APOA5 Human, HEK

Apolipoprotein A-V Human Recombinant, HEK
Shipped with Ice Packs
In Stock

Description

Functional Role in Lipid Metabolism

APOA5 regulates plasma triglyceride (TG) levels through multiple mechanisms:

Triglyceride Regulation

  • ANGPTL3/8 Suppression: APOA5 binds to the angiopoietin-like protein 3/8 (ANGPTL3/8) complex, inhibiting its ability to suppress lipoprotein lipase (LPL) activity . This interaction is critical for hydrolyzing TG-rich lipoproteins (TRLs).

  • LPL Activation: APOA5 enhances LPL-mediated TG catabolism by stabilizing LPL on endothelial surfaces, particularly in heart and adipose tissue .

Production in HEK293 Cells

HEK293 cells are preferred for recombinant APOA5 production due to their high transfection efficiency and scalability. Key features include:

ParameterDetailSource
Expression SystemTransient or stable transfection with APOA5 cDNA
TagFlag-tag (DDDK) for purification
Yield~0.5 mg/mL in culture supernatant
PurificationHeparin-Sepharose chromatography, followed by Flag-tag affinity purification

The recombinant protein retains functional activity, as demonstrated by its ability to suppress ANGPTL3/8 and activate LPL in vitro .

Hypertriglyceridemia Models

  • APOA5 Knockout Mice: APOA5 deficiency results in a 4-fold increase in plasma TG levels due to delayed TRL processing and reduced LPL activity .

  • Postheparin LPL Activity: In APOA5-deficient mice, LPL mass and activity in postheparin plasma are reduced by ~34–37%, correlating with impaired TG clearance .

Mechanistic Studies

  • GPIHBP1 Interaction: APOA5 stabilizes LPL on GPIHBP1, a endothelial receptor that anchors LPL to capillaries. Deficiency reduces LPL/GPIHBP1 ratios by ~40% in heart and brown adipose tissue .

  • ANGPTL3/8 Binding: Truncation of APOA5’s C-terminal helix (e.g., APOA5Δ40) abolishes ANGPTL3/8 binding, highlighting its role in regulating LPL activity .

Clinical Relevance

  • Hypertriglyceridemia: Mutations in APOA5 are linked to familial hypertriglyceridemia and hyperlipoproteinemia type V .

  • Cardiovascular Risk: Low plasma APOA5 levels correlate with coronary artery disease, as it modulates VLDL-TG production and LPL activity .

Future Directions

  • Therapeutic Applications: Recombinant APOA5 may serve as a therapeutic agent to lower TG levels in hypertriglyceridemia.

  • Structural Studies: Resolving APOA5’s crystal structure could elucidate its interaction with ANGPTL3/8 and LPL, aiding drug design .

Product Specs

Introduction
ApoA5 plays a crucial role in regulating triglyceride levels in the blood. Studies using mouse models have demonstrated that the absence of ApoA5 leads to a significant increase in serum triglycerides, while the introduction of human ApoA5 results in a substantial decrease. Additionally, medications used to lower triglyceride levels have been shown to increase ApoA5 expression. However, our understanding of human serum ApoA5 protein remains limited. Recent studies have confirmed its presence in human serum, detectable by specific antibodies, although at lower concentrations compared to other apolipoproteins.
Description
This product consists of the human APOA5 protein with a Flag-tag fused to it. The resulting recombinant protein has a molecular weight of 40.1 kDa and comprises 354 amino acids from the APOA5 protein and an additional 11 amino acids from the Flag-tag.
Physical Appearance

The product appears as a white powder that has been freeze-dried and filtered.

