FABP1 Mouse

Fatty Acid Binding Protein-1 Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Characterization of Murine FABP1

Structure:

  • Mouse FABP1 is a 127-amino acid cytoplasmic protein with a β-barrel structure containing a large ligand-binding pocket .

  • Exhibits unique capacity to bind two long-chain fatty acids (LCFAs) simultaneously alongside other ligands (e.g., heme, bile acids, prostaglandins) .

Expression:

  • Highly expressed in hepatocytes (>7% cytosolic protein)

  • Detected in kidney proximal tubules, intestinal epithelium, and lung alveolar type II cells

Functional Roles in Mice

Key biological activities include:

FunctionMechanismPhenotypic Evidence
Lipid TransportShuttles LCFAs to mitochondria/peroxisomes for β-oxidationKO mice show 50% reduced hepatic fatty acid uptake
CytoprotectionBinds cytotoxic molecules (heme, oxidized lipids)FABP1⁻/⁻ mice exhibit increased oxidative liver/kidney injury
Metabolic RegulationDelivers ligands to PPARα nuclear receptorPPARα target genes downregulated 40-60% in KO models
Cell ProliferationModulates mitotic signaling pathwaysLiver regeneration reduced 30% post-hepatectomy in FABP1⁻/⁻ mice

Research Models and Phenotypic Findings

Knockout Mouse Lines:

  1. FABP1⁻/⁻-GFP:

    • Impaired VLDL secretion (plasma triglycerides ↓25%)

    • Resistance to diet-induced hepatic steatosis

  2. Double KO (A-FABP/E-FABP⁻/⁻):

    • Delayed EAE onset (19.5 vs 15.3 days in WT)

    • Reduced CNS inflammation (CD45⁺ cells ↓60%)

Transgenic Overexpression:

  • AAV9-FABP1 delivery:

    • Reduced pulmonary fibrosis severity by 45% in bleomycin model

    • Increased alveolar epithelial cell survival (+32%)

Disease Implications

Liver Pathologies:

  • Steatosis: FABP1 expression ↓4.7x in diabetic mice; restored by fish oil

  • Hepatocellular Carcinoma:

    • FABP1⁺ TAMs promote M2 polarization via PPARγ/CD36

    • KO mice show enhanced anti-PD1 response (tumor volume ↓58%)

Extrahepatic Effects:

  • Pulmonary Fibrosis:

    • FABP1⁰ᵛᵉʳ mice: Collagen deposition ↓33%, epithelial regeneration ↑2.1x

  • Kidney Injury:

    • Urinary FABP1 predicts AKI 1hr post-ischemia (AUC=0.92 vs 0.65 for BUN)

Therapeutic Targeting Potential

Emerging strategies include:

  • Gene Therapy: AAV9-FABP1 reduced BLM-induced lung scarring by 40%

  • Small Molecule Modulators: T94A variant linked to dyslipidemia in humanized mice

  • Immune Modulation: Combined FABP1 KO + anti-PD1 eliminated 80% HCC tumors

Product Specs

Introduction
The fatty acid binding protein found in the liver is encoded by the FABP1 gene (Fatty acid binding protein1). Ten antiparallel beta strands make up FABP1, which form a barrel with a binding pocket larger than that of other FABPs, enabling it to accommodate two fatty acids. In the cytoplasm, this protein attaches to free fatty acids and their coenzyme A derivatives, bilirubin, and a few other tiny molecules. It might be involved in the metabolism and intracellular transport of lipids.
Description
E. coli is used to produce recombinant mouse Fatty Acid Binding Protein-1, a single, non-glycosylated polypeptide chain with 127 amino acids and a molecular weight of 14.2 kDa. The FABP1 is purified using patented chromatographic methods.
Physical Appearance
White lyophilized (freeze-dried) powder that has been sterile filtered.
Formulation
Lyophilized in PBS, pH 7.4, at a concentration of 0.2 µm after being filtered.
Solubility
It is advised to reconstitute the lyophilized FABP1 in sterile 18 MΩ-cm H2O to a final concentration of at least 100 µg/ml. This solution can then be diluted further into other aqueous solutions.
Stability
Although lyophilized FABP1 is stable at room temperature for 3 weeks, it should be kept dry below -18°C. FABP1 should be kept at 4°C for 2–7 days after reconstitution, and it should be kept below -18°C for longer-term storage. Avoid freeze-thaw cycles.
Purity
Greater than 95.0% as determined by:
(a) RP-HPLC analysis.
(b) SDS-PAGE analysis.
Synonyms
Fatty acid-binding protein 1 liver, L-FABP, FABPL, FABP-1, FABP1, Z-protein.
Source
Escherichia Coli.
Amino Acid Sequence
MNFSGKYQLQ SQENFEPFMK AIGLPEDLIQ KGKDIKGVSE IVHEGKKIKL TITYGPKVVR NEFTLGEECE LETMTGEKVK AVVKLEGDNK MVTTFKGIKS VTELNGDTIT NTMTLGDIVY KRVSKRI.

