MAFG (MAF BZIP Transcription Factor G) is a human recombinant protein belonging to the small Maf transcription factor family. These proteins regulate gene expression by dimerizing with other transcription factors, influencing processes such as oxidative stress response, hematopoiesis, and metabolic regulation . MAFG lacks a transactivation domain, functioning as a transcriptional repressor when homodimerized but acting as an activator when heterodimerized with partners like NFE2 or NRF2 . Its recombinant form (MAFG Human) is widely used in biochemical and biomedical research to study gene regulation mechanisms and disease pathways .
Protein structure:
Production: Recombinantly expressed in Escherichia coli and purified via chromatographic techniques .
MAFG partners with proteins such as:
NFE2: Regulates hematopoietic gene expression and platelet production .
NRF2: Activates antioxidant response elements (ARE) under oxidative stress .
BACH1: Represses heme oxygenase-1 (HMOX1) and other cytoprotective genes .
Cancer: MAFG overexpression in non-small-cell lung cancer (NSCLC) confers resistance to cisplatin by enhancing reactive oxygen species (ROS) detoxification .
Metabolic Disorders: MAFG modulates hepatic glucose metabolism and mTOR signaling, influencing insulin resistance and diabetes .
Neurological Defects: Deficiencies in MAFG and related proteins (e.g., MAFK) cause progressive neuronal degeneration and cataracts in mice .
Genotype | Phenotype |
---|---|
Mafg⁻/⁻ | Mild thrombocytopenia, motor ataxia |
Mafg⁻/⁻::Mafk⁺/⁻ | Severe thrombocytopenia, neuronal degeneration, cataracts |
Mafg⁻/⁻::Mafk⁻/⁻ | Perinatal lethality, severe developmental defects |
MAFG upregulation in NSCLC correlates with poor prognosis and cisplatin resistance .
MAFG aptamers (e.g., apMAFG6F) reduce ROS detoxification, restoring chemosensitivity .
Hepatic MAFG represses bile acid synthesis genes (e.g., Cyp7a1, Cyp8b1) via FXR signaling, altering bile acid composition .
Mafg knockdown in mice increases gluconeogenic gene expression (e.g., G6pc, Pck1), elevating blood glucose .
MAFG-associated loci (e.g., rs6706649) influence gene expression diversity across populations, with 1,310 eQTLs specific to non-European groups .
MAFG recruits DNMT3B to promote CpG island hypermethylation in cancer .
MAFG interacts with key proteins involved in transcription and chromatin remodeling:
Partner | Function | Interaction Score |
---|---|---|
NFE2L2 | Antioxidant response activation | 0.999 |
BACH2 | Transcriptional repression | 0.981 |
DNMT3B | DNA methylation | 0.952 |
MAFK | Redundant transcriptional regulation | 0.975 |
MAFG is a basic-leucine zipper (bZIP) transcription factor belonging to the family of proteins related to the v-maf oncogene. The human MAFG gene consists of at least three exons separated by small introns, with the first exon being untranslated. The genomic structure is highly conserved between human and chicken, suggesting evolutionary importance of this transcription factor . MAFG is mapped to human chromosome region 17q25 through fluorescence in situ hybridization techniques, a telomeric region associated with several potential human disease loci .
MAFG functions through protein-protein interactions, forming heterodimers with various partners to regulate transcription. One key partner is the blood cell-specific bZIP factor p45 NF-E2, indicating MAFG's role in regulating hematopoietic gene expression . In melanoma research, MAFG has been shown to interact with MITF (Microphthalmia-associated transcription factor), with this MAFG~MITF complex co-occupying numerous genomic sites to influence target gene expression . These protein interactions can be studied using co-immunoprecipitation, proximity ligation assays, and chromatin immunoprecipitation followed by sequencing (ChIP-seq).
To quantify MAFG expression, researchers employ multiple complementary techniques:
RT-qPCR for mRNA quantification, which showed induction of MAFG following treatment with FXR agonists (GW4064 and GSK2324) in liver tissues
Western blotting for protein detection, which demonstrated 2-3 fold induction of MAFG protein after FXR agonist treatment
Immunohistochemistry for tissue localization studies
RNA-seq for transcriptome-wide expression analysis
In melanoma studies, researchers have documented increased MAFG expression correlating with advancing human melanoma stages, requiring careful normalization to housekeeping genes and statistical analysis of expression data .
