GNPAT Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Liquid in PBS containing 50% glycerol, 0.5% BSA, and 0.02% sodium azide.
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchasing method and location. Please consult your local distributors for specific delivery details.
Synonyms
Acyl CoA dihydroxyacetonephosphateacyltransferase antibody; Acyl-CoA:dihydroxyacetonephosphateacyltransferase antibody; DAP AT antibody; DAP-AT antibody; DAPAT antibody; DHAP AT antibody; DHAP-AT antibody; DHAPAT antibody; Dihydroxyacetone phosphate acyltransferase antibody; EC 2.3.1.42 antibody; Glycerone-phosphate O-acyltransferase antibody; Glyceronephosphate O acyltransferase antibody; GNPAT antibody; GNPAT_HUMAN antibody; OTTHUMP00000036147 antibody
Target Names
GNPAT
Uniprot No.

Target Background

Function
Dihydroxyacetonephosphate acyltransferase is an enzyme involved in the biosynthesis of plasmalogens.
Gene References Into Functions

GNPAT is a gene encoding for Dihydroxyacetonephosphate acyltransferase. Research findings have highlighted the significant roles of GNPAT in various physiological processes, particularly in relation to iron metabolism, lipid biosynthesis, and neurological functions.

  • GNPAT p.D519G is strongly associated with increased iron stores in individuals homozygous for the p.C282Y mutation in the HFE gene, even after accounting for age, iron-related variables, and alcohol consumption. PMID: 27936396
  • The rs11558492 SNP in GNPAT is not a primary factor influencing iron status or associated with liver fibrosis in patients with HFE-related hemochromatosis. PMID: 28425416
  • A study characterized recombinant human DHAP acyl-transferase, which catalyzes the initial step in alkyl-DHAP synthesis. PMID: 27836547
  • Reduction of GNPAT activity activated NF-kappaB in glial cell lines and microglia in the cortex. PMID: 28292831
  • Individuals homozygous for the C282Y mutation in the HFE gene, who are referred for HFE testing, often carry a GNPAT variant. While this variant does not seem to be a major modifier of HFE-related hemochromatosis expression, it does not reliably predict the severity of iron overload. PMID: 27740525
  • A variant in the GNPAT gene exhibited a strong association with severe iron overload. PMID: 25605615
  • Silencing of ACOX1 and GNPAT genes resulted in an upregulation of ceramide galactosyltransferase (UGT8) mRNA expression and a downregulation of UDP-glucoseceramide glucosyltransferase (UGCG). PMID: 23933200
  • Novel mutations in GNPAT are associated with rhizomelic chondrodysplasia punctata (RCDP) type 2. PMID: 21990100
  • Peroxisomal DHAPAT is crucial for plasmalogen biosynthesis in animal cells. PMID: 15687349
  • Chromosome 1q42.1 contains GNPAT and DISC1, which are candidate genes for schizophrenia. PMID: 16997000
Database Links

HGNC: 4416

OMIM: 222765

KEGG: hsa:8443

STRING: 9606.ENSP00000355607

UniGene: Hs.498028

Involvement In Disease
Rhizomelic chondrodysplasia punctata 2 (RCDP2)
Protein Families
GPAT/DAPAT family
Subcellular Location
Peroxisome membrane; Peripheral membrane protein; Matrix side.

Q&A

What is GNPAT and why is it important in cellular metabolism?

GNPAT (glyceronephosphate O-acyltransferase) is a key enzyme exclusively localized within peroxisomes that plays a critical role in the biosynthesis of ether phospholipids, particularly plasmalogens. With a calculated molecular weight of 77 kDa (though typically observed at 65-69 kDa in experimental conditions), GNPAT represents the first step in plasmalogen synthesis . The importance of GNPAT in cellular metabolism stems from its role in producing plasmalogens, which are essential membrane components that protect cells against oxidative stress, maintain membrane fluidity, and support various signaling functions. Full GNPAT activity depends not only on its expression but also on the presence of AGPS (alkylglycerone phosphate synthase) and the integrity of substrate channeling between these enzymes . Dysfunction in this pathway has been implicated in various pathological conditions, including neurodegenerative diseases and inflammatory responses.

What applications can GNPAT antibodies be used for in experimental research?

GNPAT antibodies have been validated for multiple experimental applications:

ApplicationDetailsRecommended Dilution
Western Blot (WB)Detects GNPAT in various cell lines and tissues including HepG2, HeLa, PC-3, and mouse brain tissue1:500-1:1000
Immunoprecipitation (IP)Successfully detects GNPAT in samples like COLO 320 cells0.5-4.0 μg for 1.0-3.0 mg of total protein lysate
Immunohistochemistry (IHC)Used to detect GNPAT in tissue sectionsVaries by protocol
Immunofluorescence (IF)Used for localization studies, including double staining with cellular markersTypically 1:200-1:300
ELISAUsed for quantitative detectionVaries by protocol

For optimal results, the antibody should be titrated for each specific experimental system. GNPAT antibody has demonstrated reactivity with human, mouse, and rat samples, making it versatile for cross-species studies .