Formulation
The APOA5 human protein was purified through filtration (0.4µm) and subsequently lyophilized. The lyophilized product is provided in a solution containing 20mM TRIS and 50mM NaCl, pH 7.5, at a concentration of 0.5 mg/ml.
Solubility
To prepare a working solution, add deionized water to the lyophilized pellet to achieve a concentration of about 0.5mg/ml, ensuring the pellet is completely dissolved. Note that this product is not sterile. Prior to use in cell culture, it is essential to filter the solution using a sterile filter of appropriate pore size.
Stability
For long-term storage, store the lyophilized APOA5 human protein at -20°C. Once reconstituted, divide the product into smaller portions (aliquots) to minimize repeated freezing and thawing. The reconstituted APOA5 human protein can be stored at 4°C for a short period, showing no significant changes for at least two weeks.
Purity
Analysis by SDS-PAGE confirms that the purity of this product is greater than 70%.
Synonyms
Apolipoprotein A-V, Apo-AV, ApoA-V, Apolipoprotein A5, Regeneration-associated protein 3, APOA5, RAP3, APOAV.
Source
HEK293
Amino Acid Sequence
RKGFWDYFSQ TSGDKGRVEQ IHQQKMAREP ATLKDSLEQD LNNMNKFLEK LRPLSGSEAP RLPQDPVGMR RQLQEELEEV KARLQPYMAE AHELVGWNLE GLRQQLKPYT MDLMEQVALR VQELQEQLRV VGEDTKAQLL GGVDEAWALL QGLQSRVVHH TGRFKELFHP YAESLVSGIG RHVQELHRSV APHAPASPAR LSRCVQVLSR KLTLKAKALH ARIQQNLDQL REELSRAFAG TGTEEGAGPD PQMLSEEVRQ RLQAFRQDTY LQIAAFTRAI DQETEEVQQQ LAPPPPGHSA FAPEFQQTDS GKVLSKLQAR LDDLWEDITH SLHDQGHSHL GDPAAADYKD DDDK

Q&A

What is APOA5 and what is its physiological significance?

APOA5 is an apolipoprotein localized in the APOA4/APOC3/APOA1 gene cluster on human chromosome 11q23 . It plays a critical role in modulating triacylglycerol (TG) levels in humans and animal models, with transgenic mice expressing human APOA5 showing decreased plasma TG levels while APOA5 knock-out mice display increased plasma TG levels . The protein is present in low amounts in plasma yet has a significant impact on lipoprotein metabolism . Genetic variations in the human APOA5 locus correlate with changes in plasma lipoprotein levels, and certain polymorphisms are significantly associated with increased risk for metabolic syndrome . More dramatically, mutations in the APOA5 gene that result in truncated proteins lacking lipid-binding domains have been demonstrated to cause severe hyperlipidemia in homozygous patients . Despite its established importance in lipid metabolism, the precise mechanism whereby APOA5 exerts its effect on plasma TG levels remains incompletely understood, making it an important subject for ongoing research.

Why are HEK-293 cells commonly used for APOA5 expression studies?

HEK-293 cells represent an ideal expression system for APOA5 research due to several advantageous characteristics that facilitate both basic and translational investigations. These cells provide an efficient mammalian expression system with proper post-translational modifications necessary for functional studies of human proteins . They exhibit high transfection efficiency and protein expression levels, making them suitable for recombinant protein production, as evidenced by their use in manufacturing APOA5 Human Recombinant Flag-Tagged Fusion Protein . HEK-293 cells are particularly valuable for studies involving receptor interactions and trafficking, as demonstrated in research examining APOA5 interactions with receptors such as sortilin and SorLA . The cellular machinery in HEK-293 cells supports proper folding and secretion of lipoproteins, allowing researchers to study APOA5 in a physiologically relevant context . Furthermore, these cells permit the investigation of intracellular trafficking and degradation of APOA5, as shown in studies using 125I-labeled APOA5 to quantify internalization processes . These attributes collectively make HEK-293 cells a preferred system for studying various aspects of APOA5 biology and function.

What are the typical characteristics of recombinant APOA5 produced in HEK cells?

