ANGPTL3 (17-460) Human

Angiopoietin-like Protein 3 (17-460 a.a.) Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to ANGPTL3 (17-460) Human

ANGPTL3 (Angiopoietin-like Protein 3), spanning amino acids 17–460, is a 453-amino acid hepatokine primarily secreted by the liver. It regulates plasma lipid metabolism by modulating lipoprotein lipase (LPL) and endothelial lipase (EL) activity, thereby influencing triglyceride (TG) and cholesterol homeostasis . The protein’s structure includes an N-terminal domain (17–207) critical for LPL inhibition and a C-terminal fibrinogen-like domain (207–460) with potential roles in protein interactions .

Biological Role in Lipid Metabolism

ANGPTL3 suppresses LPL in oxidative tissues (e.g., muscle), directing TG-rich lipoproteins to white adipose tissue (WAT) for storage. This regulation balances energy availability during feeding and fasting states . Key mechanisms include:

  1. LPL Inhibition: Reduces TG hydrolysis in oxidative tissues, lowering free fatty acid (FFA) release .

  2. VLDL and LDL Regulation: Lowers circulating VLDL and LDL cholesterol by inhibiting LPL-mediated TG breakdown .

  3. HDL Modulation: ANGPTL3 downregulation correlates with reduced HDL levels, particularly in loss-of-function mutation carriers .

Genetic Variants and Lipid Phenotypes

Loss-of-function (LOF) mutations in ANGPTL3 (e.g., S17X, E129X) are associated with marked hypolipidemia. Below are key findings from human studies:

Table 1: Lipid Profiles in ANGPTL3 Mutation Carriers

ParameterNoncarriersHeterozygotesHomozygotes
Total Cholesterol (mg/dL)187.5 ± 25.5167.2 ± 31.682.4 ± 12.1
Triglycerides (mg/dL)80.8 ± 39.174.3 ± 40.632.4 ± 4.1
LDL Cholesterol (mg/dL)102.2 ± 24.797.3 ± 28.148.3 ± 15.3
HDL Cholesterol (mg/dL)69.6 ± 15.155.4 ± 13.727.8 ± 8.1

Data from carriers of ANGPTL3 S17X mutation .

Key Observations:

  • Homozygotes: Near-absent ANGPTL3 levels, 50–60% reduction in LDL-C and TGs, and 60% lower HDL-C .

  • Threshold Effects: ANGPTL3 levels <60 ng/dL correlate strongly with lipid reductions, suggesting dose-dependent effects .

Intracellular and Thyroid Hormone Regulation

ANGPTL3 downregulation in hepatocytes increases intracellular TGs by:

  1. Reducing Beta-Oxidation: Lower expression of DIO1 (thyroid hormone T4 to T3 converter) impairs mitochondrial fatty acid metabolism .

  2. Disrupting Lipid Droplet Remodeling: Alters peroxisomal beta-oxidation and lipid storage dynamics .

Thyroid hormone (T3) supplementation rescues this phenotype, indicating ANGPTL3’s role in maintaining hepatic T3 levels and metabolic activity .

Cardiovascular Risk and HDL Function

  • HDL Cholesterol Uptake Capacity (CUC): Polymorphisms like rs1748195 correlate with reduced CUC, linking ANGPTL3 to HDL functionality .

  • ASCVD Risk: Rare ANGPTL3 LOF mutations are associated with lower ASCVD risk, though this requires confirmation in larger cohorts .

Therapeutic Strategies

ApproachMechanismOutcome
Antisense OligonucleotidesReduce hepatic ANGPTL3 expressionLower circulating ANGPTL3 and lipids
Monoclonal AntibodiesNeutralize circulating ANGPTL3Limited impact on liver TGs; effective for plasma lipid reduction

Challenges: Intracellular vs. extracellular inhibition yields divergent effects (e.g., liver vs. plasma lipid changes) .

Genetic and Lipidomic Associations

ANGPTL3 variants influence specific lipid species:

  • ApoE and ApoA-II: Correlate with ANGPTL3 levels, reflecting shared regulatory pathways .