MAFG functions as a direct target gene of FXR (Farnesoid X Receptor) and operates as a transcriptional repressor in bile acid synthesis pathways. Research has shown that hepatic overexpression of MAFG in mice represses genes encoding enzymes of both the classic and alternative bile acid synthesis pathways . This repression particularly affects Cyp8b1 expression, resulting in decreased cholic acid and increased muricholic acid levels in bile. These findings were validated through multiple experimental approaches:
Adenoviral overexpression of MAFG in mouse liver
Measurement of bile acid composition using liquid chromatography-mass spectrometry
Gene expression analysis by qPCR
Comparative studies using control adenovirus and other transcription factors (Ad-Crip2, Ad-Zfp385a)
MAFG expression is directly regulated by FXR activation. Experimental data shows:
Treatment of wild-type mice with FXR agonists (GW4064 or GSK2324) induced MAFG mRNA and protein levels 2-3 fold
This induction was absent in FXR knockout (FXR^-/-) mice, confirming FXR dependence
CDCA (chenodeoxycholic acid), a natural FXR agonist, increased MAFG expression in human HepG2 cells in a dose-dependent manner
The MAFG-dependent pathway appears conserved between mice and human cells
Researchers studying MAFG's role in bile acid metabolism employ these methodological approaches:
Loss-of-function studies: Knockdown of MAFG with antisense oligonucleotides or using MAFG+/- mice resulted in de-repression of Cyp8b1 and increased biliary cholic acid levels
Gain-of-function studies: Adenoviral-mediated MAFG overexpression decreased cholic acid and increased muricholic acid levels
Comparative analysis: Total bile acid levels in liver, intestine, and gallbladder remained unchanged despite MAFG modulation, indicating qualitative rather than quantitative changes
Cross-species validation: Confirmation of similar regulatory mechanisms in human HepG2 cells demonstrates evolutionary conservation of this pathway
Recent research has identified MAFG as a potent driver of melanoma progression through multiple lines of evidence:
MAFG expression increases with advancing human melanoma stages
Ectopic MAFG expression enhances malignant behavior of human melanoma cells in:
In vitro assays
Xenograft models
Genetic mouse models of spontaneous melanoma
MAFG induces a phenotype switch from a melanocytic state to a more dedifferentiated state, which is associated with increased aggressiveness and therapy resistance
The mechanistic interaction between MAFG and MITF represents a novel regulatory pathway in melanoma:
MAFG physically interacts with MITF, the master regulator of melanocyte development and function
This interaction is required for the pro-tumorigenic effects of MAFG
MAFG and MITF co-occupy numerous genomic sites
MAFG overexpression influences the expression of genes harboring binding sites for the MAFG~MITF complex
This interaction facilitates a shift toward a dedifferentiated melanoma cell state
This research reveals an unappreciated mechanism of MITF regulation with significant implications for understanding melanoma progression.
Researchers investigating MAFG in cancer utilize multiple complementary models:
Cell line studies: Human melanoma cell lines with controlled MAFG expression
Xenograft models: Implantation of MAFG-manipulated cancer cells into immunocompromised mice
Genetic mouse models: Engineered to develop spontaneous melanoma with modulated MAFG expression
Patient-derived samples: For correlation of MAFG expression with clinical parameters and outcomes
Each model provides unique insights, with genetic mouse models offering the most physiologically relevant system for studying MAFG's role in tumor initiation and progression.
To map MAFG binding sites across the genome, researchers typically employ:
ChIP-seq (Chromatin Immunoprecipitation followed by sequencing): This technique identified co-occupancy of MAFG and MITF at numerous genomic loci in melanoma studies
CUT&RUN or CUT&Tag: These more sensitive alternatives to ChIP-seq can be used with lower cell numbers
ATAC-seq: To identify accessible chromatin regions where MAFG might bind
Motif analysis: Computational approaches to identify potential MAFG binding motifs (MARE sites) within ChIP-seq peaks
The integration of these approaches provides a comprehensive map of MAFG's genomic occupancy and potential transcriptional targets.