Q&A

What is FABP1 and what are its primary functions in mice?

FABP1 (Fatty Acid Binding Protein 1), also known as L-FABP (Liver Fatty Acid Binding Protein), plays a crucial role in regulating fatty acid metabolism and intracellular transport. Unlike other FABPs, FABP1 has the distinctive capability of binding two long-chain fatty acids simultaneously and transporting more fatty acids to mitochondria or peroxisomes than other FABP family members . In mice, FABP1 is predominantly expressed in the liver but also appears in other tissues such as the lung, where it has been detected in type II alveolar epithelial cells . Its primary functions include:

  • Facilitating the uptake and transport of long-chain fatty acids

  • Protecting cells from lipotoxicity caused by excess free fatty acids

  • Mediating fatty acid oxidation processes

  • Contributing to energy homeostasis through lipid metabolism regulation

  • Protecting specific cells (such as alveolar epithelium) from injury

How is FABP1 expression typically detected in mouse tissues?

FABP1 expression in mouse tissues can be detected through several complementary techniques:

  • Western Blotting: Using specific antibodies such as Mouse Anti-Human/Mouse/Rat FABP1/L-FABP Monoclonal Antibody (like MAB29641), FABP1 appears as a band at approximately 14 kDa under reducing conditions . This method works well with various mouse tissues including liver, lung, and kidney samples.

  • Immunohistochemistry and Immunofluorescence: These techniques are valuable for visualizing the spatial distribution of FABP1 in tissue sections. Double immunofluorescence staining has revealed that FABP1 is mainly localized in type II alveolar epithelial cells in mouse lung tissue .

  • qPCR Analysis: For quantifying FABP1 mRNA expression levels, real-time qPCR using SYBR Green and FABP1-specific primers, with normalization against GAPDH, provides reliable results .

  • Proteomics Analysis: Mass spectrometry-based proteomics can detect changes in FABP1 protein levels, as demonstrated in bleomycin and SiO₂-induced pulmonary fibrosis models .

What are the validated mouse models used to study FABP1 function?

Several validated mouse models have been developed to study FABP1 function:

  • FABP1 Knockout Mice: C57BL/6 J mice with FABP1 gene deletion have been established and are available from institutions like the Model Animal Research Center of Nanjing University . These models provide valuable insights into the physiological roles of FABP1 through loss-of-function studies.

  • FABP1-Overexpressing Mice: Generated through intratracheal instillation with AAV9-FABP1 viral vectors to increase FABP1 expression in specific tissues like the lung . Control mice typically receive AAV9-GFP for comparative analysis.

  • Disease-Specific Models:

    • Bleomycin (BLM)-induced pulmonary fibrosis model (2.5 mg/kg administered intratracheally)

    • SiO₂-induced pulmonary fibrosis model

    • Hepatocellular carcinoma models where FABP1's role in tumor-associated macrophages can be studied

These models can be combined with FABP1 genetic modifications to investigate the protein's role in disease pathogenesis and potential therapeutic interventions.

How does FABP1 contribute to the pathogenesis of pulmonary fibrosis in mouse models?

FABP1's role in pulmonary fibrosis appears to be protective, with its expression significantly reduced during disease progression. Proteomics analyses of both bleomycin and SiO₂-induced pulmonary fibrosis models have demonstrated a consistent downregulation of FABP1 compared to control mice . The mechanism through which FABP1 contributes to pulmonary fibrosis pathogenesis involves multiple pathways:

  • Alveolar Epithelial Protection: FABP1 protects type II alveolar epithelial cells from injury and promotes their survival. Overexpression of FABP1 through AAV9-FABP1 delivery alleviates pulmonary fibrosis by enhancing this protective effect .

  • Fatty Acid Metabolism Regulation: In IPF (Idiopathic Pulmonary Fibrosis), dysfunctional type II alveolar epithelial cells become vulnerable to apoptosis due to endoplasmic reticulum stress caused by elevated saturated fatty acid concentrations. FABP1 may mitigate this process by facilitating proper fatty acid transport and metabolism .