  • Lipidome Profiling: Rare ANGPTL3 variants in the San Antonio Family Heart Study linked to altered phospholipids and sphingolipids .

Product Specs

Introduction
ANGPTL3 and ANGPTL4, primarily produced by the liver, are angiopoietin-like proteins that play a crucial role in regulating triglyceride metabolism. They achieve this by inhibiting the breakdown of triglyceride-rich lipoproteins. Experimental evidence suggests that Angptl3 and Angptl4 differentially inhibit Lipoprotein lipase (LPL) to control circulating triglyceride levels during different nutritional states, such as feeding and fasting. ANGPTL3's molecular structure resembles angiopoietins, which are vascular endothelial growth factors. Studies using deletion mutants of human Angiopoietin 5 have shown that the N-terminal domain (fragment 17-207) is responsible for increasing plasma triglyceride levels in mice, while the C-terminal fibrinogen-like domain (fragment 207-460) is not.
Description
ANGPTL3, produced in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain with 453 amino acids (17-460 a.a.). It has a molecular mass of 52.9kDa and appears between 25-70kDa on SDS-PAGE under reducing conditions. This protein is expressed with a 6 amino acid His tag at its C-Terminus and purified using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The ANGPTL3 protein solution is provided at a concentration of 0.25mg/ml and contains Buffered Saline (pH 7.4), 30% glycerol, and 1mM DTT.
Stability
For short-term storage (up to 4 weeks), the ANGPTL3 solution can be stored at 4°C. For longer periods, it should be stored frozen at -20°C. To ensure optimal stability during long-term storage, adding a carrier protein (0.1% HSA or BSA) is recommended. Repeated freezing and thawing of the solution should be avoided.
Purity
The purity of ANGPTL3 is greater than 90%, as determined by SDS-PAGE analysis.
Synonyms
Angiopoietin Like 3, Angiopoietin 5, ANGPT5, ANG-5, Angiopoietin-Related Protein 3 , Angiopoietin-Like Protein 3, Angiopoietin-Like 3, Angiopoietin-5, FHBL2, ANL3.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
ADPSRIDQDN SSFDSLSPEP KSRFAMLDDV KILANGLLQL GHGLKDFVHK TKGQINDIFQ KLNIFDQSFY DLSLQTSEIK EEEKELRRTT YKLQVKNEEV KNMSLELNSK LESLLEEKIL LQQKVKYLEE QLTNLIQNQP ETPEHPEVTS LKTFVEKQDN SIKDLLQTVE DQYKQLNQQH SQIKEIENQL RRTSIQEPTE ISLSSKPRAP RTTPFLQLNE IRNVKHDGIP AECTTIYNRG EHTSGMYAIR PSNSQVFHVY CDVISGSPWT LIQHRIDGSQ NFNETWENYK YGFGRLDGEF WLGLEKIYSI VKQSNYVLRI ELEDWKDNKH YIEYSFYLGN HETNYTLHLV AITGNVPNAI PENKDLVFST WDHKAKGHFN CPEGYSGGWW WHDECGENNL NGKYNKPRAK SKPERRRGLS WKSQNGRLYS IKSTKMLIHP TDSESFEHHH HHH.

Q&A

What is the structural organization of ANGPTL3 and how does this relate to the (17-460) fragment?

ANGPTL3 contains an N-terminal coiled-coil domain and a C-terminal fibrinogen-like domain (FLD). The N-terminal domain includes the specific epitope 1 (SE1) region known to inhibit LPL enzymatic activity. ANGPTL3 circulates both as full-length protein and as truncated N-/C-terminal fragments . The (17-460) fragment would encompass most of the protein excluding the signal peptide, containing critical functional domains responsible for LPL inhibition and protein-protein interactions.

What experimental methods are recommended for initial characterization of ANGPTL3 (17-460)?

For initial characterization, researchers should employ:

  • Size-exclusion chromatography to verify oligomeric state

  • Circular dichroism to assess secondary structure content

  • In vitro LPL activity assays using purified LPL and triglyceride substrates

  • Binding assays with potential interaction partners (ANGPTL8, ApoA5)

  • Western blotting to verify integrity of the fragment

When designing such experiments, it's important to include full-length ANGPTL3 as a comparative control to understand how the fragment's function may differ from the complete protein .