How should Western blot protocol be optimized for GNPAT detection?

For optimal Western blot detection of GNPAT, follow these methodological guidelines:

  • Sample Preparation: Extract total protein using RIPA lysis buffer and determine concentration using BCA protein assay .

  • Protein Loading: Load approximately 30 μg of protein per lane after mixing with loading buffer and boiling .

  • Gel Electrophoresis: Use SDS-PAGE to separate proteins, looking for GNPAT at 65-69 kDa (observed molecular weight) .

  • Blocking: Block membranes with 5% non-fat milk for 2 hours at room temperature .

  • Primary Antibody Incubation: Use GNPAT antibody at a dilution of 1:500-1:1000 and incubate overnight at 4°C .

  • Secondary Antibody: Depending on your detection system, use either:

    • Horseradish peroxidase-conjugated secondary antibody (1:5000) for chemiluminescent detection

    • Infrared dye-conjugated secondary antibody (1:5000) for infrared imaging systems

  • Detection: Visualize using enhanced chemiluminescence reagent or an infrared imaging system .

  • Normalization: Normalize GNPAT expression to β-actin or other housekeeping proteins for quantitative analysis .

When troubleshooting, consider that the observed molecular weight (65-69 kDa) differs from the calculated weight (77 kDa), which is important for accurate band identification .

What are the recommended storage conditions for GNPAT antibody?

To maintain optimal antibody performance, GNPAT antibody should be stored at -20°C, where it remains stable for one year after shipment . The antibody is typically provided in PBS with 0.02% sodium azide and 50% glycerol at pH 7.3 . Importantly, aliquoting is unnecessary for -20°C storage, which simplifies laboratory handling procedures . For smaller sized aliquots (20 μL), the preparation may contain 0.1% BSA as a stabilizing agent . These storage conditions are critical for maintaining antibody reactivity and specificity, particularly for sensitive applications like immunofluorescence and immunoprecipitation. Proper storage prevents antibody degradation and ensures consistent experimental results across multiple studies.

How can I validate the specificity of my GNPAT antibody?

Validating GNPAT antibody specificity requires a multi-faceted approach:

  • Knockdown/Knockout Validation: Use shRNA-mediated knockdown of GNPAT (as demonstrated in studies using sh-GNPAT lentivirus) to confirm antibody specificity by demonstrating reduced signal compared to controls . This provides strong evidence that the antibody is detecting the intended target.

  • Western Blot Analysis: Verify that the antibody detects a protein of the expected molecular weight (65-69 kDa for GNPAT) across multiple cell lines or tissues known to express the protein, such as HepG2 cells, human placenta tissue, COLO 320 cells, and mouse brain tissue .

  • Immunoprecipitation: Perform IP followed by Western blot to confirm that the antibody can both capture and detect GNPAT protein .

  • Multiple Antibody Comparison: Compare results using different GNPAT antibodies (e.g., comparing Proteintech 14931-1-AP and Abcam ab75060) .

  • Positive Controls: Include positive control samples with known GNPAT expression, such as Transfected HEK-293 cells overexpressing GNPAT .

  • Application-specific Controls: For immunofluorescence, perform negative controls by omitting primary antibody and include double staining with known peroxisomal markers to confirm subcellular localization .

How is GNPAT expression regulated at the post-translational level?

Recent research has revealed significant insights into the post-translational regulation of GNPAT, particularly through acetylation/deacetylation mechanisms:

  • SIRT4-Mediated Deacetylation: SIRT4, a mitochondrial sirtuin with deacetylase activity, has been identified as a key regulator of GNPAT. Studies show that SIRT4 can deacetylate GNPAT, affecting its function and stability . This regulatory mechanism appears particularly important in cellular stress responses.

  • Acetylation Status Detection: The acetylation status of GNPAT can be assessed through immunoprecipitation assays using GNPAT antibody (e.g., Abcam ab75060) followed by Western blot analysis with an acetyl-lysine antibody . This approach allows researchers to monitor changes in GNPAT acetylation under different experimental conditions.

  • CSE-Induced Changes: Cigarette smoke extract (CSE) exposure has been shown to reduce SIRT4 expression while increasing GNPAT acetylation levels in a concentration-dependent manner . This suggests that environmental factors can influence GNPAT activity through modulation of its acetylation status.

  • Functional Consequences: Changes in GNPAT acetylation appear to affect its enzymatic activity and role in pathological processes such as ferroptosis . This provides a potential therapeutic target for conditions where GNPAT dysregulation contributes to disease pathogenesis.