Recombinant APOA5 produced in HEK cells typically manifests as a protein of approximately 40.1 kDa, containing 354 amino acid residues of the human APOA5 sequence . When expressed as a tagged fusion protein, it may include additional amino acid residues such as the 11 residues of a FLAG tag that facilitate purification and detection in experimental systems . The protein produced in HEK-293 cells undergoes proper folding and post-translational modifications that are essential for its biological function, particularly its ability to bind lipids and interact with receptors . In experimental settings, researchers often incorporate APOA5 into dimyristoylphosphatidylcholine (DMPC) disks to mimic its physiological lipid-associated state, which is critical for studying its interactions with cellular receptors like sortilin and SorLA . For visualization in cellular assays, APOA5 can be conjugated with fluorescent labels such as Alexa Fluor 488, enabling researchers to track its binding, internalization, and trafficking in live cells . The recombinant protein retains its ability to interact with members of the low-density lipoprotein receptor family, making it suitable for mechanistic studies investigating APOA5's role in lipid metabolism . These characteristics make HEK-expressed APOA5 a valuable tool for investigating this protein's structure-function relationships and cellular interactions.

How can researchers effectively express and purify APOA5 in HEK cell systems?

The expression and purification of APOA5 in HEK cell systems requires a carefully optimized protocol to ensure high yield and biological activity of the target protein. Begin by selecting an appropriate expression vector containing a strong promoter (such as CMV) and incorporating a purification tag (commonly FLAG or His6) to facilitate downstream isolation . Transfect HEK-293 cells using either calcium phosphate precipitation or lipid-based transfection reagents, with optimization of DNA:transfection reagent ratios being critical for maximizing expression efficiency . For stable expression, select transfected cells using appropriate antibiotics and validate expression through Western blotting or ELISA before scaling up production . The purification process typically involves affinity chromatography using the incorporated tag, with FLAG-tagged APOA5 being purified on anti-FLAG columns and His-tagged proteins on metal affinity resins . Consider incorporating a protease cleavage site between the tag and APOA5 sequence if tag removal is desired for downstream applications . For studies requiring lipid-associated APOA5, reconstitute the purified protein with dimyristoylphosphatidylcholine (DMPC) to form protein-lipid disks that better mimic the physiological state of the protein . Finally, verify the purity and integrity of the purified protein using SDS-PAGE, Western blotting, and mass spectrometry, and confirm its biological activity through functional assays such as receptor binding studies .

What are the most effective methods for studying APOA5 interactions with cellular receptors?

Investigating APOA5 interactions with cellular receptors requires multiple complementary methodologies to establish binding parameters and functional outcomes. Surface plasmon resonance (SPR) represents a powerful technique for quantifying binding kinetics and affinities, as demonstrated in studies where sortilin and other receptors were immobilized on CM5 sensor chips to measure direct interactions with APOA5 . Cellular binding and internalization assays using fluorescently labeled APOA5 (e.g., Alexa Fluor 488-conjugated APOA5) provide valuable insights into the spatial and temporal dynamics of receptor-mediated endocytosis . For quantitative assessment of internalization and degradation pathways, researchers can employ 125I-labeled APOA5 followed by acid precipitation of media to distinguish between intact protein and degraded fragments . Fluorescence resonance energy transfer (FRET) offers a sophisticated approach for confirming direct molecular interactions, as evidenced by studies measuring energy transfer between labeled APOA5 and receptors like sortilin and SorLA . The sensitized acceptor emission PFRET procedure with subsequent calculation of apparent FRET efficiencies (Eapp) can determine the proximity of interacting proteins at the nanometer scale . Complementary approaches should include co-immunoprecipitation studies and competition assays with known ligands to establish binding specificity . For functional validation, researchers should perform studies with receptor variants harboring mutations in their internalization motifs (such as the tyrosine and dileucine motifs in sortilin) to determine the mechanistic basis of APOA5-receptor interactions .

How can researchers effectively study APOA5 polymorphisms and their functional implications?

