APO D Human, GST

APO-D Human Recombinant, GST Tag
Shipped with Ice Packs
In Stock

Description

Overview of APO D Human, GST

APO D Human, GST refers to recombinant human apolipoprotein D (APOD) fused with a glutathione S-transferase (GST) tag. This fusion protein is engineered for enhanced solubility and purification efficiency, enabling downstream applications in biochemical research. APOD itself is a member of the lipocalin family, characterized by a β-barrel tertiary structure that facilitates the transport of small hydrophobic ligands such as cholesterol, progesterone, and arachidonic acid . The GST tag, derived from Schistosoma japonicum, binds glutathione, allowing affinity chromatography-based purification .

Production and Purification

APOD-GST is produced via recombinant DNA technology:

  1. Cloning: APOD cDNA (e.g., residues 1–189) is inserted into a GST-fusion vector .

  2. Expression: Induced in E. coli (e.g., BL21(DE3) strains) with IPTG .

  3. Purification:

    • Step 1: Glutathione-agarose affinity chromatography to capture GST-APOD .

    • Step 2: Proprietary chromatographic techniques (e.g., ion-exchange) to achieve >95% purity .

Quality Control:

  • SDS-PAGE: Single band at ~48 kDa .

  • Endotoxin Levels: <0.1 ng/μg (LAL test) .

Research Applications

APOD-GST is utilized in studies exploring lipid transport, neurodegeneration, and pathogen interactions:

Lipid Metabolism and Neuroprotection

APOD-GST models APOD’s role in HDL-mediated cholesterol transport and neuroprotection. In Alzheimer’s disease models, APOD accumulates in cerebrospinal fluid, suggesting a role in lipid scavenging and neuronal repair . GST-tagged APOD facilitates binding assays to study interactions with lipids or receptors .

Virology and Pathogen Interactions

Recent studies reveal APOD’s interaction with rabies virus (RABV) glycoproteins. APOD-GST binds RABV G proteins, modulating viral propagation. This interaction is critical for understanding viral entry mechanisms and therapeutic targeting .

Cell Migration and Signaling

APOD-GST is used to study smooth muscle cell (SMC) migration. In PDGF-BB-stimulated SMCs, APOD knockdown suppresses migration, while overexpression enhances Rac1 activation, promoting motility .

Disease Associations and Genetic Polymorphisms

DiseaseAPOD’s Role
Breast Cysts/CancerElevated in cyst fluid of women with gross cystic disease
Neurodegenerative DisordersAccumulates in Alzheimer’s and Parkinson’s brains; neuroprotective
AtherosclerosisExpressed in atherosclerotic plaques; regulates SMC migration

Genetic Variants:
APOD polymorphisms (e.g., APOD1 and APOD2) are observed in populations of African ancestry, serving as anthropogenetic markers .

Future Directions

APOD-GST’s utility extends to:

  • Therapeutic Development: Targeting APOD for neurodegenerative diseases or lipid disorders .

  • Structural Studies: Crystallization with GST tag to resolve ligand-binding pockets .

  • Viral Pathogenesis: Exploring APOD-GST interactions with other pathogens (e.g., coronaviruses) .

Data Tables and Key Research Findings

Table 2: Research Applications

Study FocusKey FindingsSource
NeuroprotectionAPOD scavenges cholesterol in Alzheimer’s; GST-tagged APOD models neuroprotective roles
Rabies Virus InteractionAPOD binds RABV G protein, enhancing viral propagation; GST-APOD used in pulldown assays
SMC MigrationAPOD knockdown suppresses PDGF-BB-induced migration; overexpression activates Rac1

