APOM Human, HEK

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

Description

Functional Role in Lipid Metabolism

APOM is a lipocalin-family protein that binds hydrophobic ligands such as retinol and retinoic acid (Kd=23 μMK_d = 2–3\ \mu M) . Its primary functions include:

  • HDL Modulation: Enhances the formation of larger nascent preβ-HDL particles by interacting with ABCA1, a cholesterol transporter .

  • Ligand Binding: Binds retinoids but not cholesterol or arachidonic acid, supporting its classification as a lipocalin .

Key Research Findings:

  • Preβ-HDL Enlargement: HEK293 cells expressing APOM and ABCA1 generated preβ-HDL particles 40% larger than controls .

  • Insulin Sensitivity: Overexpression in diabetic GK rats reduced fasting blood glucose by 1.6-fold and increased muscle AKT phosphorylation, improving insulin sensitivity .

Table: Experimental Models and Outcomes

StudyModelKey Result
Lipid Metabolism HEK293 + ABCA1APOM increased preβ-HDL size without direct physical association with apoA-I.
Diabetes GK RatsAPOM overexpression reduced blood glucose and enhanced glucose uptake.
Intralipid Effects SD RatsIntralipid infusion inhibited hepatic APOM expression by 40.4%.

Therapeutic Implications

APOM’s roles in lipid transport and glucose metabolism highlight its potential as a:

  • Biomarker for cardiovascular and metabolic diseases .

  • Therapeutic Target: Modulating APOM expression could improve HDL functionality or insulin resistance .

Product Specs

Introduction
Apolipoprotein M (APOM) is a member of the lipocalin protein family and associates with high-density lipoproteins (HDL). It is also found, to a lesser extent, with low-density lipoproteins (LDL) and triglyceride-rich lipoproteins. APOM is secreted through the plasma membrane but remains membrane-bound, where it participates in lipid transport.
Description
Recombinant human APOM protein, expressed in HEK cells, is a 20 kDa protein. It comprises 179 amino acid residues of the human APOM sequence and an additional 13 amino acid residues at the C-terminus constituting a flag tag.
Physical Appearance
Sterile, white, lyophilized (freeze-dried) powder.
Formulation
Recombinant human APOM protein solution (0.5 mg/ml) was filtered through a 0.4 μm filter and subsequently lyophilized. The lyophilized protein is supplied in 20 mM TRIS and 50 mM NaCl, pH 7.5.
Solubility
To prepare a working stock solution, add deionized water to the lyophilized protein to achieve a concentration of approximately 0.5 mg/ml. Allow the lyophilized pellet to dissolve completely. The product is not sterile. Prior to using in cell culture, filter the reconstituted protein using an appropriate sterile filter.
Stability
Store lyophilized recombinant human APOM protein at -20°C. After reconstitution, aliquot the protein and store at -20°C to prevent repeated freeze/thaw cycles. Reconstituted protein can be stored at 4°C for a short period (up to two weeks) without noticeable degradation.
Applications
Recombinant human APOM protein is suitable for various research applications, including cell culture, animal studies, enzyme-linked immunosorbent assays (ELISA), and Western blotting.
Purity
The purity of recombinant human APOM protein is greater than 90% as determined by SDS-PAGE analysis.
Synonyms
G3a, HSPC336, NG20, Apolipoprotein M, APOM, Apo-M, MGC22400.
Source
HEK293.
Amino Acid Sequence
HVDYKDDDDK PAGCPEHSQL TTLGVDGKEF PEVHLGQWYF IAGAAPTKEE LATFDPVDNI VFNMAAGSAP MQLHLRATIR MKDGLCVPRK WIYHLTEGST DLRTEGRPDM KTELFSSSCP GGIMLNETGQ GYQRFLLYNR SPHPPEKCVE EFKSLTSCLD SKAFLLTPRN QEACELSNN

Q&A

What is Apolipoprotein M (APOM) and what are its primary biological functions?

Apolipoprotein M (APOM) is a member of the lipocalin superfamily primarily expressed on high-density lipoprotein (HDL). It serves as the carrier protein for sphingosine-1-phosphate (S1P) in lipoproteins and plays significant roles in both lipid transport and glucose metabolism . Research demonstrates that APOM affects hepatic lipid and glucose metabolism, making it a protein of interest in metabolic disease studies. When investigating APOM in experimental systems, researchers should consider its dual role in both lipid transport mechanisms and glucose homeostasis pathways.

How does APOM expression in HEK systems compare to native human tissue expression?

When expressing human APOM in HEK cell systems, researchers should be aware that expression patterns may differ from those observed in vivo. In native conditions, APOM is predominantly expressed in liver and, to a lesser extent, in kidney and pancreatic tissues. When utilizing HEK cells for APOM production, expression vectors with appropriate promoters (such as CAG promoters) should be selected to achieve physiologically relevant expression levels . For accurate comparisons, quantification of both mRNA and protein levels using qPCR and Wes-ProteinSimple systems respectively is recommended, as demonstrated in ApoM research protocols.

What experimental models are most appropriate for studying APOM's metabolic effects?

When investigating APOM's metabolic effects, several experimental models have demonstrated utility. The Goto-Kakizaki (GK) rat model, being non-obese and spontaneously developing type 2 diabetes, represents an advantageous system for studying APOM's effects without confounding factors like obesity . This model closely resembles human type 2 diabetes in terms of insulin secretion defects and complications. For cell-based models, pancreatic β-cell lines provide insights into APOM's role in insulin secretion. When designing APOM studies, researchers should carefully select models based on specific research questions, considering that different models may yield different results regarding APOM's metabolic effects.

What viral vector systems are most efficient for APOM gene transfer in metabolic research?

For optimal gene transfer of human APOM in metabolic research, adeno-associated virus (AAV) vectors have demonstrated significant efficacy. Specifically, AAV2 vector genomes pseudo-serotyped with type 8 capsid (AAV2/8) have shown promising results as effective gene transfer agents, particularly for targeting pancreatic tissues . The methodology should include:

  • Design of expression constructs containing the APOM gene under strong promoters (e.g., CAG promoter)

  • Incorporation of reporter genes (such as EGFP) to visualize successful transduction

  • Production of high-titer viral particles (recommended: 5 × 10^11 vg/200 μL/animal for in vivo applications)

  • Verification of tissue-specific expression through fluorescence microscopy and quantification of mRNA/protein levels

When selecting AAV serotypes, researchers should note that AAV8 shows superior transduction efficiency in pancreatic tissue compared to other serotypes, making it particularly valuable for diabetes research .

How can researchers effectively measure APOM-induced changes in insulin sensitivity?

To rigorously assess APOM-induced changes in insulin sensitivity, researchers should implement a complementary set of methodologies:

  • Hyperinsulinemic-Euglycemic Clamp (HEC): This gold standard technique provides the most direct measurement of insulin sensitivity. In APOM studies, glucose infusion rates should be carefully monitored at multiple timepoints (1-3 weeks post-intervention) to capture dynamic changes in insulin sensitivity. Research has shown that APOM overexpression can increase glucose infusion rates up to 1.95-fold compared to controls .

  • Oral Glucose Tolerance Test (OGTT): This complements HEC data by assessing whole-body glucose disposal. Blood samples should be collected at baseline (fasting), 30, 60, and 120 minutes post-glucose challenge (2.5 g/kg body weight) .

  • Molecular Pathway Analysis: Insulin signaling should be assessed through quantification of phosphorylated protein kinase B (p-AKT)/total AKT ratios in insulin-responsive tissues. The Wes-ProteinSimple assay has proven effective for this purpose in APOM research .

These methodologies together provide comprehensive assessment of both physiological responses and underlying molecular mechanisms in APOM-mediated insulin sensitivity.

What are the critical considerations when investigating APOM's tissue-specific effects?

When investigating tissue-specific effects of APOM, researchers must address several methodological challenges:

  • Tissue Tropism of Delivery Systems: Different AAV serotypes demonstrate varying tissue tropism. While AAV8 shows effective pancreatic tissue transduction, other tissues may require alternative serotypes. Researchers should verify expression patterns through tissue-specific analyses of both mRNA and protein levels .

  • Temporal Expression Patterns: APOM expression following gene transfer shows tissue-dependent temporal dynamics. In pancreatic tissues, significant overexpression may only be detectable at specific timepoints (e.g., three weeks post-transfection) . Therefore, experimental timelines should include multiple assessment points.

  • Functional Readouts: Each tissue requires specific functional assessments. For pancreatic expression, insulin secretion measurements are critical. For muscle tissues, p-AKT/AKT ratios provide insights into insulin signaling efficacy .

  • Systemic vs. Local Effects: Distinguishing between direct tissue-specific effects and secondary systemic effects requires careful experimental design, potentially including tissue-specific knockout models alongside overexpression systems.

Comprehensive tissue analysis should include liver, muscle, adipose, pancreatic, kidney and heart tissues to fully characterize APOM's differential effects across metabolic tissues .

How should researchers address contradictory findings regarding APOM's role in glucose metabolism?

The literature presents apparently contradictory findings regarding APOM's metabolic effects. For example, some studies indicate APOM deficiency protects against diet-induced obesity and improves glucose tolerance, while others demonstrate APOM overexpression improves insulin sensitivity . To address these contradictions, researchers should:

  • Consider Sex-Specific Effects: Research indicates estrogen upregulates APOM expression through estrogen receptor α, potentially explaining differential effects in male versus female models. Experimental designs should explicitly control for sex as a biological variable .

  • Account for Model Differences: Diet-induced obesity models may yield different results compared to spontaneous diabetes models like GK rats. Study designs should acknowledge model-specific limitations .

  • Evaluate Temporal Dynamics: APOM's effects may vary temporally, with different outcomes observed at various timepoints. Longitudinal assessments are recommended (e.g., measurements at weeks 1-7 post-intervention) .

  • Assess Dose-Dependency: Different expression levels may produce varying or even opposing physiological effects. Dose-response studies with careful quantification of APOM levels are essential.

  • Examine Pathway Integration: Investigation of how APOM interacts with other metabolic regulators, including leptin and insulin sensitizers like rosiglitazone, may help reconcile contradictory findings .

What methodological approaches best capture the relationship between APOM and insulin secretion?

To effectively investigate APOM's influence on insulin secretion, researchers should implement a multi-faceted approach:

  • In Vivo Assessment: Measure serum insulin levels at multiple timepoints following APOM intervention (e.g., at weeks 2 and 6 post-AAV injection). ELISA-based methods with sensitivity appropriate for rodent insulin have proven effective .

  • Ex Vivo Pancreatic Islet Studies: Isolated islets from APOM-overexpressing models can be challenged with varying glucose concentrations to assess glucose-stimulated insulin secretion directly.

  • S1P Dependency Analysis: Since APOM functions as an S1P carrier, experiments should determine whether APOM's effects on insulin secretion are S1P-dependent. This can be accomplished through S1P receptor antagonists or S1P-binding deficient APOM mutants .

  • Molecular Mechanism Investigation: Assess pancreatic β-cell health markers, calcium signaling, and exocytosis machinery function to elucidate underlying mechanisms.

This comprehensive approach has revealed that APOM augments insulin secretion by maintaining S1P concentration under both in vivo and in vitro conditions .

What are the optimal transfection and expression conditions for human APOM in HEK cells?

For optimal transfection and expression of human APOM in HEK cells, researchers should consider:

  • Vector Design:

    • Utilize pAAV-CAG-APOM-EGFP-3FLAG constructs for efficient expression and detection

    • Include reporter genes (like EGFP) to visually confirm successful transfection

    • Consider inducible promoter systems for controlled expression timing

  • Transfection Protocol:

    • For transient expression: Lipofection or calcium phosphate methods with optimized DNA:reagent ratios

    • For stable cell line generation: Viral transduction followed by appropriate selection

    • Transfection efficiency assessment through flow cytometry of reporter gene expression

  • Expression Verification:

    • Quantify mRNA levels through RT-qPCR

    • Assess protein expression using Wes-ProteinSimple system for sensitive detection

    • Verify functionality through binding assays with S1P

  • Protein Purification Considerations:

    • Utilize FLAG or similar epitope tags for affinity purification

    • Consider secreted versus cellular fractions, as APOM is predominantly secreted

    • Optimize purification buffers to maintain protein stability and lipid binding capacity

These methodological considerations ensure reproducible and physiologically relevant expression of human APOM in HEK systems.

How should researchers interpret changes in APOM expression relative to metabolic parameters?

When analyzing the relationship between APOM expression and metabolic parameters, researchers should implement a comprehensive analytical approach:

  • Correlation Analysis: Calculate Pearson or Spearman correlation coefficients between APOM levels and key metabolic parameters (fasting glucose, insulin levels, HOMA-IR, etc.). Research indicates significant inverse correlations between pancreatic APOM expression and blood glucose levels .

  • Temporal Relationship Analysis: Assess time-course data to determine whether APOM expression changes precede or follow metabolic improvements. For example, significant decreases in random blood glucose have been observed starting from the second week after APOM overexpression .

  • Dose-Response Assessment: Quantify the relationship between degree of APOM overexpression and magnitude of metabolic parameter changes. In experimental models, 1.04- to 1.95-fold increases in glucose infusion rates have been observed with APOM overexpression .

  • Multivariate Analysis: Apply principal component or factor analysis to determine how APOM relates to clusters of metabolic variables, potentially revealing pathway-specific effects.

  • Confounding Variable Control: Account for potential confounders such as body weight changes, which have been observed to increase significantly with APOM overexpression in weeks 5-6 .

What statistical approaches are most appropriate for analyzing APOM intervention studies?

For robust statistical analysis of APOM intervention studies, researchers should consider:

  • Sample Size Determination: Power analysis should guide experimental design, with consideration for expected effect sizes. Previous APOM studies have utilized approximately 18 animals per experimental group .

  • Appropriate Statistical Tests:

    • For comparing two groups: Student's t-test for normally distributed data

    • For multiple timepoints or conditions: One-way ANOVA followed by appropriate post-hoc tests

    • For repeated measures: Mixed-effects models to account for within-subject correlations

  • Normalization Strategies: When analyzing molecular signaling data (e.g., p-AKT/AKT ratios), appropriate normalization to housekeeping proteins and baseline conditions is essential .

  • Handling Non-Normal Data: For metabolic parameters that often show skewed distributions (e.g., insulin levels), consider log transformation before analysis or non-parametric alternatives.

  • Multiple Testing Correction: When assessing APOM's effects across multiple tissues or parameters, implement Bonferroni or false discovery rate corrections to maintain appropriate familywise error rates.

Data should be presented as mean ± standard error of the mean, with significance established at P<0.05, consistent with established conventions in metabolic research .

What emerging technologies might enhance human APOM research in HEK and other expression systems?

Several cutting-edge technologies hold promise for advancing human APOM research:

  • CRISPR/Cas9 Gene Editing: Precise modification of endogenous APOM in HEK cells can create physiologically relevant models without overexpression artifacts. This approach allows:

    • Introduction of specific polymorphisms associated with metabolic disease

    • Creation of reporter knock-in lines for real-time APOM expression monitoring

    • Generation of conditional expression systems for temporal control

  • Single-Cell Transcriptomics: Application to APOM-expressing tissues can reveal:

    • Cell-type specific responses to APOM overexpression

    • Heterogeneity in metabolic pathway activation

    • Novel cellular targets previously unrecognized in bulk analysis

  • Organoid Culture Systems: Development of pancreatic, liver, or multi-organ-on-chip systems expressing human APOM can provide more physiologically relevant models than traditional HEK monocultures.

  • Advanced AAV Engineering: Next-generation AAV capsids with enhanced tissue specificity could improve targeted delivery of APOM to metabolic tissues of interest .

  • Proteomics and Interactome Analysis: Mass spectrometry-based approaches to identify APOM-interacting proteins in different metabolic states could reveal novel mechanistic insights beyond the established S1P carrier function .

These emerging technologies will help address current limitations in understanding APOM's tissue-specific functions and molecular mechanisms.

How can researchers best integrate APOM findings into broader metabolic disease research?

To maximize the impact of APOM research findings on metabolic disease understanding:

  • Translational Research Integration:

    • Correlate findings from HEK and animal models with human biobank data

    • Investigate APOM polymorphisms identified in diabetic populations (e.g., rs707922 and T-778C in the APOM promoter)

    • Develop potential therapeutic strategies based on APOM's beneficial metabolic effects

  • Pathway Interconnection Analysis:

    • Investigate how APOM-S1P signaling interfaces with established metabolic regulators

    • Explore connections between APOM and insulin signaling pathways, particularly the AKT pathway identified in muscle tissues

    • Examine how APOM relates to other apolipoproteins in metabolic regulation

  • Biomarker Development:

    • Assess APOM's potential as a biomarker for metabolic disease progression or treatment response

    • Develop standardized assays for APOM and APOM-associated S1P measurement in clinical samples

  • Therapeutic Target Evaluation:

    • Investigate whether modulation of APOM expression or function could provide metabolic benefits

    • Explore the potential of AAV-mediated APOM gene therapy approaches for metabolic disorders

    • Assess whether APOM-mimetic peptides could recapitulate beneficial effects

  • Contradictory Finding Resolution:

    • Design experiments specifically addressing the paradox of both APOM deficiency and overexpression showing metabolic benefits in different contexts

    • Investigate tissue-specific and temporal aspects of APOM function

Product Science Overview

Introduction

Apolipoprotein M (ApoM) is a member of the apolipoprotein family, which plays a crucial role in lipid metabolism and cardiovascular health. ApoM is predominantly associated with high-density lipoproteins (HDL) and, to a lesser extent, with low-density lipoproteins (LDL) and triglyceride-rich lipoproteins . The recombinant form of ApoM, expressed in Human Embryonic Kidney (HEK) cells, has been extensively studied for its biological properties and potential therapeutic applications.

Structure and Expression

ApoM is a lipocalin protein that is secreted through the plasma membrane but remains membrane-bound, participating in lipid transport . The human recombinant ApoM produced in HEK293 cells is a single, non-glycosylated polypeptide chain containing 166 amino acids, with a molecular mass of approximately 19-24 kDa . It is expressed without a signal peptide sequence and includes a His tag at the N-terminus for purification purposes .

Biological Functions

ApoM has several important biological functions, including:

  • Lipid Transport: ApoM is involved in the transport of lipids, particularly in association with HDL particles .
  • Binding to Retinoic Acid: ApoM can bind to all-trans-retinoic acid, a derivative of vitamin A, which is essential for various physiological processes .
  • Neuroprotection: ApoM secreted by HEK cells stably expressing apoE3 or apoE4 can bind amyloid-beta (Aβ) and inhibit Aβ-induced neurotoxicity, suggesting a potential role in neuroprotection .
Tissue Distribution

ApoM is primarily found in the liver and kidneys, where it is synthesized and secreted into the bloodstream. Its presence in HDL particles suggests a significant role in cholesterol metabolism and cardiovascular health .

Regulatory Mechanisms

The expression of ApoM is regulated by various factors, including:

  • Liver X Receptors (LXR): These nuclear receptors play a key role in the regulation of ApoM expression in response to lipid levels .
  • Peroxisome Proliferator-Activated Receptors (PPARs): These receptors are involved in the regulation of lipid metabolism and can influence ApoM expression .
Potential Therapeutic Applications

Given its role in lipid metabolism and neuroprotection, ApoM has been investigated as a potential therapeutic target for cardiovascular diseases and neurodegenerative disorders. Its ability to bind and transport retinoic acid also highlights its importance in vitamin A metabolism and related physiological processes .

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.