FABP1 Mouse, His is widely used in studies investigating lipid metabolism, disease pathogenesis, and therapeutic interventions. Key applications include:
Lipid transport assays: Quantifying fatty acid uptake and intracellular trafficking .
Inflammation and fibrosis models: Evaluating FABP1's role in mitigating pulmonary fibrosis and liver injury .
Protein-protein interaction studies: Identifying binding partners using affinity purification .
Downregulation in fibrosis: FABP1 expression is significantly reduced in bleomycin- and silica-induced pulmonary fibrosis models. Overexpression via AAV9-FABP1 attenuated fibrosis by promoting alveolar epithelial cell survival and regeneration .
Mechanistic insights: FABP1 deficiency exacerbates endoplasmic reticulum stress in alveolar type II cells, increasing apoptosis susceptibility .
Knockout (KO) mouse studies:
Parameter | Wild-Type Mice | FABP1-KO Mice |
---|---|---|
Hepatic fatty acid uptake | Normal | Reduced by ~40% |
Plasma free fatty acids | Baseline | Elevated by ~50% |
Insulin sensitivity | Normal | Impaired |
Data compiled from PMC studies . |
Fatty acid-binding protein 1 liver, L-FABP, FABPL, FABP-1, FABP1, Z-protein, Fatty acid-binding protein, liver.
MGSSHHHHHH SSGLVPRGSH MGSMNFSGKY QLQSQENFEP FMKAIGLPED LIQKGKDIKG VSEIVHEGKK IKLTITYGPK VVRNEFTLGE ECELETMTGE KVKAVVKLEG DNKMVTTFKG IKSVTELNGD TITNTMTLGD IVYKRVSKRI.
FABP1 is a cytoplasmic protein that binds long-chain fatty acids and facilitates their intracellular transport from the cytoplasm to mitochondria or peroxisomes for energy production. In mice, FABP1 is encoded by the Fabp1 gene and consists of 127 amino acids (Met1-Ile127) . The protein has a molecular weight of approximately 14-16 kDa as detected by Western blot analysis under reducing conditions . FABP1 is unique among FABPs in its capacity to bind two long-chain fatty acids simultaneously, allowing it to transport more fatty acids to mitochondria than other FABPs . Mouse FABP1 shares high sequence homology with human and rat FABP1, making it valuable for translational research in metabolism and disease modeling.
FABP1 demonstrates a specific tissue distribution pattern in mice. It is predominantly expressed in the liver (hence its alternative name L-FABP), but is also found in other tissues:
Lung: Expressed primarily in type II alveolar epithelial cells (ATII cells)
Small intestine: Present in enterocytes
Immunohistochemical and immunofluorescence studies have confirmed this distribution pattern. For instance, FABP1 can be detected in paraffin-embedded sections of mouse liver using specific antibodies such as Mouse Anti-Human/Mouse/Rat FABP1/L-FABP Monoclonal Antibody (at concentrations of 0.25 μg/mL for Western blot) . Western blot analysis of mouse liver tissue consistently reveals a specific band at approximately 14-16 kDa representing FABP1 .
FABP1 expression varies throughout mouse development, with changes corresponding to metabolic demands. During embryonic development, FABP1 expression begins in the fetal liver, correlating with hepatocyte maturation and the onset of lipid metabolism capabilities. Expression levels increase postnatally as the liver assumes full metabolic functionality. In adult mice, FABP1 expression remains relatively stable but can be modulated by nutritional status, hormonal regulation, and pathological conditions.
Age-related changes in FABP1 expression have been observed, with some studies suggesting altered lipid homeostasis in aging mice correlates with changes in FABP1 levels. The MGI database indicates that FABP1 is involved in multiple biological processes including homeostasis/metabolism and liver/biliary system development, suggesting its importance throughout the lifespan .