The study of APOA5 polymorphisms requires a systematic approach integrating genetic, biochemical, and clinical methodologies to elucidate their functional significance. Begin with comprehensive genetic screening through sequencing or genotyping methods to identify polymorphisms of interest, as exemplified in studies examining 16 APOA5 polymorphisms in the CARDIA cohort . Employ statistical analyses to assess associations between identified polymorphisms and phenotypic characteristics such as plasma triglyceride levels, calculating the percentage of triglyceride variation attributable to specific single-nucleotide polymorphisms (SNPs) . For functional characterization, express wild-type and variant APOA5 proteins in HEK-293 cells to compare expression levels, secretion efficiency, and post-translational modifications . Lipid association studies should be conducted to determine whether polymorphisms affect the protein's ability to bind lipids, which is crucial for its physiological function . Receptor interaction assays using techniques like surface plasmon resonance and cellular internalization studies can reveal whether polymorphisms alter APOA5's ability to engage with its physiological receptors . For population-level analyses, stratify results by demographic factors such as age, gender, and ethnicity, as research has shown differential associations of APOA5 polymorphisms with triglyceride levels across different populations . Finally, conduct longitudinal studies when possible to determine whether the influence of APOA5 polymorphisms changes with age or under different physiological conditions, building on observations that polymorphisms affect young adults (18-30 years) even in the absence of disease .

What experimental controls are critical when studying APOA5-receptor interactions in HEK cell systems?

Implementing rigorous controls is essential for ensuring the validity and specificity of observed APOA5-receptor interactions in HEK cell systems. Include untransfected HEK-293 cells as negative controls to establish baseline levels of non-specific binding and internalization, as these cells have been shown to exhibit significantly lower degradation of 125I-labeled APOA5 compared to cells transfected with receptors like SorLA or sortilin . Employ receptor variants with mutations in key internalization motifs (such as the altered tyrosine and dileucine motifs in sortilin or the mutated DD, EDDED cluster in SorLA) to confirm the specificity of receptor-mediated endocytosis, as these mutations should significantly reduce APOA5 internalization . For FRET experiments measuring direct molecular interactions, prepare appropriate donor-only and acceptor-only samples to enable proper correction of spectral bleed-through using standardized algorithms like the PFRET method . Incorporate negative control proteins that are structurally similar but functionally distinct from APOA5, such as apoC-II labeled with the same fluorescent tags (apoC-II488), which has been demonstrated not to interact with the cells under investigation . When studying lipid-associated APOA5, include control dimyristoylphosphatidylcholine (DMPC) liposomes without protein but processed through the same labeling protocol (DMPC488) to rule out non-specific effects of the lipid component . For competition assays, use known ligands of the receptors under study (such as RAP for LRP1) at varying concentrations to confirm binding specificity and establish whether APOA5 interacts with the same or different binding sites .

How should researchers address data inconsistencies in APOA5 genetic association studies?

Addressing inconsistencies in APOA5 genetic association studies requires a multi-faceted approach that considers methodological variations, population characteristics, and statistical analysis techniques. Begin by critically evaluating sample size and power calculations, as insufficiently powered studies may yield false negative results, particularly for polymorphisms with modest effect sizes like those observed in APOA5 research where individual SNPs account for only 0-2.46% of triglyceride variation . Consider stratification by demographic factors including age, sex, and ethnicity, as APOA5 polymorphisms have shown context-dependent influences; for example, significant associations were observed in white females and males but not consistently in African American males in the CARDIA study . Examine covariates and confounding factors such as diet, physical activity, and concurrent medications, which may mask or amplify genetic associations if not properly controlled . Harmonize phenotype definitions and measurement methods across studies, ensuring that triglyceride measurements follow standardized protocols with appropriate fasting conditions . Implement rigorous quality control for genotyping, including Hardy-Weinberg equilibrium testing and assessment of linkage disequilibrium patterns, which may explain apparent discrepancies if different marker SNPs are in variable linkage with functional variants . Employ appropriate statistical methodologies including correction for multiple testing and consideration of gene-environment interactions, which may account for differential associations observed across populations . Finally, conduct meta-analyses or systematic reviews when available data permit, as these approaches can provide more robust evidence of genetic associations by increasing effective sample size and mitigating the influence of individual study biases .

What technical challenges arise when using fluorescence-based approaches to study APOA5 trafficking, and how can they be overcome?