To investigate how MAFG influences gene expression:
RNA-seq following MAFG modulation: Reveals genome-wide transcriptional changes
MAFG overexpression/knockdown experiments: In hepatocytes, MAFG overexpression repressed bile acid synthesis genes, while knockdown led to their de-repression
Reporter assays: Using constructs containing MAFG binding sites linked to luciferase reporters
Nascent RNA analysis: Techniques like GRO-seq or PRO-seq to distinguish direct from indirect transcriptional effects
Single-cell approaches: To capture heterogeneity in MAFG-mediated responses across cell populations
When facing contradictory results in MAFG research:
Context specificity analysis: MAFG may function differently depending on:
Cell/tissue type (e.g., liver vs. melanoma)
Dimerization partners available (e.g., MITF in melanoma, NF-E2 in hematopoietic cells)
Metabolic state or disease context
Technical validation across platforms: Verify findings using multiple:
Expression modulation approaches (siRNA, shRNA, CRISPR, overexpression)
Detection methods (antibodies, tagged constructs)
Model systems (cell lines, primary cells, animal models)
Dose-response relationships: MAFG effects may vary with expression level, as seen in dose-dependent CDCA studies
Temporal considerations: Acute vs. chronic MAFG modulation may yield different outcomes
Human-based NAMs could significantly enhance MAFG research through:
Advanced in vitro models: Human-derived microphysiological systems (organ-on-chip) could better recapitulate MAFG function in complex tissue environments like liver or skin
In silico approaches: Computational models predicting MAFG binding and functional outcomes across diverse genomic contexts
Integration of diverse methodologies: Combining in vitro, in silico, and in chemico approaches as outlined in NIH's human-based NAM initiative
Patient-derived models: Using cells from diverse populations to understand variability in MAFG function across human populations
These approaches align with the NIH Common Fund's goal to develop transformative methodologies with human relevance in biomedical research .
As highlighted in human augmentation research, diverse technological approaches could advance MAFG understanding:
Single-cell multi-omics: To capture heterogeneity in MAFG function across cell populations
Spatial transcriptomics: To map MAFG expression and activity in complex tissues
CRISPR-based epigenome editing: For precise modulation of MAFG expression and target genes
Human organoid models: To study MAFG in more physiologically relevant 3D contexts
AI-driven predictive modeling: To anticipate MAFG's role in diverse human populations and disease states
These approaches support both inclusion and diversity initiatives by enabling more personalized understanding of MAFG function across different human populations and conditions.
MAFG is a small basic leucine zipper (bZIP) transcription factor that forms homodimers or heterodimers with other bZIP proteins. These dimers bind to specific DNA sequences, regulating the expression of target genes involved in oxidative stress response, detoxification, and cellular differentiation . MAFG, along with other MAF proteins, is known to interact with the antioxidant response element (ARE) in the promoter regions of genes encoding detoxifying enzymes and antioxidant proteins.
MAFG has been implicated in various diseases, including cancer and neurodegenerative disorders. Its overexpression has been observed in certain types of cancer, where it may contribute to tumor progression by promoting cell survival and proliferation . Additionally, MAFG has been linked to the regulation of genes involved in the oxidative stress response, which is a critical factor in the pathogenesis of neurodegenerative diseases such as Alzheimer’s and Parkinson’s .
Human recombinant MAFG is a synthetically produced version of the protein, often used in research to study its function and potential therapeutic applications. One notable application of human recombinant MAFG is in the reprogramming of human pancreatic duct-derived cells (HDDCs) into insulin-secreting cells. This innovative approach involves the use of synthetic modified mRNA encoding MAFG, which is transfected into HDDCs to induce their differentiation into β-like cells capable of producing insulin .
The ability to reprogram HDDCs into insulin-secreting cells using human recombinant MAFG holds significant promise for the treatment of type 1 diabetes. This approach offers a potential alternative to β-cell replacement therapy, which aims to restore β-cell mass and glucose homeostasis in diabetic patients . The use of non-integrative RNA-based reprogramming techniques ensures that the genetic modifications are transient and do not integrate into the host genome, reducing the risk of adverse effects .