For reliable detection of His-tagged FABP1 in mouse tissue samples, several complementary approaches yield optimal results:
Western Blot Analysis:
Sample preparation: Homogenize mouse tissues (liver, kidney, lung) in appropriate lysis buffers containing protease inhibitors
Protein separation: Use reducing conditions and Immunoblot Buffer Group 1
Antibody selection: Either:
Expected band size: Approximately 14-16 kDa (slightly higher for His-tagged versions)
Immunohistochemistry/Immunofluorescence:
Fixation: For optimal results with His-tagged FABP1, use either:
Detection systems: HRP-DAB Cell & Tissue Staining Kit for chromogenic detection
Controls: Include tissues from FABP1-knockout mice or omit primary antibody as negative controls
Simple Western™ Analysis:
This automated capillary-based Western blot alternative provides high sensitivity for FABP1 detection:
Sample concentration: 0.2 mg/mL of tissue lysate
Antibody concentration: 5-10 μg/mL
System parameters: Use 12-230 kDa separation system under reducing conditions
Optimizing immunohistochemical detection of FABP1 in mouse lung sections requires specific considerations due to the protein's localization primarily in type II alveolar epithelial cells:
Protocol Optimization:
Fixation and processing:
For paraffin sections: 10% neutral buffered formalin fixation for 24 hours
For frozen sections: Perfusion fixation followed by OCT embedding
Section thickness: 5 μm for optimal antibody penetration
Antigen retrieval:
Heat-induced epitope retrieval in citrate buffer (pH 6.0) for 20 minutes
Allow cooling to room temperature before proceeding
Blocking and antibody incubation:
Counterstaining and interpretation:
For dual immunofluorescence to confirm FABP1 localization in type II alveolar epithelial cells, combine FABP1 antibody with surfactant protein C antibody (SP-C, a specific marker for type II cells) as demonstrated in pulmonary fibrosis research .
When conducting mouse cell culture experiments with His-tagged FABP1, several important considerations ensure experimental success:
Expression System Selection:
Bacterial expression: E. coli systems have successfully produced recombinant FABP1 as evidenced by the production of antibodies against E. coli-derived recombinant rat FABP1
Mammalian expression: For proper post-translational modifications, consider expression in HEK293 or CHO cells
Viral vectors: AAV9 vectors have been successfully used for FABP1 overexpression in mice for pulmonary fibrosis studies
Purification Strategy:
IMAC (Immobilized Metal Affinity Chromatography): Optimize binding and elution conditions (typically 250-300 mM imidazole)
Buffer composition: Include appropriate detergents if needed to maintain FABP1 fatty acid binding capacity
Quality control: Verify purity by SDS-PAGE and functionality by fatty acid binding assays
Functional Considerations:
Tag position: N-terminal His-tags generally preserve FABP1 function better than C-terminal tags
Fatty acid loading: Pre-load with specific fatty acids of interest before cellular experiments
Cell type specificity: Consider the appropriate mouse cell lines:
Verification of Expression and Function:
Western blot detection using either anti-His antibodies or FABP1-specific antibodies (0.25 μg/mL)
Immunofluorescence to confirm cellular localization
Functional assays to verify fatty acid binding and transport capacity
FABP1 plays a protective role against pulmonary fibrosis development in mouse models, with its downregulation contributing to disease progression through several mechanisms:
Expression Patterns and Localization:
Proteomics screening of two different mouse pulmonary fibrosis models (bleomycin-induced and silica-induced) revealed significantly reduced FABP1 expression compared to control mice . Double immunofluorescence staining demonstrated that FABP1 is primarily localized in type II alveolar epithelial cells (ATII cells) in the lung . The expression level of FABP1 was found to be negatively correlated with the progression of pulmonary fibrosis, suggesting its protective role .
Protective Mechanisms:
Alveolar epithelial cell protection: FABP1 appears to protect ATII cells from injury and promotes their survival
Fatty acid metabolism regulation: FABP1 facilitates the transport of long-chain fatty acids from the cytoplasm to mitochondria for energy production
Alveolar epithelial regeneration: Overexpression of FABP1 alleviates pulmonary fibrosis by promoting alveolar epithelial regeneration
Experimental Evidence:
In vivo experiments utilizing AAV9-mediated FABP1 overexpression demonstrated significant therapeutic effects in bleomycin-induced pulmonary fibrosis. Mice were divided into three groups: control, AAV9-GFP+BLM (GFP+BLM), and AAV9-FABP1+BLM (BLM+FABP1). FABP1 overexpression resulted in:
Improved lung pathology
Reduced fibrotic changes on micro-CT imaging
This research suggests that strategies aimed at activating or upregulating FABP1 could represent viable therapeutic approaches for treating pulmonary fibrosis, addressing the current lack of effective treatments for this condition .