Fluorescence-based approaches for studying APOA5 trafficking present several technical challenges that require careful methodological considerations to overcome. Photobleaching represents a significant issue, particularly during extended live-cell imaging experiments tracking APOA5 internalization; researchers should minimize light exposure by reducing excitation intensity, using antifade reagents, and employing sensitive detectors that allow lower excitation power . Background autofluorescence from HEK cells and culture media can obscure specific signals, necessitating proper background subtraction, the use of fluorophores with emission spectra distinct from cellular autofluorescence, and implementation of spectral unmixing algorithms when appropriate . For FRET experiments measuring direct APOA5-receptor interactions, spectral bleed-through can lead to false-positive results; this should be addressed by acquiring proper donor-only and acceptor-only control images and implementing correction algorithms such as those in the PFRET ImageJ plugin . Signal quantification challenges arise when defining regions of interest (ROIs), particularly for membrane-localized interactions; researchers should establish consistent criteria for ROI selection, such as minimum signal thresholds (e.g., average signal of at least 25) and fulfillment of lower bound settings for at least 80% of pixels within an ROI . Signal saturation in bright cellular regions can lead to data loss or inaccurate measurements; researchers should carefully set upper bounds in image acquisition (e.g., one unit lower than saturated intensity) and employ high dynamic range imaging when necessary . For co-localization studies, optical aberrations and the diffraction limit of light microscopy may lead to false co-localization; super-resolution microscopy techniques or proximity ligation assays can provide more accurate assessment of protein interactions at the nanometer scale .

How can APOA5-HEK expression systems contribute to understanding metabolic disorders?

APOA5-HEK expression systems offer powerful platforms for elucidating the molecular mechanisms underlying metabolic disorders, particularly those involving dysregulated lipid metabolism. These systems enable detailed investigation of APOA5 mutations associated with severe hyperlipidemia, such as those resulting in truncated proteins lacking lipid-binding domains, providing insights into structure-function relationships critical for normal lipid homeostasis . By expressing APOA5 variants identified in patient populations within HEK cells, researchers can systematically evaluate how specific genetic alterations affect protein folding, secretion, lipid binding, and receptor interactions, thus establishing mechanistic links between genotype and phenotype in disorders such as hypertriglyceridemia . The ability to study APOA5 interactions with members of the low-density lipoprotein receptor family in HEK cell systems reveals potential therapeutic targets, as these receptors mediate the endocytosis and degradation of APOA5-containing lipoprotein particles . Metabolic labeling studies using HEK cells expressing APOA5 can trace the intracellular trafficking and degradation pathways of the protein, providing insights into how these processes may be dysregulated in disease states . The quantitative nature of HEK cell-based assays allows researchers to determine the relative impact of different APOA5 polymorphisms on protein function, corresponding to their clinical associations with plasma triglyceride levels and metabolic syndrome risk . Furthermore, these expression systems can be adapted for high-throughput screening of compounds that modulate APOA5 function or stability, potentially identifying novel therapeutic approaches for disorders characterized by elevated triglycerides .

What are the emerging methodologies for studying APOA5 structure-function relationships?

The field of APOA5 research is being transformed by emerging methodologies that provide unprecedented insights into structure-function relationships of this important apolipoprotein. Cryo-electron microscopy (cryo-EM) offers a revolutionary approach for visualizing APOA5 in its lipid-associated state at near-atomic resolution, revealing the conformational changes that occur upon lipid binding and receptor interaction . Advanced molecular dynamics simulations complement experimental approaches by predicting how APOA5 polymorphisms might alter protein flexibility, lipid binding capacity, and interaction with receptor binding domains . CRISPR-Cas9 genome editing in HEK cells enables precise modification of endogenous APOA5 or receptor genes, allowing researchers to study protein variants in their native genomic context rather than through overexpression systems . Single-molecule tracking techniques using quantum dots or photoactivatable fluorophores provide detailed information about the dynamics of APOA5-receptor interactions at the cell surface and during endocytic trafficking, revealing kinetic parameters not accessible through bulk measurements . Proximity-dependent labeling methods such as BioID or APEX2 can identify novel interaction partners of APOA5 in living cells, potentially uncovering previously unknown functions or regulatory mechanisms . Mass spectrometry-based approaches including hydrogen-deuterium exchange mass spectrometry (HDX-MS) and crosslinking mass spectrometry (XL-MS) provide structural information about APOA5 in solution and in complex with its binding partners, helping to map interaction interfaces at the amino acid level . Integration of these methodologies with traditional biochemical and cellular approaches will provide a comprehensive understanding of how APOA5 structure determines its function in lipid metabolism and how alterations in this structure contribute to metabolic disorders .

