The MLXIP Antibody (catalog number 13614-1-AP) is a polyclonal rabbit antibody developed to target MLXIP (MLX interacting protein), also known as MondoA. It is widely used in research to study glucose-responsive gene regulation and metabolic pathways. Below is a comprehensive analysis of its characteristics, applications, and biological implications, supported by experimental data and research findings.
MLXIP forms heterodimers with MLX to regulate glycolytic gene expression via CACGTG E-box motifs. Key functional insights include:
N-Terminus Role: Acts as a CRM1-dependent nuclear export signal, ensuring cytoplasmic localization of the MLX-MLXIP complex. It also binds the 13 protein .
C-Terminus Function: Mediates cytoplasmic localization signals and interacts with MONDOA/MLX to activate transcription .
Recent studies reveal MLXIP’s localization to lipid droplets (LDs), where it regulates metabolic gene expression in response to lipid storage levels. Binding to LDs attenuates transcriptional activity, while LD absence hyperactivates target genes .
MLXIP partners with MLX to recruit transcriptional machinery to promoters of glucose-responsive genes (e.g., Txnip). ChIP-qPCR experiments demonstrate glucose-dependent recruitment of MLX-MLXIP complexes to the Txnip promoter, which is disrupted by MLX-DN (dominant-negative) variants .
Proteomic studies identify MLXIP’s association with LDs in multiple cell types (e.g., THP-1 macrophages, SUM159 carcinoma cells). Endogenous MLXIP is enriched in LD fractions, suggesting a role in lipid metabolism .
The MLXIP-MLX complex activates glycolytic genes (e.g., HK2, PFKFB3) and integrates glucose/lipid signals. Its activity is modulated by post-translational modifications, including ubiquitination .
A prominent 130 kDa band is detected in lysates from human, mouse, and rat tissues, confirming the antibody’s specificity .
MLXIP antibody successfully pulls down chromatin-bound complexes at the Txnip promoter in glucose-stimulated cells .
Co-localization of MLXIP with LD markers (e.g., ADRP) in oleate-treated cells highlights its LD-binding capacity .
MLXIP (MLX Interacting Protein, also known as MondoA) is a member of the Myc superfamily of transcription factors that functions as part of a heterodimer with Max-like protein X (MLX) to activate transcription. It binds to the canonical E box sequence 5'-CACGTG-3' and plays a crucial role in transcriptional activation of glycolytic target genes and glucose-responsive gene regulation . MLXIP contributes to the regulation of glucose homeostasis as the MLXIP/MLX heterodimer activates expression of thioredoxin-interacting protein (TXNIP), a potent inhibitor of cellular glucose uptake and aerobic glycolysis . Additionally, the mammalian target of rapamycin (mTOR) interacts with this pathway by binding to MLXIP, thereby preventing MLXIP/MLX heterodimer formation . MLXIP interaction with Myc may also be involved in metabolic reprogramming and tumorigenesis, making it an important target for cancer research .
MLXIP antibodies have been validated for multiple applications across different research methodologies:
It is recommended to optimize dilutions for each specific experimental system to obtain optimal results, as antibody performance can be sample-dependent .
When selecting an MLXIP antibody, researchers should consider:
Target epitope region: Different antibodies target distinct regions of MLXIP (N-terminal, middle region, C-terminal). For example, some antibodies target amino acids 1-217 , others target regions near the N-terminus , and some target the C-terminal region . The epitope location can affect detection of specific isoforms or post-translationally modified forms.
Isoform detection: MLXIP has multiple isoforms, including isoform 1 (~110 kDa) and isoform 3 (~69 kDa) . Some antibodies can recognize multiple isoforms while others may be more specific.
Observed molecular weight: The calculated molecular weight of MLXIP is approximately 100-101 kDa, but the observed molecular weight in Western blots is often around 130 kDa , suggesting post-translational modifications.
Host species and clonality: Most MLXIP antibodies are rabbit polyclonals , though some mouse monoclonal options exist . Consider the host species in relation to your secondary detection systems and other antibodies in multiplexed experiments.
Validated applications: Ensure the antibody has been validated for your specific application through published literature or manufacturer validation data .