FABP1-deficient mice exhibit significant alterations in several metabolic pathways, particularly those related to lipid metabolism and energy homeostasis:
Fatty Acid Metabolism:
Impaired intracellular fatty acid transport: Without FABP1's ability to bind and transport long-chain fatty acids, there is reduced delivery to mitochondria and peroxisomes
Altered fatty acid oxidation: Decreased capacity to oxidize fatty acids for energy production
Disrupted fatty acid uptake: Changes in cellular fatty acid uptake patterns affecting lipid distribution
Lipid Homeostasis:
Hepatic lipid accumulation: FABP1-deficient mice often develop hepatic steatosis due to altered lipid trafficking
Altered bile acid metabolism: Changes in cholesterol and bile acid synthesis pathways
Modified lipid profiles: Alterations in plasma lipid levels and composition
Energy Metabolism:
Reduced mitochondrial function: Decreased capacity for fatty acid-derived energy production
Metabolic adaptation: Increased reliance on glucose metabolism
Impaired response to fasting: Limited ability to mobilize and utilize fatty acids during periods of fasting
Oxidative Stress and Inflammation:
Increased oxidative stress: Higher levels of reactive oxygen species due to metabolic dysregulation
Enhanced inflammatory responses: Altered lipid mediator production affecting inflammatory pathways
Increased susceptibility to hepatocellular damage: FABP1-deficient mice are more vulnerable to various hepatic insults
These metabolic alterations are particularly relevant in contexts like pulmonary fibrosis, where FABP1 expression is significantly reduced in type II alveolar epithelial cells. The resulting dysfunctional ATII cells become more vulnerable to apoptosis due to endoplasmic reticulum stress caused by elevated concentrations of saturated fatty acids , highlighting the importance of FABP1 in maintaining cellular lipid homeostasis and energy metabolism.
Several approaches have been successfully employed to modulate FABP1 expression in mouse models for therapeutic investigation, particularly in the context of pulmonary fibrosis:
Viral Vector-Mediated Overexpression:
Adeno-associated virus (AAV) delivery: AAV9-FABP1 vectors administered via intratracheal instillation have successfully increased FABP1 expression in mouse lungs
Protocol effectiveness: In pulmonary fibrosis studies, mice were treated with AAV9-FABP1 or control AAV9-GFP three weeks before bleomycin challenge (2.5 mg/kg), resulting in significant protection against fibrosis development
Tissue targeting: AAV9 serotype demonstrates good tropism for lung epithelial cells, particularly ATII cells where FABP1 is normally expressed
Pharmaceutical Induction:
PPAR agonists: Peroxisome proliferator-activated receptor (PPAR) agonists can upregulate FABP1 expression
Natural compounds: Certain dietary compounds have shown ability to increase FABP1 expression through transcriptional regulation
Small molecule screening: High-throughput screening approaches to identify compounds that enhance FABP1 expression or activity
Genetic Modification Approaches:
Transgenic overexpression: Development of mouse lines with tissue-specific FABP1 overexpression
CRISPR/Cas9 enhancement: Targeted enhancement of endogenous FABP1 expression through promoter modification
Conditional expression systems: Inducible FABP1 expression using tetracycline-responsive or similar systems
Combined Therapeutic Strategies:
Dual targeting: Combining FABP1 modulation with anti-fibrotic agents
Metabolic support: Supplementation with specific fatty acids or metabolic substrates alongside FABP1 enhancement
Cell-based therapies: Delivery of ATII cells overexpressing FABP1 to injured lungs
Research has demonstrated that FABP1 overexpression significantly alleviates pulmonary fibrosis by protecting alveolar epithelium from injury and promoting cell survival . This provides strong evidence that activating or upregulating FABP1 could be a viable therapeutic strategy for managing pulmonary fibrosis and potentially other conditions characterized by altered lipid metabolism.