  • Disease Progression Correlation: FABP1 expression is negatively correlated with pulmonary fibrosis progression, suggesting its potential as a biomarker or therapeutic target .

Importantly, in vivo experiments have shown that FABP1 knockout mice (FABP1⁻/⁻) develop more severe fibrosis when challenged with bleomycin compared to wild-type counterparts, while FABP1 overexpression has demonstrated therapeutic effects .

What is the role of FABP1 in tumor-associated macrophages in mouse models of hepatocellular carcinoma?

FABP1 plays a critical role in establishing an immunosuppressive environment in hepatocellular carcinoma (HCC) through its effects on tumor-associated macrophages (TAMs). Single-cell RNA sequencing has revealed a unique FABP1-expressing macrophage population (Mac-FABP1) with distinct functional properties :

  • Phenotype Regulation: FABP1 promotes the M2 (immunosuppressive) phenotype of TAMs. Silencing FABP1 (si-FABP1) in these cells causes reversion from M2 to M1 (pro-inflammatory) phenotype, demonstrated by decreased expression of M2 markers including PD-L1 .

  • Metabolic Programming: FABP1 interacts with PPARG (peroxisome proliferator-activated receptor gamma) to increase fatty acid oxidation (FAO) in TAMs. This metabolic adaptation is crucial for maintaining the M2 phenotype:

    • FABP1 knockdown decreases expression of PPARG and CD36 (a scavenger receptor involved in lipid uptake)

    • Co-immunoprecipitation experiments have confirmed direct interaction between FABP1 and PPARG

    • Lipid droplet accumulation in TAMs is significantly reduced following FABP1 silencing

  • Immunosuppressive Environment: In wild-type versus FABP1⁻/⁻ mouse tumors, mass cytometry revealed that FABP1 deficiency altered immune cell populations:

    • Decreased regulatory T cells and natural killer cells

    • Increased dendritic cells, M1 macrophages, and B cells

    • Reduced expression of immune checkpoint molecules including PD-1 and CTLA-4

These findings suggest that FABP1 inhibition represents a potential therapeutic strategy for HCC, particularly in combination with immune checkpoint inhibitors.

How do genetic modifications of FABP1 in mice affect response to therapeutic interventions?

Genetic modifications of FABP1 in mice significantly impact their response to therapeutic interventions across multiple disease contexts:

  • Pulmonary Fibrosis Treatment:

    • FABP1⁻/⁻ knockout mice show exacerbated fibrotic responses to bleomycin challenge (1.7 mg/kg administered intratracheally), suggesting that strategies to increase FABP1 expression might be therapeutically beneficial .

    • Conversely, FABP1 overexpression via AAV9-FABP1 viral vectors administered 3 weeks before bleomycin challenge (2.5 mg/kg) demonstrates protective effects against fibrosis development .

  • Hepatocellular Carcinoma Immunotherapy:

    • FABP1 deficiency creates an immune-activated tumor microenvironment with increased infiltration of immune effector cells and decreased immunosuppressive cell populations .

    • This altered immune landscape enhances tumor sensitivity to immune checkpoint inhibitor therapy, particularly anti-PD-1 treatment .

    • FABP1⁻/⁻ mice consistently show attenuated tumor growth compared to wild-type C57BL/6 mice .

  • Pharmacological Inhibition Synergy:

    • Orlistat (a lipase inhibitor) has been identified as an effective FABP1 inhibitor that can replicate many effects of genetic deletion .

    • Combination therapy using orlistat with anti-PD-1 antibodies shows synergistic effects in treating HCC progression .

    • Advanced drug delivery systems, such as liposomes loaded with orlistat and connected with IR780 probe, enhance therapeutic efficacy and enable visualization of drug metabolism in vivo .

These findings highlight how understanding the genetic impacts of FABP1 modification can inform rational therapeutic strategies that may vary depending on the disease context.

What are the optimal techniques for detecting FABP1 protein in mouse tissue samples?

The detection of FABP1 protein in mouse tissue samples requires careful consideration of sample preparation and detection methods:

Western Blot Protocol Optimization:

  • Protein extraction should be performed under reducing conditions using Immunoblot Buffer Group 1 .

  • PVDF membrane is recommended for optimal protein transfer .

  • Mouse Anti-Human/Mouse/Rat FABP1/L-FABP Monoclonal Antibody (e.g., MAB29641) at 0.25 μg/mL concentration provides specific detection .

  • HRP-conjugated Anti-Mouse IgG Secondary Antibody (e.g., HAF018) is appropriate for visualization .