Product Specs

Introduction
Apolipoprotein-D, a member of the lipocalin family, primarily associates with high-density lipoproteins in human plasma. Unlike typical apolipoproteins, it exhibits a beta-barrel structure characteristic of lipocalins, enabling it to transport small hydrophobic molecules like cholesterol, progesterone, pregnenolone, bilirubin, and arachidonic acid. Notably expressed in the spleen, testes, and brain, Apolipoprotein-D is found in elevated concentrations within the cyst fluid of individuals with gross cystic breast disease, a condition linked to heightened breast cancer risk. Its accumulation is also observed in regenerating peripheral nerves and the cerebrospinal fluid of patients with neurodegenerative disorders such as Alzheimer's disease. Functionally, Apolipoprotein-D contributes to the maintenance and repair processes within the central and peripheral nervous systems, acting as a versatile transporter capable of shuttling ligands between cells within an organ, scavenging ligands for transport to the bloodstream, or delivering ligands from circulation to specific tissues.
Description
Recombinant human APO-D, expressed in E. coli, exhibits a molecular weight of 48 kDa as determined by SDS-PAGE. The protein undergoes purification using proprietary chromatographic techniques.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
The APO-D protein is supplied at a concentration of 100 µg/ml in a buffer composed of 50 mM Tris-Acetate (pH 7.5), 1 mM EDTA, and 20% glycerol.
Stability
For long-term storage, the APO-D protein should be stored in a vial at a temperature of -20°C to -80°C. Under these conditions, the protein remains stable for a period of 12 months.
Synonyms
Apolipoprotein D, Apo-D, ApoD.
Source
Escherichia Coli.

Q&A

What is Apolipoprotein D (APOD) and how is it identified in human tissues?

Apolipoprotein D (APOD) is a protein that belongs to the lipocalin family and plays various roles in lipid transport and cell signaling pathways. In research settings, APOD can be identified through several complementary approaches. RT-PCR is commonly used to clone APOD cDNA from specific tissues, as demonstrated in studies with ovine aortic smooth muscle cells (SMCs) . For protein detection, western blotting techniques can quantify APOD expression levels across different experimental conditions. Confocal microscopy serves as a valuable method for visualizing APOD's subcellular localization, revealing distinctive patterns such as perinuclear and trailing edge expression in migrating cells versus confluent monolayers . When investigating APOD's functional significance, researchers often employ both overexpression and knockdown strategies, with RNAi being particularly effective for suppressing APOD expression to assess resulting phenotypic changes .

How does APOD expression change during cell migration?

APOD expression undergoes significant changes during cell migration processes. Research using multiwound migration assays has demonstrated a 2.5-fold increase in APOD protein levels in aortic smooth muscle cells (SMCs) 48 hours after initiating migration . Interestingly, this upregulation occurs without corresponding increases in APOD mRNA levels, indicating post-transcriptional regulatory mechanisms . Confocal microscopy has revealed that APOD adopts a specific subcellular distribution pattern in actively migrating cells, with prominent expression in perinuclear regions and at the trailing edge of the cell . This localization pattern is notably absent in confluent, non-migrating cell monolayers, suggesting that APOD's spatial organization within the cell is functionally linked to the migratory phenotype . These observations provide important methodological considerations for researchers studying APOD in migration contexts, highlighting the importance of examining both protein levels and subcellular localization rather than relying solely on mRNA expression analysis.

What is the relationship between APOD and growth factors in cell function?

APOD exhibits a synergistic relationship with specific growth factors, particularly platelet-derived growth factor BB (PDGF-BB). When smooth muscle cells are stimulated with PDGF-BB at a concentration of 10 ng/mL, APOD protein expression significantly increases . This relationship appears to be functionally important, as PDGF-BB-stimulated migration of human pulmonary artery SMCs is substantially suppressed when APOD is knocked down using RNAi techniques . Furthermore, the presence of APOD, either through overexpression or exogenous addition, enables cell migration responses to subthreshold doses of PDGF-BB that would otherwise be insufficient to trigger migration . This synergistic effect does not involve enhanced phosphorylation of ERK1/2 or phospholipase C-γ1 signaling pathways, but instead correlates with increased Rac1 activation . These findings highlight the importance of considering growth factor interactions when designing experiments to study APOD function, suggesting that optimal experimental conditions should include appropriate growth factor stimulation to fully reveal APOD's physiological roles.