How can APOA5 polymorphism studies inform personalized medicine approaches for dyslipidemia?

APOA5 polymorphism studies hold significant potential for advancing personalized medicine approaches in the management of dyslipidemia by enabling risk stratification, treatment selection, and preventive interventions tailored to individual genetic profiles. Population-based studies have demonstrated that specific APOA5 polymorphisms (such as -3A/G, -1131T/C, and S19W) are associated with variations in plasma triglyceride levels, with some markers showing consistent associations across different demographic groups while others display context-dependent effects . These genetic markers can be incorporated into risk prediction models that help identify individuals at increased risk for hypertriglyceridemia even at a young age and in the absence of overt disease, as evidenced by significant associations observed in young adults (18-30 years) in the CARDIA study . The differential impact of APOA5 polymorphisms across ethnic groups—with some markers showing stronger associations in white individuals than in African Americans—highlights the importance of developing population-specific genetic screening panels for dyslipidemia risk assessment . Functional characterization of APOA5 variants in HEK cell systems can reveal the molecular mechanisms underlying their clinical effects, allowing for more precise classification of variants as pathogenic, likely pathogenic, or benign . These mechanistic insights can guide the development of targeted therapies that address specific defects in APOA5 function, such as impaired receptor binding or altered lipid association . Pharmacogenetic studies can determine whether APOA5 polymorphisms predict differential responses to lipid-lowering therapies, potentially enabling physicians to select the most effective treatment based on a patient's genetic profile . Finally, by identifying APOA5 variants that influence triglyceride levels independently of traditional risk factors, these studies help define patient subgroups that may benefit from more aggressive lipid management or novel therapeutic approaches targeting APOA5-mediated pathways .

Product Science Overview

Introduction

Apolipoprotein A-V (ApoA-V) is a protein that plays a crucial role in lipid metabolism, particularly in the regulation of plasma triglyceride levels. Elevated triglyceride levels are a significant risk factor for cardiovascular diseases, making ApoA-V an important target for research and therapeutic interventions. The recombinant form of this protein, expressed in Human Embryonic Kidney (HEK) cells, is widely used in scientific studies to understand its function and potential applications.

Structure and Expression

Apolipoprotein A-V is a 40.1 kDa protein consisting of 354 amino acid residues . The recombinant form is often tagged with a flag tag for ease of purification and detection. The protein is expressed in HEK 293 cells, a human cell line that provides a suitable environment for proper protein folding and post-translational modifications .

Function

ApoA-V is primarily involved in the regulation of plasma triglyceride levels. It acts by enhancing the hydrolysis of triglycerides in very low-density lipoproteins (VLDL) and chylomicrons, facilitating their clearance from the bloodstream. This function is critical in maintaining lipid homeostasis and preventing hypertriglyceridemia, a condition associated with an increased risk of cardiovascular diseases .

Clinical Significance

Mutations in the APOA5 gene, which encodes ApoA-V, have been linked to various lipid disorders. For instance, certain polymorphisms are associated with hypertriglyceridemia and an increased risk of coronary artery disease. Understanding the role of ApoA-V in lipid metabolism can lead to the development of novel therapeutic strategies for managing dyslipidemia and related cardiovascular conditions .

Applications

The recombinant form of ApoA-V is used in various research applications, including:

  • Western Blotting (WB): To detect and quantify the protein in different samples.
  • Enzyme-Linked Immunosorbent Assay (ELISA): For measuring the protein’s concentration in biological fluids.
  • Sodium Dodecyl Sulfate Polyacrylamide Gel Electrophoresis (SDS-PAGE): To analyze the protein’s purity and molecular weight .
Stability and Storage

Recombinant ApoA-V is typically lyophilized and can be reconstituted in deionized water to prepare a working stock solution. It is recommended to store the lyophilized protein at -20°C and avoid repeated freeze-thaw cycles to maintain its stability. Once reconstituted, the protein can be stored at 4°C for a limited period without significant degradation .

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.