The ability to manipulate and measure GNPAT acetylation status represents an important methodological advance in understanding its regulation beyond transcriptional control mechanisms.

What is the relationship between GNPAT and cellular signaling pathways?

GNPAT plays a critical role in modulating several key cellular signaling pathways:

  • ERK and Akt Signaling: GNPAT knockdown using sh-GNPAT lentivirus significantly reduces phosphorylated ERK and Akt protein levels in neuronal cells, indicating that GNPAT activity is necessary for the maintenance of these signaling pathways . Quantitative analysis showed statistically significant reductions (p < 0.01) in the activation of both pathways.

  • Plasmalogen-Mediated Signaling: Treatment with plasmalogens (Pls, 500 ng/ml) activates ERK and Akt phosphorylation, while phosphatidylethanolamine (PtdEtn) at the same concentration does not produce comparable effects . This suggests that the products of GNPAT enzymatic activity specifically trigger these signaling cascades.

  • G-Protein Coupled Receptor Interactions: GPCRs-mediated increases in ERK phosphorylation observed in control conditions are abolished in sh-GNPAT treated cells . This demonstrates that GNPAT function is required for GPCR-dependent activation of downstream signaling.

  • Functional Recovery: The signaling defects observed in GNPAT-deficient cells can be rescued by exogenous plasmalogen supplementation, confirming the specific role of these lipids in signal transduction .

These findings provide mechanistic insights into how GNPAT-dependent lipid metabolism integrates with cellular signaling networks, with important implications for understanding neuronal function and potential therapeutic interventions in conditions where these pathways are dysregulated.

How does GNPAT contribute to ferroptosis and what experimental approaches can be used to study this connection?

GNPAT plays a significant role in ferroptosis, a form of regulated cell death characterized by iron-dependent lipid peroxidation. Recent research has established experimental approaches to investigate this connection:

  • Knockdown Studies: Silencing GNPAT using sh-GNPAT transfection significantly alleviates cigarette smoke extract (CSE)-induced ferroptosis in A549 cells . Successful knockdown can be confirmed through:

    • Quantitative real-time PCR

    • Western blot analysis

    • Immunofluorescence staining using GNPAT antibody (14931-1-AP, Proteintech)

  • Functional Assays to Measure Ferroptosis Parameters:

    • Cell viability assays

    • LDH activity measurements

    • ROS production quantification

    • Cell apoptosis rate determination

    • Transmission electron microscopy to observe mitochondrial inner ridge morphology

    • Lipid peroxidation assessment

  • Molecular Markers Analysis: GNPAT inhibition results in:

    • Increased levels of GPX4 (a key anti-ferroptotic protein)

    • Elevated GSH (glutathione) levels

    • Decreased MDA (malondialdehyde, a product of lipid peroxidation)

  • SIRT4-GNPAT Interaction: The acetylation status of GNPAT affects its role in ferroptosis:

    • Higher CSE concentrations reduce SIRT4 expression

    • This correlates with increased GNPAT acetylation

    • Acetylation levels can be measured by immunoprecipitating GNPAT and blotting with acetyl-lysine antibody

These methodological approaches provide a comprehensive framework for investigating GNPAT's contribution to ferroptosis and potential intervention strategies for conditions where ferroptosis plays a pathological role.

How is GNPAT involved in neurodegeneration and what techniques are most suitable for studying this relationship?

GNPAT plays a crucial role in neurodegeneration through its function in plasmalogen biosynthesis. Several techniques have proven valuable for investigating this relationship:

  • Time-Dependent Expression Analysis: Studies in Alzheimer's disease models (J20 mice) have tracked GNPAT expression changes over disease progression using Western blot analysis with anti-GNPAT antibodies, normalized to β-actin . This approach allows for correlation between GNPAT expression levels and disease progression.

  • Cellular Localization in Neural Tissues: Double immunofluorescence staining techniques can determine the localization of GNPAT relative to:

    • Iba1+ microglia (using anti-Iba1 IgG, 1:200)

    • GFAP+ astrocytes (using anti-GFAP IgG, 1:200)

    • Followed by fluorescence-conjugated secondary antibodies and nuclear counterstaining with Hoechst 33258

  • Quantification of Immunoreactive Cells: The number of GNPAT-immunoreactive cells can be counted in consecutive brain sections using NIH ImageJ analysis software, providing quantitative assessment of expression changes .

  • Correlation with Plasmalogen Levels: Mass spectrometry techniques can measure plasmalogen molecular species in hippocampal brain tissues, allowing researchers to connect GNPAT expression with its enzymatic products .

  • Neuronal Signaling Analysis: GNPAT knockdown affects ERK and Akt signaling pathways in neuronal cells, which can be quantified by Western blotting assays. This approach helps understand how GNPAT-dependent lipid metabolism influences neuronal signaling and survival .