What experimental approaches are used to create GST fusion proteins for APOD research?

GST fusion proteins represent valuable tools for APOD research, enabling protein purification, interaction studies, and functional analyses. The experimental approach begins with cloning the APOD cDNA into a GST expression vector, which contains the GST tag sequence upstream of a multiple cloning site. Researchers typically use bacterial expression systems, most commonly E. coli BL21 or similar strains optimized for protein expression. After transformation, bacterial cultures are grown to appropriate density before protein expression is induced with IPTG. The expressed fusion proteins can be efficiently purified using glutathione-affinity chromatography, where the GST portion binds to glutathione-immobilized resin while contaminants are washed away. For functional studies, the purified GST-APOD fusion protein can be eluted under mild conditions using reduced glutathione. Quality control steps include SDS-PAGE and western blotting to confirm protein size, purity, and immunoreactivity. When studying protein-protein interactions, GST-APOD can be immobilized on glutathione beads and used for pull-down assays to identify binding partners from cell lysates. This methodological approach provides researchers with purified APOD protein that maintains functional properties for downstream applications.

How does APOD overexpression affect cellular migration and what methodologies best capture these effects?

APOD overexpression significantly enhances cellular migration through mechanisms that require specialized methodological approaches to properly quantify. Studies with stable APOD overexpression in aortic smooth muscle cells (Ao SMCs) have demonstrated a 62% increase in random migration compared to vector-transfected control cells . When designing experiments to capture these effects, researchers should employ multiple complementary migration assays. Random migration assays, which track individual cell movements across a substrate without directional cues, effectively capture APOD's baseline effects on motility . For growth factor response studies, modified Boyden chamber or transwell assays with varying PDGF-BB concentrations can reveal APOD's synergistic effects, particularly at subthreshold growth factor doses that become effective only in APOD-overexpressing cells . Time-lapse microscopy combined with cell tracking software provides quantitative data on migration speed, directionality, and persistence. For mechanistic insights, researchers should complement these functional assays with molecular analyses examining Rac1 activation states rather than ERK1/2 or PLC-γ1 phosphorylation, as APOD appears to specifically enhance Rac1 signaling . Control experiments should include both vector-transfected cells and dose-response curves for growth factors to properly contextualize APOD's effects across varying stimulation conditions.

What are the technical considerations for RNAi-mediated APOD knockdown experiments?

RNAi-mediated APOD knockdown experiments require careful technical considerations to ensure specificity, efficiency, and physiological relevance. When designing siRNA sequences, researchers should target regions unique to APOD while avoiding sequences with homology to other apolipoproteins, particularly APOE, which shares structural features . Multiple independent siRNA sequences should be tested to confirm that observed phenotypes are due to APOD suppression rather than off-target effects. Transfection optimization is crucial, with lipid-based transfection reagents typically providing good efficiency in smooth muscle cells, though electroporation may be preferable for difficult-to-transfect primary cells . Knockdown efficiency should be verified at both mRNA (RT-qPCR) and protein (western blot) levels, with 70-90% reduction considered effective for functional studies . The timing of knockdown assessment is critical, as the post-transcriptional regulation of APOD means protein levels may persist longer than mRNA . For migration studies, knockdown should be confirmed immediately before and after the assay period. Control experiments must include non-targeting siRNA sequences that activate the RNAi machinery without targeting any specific transcript. Additionally, rescue experiments, where siRNA-resistant APOD is re-expressed, provide compelling evidence that observed phenotypes are specifically due to APOD depletion rather than off-target effects or general disruption of cellular homeostasis.

How can researchers effectively investigate the synergism between APOD and PDGF-BB in experimental systems?