For optimal Western blot results with MLXIP antibodies:
Sample preparation:
Electrophoresis and transfer:
Use standard SDS-PAGE conditions, ensuring adequate resolution in the 100-130 kDa range
For complete detection of MLXIP isoforms, use gels with appropriate resolution range (e.g., 6-12% acrylamide)
Antibody incubation:
Block membrane using standard blocking buffer (e.g., 5% non-fat dry milk or BSA in TBST)
Dilute primary MLXIP antibody according to manufacturer recommendations (typically 1:200-1:1000 or 1-2 μg/mL )
Incubate at 4°C overnight for optimal results
Use appropriate HRP-conjugated secondary antibodies at recommended dilutions
Detection:
Controls:
Include positive control lysates from cells known to express MLXIP
Consider using MLXIP knockdown or knockout samples as negative controls when available
For immunohistochemistry with MLXIP antibodies:
Sample preparation:
Antigen retrieval:
Antibody incubation:
Detection system:
Use polymer-based or biotin-streptavidin detection systems with appropriate secondary antibodies
Include proper controls (primary antibody omission, isotype controls)
Counterstaining and mounting:
Use standard hematoxylin counterstaining
Mount with compatible mounting medium
Evaluation:
For effective immunoprecipitation of MLXIP:
Antibody selection:
Lysate preparation:
Pre-clearing step:
Pre-clear lysates with appropriate control beads/resin to reduce non-specific binding
Use protein A/G beads for rabbit polyclonal antibodies
Immunoprecipitation procedure:
Incubate lysates with MLXIP antibody overnight at 4°C with gentle rotation
Add appropriate beads and continue incubation
Wash stringently to reduce background while preserving specific interactions
Elute under appropriate conditions for downstream applications
Controls and validation:
Include isotype control antibodies to identify non-specific interactions
Validate IP success by Western blot, using a portion of the IP product
Consider using a second MLXIP antibody recognizing a different epitope for validation in Western blot
Co-IP considerations:
Chromatin immunoprecipitation (ChIP) with MLXIP antibodies requires careful optimization:
Experimental design:
Chromatin preparation:
Optimize crosslinking conditions (typically 1% formaldehyde for 10-15 minutes)
Ensure effective sonication/fragmentation to generate appropriate DNA fragment sizes (200-500 bp)
Verify fragmentation efficiency by agarose gel electrophoresis
Immunoprecipitation:
Use optimized antibody amounts based on preliminary titration experiments
Include appropriate controls (IgG control, input DNA, positive control antibody)
Extend incubation times (overnight at 4°C) to enhance binding efficiency
Washing and elution:
Use stringent washing conditions to reduce background
Optimize reverse crosslinking conditions
PCR primers design:
Design primers around known or predicted MLXIP binding sites
Include primers for established MLXIP target genes as positive controls
Consider primers for known E-box containing promoters
Data analysis:
Normalize ChIP-qPCR data to input DNA and IgG controls
For ChIP-seq applications, use appropriate peak calling algorithms
Consider integrated analysis with transcriptomic data to correlate binding with expression changes
Validation:
Confirm findings with reporter assays or directed mutagenesis of binding sites
Consider comparing wildtype and MLXIP knockdown/knockout conditions
MLXIP exhibits complex subcellular localization patterns, including cytoplasmic, nuclear, and mitochondrial outer membrane localization . To effectively study its shuttling:
Antibody selection and optimization:
Experimental conditions to consider:
Co-localization studies:
Mitochondrial markers (e.g., MitoTracker or TOMM20) to confirm outer mitochondrial membrane localization
Nuclear markers (e.g., DAPI) to assess nuclear translocation
MLX co-staining to examine heterodimer formation and co-localization
Live-cell imaging considerations:
For dynamic studies, consider GFP-tagged MLXIP constructs
Time-lapse imaging following glucose level changes
Fixation and permeabilization optimization:
Test multiple fixation methods (paraformaldehyde, methanol) as they may differentially preserve subcellular structures
Optimize permeabilization to ensure antibody access while preserving cellular architecture
Quantitative analysis:
Measure nuclear/cytoplasmic ratios across conditions
Quantify co-localization coefficients with mitochondrial markers
Track dynamic changes in response to perturbations
Advanced microscopy techniques:
Consider super-resolution microscopy for detailed subcellular localization
FRET analysis for protein-protein interactions if using fluorescently tagged constructs
To study MLXIP's role in glucose-responsive gene regulation:
Experimental design considerations:
Combined methodological approaches:
ChIP or ChIP-seq to identify MLXIP binding sites under different glucose conditions
RNA-seq to correlate binding with transcriptional changes
Western blotting to assess MLXIP and target protein levels
Co-IP to examine dynamic interaction partners under different metabolic states
Key pathway components to monitor:
MLXIP and MLX levels and heterodimer formation
TXNIP expression as a primary downstream target
mTOR interaction with MLXIP under varying conditions
Glucose uptake and glycolytic flux measurements as functional readouts
Perturbation strategies:
MLXIP knockdown or knockout using siRNA or CRISPR-Cas9
mTOR inhibitors to assess pathway regulation
Glucose level manipulation protocols
Hypoxia conditions that may affect metabolic programming
Temporal considerations:
Time-course experiments to capture dynamic responses
Acute vs. chronic glucose level changes
Cell type selection:
MLXIP exists in multiple isoforms and undergoes various post-translational modifications, which can significantly impact antibody recognition:
Isoform considerations:
MLXIP has multiple isoforms, including isoform 1 (~110 kDa) and isoform 3 (~69 kDa)
The calculated molecular weight is approximately 100-101 kDa, but the observed molecular weight is often around 130 kDa, suggesting extensive post-translational modifications
Antibody epitope location determines which isoforms will be detected
N-terminal targeting antibodies may detect different isoform subsets than C-terminal antibodies
Post-translational modifications:
Experimental strategies:
Use antibodies targeting different epitopes to compare detection patterns
Consider phosphatase treatment of samples to assess phosphorylation impact
When interpreting localization studies, consider that modifications may affect shuttling between compartments
Data interpretation considerations:
Always report both calculated and observed molecular weights
Specify which antibody (targeting which epitope) was used for detection
Consider running parallel experiments with antibodies recognizing different regions
For functional studies, determine which isoforms are being measured and their relative contributions
To ensure robust and reproducible MLXIP antibody experiments:
Essential controls:
Positive controls: Cell lines with known MLXIP expression (K-562, HeLa, Jurkat, 293T)
Negative controls: MLXIP knockdown or knockout samples where available
Isotype controls: Particularly important for IP, ChIP, and flow cytometry applications
Secondary antibody-only controls: To assess non-specific binding of secondary detection systems
Antibody validation strategies:
Multiple antibody approach: Use antibodies targeting different epitopes to confirm results
Peptide competition: Pre-incubation with immunizing peptide should abolish specific signal
Genetic validation: Use samples with MLXIP genetic modification (siRNA, CRISPR)
Cross-application validation: Confirm findings using multiple techniques (e.g., WB and IF)
Reproducibility considerations:
Antibody lot testing: Test new lots against previous ones before adoption
Detailed protocol documentation: Record all experimental variables
Consistency in sample preparation: Standardize lysis buffers, fixation protocols, etc.
Quantitative assessment: Use quantitative measures rather than subjective assessments
Advanced validation approaches:
Mass spectrometry validation of immunoprecipitated proteins
CRISPR-Cas9 knockout followed by antibody testing
Recombinant protein controls at known concentrations
Cross-reactivity testing across species if working with non-human models
MLXIP's role in glucose metabolism makes it particularly relevant to cancer research, where metabolic reprogramming is a hallmark:
Current research applications:
MLXIP interaction with Myc may be involved in metabolic reprogramming and tumorigenesis
MLXIP/MLX regulation of TXNIP affects glucose uptake and glycolysis, processes often dysregulated in cancer
Antibody-based detection of MLXIP expression and localization in tumor samples
Investigation of mTOR-MLXIP interactions in cancer contexts
Methodological approaches:
IHC analysis of MLXIP expression in tumor microarrays
Co-localization studies with metabolic enzymes in cancer cell lines
ChIP-seq to identify altered MLXIP binding patterns in cancer models
IP-mass spectrometry to identify cancer-specific interaction partners
Key research questions being addressed:
How does MLXIP expression correlate with cancer progression and prognosis?
Does MLXIP subcellular localization differ in cancer cells compared to normal cells?
Can targeting MLXIP-dependent pathways provide therapeutic opportunities?
How do oncogenic signals modify MLXIP function and target gene selection?