Designing a comprehensive experiment to study FABP1-His interactions with fatty acids in mouse models requires a multifaceted approach combining in vitro, cellular, and in vivo techniques:
In Vitro Binding Characterization:
Protein preparation:
Binding assays:
Fluorescent displacement assays using ANS (8-anilino-1-naphthalenesulfonic acid) or DAUDA
Isothermal titration calorimetry (ITC) to determine binding constants for different fatty acids
Surface plasmon resonance to measure binding/dissociation kinetics
Cellular Fatty Acid Transport Studies:
Cell model selection:
Transport assays:
Fluorescently labeled fatty acid uptake assays
Mitochondrial targeting of fatty acids (using MitoTracker co-localization)
Fatty acid oxidation rate measurements using radiolabeled substrates
In Vivo Mouse Models:
Genetic approaches:
Compare wild-type, FABP1-knockout, and FABP1-His transgenic mice
Use tissue-specific expression systems (e.g., liver-specific or lung-specific promoters)
Metabolic challenge experiments:
Analysis methods:
Advanced Imaging Approaches:
Intravital microscopy:
Track fluorescently labeled fatty acids in real-time in living mice
Monitor FABP1-His-fatty acid complexes using FRET-based reporters
Multi-omics integration:
Combine lipidomics, proteomics, and transcriptomics data
Correlate FABP1 expression with lipid profiles and metabolic outcomes
This comprehensive experimental design allows for detailed characterization of FABP1-His interactions with fatty acids across multiple biological scales, providing insights into both molecular mechanisms and physiological relevance.
Investigating FABP1's role in fatty acid transport to mitochondria in mouse models requires specialized techniques that span subcellular to organismal levels:
Subcellular Fractionation and Mitochondrial Isolation:
Preparation of pure mitochondrial fractions:
Transport assays with isolated mitochondria:
Radiolabeled fatty acid uptake by isolated mitochondria with/without FABP1
Fatty acid oxidation rates in isolated mitochondria supplemented with purified FABP1
Live Cell Imaging Approaches:
FRET-based interaction studies:
Generate fluorescently tagged FABP1-His and mitochondrial markers
Measure FRET signal to detect proximity/interaction at mitochondrial surfaces
Photoactivatable fatty acid analogs:
Track fatty acid movement from cytosol to mitochondria in real-time
Compare dynamics in cells expressing varying levels of FABP1
Genetic Manipulation Strategies:
Mitochondrial targeting sequences:
Create FABP1 variants with mitochondrial targeting sequences
Assess impact on fatty acid metabolism when FABP1 is artificially localized to mitochondria
FABP1 domain mutation analysis:
Generate mouse models with mutations in specific FABP1 domains
Identify regions critical for mitochondrial fatty acid delivery
Metabolic Flux Analysis:
Stable isotope tracing:
Administer 13C-labeled fatty acids to mice (wild-type vs. FABP1-deficient)
Track isotopomer distributions in TCA cycle intermediates to assess mitochondrial fatty acid utilization
Respirometry studies:
Measure oxygen consumption rates in isolated mitochondria or permeabilized cells
Compare fatty acid-driven respiration between FABP1-expressing and FABP1-deficient samples
In Vivo Assessment:
MitoTracker imaging in tissue sections:
Visualize mitochondrial networks in tissues from wild-type and FABP1-deficient mice
Correlate with lipid distribution using Oil Red O staining
PET imaging with fatty acid tracers:
Use 18F-labeled fatty acid analogs to track in vivo fatty acid utilization
Compare uptake and oxidation patterns between wild-type and FABP1-modified mice
These approaches collectively provide comprehensive insights into FABP1's role in facilitating fatty acid transport to mitochondria, a critical function given FABP1's unique capacity to bind two long-chain fatty acids simultaneously and carry more fatty acids to mitochondria than other FABPs .
Developing transgenic mouse models expressing His-tagged FABP1 requires careful consideration of multiple factors to ensure physiological relevance and experimental utility:
Design Considerations:
Construct design:
Promoter selection:
Endogenous Fabp1 promoter for native expression pattern
Tissue-specific promoters (e.g., albumin promoter for liver-specific expression)
Inducible systems (tetracycline-responsive) for temporal control
His-tag positioning:
N-terminal tag: Less likely to interfere with fatty acid binding
C-terminal tag: May alter protein folding or ligand binding
Include flexible linker sequences to minimize interference
Reporter elements:
Include fluorescent reporter (separated by IRES or 2A peptide) for tracking expression
Consider adding epitope tags for detection flexibility
Transgenic strategies:
Pronuclear injection:
Random integration of the His-FABP1 expression cassette
Simple but may have position effects and variable expression
CRISPR/Cas9 knock-in:
Precise insertion of His-tag into endogenous Fabp1 locus
Maintains physiological regulation but more technically challenging
Conditional systems:
Floxed constructs for tissue-specific or inducible expression
Allows for developmental or temporal studies
Validation and Characterization:
Expression verification:
Western blot analysis:
Immunohistochemistry:
Functional validation:
Fatty acid binding assays:
Extract His-FABP1 from transgenic tissues and assess binding capacity
Compare binding affinity to untagged FABP1
Metabolic phenotyping:
Experimental Applications:
Tissue-specific overexpression:
Pull-down experiments:
In vivo crosslinking followed by His-tag purification
Identify novel FABP1 protein interaction partners
Characterize the FABP1 interactome under different physiological conditions
Rescue experiments:
Cross with FABP1-knockout mice to verify functional complementation
Assess whether His-tagged FABP1 restores normal phenotype
Determine if His-tag affects protein functionality in vivo
For AAV-mediated expression as an alternative to germline transgenesis, research has shown success with AAV9-FABP1 delivery via intratracheal instillation, providing therapeutic effects in pulmonary fibrosis models .