Investigating the synergism between APOD and PDGF-BB requires strategic experimental design that captures their cooperative effects on cellular function. Researchers should initially establish dose-response curves for PDGF-BB alone to identify both optimal stimulatory concentrations (typically 10 ng/mL) and subthreshold doses that produce minimal effects in standard conditions . These subthreshold concentrations become critical for revealing synergistic interactions, as APOD has been shown to enable cellular responses to otherwise ineffective PDGF-BB levels . Experimental approaches should include: (1) Combined stimulation assays where cells receive both exogenous APOD and varying PDGF-BB concentrations; (2) Migration assays with APOD-overexpressing and control cells across a PDGF-BB gradient; (3) Signaling pathway analyses focusing specifically on Rac1 activation rather than ERK1/2 or PLC-γ1 phosphorylation events . Time-course experiments are essential, as the synergistic effects may have distinct temporal dynamics compared to either factor alone. For mechanistic insights, researchers should employ selective pathway inhibitors and constitutively active/dominant negative signaling components to dissect the precise intersection points between APOD and PDGF-BB signaling cascades. Proximity ligation assays or co-immunoprecipitation can determine whether APOD physically interacts with PDGF-BB or its receptors, while super-resolution microscopy may reveal co-localization patterns during cellular responses.

What experimental approaches can distinguish between intracellular synthesis and extracellular uptake of APOD?

Distinguishing between intracellular synthesis and extracellular uptake of APOD requires sophisticated experimental approaches that track protein origin and trafficking. Research has demonstrated that APOD can either be expressed endogenously by cells or taken up from the extracellular environment, with both sources contributing to its cellular functions . To differentiate these pathways, researchers should employ pulse-chase experiments with metabolic labeling (e.g., 35S-methionine/cysteine) to track newly synthesized APOD, while using differentially labeled exogenous APOD (e.g., fluorescently tagged or isotope-labeled) to monitor uptake. Selective inhibition approaches provide complementary insights: blocking protein synthesis with cycloheximide while adding exogenous APOD can isolate uptake mechanisms, while culturing cells in APOD-depleted serum can emphasize endogenous production. Molecular approaches include creating reporter constructs with the APOD promoter driving fluorescent protein expression to visualize transcriptional activity in real-time. For uptake studies, researchers should investigate receptor-mediated endocytosis by employing endocytosis inhibitors or performing knockdown of candidate receptors. Subcellular fractionation combined with western blotting can track APOD distribution across cellular compartments, while immunofluorescence microscopy with antibodies distinguishing modified (e.g., tagged) exogenous APOD from native protein can visualize the spatial distribution of each pool within the cell.

How should researchers approach experimental design when studying APOD's role in Rac1 activation pathways?

When studying APOD's role in Rac1 activation pathways, researchers should implement a comprehensive experimental design that captures both direct and indirect mechanisms. Evidence indicates that APOD enhances cellular migration through Rac1 activation rather than through ERK1/2 or phospholipase C-γ1 phosphorylation . To thoroughly investigate this relationship, pull-down assays using GST-fusion proteins containing the p21-binding domain of PAK1, which specifically binds active (GTP-bound) Rac1, should be employed to quantify Rac1 activation levels in response to APOD manipulation . Time-course experiments are essential to determine whether APOD affects the magnitude or duration of Rac1 activation following PDGF-BB stimulation. Researchers should complement these biochemical assays with live-cell imaging using FRET-based Rac1 biosensors to visualize spatial and temporal dynamics of activation in single cells. For mechanistic dissection, the experimental approach should include knockdown or inhibition of guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs) that regulate Rac1, to identify specific mediators influenced by APOD. Pathway verification requires rescue experiments where constitutively active or dominant negative Rac1 mutants are expressed in APOD-manipulated cells to confirm the necessity and sufficiency of Rac1 in mediating APOD's effects. Pharmacological approaches using Rac1 inhibitors (e.g., NSC23766) at varying concentrations can establish dose-dependent relationships between Rac1 activity and APOD-induced phenotypes.

How do post-transcriptional regulatory mechanisms affect APOD expression in different cellular contexts?