These methodological approaches provide a comprehensive toolkit for investigating GNPAT's involvement in neurodegenerative processes and potential therapeutic strategies.

What are the methodological considerations for studying GNPAT's interaction with other peroxisomal proteins?

Studying GNPAT's interactions with other peroxisomal proteins requires careful methodological considerations:

  • Co-Immunoprecipitation Approaches:

    • Use GNPAT antibody (such as ab75060, Abcam) cross-linked to resin

    • Incubate cell lysates with the antibody-resin complex overnight at 4°C

    • Elute protein complexes using appropriate buffers

    • Analyze by Western blot for interacting partners

  • Double Immunofluorescence Staining:

    • Primary antibodies against GNPAT and other peroxisomal proteins (e.g., AGPS, FAR1)

    • Use species-specific secondary antibodies with different fluorophores

    • Include appropriate controls to ensure specificity

    • Analyze co-localization using confocal microscopy and quantitative image analysis

  • Proximity Ligation Assays:

    • This technique can detect protein-protein interactions in situ

    • Provides spatial resolution of interactions within peroxisomes

    • Can help determine if GNPAT interactions change under different physiological or pathological conditions

  • Validation of Functional Interactions:

    • Study whether GNPAT activity depends on the presence of interacting partners

    • For example, research shows that full GNPAT activity depends not only on AGPS presence but also on intact substrate channeling between these enzymes

    • Knockdown experiments of potential interacting partners can help establish functional relationships

  • Examination of Subcellular Fractionation:

    • Isolate peroxisomal fractions to enrich for GNPAT and interacting partners

    • Western blot analysis of fractions can reveal co-purification of interacting proteins

    • Compare protein associations across different cell types or disease models

These approaches together provide a comprehensive strategy for investigating GNPAT's peroxisomal protein interactions and their functional significance in health and disease.

How can I optimize immunofluorescence protocols for GNPAT antibody?

For optimal immunofluorescence detection of GNPAT, follow these detailed optimization steps:

  • Sample Preparation:

    • For cells: Fix A549 or similar cell lines with 4% paraformaldehyde for 30 minutes

    • For tissue sections: Use appropriate fixation method (typically 4% paraformaldehyde)

    • Permeabilize with 1% Triton X-100 for 20 minutes to ensure antibody access to peroxisomal proteins

  • Blocking:

    • Block with 5% BSA for 1 hour at 37°C to reduce nonspecific binding

    • For tissue sections, longer blocking times may be necessary

  • Primary Antibody Incubation:

    • For GNPAT antibody: Use Proteintech 14931-1-AP at 1:200-1:300 dilution

    • For double staining with peroxisomal markers: Include appropriate antibodies (e.g., anti-Iba1 IgG or anti-GFAP IgG at 1:200)

    • Incubate overnight at 4°C for optimal antibody binding

  • Secondary Antibody Selection:

    • For GNPAT detection: Use fluorescence-conjugated goat anti-rabbit IgG FITC (1:200)

    • For co-staining partners: Use contrasting fluorophores (e.g., Texas Red conjugated secondary antibodies)

    • Incubate for 1-2 hours at room temperature

  • Nuclear Counterstaining:

    • Use Hoechst 33258 (10 μg/mL) for 5 minutes to identify cell nuclei

  • Controls and Validation:

    • Include negative controls (primary antibody omission)

    • Use GNPAT knockdown cells as specificity controls

    • Compare staining patterns with published literature

  • Imaging Parameters:

    • Capture images at appropriate magnification (40× for counting, higher for detailed localization)

    • Use confocal microscopy for precise co-localization studies

    • Process images with software like NIH ImageJ for quantitative analysis

This optimized protocol ensures reliable detection of GNPAT and its spatial relationship with other cellular components in both normal and experimental conditions.

What controls should be included when using GNPAT antibody in knockout/knockdown experiments?

For rigorous GNPAT knockout/knockdown experiments, the following controls are essential:

  • Knockdown Validation Controls:

    • Quantitative real-time PCR to confirm reduction at mRNA level

    • Western blot analysis using GNPAT antibody (e.g., 14931-1-AP or ab75060) to confirm protein reduction

    • Immunofluorescence staining to visualize decreased expression at cellular level

  • Vector Controls:

    • Include sh-Luciferase (sh-Luc) or other non-targeting shRNA lentivirus as negative control

    • This controls for non-specific effects of viral transduction and shRNA expression

  • Rescue Experiments:

    • Re-introduce GNPAT expression in knockdown cells to restore function

    • Alternatively, add purified plasmalogens (500 ng/ml) to rescue GNPAT-dependent phenotypes

    • These controls confirm that observed effects are specifically due to GNPAT deficiency

  • Dose-Response Analysis:

    • Use varying concentrations of knockdown vectors to establish relationship between GNPAT levels and phenotypic effects