Detecting low levels of FABP1 in mouse tissue samples can be challenging, particularly in tissues where expression is naturally low or downregulated in disease states, such as in pulmonary fibrosis . Here are common challenges and evidence-based solutions:
Solutions:
Enhanced extraction methods:
Use specialized lysis buffers containing mild detergents (0.5-1% Triton X-100)
Include protease inhibitor cocktails to prevent degradation
Consider membrane-enrichment protocols for improved recovery
Signal amplification techniques:
Antibody optimization:
Solutions:
Optimization of tissue fixation:
Enhanced blocking protocols:
Extended blocking (2 hours) with 5-10% normal serum
Include 0.3% Triton X-100 in blocking solution for better penetration
Use commercial background reducers specific to the host species
Detection system selection:
Solutions:
Antibody validation:
Preabsorption techniques:
Pre-incubate antibodies with recombinant proteins from related FABP family members
Use immunoprecipitation to verify antibody specificity
Solutions:
Sample normalization:
Include multiple housekeeping proteins as loading controls
Consider tissue-specific reference proteins rather than ubiquitous markers
Employ total protein normalization methods (Ponceau S, REVERT)
Multi-tissue analysis:
These solutions have been validated in experimental settings, with successful detection of FABP1 in challenging contexts such as pulmonary fibrosis models where protein expression is significantly reduced .
Resolving inconsistencies between FABP1 protein detection and gene expression data requires systematic investigation of potential discrepancies across multiple biological levels:
Potential Causes and Solutions:
Post-transcriptional regulation:
MicroRNA regulation:
Analyze miRNA expression profiles that target FABP1 mRNA
Perform AGO-CLIP to identify actual miRNA binding events
mRNA stability differences:
Measure FABP1 mRNA half-life using actinomycin D chase experiments
Compare stability across different tissues/conditions
Translational efficiency:
Polysome profiling:
Isolate polysome-bound FABP1 mRNA to assess translation rates
Compare translational efficiency between experimental conditions
Ribosome profiling:
Perform Ribo-seq to precisely measure translation efficiency
Identify potential translational pausing or regulation
Protein stability and turnover:
Pulse-chase experiments:
Label FABP1 protein to track its half-life
Compare degradation rates across conditions
Proteasome/lysosome inhibition:
Test whether protein degradation pathways account for discrepancies
Use MG132 (proteasome) or chloroquine (lysosome) inhibitors
Technical Validation Approaches:
Cross-methodology validation:
Protein detection methods:
RNA measurement approaches:
Validate with multiple techniques: RT-qPCR, RNA-seq, in situ hybridization
Design primers spanning different exon junctions
Spatial considerations:
Tissue heterogeneity:
Use laser capture microdissection to isolate specific cell populations
Perform single-cell RNA-seq and compare to bulk measurements
Subcellular localization:
Temporal dynamics:
Time-course experiments:
Collect samples across multiple timepoints to capture expression dynamics
Consider protein production and degradation kinetics
Circadian variations:
Control for time-of-day effects in sample collection
Assess whether FABP1 exhibits circadian regulation
Integrative Analysis Solutions:
Multi-omics integration:
Correlative analysis:
Calculate protein-mRNA correlations across multiple samples/conditions
Identify patterns of concordance/discordance
Pathway analysis:
Examine regulatory networks affecting FABP1 expression
Look for feedback mechanisms or compensatory regulation
Mathematical modeling:
Kinetic models:
Develop models incorporating transcription, translation, and degradation rates
Use to predict and explain observed discrepancies
Machine learning approaches:
Train algorithms to identify features predicting protein-mRNA discordance
Apply to FABP1 data to identify key regulatory factors
In pulmonary fibrosis research, both proteomic and immunological methods demonstrated concordant reductions in FABP1 protein levels , suggesting that in some disease contexts, transcriptional regulation may be the primary driver of FABP1 expression changes.