How does ANGPTL3 form complexes with ANGPTL8, and what experimental approaches can detect this interaction?

ANGPTL3 forms a complex with ANGPTL8 (known as ANGPTL3/8) that exhibits significantly higher LPL inhibitory activity than ANGPTL3 alone. This complex formation explains the paradox of why ANGPTL3 knockout produces dramatic metabolic effects despite its weak inhibitory activity in isolation .

To study this complex formation, researchers have employed:

  • Co-expression of both proteins in HEK293 cells

  • Purification through nickel-nitrilotriacetic acid affinity followed by size-exclusion chromatography

  • Bio-layer interferometry and surface plasmon resonance to characterize binding kinetics

  • Hydrogen-deuterium exchange mass spectrometry (HDXMS) and molecular modeling to map interaction sites

How can researchers distinguish between the activities of ANGPTL3 (17-460) and other ANGPTL proteins in experimental settings?

When designing experiments to distinguish between different ANGPTL proteins:

  • Use specific antibodies that recognize only ANGPTL3 (and not ANGPTL4 or ANGPTL8)

  • Compare results from wild-type versus specific knockout models (ANGPTL3-/-, ANGPTL4-/-, ANGPTL8-/-, and double knockouts)

  • Employ recombinant proteins of defined composition

  • Perform concentration-response studies (ANGPTL4 is ~100x more potent than ANGPTL3 in LPL inhibition)

  • Include appropriate controls in all experiments

Research has demonstrated that ANGPTL3 and ANGPTL4 operate via independent mechanisms, as evidenced by additive effects when neutralizing antibodies against both proteins are administered in appropriate knockout models .

What is the tripartite relationship between ANGPTL3, ApoA5, and LPL?

ApoA5 (Apolipoprotein A5) plays a crucial role in modulating the ANGPTL3/8 complex's inhibitory effect on LPL. While circulating at ng/ml levels (much lower than other apolipoproteins but higher than ANGPTL proteins), ApoA5 can bind to the ANGPTL3/8 complex and reduce its LPL inhibitory activity .

This creates a complex regulatory system where:

  • ANGPTL3/8 complex potently inhibits LPL

  • ApoA5 binds to ANGPTL3/8 complex, reducing this inhibition

  • The balance between these proteins contributes to appropriate tissue-specific regulation of triglyceride metabolism

Researchers have used HDXMS and molecular modeling to map the exact sites on the ANGPTL3/8 complex to which LPL and ApoA5 bind, revealing distinct epitopes for each interaction partner .

What are the optimal expression systems and purification strategies for producing ANGPTL3 (17-460)?

Based on published methodologies for ANGPTL proteins:

  • Expression system: Mammalian expression (typically HEK293 cells) is preferred over bacterial expression to ensure proper folding and post-translational modifications

  • Expression vector: Mammalian expression vector containing a cytomegalovirus promoter

  • Purification steps:

    • Initial capture via nickel-nitrilotriacetic acid affinity chromatography (using His-tagged constructs)

    • Further purification by size-exclusion chromatography

    • Optional removal of fusion tags as needed for specific applications

When expressing ANGPTL3/8 complex, co-transfection of both expression constructs in HEK293 cells cultured in serum-free media has been successful, with harvest 5 days post-transfection .

How can researchers accurately measure the inhibitory activity of ANGPTL3 (17-460) on LPL?

To accurately measure ANGPTL3's inhibitory effects on LPL:

  • LPL source options:

    • Purified recombinant human GPIHBP1-LPL complex (more stable than LPL alone)

    • LPL produced through co-expression with GPIHBP1 in HEK293 cells

  • Assay considerations:

    • Include concentration ranges spanning physiological levels

    • Test both ANGPTL3 alone and in complex with ANGPTL8

    • Include ANGPTL4 as a positive control inhibitor

    • Test in the presence and absence of ApoA5 to assess modulation

    • Consider time-dependent effects, as ANGPTL3 may accelerate LPL inactivation over time

  • Data analysis:

    • Calculate IC50 values and compare between different conditions

    • Consider kinetic parameters rather than endpoint measurements

    • Account for potential cooperativity in inhibition

What are the critical controls when developing antibodies against ANGPTL3 (17-460)?

When developing antibodies against ANGPTL3:

  • Specificity controls:

    • Test cross-reactivity with related proteins (ANGPTL4, ANGPTL8)

    • Verify binding to both native and denatured forms if relevant

    • Test species cross-reactivity (human vs. mouse vs. cynomolgus monkey ANGPTL3)

  • Functional validation:

    • Assess ability to block ANGPTL3-mediated LPL inhibition in vitro

    • Evaluate effects on ANGPTL3/8 complex formation and activity

    • Determine whether the antibody recognizes ANGPTL3 alone, the ANGPTL3/8 complex, or both

  • In vivo validation:

    • Measure effects on serum triglyceride levels in appropriate animal models

    • Compare efficacy to established anti-ANGPTL3 antibodies

    • Assess tissue distribution and pharmacokinetics

How can researchers reconcile the apparent discrepancy between ANGPTL3's weak in vitro activity and strong in vivo effects?

This apparent paradox has been partially resolved through several discoveries:

  • ANGPTL3/8 complex formation: ANGPTL3 alone is a weak inhibitor, but becomes significantly more potent when complexed with ANGPTL8 .

  • Tissue-specific effects: ANGPTL3's activity may vary between tissues due to differences in local concentrations and the presence of modulatory factors.

  • Concentration-dependent oligomerization: Higher-order ANGPTL3 oligomers may exhibit different inhibitory properties than monomeric protein.

  • Methodological considerations: In vitro assay conditions may not fully recapitulate the physiological environment where ANGPTL3 functions.

To address these discrepancies, researchers should:

  • Compare in vitro results using both ANGPTL3 alone and ANGPTL3/8 complex

  • Use physiologically relevant concentrations

  • Include modulatory factors like ApoA5 in experimental designs

  • Correlate biochemical findings with genetic models (knockout mice)

What approaches can resolve contradictory data on ANGPTL3 function in different experimental systems?

When facing contradictory data:

  • Compare protein preparations:

    • Full-length vs. truncated fragments

    • Different expression systems

    • Presence of tags or fusion partners

    • Oligomeric state

  • Evaluate experimental conditions:

    • Buffer composition and pH

    • Presence of stabilizing factors

    • Temperature and incubation times

    • Concentration ranges tested

  • Integrate multiple approaches:

    • In vitro biochemical assays

    • Cell-based functional assays

    • In vivo models (genetic and pharmacological)

    • Structural studies (HDXMS, crystallography)

  • Consider biological complexity:

    • Tissue-specific effects

    • Compensatory mechanisms in knockout models

    • Species differences

    • Interplay with other regulatory proteins

How should researchers interpret the relative contributions of ANGPTL3, ANGPTL4, and ANGPTL8 to LPL regulation?

The current literature suggests a complex, context-dependent regulatory system:

  • ANGPTL4 is the more potent direct inhibitor of LPL (~100x more potent than ANGPTL3)

  • ANGPTL3's primary role may be in forming the ANGPTL3/8 complex, which is a more potent inhibitor than ANGPTL3 alone

  • Tissue specificity appears important:

    • ANGPTL4 primarily inhibits adipocyte-localized LPL activity

    • ANGPTL3/8 complex may regulate LPL activity in other tissues

    • This differential regulation contributes to appropriate energy partitioning between storage and utilization

  • Fasting vs. fed states affect expression patterns:

    • ANGPTL8 production changes with nutritional status, affecting ANGPTL3/8 complex formation

    • This creates a dynamic system responsive to metabolic needs

When designing experiments, researchers should account for these complex interactions and nutritional state effects rather than studying individual proteins in isolation .

What are the emerging therapeutic approaches targeting ANGPTL3 (17-460)?

Several therapeutic strategies focusing on ANGPTL3 are under investigation:

  • Anti-ANGPTL3/8 antibodies:

    • Specifically target the complex rather than individual proteins

    • Block ANGPTL3/8-mediated LPL inhibition

    • Have shown dramatic triglyceride-lowering effects in hypertriglyceridemic mice

  • Advantages of targeting the ANGPTL3/8 complex vs. ANGPTL3 alone:

    • More specific inhibition of the physiologically relevant inhibitory complex

    • Potentially fewer off-target effects

    • More potent triglyceride-lowering activity

  • Considerations for therapeutic development:

    • Epitope selection for antibody development

    • Species cross-reactivity for preclinical testing

    • Potential compensatory mechanisms

    • Effects on multiple lipid parameters beyond triglycerides

How do genetic variations in ANGPTL3 affect lipid metabolism and inform therapeutic strategies?

Genetic studies of ANGPTL3 variants provide important insights:

  • Loss-of-function variants are associated with:

    • Reduced plasma triglyceride levels

    • Lower LDL and HDL cholesterol

    • Potentially decreased cardiovascular risk

  • These genetic associations validate ANGPTL3 as a therapeutic target and suggest:

    • The potential efficacy of ANGPTL3-targeting therapies

    • Possible pleiotropic effects beyond triglyceride lowering

    • Safety considerations based on the phenotype of individuals with natural loss-of-function variants

  • Research directions should include:

    • Functional characterization of naturally occurring variants

    • Structure-function studies to understand how specific mutations affect activity

    • Long-term follow-up of individuals with ANGPTL3 variants to assess health outcomes

What new methodological approaches are advancing our understanding of ANGPTL3-LPL interactions?

Advanced methodologies are providing deeper insights:

  • Structural biology techniques:

    • Hydrogen-deuterium exchange mass spectrometry (HDXMS) to map binding interfaces

    • Molecular modeling to predict protein-protein interactions

    • Potential for cryo-EM studies of the ANGPTL3/8-LPL complex

  • Systems biology approaches:

    • Integration of lipid metabolism data across multiple tissues

    • Mathematical modeling of LPL regulation

    • Multi-omics analyses to understand downstream effects

  • Novel in vivo approaches:

    • Tissue-specific conditional knockout models

    • Inducible expression systems

    • CRISPR-based screening for modulators

  • Translational methodologies:

    • Development of humanized mouse models

    • Patient-derived cells expressing variant forms of ANGPTL3

    • Biomarker development for monitoring therapeutic responses

Product Science Overview

Structure and Domains

The human recombinant ANGPTL3 protein, specifically the 17-460 amino acid (a.a.) fragment, includes several important structural domains:

  • N-terminal coiled-coil domain: This domain is involved in protein-protein interactions and is essential for the oligomerization of ANGPTL3 .
  • C-terminal fibrinogen-like domain: This domain is similar to the fibrinogen-like domains found in angiopoietins and is involved in binding to receptors and other molecules .
Function and Activity

ANGPTL3 has several key functions:

  1. Inhibition of Lipoprotein Lipase (LPL): ANGPTL3 inhibits LPL, an enzyme that hydrolyzes triglycerides in lipoproteins into free fatty acids and glycerol. This inhibition leads to increased plasma triglyceride levels .
  2. Regulation of Lipid Metabolism: By inhibiting LPL, ANGPTL3 plays a significant role in regulating lipid metabolism and maintaining energy homeostasis .
  3. Promotion of Cell Expansion: ANGPTL3 has been shown to promote the expansion of liver mononuclear cells in vitro, indicating its potential role in liver function and regeneration .
Recombinant Production

The recombinant human ANGPTL3 protein (17-460 a.a.) is produced using baculovirus expression systems in insect cells (Spodoptera frugiperda, Sf 21). This method ensures high purity and activity of the protein . The recombinant protein is typically tagged with a C-terminal 10-His tag for easy purification and detection .

Applications

Recombinant ANGPTL3 is used in various research applications, including:

  • Lipid Metabolism Studies: To investigate the role of ANGPTL3 in lipid metabolism and its potential as a therapeutic target for hyperlipidemia and cardiovascular diseases.
  • Cell Culture: As a supplement in cell culture media to study its effects on cell growth and differentiation.
  • Biochemical Assays: To measure its activity in inhibiting LPL and other related enzymes.

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.