  • FABP1 appears as a distinct band at approximately 14 kDa .

Simple Western Analysis:

  • For automated capillary-based immunoassays, 5-10 μg/mL of anti-FABP1 antibody is recommended .

  • Sample loading concentration should be optimized around 0.2 mg/mL for liver tissue .

  • 12-230 kDa separation system works effectively for FABP1 detection .

Immunohistochemistry and Immunofluorescence:

  • For localization studies, double immunofluorescence staining is particularly valuable for identifying cell-specific expression (e.g., type II alveolar epithelial cells in lung tissue) .

  • Standard fixation with 4% paraformaldehyde followed by paraffin embedding or frozen section preparation is suitable.

Sample Sources Validated for Mouse FABP1 Detection:

  • Liver tissue (highest expression)

  • Lung tissue

  • Kidney tissue

  • Colon tissue

  • HepG2 hepatocellular carcinoma cell line (for in vitro studies)

When comparing samples across experimental conditions (e.g., wild-type versus knockout, or disease versus control), consistent protein loading and normalization to housekeeping proteins are essential for accurate quantification.

What considerations should be taken when designing experiments with FABP1 knockout mice?

Designing experiments with FABP1 knockout mice requires attention to several important factors:

Genetic Background and Controls:

  • Ensure FABP1⁻/⁻ mice and wild-type controls share the same genetic background (typically C57BL/6J) .

  • Include heterozygous mice (FABP1⁺/⁻) in breeding schemes to generate littermate controls when possible.

  • Age- and sex-matching between experimental groups is critical as lipid metabolism can vary significantly between male and female mice and across development.

Experimental Group Design:

  • A comprehensive experimental design should include at minimum: wild-type control group, FABP1⁻/⁻ group, wild-type + experimental challenge, and FABP1⁻/⁻ + experimental challenge groups .

  • Sample size calculations should account for potential variability in phenotypes related to lipid metabolism.

Phenotyping Considerations:

  • Baseline characterization should include liver function tests, lipid profiles, body weight monitoring, and metabolic parameters.

  • Tissue collection protocols must be standardized, with particular attention to fasting conditions before harvest, as this significantly affects lipid metabolism.

  • For pulmonary studies, techniques such as micro-CT imaging provide valuable non-invasive assessment of disease progression .

Disease Model Adaptations:

  • For pulmonary fibrosis models, bleomycin dosage may need adjustment in FABP1⁻/⁻ mice (1.7 mg/kg has been validated) compared to standard protocols .

  • For HCC models, tumor development kinetics may differ in FABP1⁻/⁻ mice, requiring adjusted experimental timelines .

Compensatory Mechanism Assessment:

  • Evaluate potential upregulation of other FABP family members (FABP2-9) that might compensate for FABP1 loss, using qPCR or protein analysis.

  • Consider comprehensive lipidomic profiling to understand broader metabolic adaptations in knockout animals.

What protocols are recommended for studying FABP1's role in fatty acid metabolism in primary mouse cells?

To investigate FABP1's role in fatty acid metabolism in primary mouse cells, the following protocols are recommended:

Primary Cell Isolation and Culture:

  • For primary hepatocytes: Two-step collagenase perfusion method followed by Percoll gradient centrifugation to obtain pure hepatocyte populations.

  • For alveolar type II cells: Elastase digestion followed by negative selection against CD45, CD31, and Ter119 positive cells.

  • For tumor-associated macrophages: Enzymatic digestion of tumor tissue followed by CD11b positive selection using magnetic beads.

FABP1 Expression Modulation:

  • Knockdown approaches:

    • siRNA transfection using lipid-based reagents (validated sequences targeting mouse FABP1 mRNA) .

    • shRNA lentiviral vectors for stable knockdown in longer-term cultures.

  • Overexpression approaches:

    • Adenoviral or AAV vectors containing the mouse FABP1 coding sequence under a strong promoter (e.g., CMV).

    • Transfection of expression plasmids for transient studies.

Fatty Acid Metabolism Assessment:

  • Fatty acid uptake:

    • Use fluorescently labeled fatty acids (e.g., BODIPY-FA) and measure cellular accumulation by flow cytometry or fluorescence microscopy.

  • Lipid droplet visualization:

    • Oil Red O or BODIPY 493/503 staining followed by quantitative image analysis .

  • Fatty acid oxidation measurement:

    • Seahorse XF analyzer to measure oxygen consumption rate (OCR) with or without etomoxir (CPT1 inhibitor).

    • ¹⁴C-palmitate oxidation assay to measure complete oxidation (¹⁴CO₂ production) and incomplete oxidation (¹⁴C-labeled acid-soluble metabolites).

  • Lipidomics analysis:

    • Liquid chromatography-mass spectrometry (LC-MS) to profile changes in lipid species.

Protein Interaction Studies:

  • Co-immunoprecipitation experiments to detect FABP1 interactions with partners like PPARG .

  • Proximity ligation assay for in situ visualization of protein-protein interactions.

  • FRET or BRET assays for real-time monitoring of dynamic interactions.

Gene Expression Analysis:

  • qPCR to measure expression of genes involved in fatty acid metabolism (e.g., PPARG, CD36, CPT1) .

  • RNA-seq to capture genome-wide transcriptional changes in response to FABP1 modulation.

These methodological approaches provide a comprehensive toolkit for dissecting FABP1's specific functions in fatty acid metabolism across different primary mouse cell types.

How can researchers address variability in FABP1 expression across different mouse strains?

Addressing variability in FABP1 expression across different mouse strains requires systematic approaches:

Characterization of Baseline Expression:

  • Perform strain-specific FABP1 expression profiling using qPCR and western blot analysis across major inbred strains (C57BL/6J, BALB/c, C3H, FVB, etc.).

  • Create a reference database of baseline FABP1 expression levels in key tissues (liver, lung, kidney) across different strains and age groups.

Experimental Design Strategies:

  • Use littermate controls whenever possible to minimize strain-specific variation.

  • If comparing different strains is unavoidable, increase biological replicates (n≥8 per group) to account for strain-dependent variability.

  • Consider backcrossing genetically modified FABP1 models (knockouts or transgenics) onto the desired strain background for at least 8-10 generations to achieve >99% genetic homogeneity.

Normalization and Standardization:

  • Employ multiple housekeeping genes (e.g., GAPDH, β-actin, and 18S rRNA) for qPCR normalization rather than relying on a single reference gene.

  • Include a common reference sample across all western blot experiments to enable inter-blot comparisons.

  • Consider using pooled samples from multiple strains as universal reference standards.

Technical Considerations:

  • Standardize tissue collection protocols, including time of day, feeding status, and euthanasia method, as these factors can affect FABP1 expression.

  • Process all comparative samples simultaneously using identical reagent lots and protocols.

  • For antibody-based detection, validate antibody specificity in each strain using FABP1⁻/⁻ tissues as negative controls .

Statistical Approaches:

  • Apply mixed-effects models that can account for strain as a random effect when analyzing data from multiple strains.

  • Consider using strain-specific z-scores rather than raw expression values when compiling data across strains.

By implementing these strategies, researchers can better account for and address the inherent variability in FABP1 expression across different mouse genetic backgrounds.

What are the potential pitfalls when interpreting phenotypes in FABP1 knockout mice?

Interpreting phenotypes in FABP1 knockout mice presents several potential pitfalls that researchers should consider:

Compensatory Mechanisms:

  • Other FABP family members (particularly FABP2, FABP4, and FABP5) may be upregulated in FABP1⁻/⁻ mice, partially masking expected phenotypes.

  • Alternative fatty acid transport pathways involving CD36, FATPc, or passive diffusion may compensate for FABP1 deficiency.

  • These compensatory changes should be systematically evaluated before attributing phenotypes directly to FABP1 loss.

Developmental Adaptations:

  • FABP1⁻/⁻ mice may develop adaptive mechanisms during embryonic and postnatal development that would not occur with acute FABP1 inhibition.

  • Consider complementing germline knockout studies with inducible knockout systems or acute pharmacological inhibition to distinguish developmental from functional effects.

Context-Dependent Phenotypes:

  • FABP1 deficiency may manifest differently depending on dietary conditions. Standard chow versus high-fat diet can reveal distinct phenotypes.

  • Environmental stressors (e.g., fasting, cold exposure) may be required to unmask subtle metabolic phenotypes not apparent under basal conditions.

  • Disease challenges (e.g., bleomycin for pulmonary fibrosis) might be necessary to reveal FABP1's protective roles .

Tissue-Specific Effects:

  • Global FABP1 knockout affects multiple organs simultaneously, making it difficult to distinguish primary from secondary effects.

  • Consider using tissue-specific conditional knockout models to isolate FABP1's role in specific cell types (e.g., hepatocytes versus macrophages).

Background Strain Influences:

  • The same FABP1 knockout on different genetic backgrounds may yield different phenotypes.

  • The C57BL/6J background used in most studies may have specific interactions with FABP1 deficiency not generalizable to other strains.

Sex-Specific Differences:

  • Male and female FABP1⁻/⁻ mice may display different phenotypes, particularly in lipid metabolism and immune responses.

  • Always analyze and report data for both sexes separately before combining.

Age-Dependent Effects:

  • Young FABP1⁻/⁻ mice may show minimal phenotypes while aging might reveal progressive metabolic or inflammatory abnormalities.

  • Longitudinal studies across different age groups are valuable for comprehensive phenotyping.

By carefully considering these potential pitfalls, researchers can design more robust experiments and arrive at more accurate interpretations of FABP1 knockout phenotypes.

How can researchers distinguish between direct and indirect effects of FABP1 in complex disease models?

Distinguishing between direct and indirect effects of FABP1 in complex disease models requires multi-faceted experimental approaches:

Temporal Intervention Studies:

  • Implement time-course experiments with FABP1 modulation at different disease stages:

    • Pre-disease initiation (preventive)

    • Early disease (early intervention)

    • Established disease (therapeutic)

  • Different outcomes based on timing suggest stage-specific FABP1 functions.

  • For example, in pulmonary fibrosis models, administer AAV9-FABP1 before, concurrent with, or after bleomycin challenge to determine when FABP1 exerts its protective effects .

Cell Type-Specific Approaches:

  • Use conditional knockout or overexpression systems with cell type-specific promoters (e.g., SPC-Cre for type II alveolar cells, LysM-Cre for macrophages).

  • Perform adoptive transfer experiments (e.g., FABP1⁻/⁻ macrophages into wild-type mice) to isolate cell-autonomous effects.

  • Employ cell type-specific FACS sorting followed by molecular analysis to identify which cells actually express or respond to FABP1 in the disease microenvironment .

Molecular Pathway Validation:

  • Conduct epistasis experiments by modulating proposed downstream effectors (e.g., PPARG or CD36) in FABP1⁻/⁻ background to test pathway dependencies .

  • Use pharmacological inhibitors targeting specific pathways in combination with genetic FABP1 modulation:

    • If inhibiting a pathway blocks FABP1 effects, that pathway is likely downstream

    • If effects are additive, pathways may be parallel

Ex Vivo and In Vitro Validation:

  • Isolate primary cells from disease models and assess direct FABP1 effects in controlled environments.

  • Use conditioned media experiments to determine if FABP1 effects are mediated by secreted factors.

  • Employ co-culture systems to study cell-cell interactions dependent on FABP1 expression.

Biomarker and Metabolite Profiling:

  • Perform targeted metabolomics focusing on fatty acids and their metabolites.

  • Create a temporal map of metabolic changes following FABP1 modulation to identify primary versus secondary effects.

  • Validate putative direct targets using techniques like thermal shift assays or binding studies with recombinant FABP1 protein.

Multi-Omics Integration:

  • Combine transcriptomics, proteomics, and metabolomics data with computational network analysis to identify direct FABP1-dependent nodes versus downstream effects.

  • Employ single-cell approaches to resolve cellular heterogeneity within complex tissues .

By systematically employing these strategies, researchers can more confidently distinguish direct FABP1-mediated effects from secondary consequences in complex disease models.

What emerging technologies might advance our understanding of FABP1 function in mouse models?

Several cutting-edge technologies show promise for deepening our understanding of FABP1 function in mouse models:

Spatial Transcriptomics and Proteomics:

  • Technologies like 10x Visium, Slide-seq, or CODEX enable simultaneous visualization of FABP1 expression and related pathways with spatial resolution in tissue sections.

  • These approaches would be particularly valuable for mapping FABP1's role in heterogeneous tissues like tumor microenvironments or fibrotic lungs, revealing localized functions not apparent in bulk analyses .

Live Cell FABP1 Tracking:

  • CRISPR-based knock-in of fluorescent tags (e.g., mNeonGreen) to endogenous FABP1 would enable real-time visualization of protein trafficking and dynamics.

  • Advanced microscopy techniques like lattice light-sheet microscopy combined with labeled fatty acids could reveal FABP1-mediated lipid transport kinetics in living cells with unprecedented resolution.

Metabolic Flux Analysis:

  • Stable isotope tracing using ¹³C-labeled fatty acids combined with mass spectrometry would provide dynamic information about FABP1's impact on metabolic pathways.

  • Integrating these approaches with genetic FABP1 modifications would reveal substrate-specific effects on fatty acid channeling to oxidation versus storage pathways.

Engineered Mouse Models:

  • Conditional and inducible FABP1 expression systems using Cre-Lox or Tet-On/Off technology would enable temporal and spatial control of FABP1 function.

  • CRISPR-based screens targeting FABP1 regulatory elements could identify context-specific control mechanisms.

  • Mouse models with specific FABP1 mutations affecting lipid binding capacity, protein-protein interactions, or post-translational modifications would help dissect structure-function relationships in vivo.

Integrative Multi-Omics Approaches:

  • Single-cell multi-omics (e.g., CITE-seq, TEA-seq) would simultaneously profile transcriptome, proteome, and epigenome in individual cells from FABP1 wild-type and knockout mice .

  • These datasets could identify cell type-specific FABP1 functions and regulatory networks.

Advanced Imaging of Lipid Metabolism:

  • Stimulated Raman scattering microscopy enables label-free visualization of lipids in tissues and cells.

  • Mass spectrometry imaging provides spatial distribution maps of specific lipid species in tissue sections.

  • These techniques could reveal how FABP1 deficiency alters lipid distribution and metabolism across tissues with subcellular resolution.

Nanobody-Based Approaches:

  • Development of FABP1-specific nanobodies could enable acute inhibition of protein function or protein-protein interactions.

  • These tools would complement genetic approaches by allowing rapid, reversible perturbation of FABP1 activity.

Implementation of these emerging technologies will likely yield new insights into FABP1's context-specific functions and potential as a therapeutic target.

How might targeting FABP1 be developed as a therapeutic strategy based on mouse model findings?

Findings from mouse models suggest several promising approaches for developing FABP1-targeted therapeutics:

Pulmonary Fibrosis Therapeutic Strategies:

  • FABP1 Upregulation Approaches:

    • AAV-mediated FABP1 gene therapy has shown efficacy in mouse models of pulmonary fibrosis .

    • Screening for small molecule inducers of FABP1 expression could yield orally available drugs.

    • Targeting transcriptional regulators of FABP1 could enhance endogenous expression.

  • Alveolar Epithelial Cell Protection:

    • Since FABP1 protects type II alveolar epithelial cells, cell-targeted delivery systems could enhance therapeutic efficacy .

    • Nanoparticle formulations with surfactant protein recognition motifs could achieve alveolar epithelial cell-specific delivery.

Hepatocellular Carcinoma Immunotherapy Enhancement:

  • FABP1 Inhibition Strategies:

    • Orlistat has demonstrated FABP1 inhibitory activity and shows synergy with anti-PD-1 therapy in mouse models .

    • Rational design of specific FABP1 inhibitors based on protein structure could yield more selective compounds.

    • Advanced drug delivery systems like liposomes connected with IR780 probe have shown enhanced efficacy and in vivo visualization capabilities .

  • Macrophage Reprogramming:

    • Since FABP1 regulates TAM polarization through PPARG/CD36 pathways, targeted inhibition in macrophages could reprogram the tumor immune microenvironment .

    • Nanoparticles designed to target CD169+ macrophages could deliver FABP1 inhibitors specifically to TAMs.

Translational Considerations from Mouse to Human:

  • Biomarker Development:

    • FABP1 expression levels could serve as biomarkers for disease progression or treatment response.

    • For pulmonary fibrosis, declining FABP1 levels correlate with disease progression .

    • For HCC, high FABP1 expression in TAMs might predict responsiveness to combination immunotherapy .

  • Patient Stratification Strategies:

    • Genetic polymorphisms affecting FABP1 expression or function in humans should be evaluated.

    • Patients could be stratified based on FABP1 expression patterns to identify those most likely to benefit from FABP1-targeted therapies.

Combination Therapy Opportunities:

  • In HCC, combining FABP1 inhibition with immune checkpoint blockade (anti-PD-1) shows synergistic effects .

  • For pulmonary fibrosis, FABP1 upregulation might complement anti-fibrotic drugs like nintedanib or pirfenidone.

Safety Considerations Based on Mouse Models:

  • Complete FABP1 knockout mice remain viable, suggesting potential therapeutic windows for intervention .

  • Liver-specific effects should be carefully monitored given FABP1's high expression in hepatocytes.

  • Metabolic parameters require monitoring since FABP1 plays important roles in lipid homeostasis.

These therapeutic strategies derived from mouse model findings provide a roadmap for translational development of FABP1-targeted interventions for pulmonary fibrosis, HCC, and potentially other diseases involving dysregulated lipid metabolism or immune function.

What are the most promising avenues for investigating FABP1's role in immune regulation based on recent findings?

Recent findings, particularly from hepatocellular carcinoma models, highlight several promising research directions for exploring FABP1's role in immune regulation:

Macrophage Polarization and Metabolism:

  • Further investigate the molecular mechanisms by which FABP1 regulates the M1/M2 polarization switch in tumor-associated macrophages .

  • Explore whether the FABP1-PPARG-CD36 axis identified in HCC is operative in other inflammatory contexts such as atherosclerosis, obesity, or autoimmune disorders.

  • Determine how FABP1-mediated changes in fatty acid oxidation influence the epigenetic landscape and gene expression patterns in macrophages.

Adaptive Immune Cell Interactions:

  • Examine direct and indirect effects of FABP1 deficiency on regulatory T cell development and function, as these populations were decreased in FABP1⁻/⁻ tumors .

  • Investigate whether FABP1 expression in antigen-presenting cells affects T cell priming and effector functions.

  • Study the impact of FABP1-mediated lipid metabolism on dendritic cell maturation and antigen presentation capabilities.

Immune Checkpoint Regulation:

  • Further characterize the mechanisms behind reduced PD-1 and CTLA-4 expression observed in FABP1⁻/⁻ deficient mice .

  • Determine whether FABP1 directly or indirectly regulates immune checkpoint molecule expression on T cells.

  • Investigate whether FABP1 modulation affects additional immune checkpoints beyond PD-1/PD-L1 and CTLA-4.

Metabolic Immune Regulation:

  • Explore metabolic competition between immune cells and tissue cells for fatty acids in FABP1-deficient environments.

  • Investigate how FABP1-mediated changes in the lipid milieu affect immune cell recruitment, survival, and function.

  • Study potential lipid mediators (e.g., eicosanoids, specialized pro-resolving mediators) whose production is affected by FABP1 activity.

Tissue-Specific Immune Effects:

  • Compare FABP1's immune regulatory functions across different tissues (liver, lung, intestine) to identify context-specific mechanisms.

  • For pulmonary fibrosis, determine whether FABP1's protective effects involve modulation of pro-fibrotic immune populations like fibrocytes or M2 macrophages .

  • In HCC, investigate whether FABP1 expression in hepatocytes affects immune cell recruitment and function differently than its expression in TAMs .

Translational Immune Monitoring:

  • Develop comprehensive immune monitoring panels for preclinical studies of FABP1 modulators that capture changes across innate and adaptive immunity.

  • Establish clinically relevant immune signatures associated with FABP1 expression patterns that could predict response to immunotherapies.

  • Investigate whether FABP1 genetic polymorphisms in humans correlate with immune-related disease susceptibility or treatment responses.

These research directions would significantly advance our understanding of how FABP1, traditionally viewed as a metabolic protein, functions as an important regulator of immune responses in various disease contexts.

Product Science Overview

Introduction

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 small, highly conserved cytoplasmic proteins that bind long-chain fatty acids and other hydrophobic ligands. FABP1 is primarily expressed in the liver but can also be found in the intestine, kidney, pancreas, stomach, and lung .

Discovery and Gene Location

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 long-chain fatty acids . The human FABP1 gene is located on the short arm of chromosome 2, while the mouse FABP1 gene is located on chromosome 6 .

Protein Structure

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 . The “portal hypothesis” suggests that fatty acids enter the solvent-accessible area of the protein through a dynamic region consisting of α-helix II and turns between the βC-βD and βE-βF loops. The fatty acid is then bound in the protein cavity for transport .

Function

FABP1 plays a significant role in the binding, transport, and metabolism of long-chain fatty acids, endocannabinoids, phytocannabinoids, and other hydrophobic molecules . It is abundantly expressed in the liver, accounting for 7-11% of the total cytosolic protein . FABP1 is unique in its ability to bind a wide range of hydrophobic ligands, including bilirubin, monoglycerides, bile acids, and fatty acyl CoA .

Role in Metabolic Conditions

Altered expression of FABP1 has been linked to various metabolic conditions, including obesity . The protein’s ability to bind and transport fatty acids and other hydrophobic molecules is crucial in preventing cytotoxicity by binding potentially toxic molecules when unbound .

Recombinant Mouse FABP1

Recombinant mouse FABP1 is used in research to study its structure, function, and role in various metabolic processes. The recombinant protein is produced using genetic engineering techniques, allowing researchers to investigate its interactions with different ligands and its impact on metabolic pathways.

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.