Comparing FABP1 expression across different mouse strains and disease models requires rigorous standardization and careful experimental design to ensure valid cross-group comparisons:
Experimental Design Considerations:
Strain-specific baseline characterization:
Establish reference values:
Age and sex matching:
Use age-matched mice (particularly important as metabolism changes with age)
Control for sex differences in metabolic profiles
Document estrous cycle stage in female mice
Disease model standardization:
Model induction protocols:
Temporal considerations:
Sample at multiple time points to capture dynamic changes
Establish disease progression timelines for each strain
Match sampling to disease severity rather than absolute time
Analytical Methods Standardization:
Sample processing protocols:
Tissue collection:
Standardize collection methods, including time of day
Use consistent fasting/feeding status prior to collection
Employ identical tissue preservation protocols
Extraction methods:
Use identical lysis buffers and protein extraction protocols
Process all samples in parallel to minimize batch effects
Include spike-in standards for absolute quantification
Detection method optimization:
Western blot standardization:
Immunohistochemistry controls:
Process all samples in the same batch with identical reagents
Include positive control tissues (liver) from each strain
Use automated staining systems to minimize technical variation
Data Analysis and Normalization:
Multi-level normalization strategy:
Protein level normalization:
Use multiple housekeeping proteins as references
Consider total protein normalization methods
Account for strain-specific differences in reference proteins
Tissue-level normalization:
Normalize to tissue weight or protein content
Consider cell-type composition differences between strains
Use stereological principles for immunohistochemistry quantification
Statistical approaches:
Nested experimental designs:
Account for mouse-to-mouse variation within strains
Use mixed-effects models for proper hierarchy handling
Meta-analytical techniques:
Standardize effect sizes for cross-study comparisons
Use forest plots to visualize strain and model differences
Calculate confidence intervals for more informative comparisons
Validation and Cross-Referencing:
Multi-method validation:
Orthogonal techniques:
Confirm key findings with alternative methods (Western blot, IHC, mass spectrometry)
Compare protein and mRNA levels to identify discrepancies
Functional correlations:
Relate FABP1 expression differences to functional outcomes
Assess fatty acid metabolism in parallel with expression analysis
In pulmonary fibrosis research, FABP1 expression was successfully compared between control mice and two different pulmonary fibrosis models (bleomycin-induced and silica-induced) , demonstrating that with proper standardization, meaningful comparisons can be made across disease models.
Fatty Acid Binding Protein-1 (FABP1), also known as liver-type fatty acid-binding protein (L-FABP), is a member of the fatty acid-binding protein family. These proteins are involved in the binding, transport, and metabolism of long-chain fatty acids (LCFAs) and other hydrophobic molecules. FABP1 is primarily expressed in the liver but can also be found in the intestine, kidney, pancreas, stomach, and lung .
The fatty acid-binding proteins were initially discovered in 1972 through experiments using labeled oleate to identify a soluble fatty acid carrier in the enterocyte responsible for intestinal absorption of LCFAs . FABP1 has a unique structure compared to other members of the FABP family, allowing it to bind multiple ligands simultaneously. It has a larger solvent-accessible core, which enables more diverse substrate binding .
FABP1 plays a significant role in the binding and transport of LCFAs, endocannabinoids, phytocannabinoids, and other hydrophobic molecules. It is involved in various metabolic processes, including fatty acid uptake, transport, and metabolism. FABP1 is also known to bind bilirubin, monoglycerides, bile acids, and fatty acyl CoA . This protein helps prevent cytotoxicity by binding potentially toxic molecules when unbound .
The His tag is a sequence of histidine residues added to proteins to facilitate their purification and detection. Recombinant proteins, such as the mouse FABP1 with a His tag, are produced using genetic engineering techniques. These proteins are expressed in host cells, such as Escherichia coli, and purified using affinity chromatography methods that exploit the His tag .
Recombinant mouse FABP1 with a His tag is used in various research applications, including studies on fatty acid metabolism, liver function, and the role of FABP1 in metabolic diseases. The His tag allows for easy purification and detection of the protein, making it a valuable tool for researchers .