    • This helps determine the threshold of GNPAT reduction needed for biological effects

  • Time-Course Analysis:

    • Monitor changes over time post-knockdown to distinguish between direct and secondary effects

    • This is particularly important when studying signaling pathways like ERK and Akt that may have complex regulatory feedback loops

  • Pathway-Specific Controls:

    • Include positive controls for specific pathways being studied (e.g., known activators of ERK/Akt signaling)

    • Include inhibitors of downstream pathways to confirm specificity of GNPAT-dependent effects

  • Cell Viability Controls:

    • Monitor general cell health metrics to ensure observed effects aren't due to cytotoxicity

    • Include assays for LDH activity, ROS production, and apoptosis rates

How can I quantify changes in GNPAT expression across different experimental conditions?

Accurate quantification of GNPAT expression requires standardized methodological approaches:

  • Western Blot Quantification:

    • Use validated antibodies (Proteintech 14931-1-AP or Abcam ab75060)

    • Always normalize GNPAT band intensity to housekeeping proteins like β-actin

    • Use densitometry software such as NIH ImageJ version 1.52 for consistent analysis

    • Report relative expression as fold change compared to control conditions

  • Immunofluorescence Quantification:

    • Count GNPAT-immunoreactive cells in multiple fields (typically 5 random fields)

    • Use consistent magnification (40×) across all samples

    • Analyze at least 3 consecutive tissue sections spaced 2 mm apart for tissue studies

    • Apply appropriate background correction and thresholding

  • Statistical Analysis Approaches:

    • For comparing two groups: Use Student's t-test with appropriate p-value thresholds (typically p < 0.05)

    • For multiple group comparisons: Apply one-way ANOVA followed by Bonferroni's test

    • For comparisons to a single control group: Use Dunnett's post hoc test

    • Always report mean ± S.E.M. with appropriate sample sizes (n values)

  • Time-Course Considerations:

    • When examining expression changes over time (as in disease progression studies), use consistent time points

    • Normalize all time points to the same control or baseline condition

    • Consider using repeated measures analysis for longitudinal studies

  • Detection System Standardization:

    • For chemiluminescent detection: Use AMERSHAM Image Quant 800 or similar systems

    • For infrared detection: Use Odyssey infrared imaging system (LI-COR Bioscience)

    • Maintain consistent exposure settings across experimental groups

What are common issues in co-immunoprecipitation experiments with GNPAT antibody and how can they be resolved?

Co-immunoprecipitation (Co-IP) with GNPAT antibody presents several technical challenges that require specific troubleshooting approaches:

  • Non-specific Binding Issues:

    • Problem: High background or false positives in Co-IP experiments

    • Solution: Use appropriate amounts of GNPAT antibody (0.5-4.0 μg for 1.0-3.0 mg of total protein lysate)

    • Solution: Increase stringency of wash buffers or include additional washing steps

    • Solution: Pre-clear lysates with protein A/G beads before adding GNPAT antibody

  • Low Yield of Precipitated Protein:

    • Problem: Weak or undetectable GNPAT band after IP

    • Solution: Optimize cell lysis conditions to ensure complete solubilization of peroxisomal proteins

    • Solution: Extend incubation time with antibody-resin complex to overnight at 4°C

    • Solution: Ensure antibody is properly cross-linked to resin to prevent antibody leaching

  • Detection of GNPAT-Interacting Proteins:

    • Problem: Difficulty detecting physiologically relevant interactions

    • Solution: Use gentler elution conditions to preserve protein-protein interactions

    • Solution: Consider chemical crosslinking of protein complexes prior to lysis

    • Solution: Use antibodies against specific modifications (e.g., acetyl-lysine antibody) to detect post-translationally modified GNPAT

  • Validation of Interactions:

    • Problem: Determining specificity of detected interactions

    • Solution: Include IgG control immunoprecipitations

    • Solution: Perform reciprocal Co-IPs where possible

    • Solution: Confirm interactions using alternative methods (e.g., proximity ligation assays)

  • Cell-Type Specific Considerations:

    • Problem: Variable results across cell types

    • Solution: Optimize lysis buffers for specific cell types (e.g., COLO 320 cells have been successfully used for GNPAT IP)

    • Solution: Adjust protein amounts based on GNPAT expression levels in your cell type of interest

By addressing these common challenges with the suggested methodological solutions, researchers can achieve more reliable and reproducible results when performing co-immunoprecipitation experiments with GNPAT antibody.

How can I design experiments to study the relationship between GNPAT acetylation and its function?