Technical considerations:
Use of patient-derived xenografts and primary cancer samples
Integration with metabolomics approaches
Comparison across cancer types with different metabolic signatures
For comprehensive analysis of metabolic regulation networks involving MLXIP:
Multiplex immunofluorescence considerations:
Antibody selection: Ensure primary antibodies are from different host species
Cross-reactivity testing: Validate antibody specificity in multiplex context
Sequential staining protocols: Consider tyramide signal amplification for sequential detection
Spectral unmixing: Employ appropriate controls for multi-fluorophore separation
Co-immunoprecipitation strategies:
Sequential IP: First IP with MLXIP antibody followed by secondary IP for interacting partners
Combined IP: Simultaneous use of antibodies against MLXIP and potential partners
Mass spectrometry analysis of MLXIP complexes under different metabolic conditions
Proximity ligation assays to confirm protein-protein interactions in situ
Integrated ChIP approaches:
Sequential ChIP (Re-ChIP): To identify genomic regions co-bound by MLXIP and other factors
ChIP-seq with motif analysis: To identify co-occurring binding motifs for MLXIP and other factors
Integration with accessibility data (ATAC-seq) and histone modification profiles
CUT&RUN or CUT&Tag alternatives for higher resolution binding profiles
Key interaction partners to consider:
MLXIP belongs to a family of transcription factors with overlapping functions, particularly ChREBP (MondoB):
Antibody selection strategies:
Choose antibodies validated for specificity against related family members
Verify cross-reactivity profiles against recombinant proteins
Select antibodies targeting divergent regions between family members
Genetic approaches:
CRISPR-Cas9 knockout of MLXIP with rescue experiments using related factors
Selective knockdown using siRNA targeting unique sequence regions
Domain swap experiments to identify functional specificities
Binding site discrimination:
Compare ChIP-seq profiles of MLXIP and related factors
Motif analysis to identify subtle binding preference differences
In vitro DNA binding assays with purified proteins and variant binding sites
Functional readouts:
Transcriptome profiling after selective knockdown
Metabolic flux analysis to identify factor-specific metabolic impacts
Promoter-reporter assays with mutations in binding sites
Context-dependent approaches:
Temporal dynamics:
Time-course experiments to identify differences in activation/deactivation kinetics
Pulse-chase approaches to measure protein stability differences
Real-time imaging of factor recruitment to target loci
For studying MLXIP in metabolic disease contexts, consider this integrated approach:
Expression profiling:
Localization studies:
DNA binding and target gene regulation:
Protein interactions:
Functional manipulation:
CRISPR-Cas9 or siRNA-mediated knockdown/knockout
Overexpression studies with wildtype and mutant MLXIP
Metabolic flux analysis following MLXIP manipulation
Integration with clinical parameters:
Correlation of MLXIP expression/localization with disease markers
Response to therapeutic interventions
Given MLXIP's association with the mitochondrial outer membrane , specialized approaches are needed:
Localization studies:
Biochemical fractionation:
Proximity-based approaches:
Proximity ligation assays to detect MLXIP interactions with mitochondrial proteins
BioID or APEX2 proximity labeling with MLXIP as bait
FRET-based detection of protein interactions at mitochondrial membranes
Functional assays:
Mitochondrial respiration measurements after MLXIP manipulation
Assessment of glucose oxidation vs. glycolysis
Mitochondrial membrane potential in response to MLXIP perturbation
Perturbation approaches:
Mitochondrial stress induction (e.g., FCCP, antimycin A) and assessment of MLXIP response
Glucose deprivation/reintroduction experiments
Hypoxia response studies
Advanced microscopy techniques:
Live-cell imaging of fluorescently tagged MLXIP in relation to mitochondria
Time-lapse studies during metabolic transitions
Correlative light and electron microscopy for ultrastructural localization
Robust quantification and statistical analysis are essential for reproducible MLXIP research:
Western blot quantification:
Immunofluorescence quantification:
Measure nuclear/cytoplasmic ratios for localization studies
Use z-stack acquisitions to capture full signal
Implement automated, unbiased analysis workflows
Report co-localization coefficients with appropriate controls
ChIP data analysis:
For ChIP-qPCR: Normalize to input and IgG controls
For ChIP-seq: Use appropriate peak calling algorithms
Perform motif enrichment analysis for E-box sequences
Integrate with gene expression data to identify functional binding
Statistical approaches:
Use appropriate statistical tests based on data distribution
Correct for multiple testing when analyzing genome-wide data
Implement mixed-effects models for data with nested variables
Report effect sizes alongside p-values
Visualization strategies:
Present full blot images with molecular weight markers
Use consistent scales for comparative analyses
Include representative images alongside quantification
Consider dimensionality reduction for complex datasets
Replication and validation:
Distinguish technical from biological replicates
Validate key findings using complementary methodologies
Consider sample size calculations for appropriate power
Critical evaluation of MLXIP antibody specificity is crucial for reliable research: