MAGEA family members may have overlapping functions.

    • Solution: Simultaneous knockdown/knockout of multiple family members; study contexts where MAGEA6 is the predominant family member expressed .

  • Disease stage-dependent effects:

    • Challenge: MAGEA6 has opposite effects at different stages of cancer progression.

    • Solution: Use models representing both early and late-stage disease; carefully interpret results based on disease context .

  • Technical detection issues:

    • Challenge: Low abundance or rapid degradation of mutant MAGEA6 proteins.

    • Solution: Use proteasome inhibitors like MG132 to stabilize proteins; employ sensitive detection methods; use epitope tags when studying exogenous expression .

How can researchers accurately assess the impact of MAGEA6 on autophagy in complex systems?

Accurately assessing MAGEA6's impact on autophagy requires a comprehensive approach:

  • Multiple autophagy markers:

    • LC3B-I to LC3B-II conversion

    • SQSTM1/p62 accumulation

    • AMPK Thr172 phosphorylation status

    • Autophagosome formation via microscopy

  • Autophagic flux assessment:

    • Treatment with autophagy inhibitors like bafilomycin A1 to determine genuine changes in autophagy versus changes in marker turnover

    • Time-course experiments to capture dynamic changes

  • Genetic manipulation:

    • Knockdown/knockout of key autophagy genes (ATG7, VPS34) to validate MAGEA6-dependent effects

    • Expression of wild-type versus mutant MAGEA6 to assess structure-function relationships

  • In vivo validation:

    • Immunohistochemical staining of tumor tissues for autophagy markers

    • Correlation of MAGEA6 expression with autophagy status in xenograft models

  • Nutrient manipulation:

    • Assessment under both nutrient-rich and nutrient-poor conditions

    • Selective supplementation with specific nutrients (glucose, glutamine) to determine their individual contributions

By employing these approaches, researchers can more accurately determine the complex relationship between MAGEA6 expression and autophagy regulation in cancer.

Product Science Overview

Introduction

The Melanoma Antigen Family A (MAGE-A) is a group of proteins that belong to the larger family of cancer/testis (CT) antigens. These antigens are characterized by their restricted expression in immune-privileged tissues, such as the testis, and a variety of cancers. MAGE-A proteins have gained significant attention in cancer research due to their potential as biomarkers and therapeutic targets.

Discovery and Classification

The MAGE-A family was first identified in the early 1990s when researchers were searching for tumor-associated antigens. MAGE-A1 was the first member discovered, and subsequent studies identified more proteins with similar properties. The MAGE-A family consists of over 60 proteins, which are further subdivided into two groups based on the location of their genes .

Structure and Function

MAGE-A proteins share a common MAGE homology domain, which is crucial for their function. These proteins are typically presented on the cell surface by major histocompatibility complex (MHC) class I molecules, making them accessible to the immune system. The primary function of MAGE-A proteins is to promote cell survival and proliferation, which is why their expression is often associated with cancer .

Melanoma Antigen Family A, 6 (MAGE-A6)

MAGE-A6 is one of the members of the MAGE-A family. Like other MAGE-A proteins, MAGE-A6 is expressed in various cancers, including melanoma, bladder cancer, and others. Its expression is usually restricted to cancerous tissues and not found in normal tissues, except for the testis. This restricted expression pattern makes MAGE-A6 an attractive target for cancer immunotherapy .

Therapeutic Potential

The unique expression pattern of MAGE-A6 and other MAGE-A proteins has made them promising candidates for cancer immunotherapy. Several strategies are being explored to target these proteins, including cancer vaccines, adoptive T-cell transfer, and combination therapies. These approaches aim to harness the immune system to recognize and destroy cancer cells expressing MAGE-A6 .

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.