Designing experiments to investigate GNPAT acetylation and its functional consequences requires a systematic approach:

  • Detection of GNPAT Acetylation Status:

    • Immunoprecipitate GNPAT using specific antibody (e.g., ab75060, Abcam)

    • Cross-link antibody to resin for optimal results

    • Western blot using acetyl-lysine antibody (e.g., ab190479, Abcam)

    • Include appropriate controls (IgG immunoprecipitation)

  • Modulation of SIRT4 Activity:

    • Overexpress SIRT4 to enhance GNPAT deacetylation

    • Use SIRT4 siRNA or shRNA for knockdown studies

    • Apply SIRT activators or inhibitors and measure effects on GNPAT acetylation

    • Monitor SIRT4 expression levels using Western blot analysis

  • Environmental Stimuli Testing:

    • Expose cells to cigarette smoke extract (CSE) at varying concentrations

    • Measure relationship between CSE exposure, SIRT4 expression, and GNPAT acetylation

    • Include time-course studies to capture dynamic changes in acetylation status

  • Functional Readouts:

    • Assess GNPAT enzymatic activity in relation to acetylation status

    • Measure plasmalogen levels using mass spectrometry or other analytical techniques

    • Evaluate cellular phenotypes like ferroptosis markers:

      • Cell viability

      • LDH activity

      • ROS production

      • Mitochondrial morphology (using transmission electron microscopy)

      • Lipid peroxidation levels

  • Mutation Studies:

    • Generate acetylation mimetic (K→Q) or acetylation-deficient (K→R) mutants of GNPAT

    • Express these mutants in GNPAT-knockdown backgrounds

    • Compare their ability to rescue GNPAT-dependent phenotypes

    • This approach helps identify specific acetylation sites critical for function

  • Pathway Analysis:

    • Examine how GNPAT acetylation status affects downstream signaling pathways

    • Focus on ERK and Akt phosphorylation states that are known to be GNPAT-dependent

These methodological approaches provide a comprehensive experimental framework for investigating the functional significance of GNPAT acetylation and its regulation by SIRT4, with important implications for understanding cellular responses to stress and disease mechanisms.

How is GNPAT expression altered in neurodegenerative disease models and what are the implications?

GNPAT expression shows significant alterations in neurodegenerative disease models, particularly in Alzheimer's disease (AD):

  • Time-Dependent Changes in AD Models:

    • Studies in J20 mice (an AD model) have documented changes in GNPAT expression throughout disease progression

    • These changes correlate with alterations in plasmalogen levels in hippocampal brain tissues

    • Western blot analysis using GNPAT antibodies reveals expression patterns that change with disease progression

  • Cellular Localization Patterns:

    • Double immunofluorescence staining demonstrates GNPAT co-localization with:

      • Iba1+ microglial cells (indicating potential role in neuroinflammation)

      • GFAP+ astrocytes (suggesting involvement in astrocyte function)

    • This pattern of expression provides insights into cell type-specific roles of GNPAT in neurodegeneration

  • Functional Consequences:

    • GNPAT regulates ERK and Akt signaling pathways in neuronal cells

    • Knockdown of GNPAT significantly reduces phosphorylated ERK and Akt protein levels

    • These pathways are critical for neuronal survival and function

    • Disruption of these pathways may contribute to neurodegeneration

  • Plasmalogen-Dependent Signaling:

    • The products of GNPAT activity (plasmalogens) can rescue signaling defects in GNPAT-deficient cells

    • This suggests potential therapeutic approaches through plasmalogen supplementation

    • Unlike standard phospholipids (phosphatidylethanolamine), plasmalogens specifically activate key survival pathways

  • G-Protein Coupled Receptor Involvement:

    • GNPAT is required for GPCR-mediated increases in ERK phosphorylation

    • This links GNPAT function to receptor-mediated signaling in neurons

    • GPCR signaling defects in GNPAT-deficient contexts may contribute to neurodegenerative processes

These findings highlight the complex role of GNPAT in neurodegenerative diseases and suggest potential therapeutic strategies focused on restoring proper GNPAT function or compensating for its deficiency through plasmalogen supplementation.

What methodological approaches are most effective for studying GNPAT in ferroptosis-related diseases?

Studying GNPAT in ferroptosis-related diseases requires integrating multiple methodological approaches:

  • Cell Culture Models for Ferroptosis Induction:

    • A549 cells exposed to cigarette smoke extract (CSE) provide a reliable model

    • Titrate CSE concentrations to establish dose-dependent effects on ferroptosis markers

    • Monitor changes in GNPAT expression and acetylation status in response to CSE

  • GNPAT Manipulation Strategies:

    • Genetic knockdown using sh-GNPAT transfection

    • Verify knockdown efficiency through multiple techniques:

      • Quantitative real-time PCR

      • Western blot analysis using GNPAT antibody

      • Immunofluorescence staining

  • Comprehensive Ferroptosis Assessment:

    • Cell viability assays (MTT or similar)

    • LDH activity measurement to quantify cell damage

    • ROS production quantification

    • Cell apoptosis rate determination by flow cytometry

    • Transmission electron microscopy to visualize mitochondrial morphology

    • Lipid peroxidation assessment

  • Molecular Marker Analysis:

    • Monitor key ferroptosis regulators:

      • GPX4 (glutathione peroxidase 4)

      • GSH (glutathione) levels

      • MDA (malondialdehyde) as a lipid peroxidation indicator

  • SIRT4-GNPAT Axis Investigation:

    • Measure SIRT4 expression levels under disease conditions

    • Assess GNPAT acetylation status using immunoprecipitation followed by acetyl-lysine antibody detection

    • Correlate acetylation changes with ferroptosis progression

  • Rescue Experiments:

    • Test whether SIRT4 overexpression can reverse CSE-induced ferroptosis

    • Evaluate if plasmalogen supplementation mitigates ferroptotic cell death

    • Determine if GPX4 activation can compensate for GNPAT dysregulation

  • Translation to In Vivo Models:

    • Extend findings to appropriate animal models of ferroptosis-related diseases

    • Assess GNPAT expression and acetylation in affected tissues

    • Test therapeutic interventions targeting the SIRT4-GNPAT axis

These methodological approaches provide a comprehensive framework for investigating GNPAT's role in ferroptosis-related diseases and identifying potential therapeutic strategies.

How can GNPAT function be modulated for potential therapeutic applications?

GNPAT function can be modulated through several methodological approaches with therapeutic potential:

  • Genetic Modulation Strategies:

    • Knockdown/silencing: Using sh-GNPAT lentivirus vectors to reduce GNPAT expression in conditions where overactivation contributes to pathology

    • Overexpression: Introducing GNPAT expression vectors in conditions of deficiency

    • Gene editing: CRISPR/Cas9 approaches for precise modification of GNPAT expression or function

  • Post-Translational Regulation:

    • SIRT4 modulation: Since SIRT4 deacetylates GNPAT, targeting SIRT4 expression or activity can regulate GNPAT function

    • Small molecule SIRT4 activators could reduce GNPAT acetylation and potentially alter its activity in ferroptosis-related conditions

  • Substrate Availability Manipulation:

    • Provide glycerol-3-phosphate to enhance GNPAT activity

    • Modify fatty acid availability to affect the acyltransferase function of GNPAT

    • Target upstream pathways that generate GNPAT substrates

  • Plasmalogen Supplementation:

    • Direct supplementation with plasmalogens (500 ng/ml) can rescue defects in GNPAT-deficient systems

    • This approach bypasses the need for functional GNPAT and directly provides the end products

    • Particularly effective for restoring signaling pathways like ERK and Akt

  • Targeting Downstream Pathways:

    • For conditions where GNPAT dysfunction affects specific signaling pathways:

      • ERK/Akt pathway modulators could compensate for GNPAT deficiency

      • Ferroptosis inhibitors might mitigate effects of GNPAT dysregulation

  • Combination Approaches:

    • Simultaneously target GNPAT expression/function and provide plasmalogen supplementation

    • Combine SIRT4 modulation with downstream pathway interventions

    • Address both GNPAT and AGPS (the next enzyme in the plasmalogen synthesis pathway) for enhanced effects

These therapeutic modulation approaches must be tailored to specific disease contexts, as GNPAT may require upregulation in some conditions (e.g., neurodegenerative diseases where plasmalogen deficiency is problematic) but downregulation in others (e.g., certain contexts of ferroptosis where excessive GNPAT activity may be detrimental).

What experimental designs are most appropriate for studying GNPAT in peroxisomal disorders?

For comprehensive investigation of GNPAT in peroxisomal disorders, consider these methodological approaches:

  • Patient-Derived Cell Models:

    • Establish fibroblast cultures from patients with peroxisomal disorders

    • Create induced pluripotent stem cells (iPSCs) and differentiate to relevant cell types

    • Compare GNPAT expression, localization, and function between patient and control cells using antibodies like 14931-1-AP

  • Comprehensive Protein Profiling:

    • Analyze GNPAT in relation to other peroxisomal proteins:

      • FAR-1 (involved in plasmalogen synthesis)

      • AGPS (works with GNPAT in plasmalogen synthesis)

      • PEX proteins (peroxisomal biogenesis factors)

    • Use Western blot analysis with appropriate antibodies to establish expression patterns

  • Enzyme Activity Assays:

    • Measure GNPAT enzymatic activity directly using radiolabeled substrates

    • Correlate activity levels with protein expression and modification status

    • Compare activity across disease models and control conditions

  • Ultrastructural Analysis:

    • Transmission electron microscopy to examine peroxisome morphology

    • Assess changes in peroxisome number, size, and membrane integrity

    • Correlate peroxisomal structural changes with GNPAT expression/function

  • Functional Rescue Experiments:

    • Express wild-type GNPAT in deficient cells

    • Supplement with plasmalogens to bypass metabolic blocks

    • Assess recovery of peroxisomal functions and cellular phenotypes

  • Genetic Interaction Studies:

    • Perform double knockdown experiments with GNPAT and other peroxisomal genes

    • Assess synthetic lethality or rescue effects

    • This approach helps establish functional relationships between peroxisomal proteins

  • Metabolomic Profiling:

    • Measure plasmalogen levels and other peroxisome-derived lipids

    • Correlate metabolite changes with GNPAT expression/function

    • Use time-dependent analysis to track dynamic changes in metabolite profiles

  • In Vivo Modeling:

    • Study GNPAT in animal models of peroxisomal disorders

    • Use tissue-specific conditional knockout approaches

    • Assess developmental and age-related changes in GNPAT function

These experimental designs provide a comprehensive framework for investigating GNPAT's role in peroxisomal disorders, potentially leading to novel diagnostic approaches and therapeutic interventions.

How can GNPAT regulation be integrated into broader understanding of metabolic diseases?

Integrating GNPAT regulation into the broader understanding of metabolic diseases requires multifaceted experimental approaches:

  • Multi-Omics Integration:

    • Combine proteomic analysis of GNPAT expression and post-translational modifications

    • Correlate with metabolomic profiling of plasmalogens and related lipids

    • Integrate transcriptomic data to understand regulatory networks

    • This comprehensive approach reveals how GNPAT functions within global metabolic pathways

  • Tissue-Specific Analysis:

    • Compare GNPAT regulation across metabolically active tissues:

      • Liver (major site of lipid metabolism)

      • Brain (high plasmalogen content)

      • Adipose tissue (energy storage)

      • Muscle (energy utilization)

    • Use tissue-specific antibody validation data to ensure accurate detection

  • Signal Transduction Network Analysis:

    • Map GNPAT-dependent regulation of ERK and Akt signaling pathways

    • Investigate how these pathways intersect with insulin signaling and energy metabolism

    • Determine how GNPAT knockdown affects these networks across different metabolic states

  • Metabolic Stress Response Studies:

    • Examine GNPAT regulation under various metabolic challenges:

      • Oxidative stress

      • Nutrient deprivation

      • Inflammatory stimuli

      • Environmental toxins (e.g., cigarette smoke extract)

    • Monitor both expression and post-translational modifications (especially acetylation)

  • SIRT4-GNPAT Axis in Metabolic Regulation:

    • Investigate how the SIRT4-mediated deacetylation of GNPAT responds to metabolic signals

    • Determine if this regulation is altered in metabolic diseases

    • Explore whether NAD+ availability (which affects sirtuin activity) influences GNPAT function

  • Therapeutic Intervention Testing:

    • Assess how established metabolic disease treatments affect GNPAT expression and function

    • Test whether GNPAT modulation can enhance the efficacy of existing therapies

    • Develop GNPAT-targeted approaches as standalone or combination therapies

  • Longitudinal Disease Progression Studies:

    • Track GNPAT expression, modification, and function across disease development

    • Correlate changes with clinical parameters and biomarkers

    • Identify critical windows for intervention targeting GNPAT-dependent processes

This integrated approach positions GNPAT not just as a peroxisomal enzyme but as a key node in complex metabolic networks, with important implications for understanding and treating metabolic diseases ranging from diabetes to neurodegenerative disorders.

What are the key considerations for successful GNPAT antibody-based research?

Successful GNPAT antibody-based research requires attention to several critical methodological considerations:

  • Antibody Selection and Validation: Choose antibodies with proven specificity and performance in your application of interest. The search results highlight multiple validated antibodies including Proteintech 14931-1-AP and Abcam ab75060, each with documented performance across various applications . Always verify antibody specificity using positive and negative controls.

  • Experimental Design Rigor: Include appropriate controls in all experiments, particularly for knockdown/knockout studies where proper validation through multiple techniques (qPCR, Western blot, immunofluorescence) is essential . Design experiments that clearly distinguish between direct GNPAT effects and secondary consequences.

  • Technical Optimization: For each application, optimize conditions including antibody dilution (1:500-1:1000 for WB; 0.5-4.0 μg for IP), incubation times, and detection methods . Consider that GNPAT's observed molecular weight (65-69 kDa) differs from its calculated weight (77 kDa) .

  • Contextual Interpretation: Recognize that GNPAT functions within complex networks involving other peroxisomal proteins, signaling pathways, and metabolic processes. Interpret results within this broader context, considering how GNPAT affects and is affected by these networks .

  • Translational Relevance: Connect basic mechanistic findings to disease relevance, particularly in areas where GNPAT dysfunction contributes to pathology, such as neurodegenerative diseases, ferroptosis-related conditions, and peroxisomal disorders .

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 2025 TheBiotek. All Rights Reserved.