APOD expression exhibits distinctive post-transcriptional regulation that varies across cellular contexts, requiring specialized experimental approaches to fully characterize. Research has demonstrated that in motile smooth muscle cells, APOD protein levels increase 2.5-fold during migration despite no corresponding increase in mRNA levels, indicating robust post-transcriptional control mechanisms . This pattern parallels other proteins like fibronectin that are similarly regulated during cell motility . To investigate these mechanisms, researchers should employ polysome profiling to assess mRNA translation efficiency, isolating mRNA-ribosome complexes through sucrose gradient ultracentrifugation followed by RT-qPCR quantification of APOD transcripts in each fraction. RNA immunoprecipitation (RIP) assays can identify RNA-binding proteins that interact with APOD mRNA, potentially regulating its stability or translation. For detailed mechanistic studies, reporter constructs containing APOD 5' and 3' UTRs fused to luciferase coding sequences allow quantification of regulatory effects and identification of specific regulatory elements through mutational analysis. Pulse-chase experiments using 35S-methionine labeling followed by immunoprecipitation provide direct measurement of APOD protein synthesis and degradation rates. MicroRNA involvement should be assessed through AGO-RIP, in silico prediction tools, and functional validation with miRNA mimics or inhibitors. Time-course analyses across different cellular states (quiescent, proliferating, migrating) are essential to capture the dynamic nature of these regulatory mechanisms in physiologically relevant contexts.

What methodological approaches best characterize APOD's subcellular localization during cell migration?

Characterizing APOD's subcellular localization during cell migration requires sophisticated imaging approaches combined with biochemical fractionation techniques. Research has revealed distinct localization patterns in migrating cells, with prominent perinuclear and trailing edge expression that differs markedly from the distribution in non-migrating cells . To comprehensively analyze these patterns, researchers should implement high-resolution confocal microscopy with Z-stack imaging to capture three-dimensional distribution, using co-staining for organelle markers (ER, Golgi, endosomes) to identify specific subcellular compartments containing APOD. Live-cell imaging with fluorescently tagged APOD constructs allows tracking of dynamic redistribution during migration, while photoactivatable or photoconvertible fusion proteins can reveal directional trafficking routes. For quantitative assessment of localization patterns, researchers should employ digital image analysis with line-scan intensity profiles across migrating cells and quantification of co-localization coefficients with organelle markers. Complementary biochemical approaches include subcellular fractionation techniques optimized to separate membrane microdomains, followed by western blotting to quantify APOD distribution across fractions. Super-resolution microscopy techniques such as STORM or PALM provide nanoscale resolution of APOD clustering and organization at the trailing edge. For functional validation, targeted mislocalization approaches using organelle-specific targeting sequences can determine whether proper localization is required for APOD's effects on migration. These methodological approaches collectively provide a comprehensive characterization of APOD's spatial organization during the complex process of cell migration.

How can advanced genetic approaches be used to study APOD function in human research models?

Advanced genetic approaches offer powerful tools for studying APOD function in human research models, allowing precise manipulation and analysis of its expression and activity. CRISPR/Cas9 genome editing represents the gold standard approach, enabling researchers to create knockout cell lines for complete APOD ablation, knockin reporter lines for endogenous expression tracking, or precise point mutations to investigate specific functional domains. Guide RNA design should target early exons while avoiding regions with homology to other apolipoproteins to prevent off-target effects. For temporal control of APOD expression, inducible systems such as Tet-On/Off or destabilized domain technologies allow researchers to modulate APOD levels at specific experimental timepoints. Single-cell RNA sequencing provides insights into heterogeneous APOD expression patterns within tissue populations, while spatial transcriptomics can map expression relative to anatomical features. For functional genomics approaches, researchers should employ CRISPR interference (CRISPRi) or activation (CRISPRa) systems to modulate APOD expression without altering the genomic sequence. Physiologically relevant models include iPSC-derived cells that can be differentiated into tissue-specific lineages expressing APOD, potentially coupled with organoid cultures that recapitulate tissue architecture. When analyzing results, researchers should implement rigorous validation using multiple independent clones and complementation experiments where APOD is reintroduced into knockout lines to confirm phenotype specificity.

What are the methodological challenges in purifying active GST-APOD fusion proteins for functional studies?

Purifying active GST-APOD fusion proteins for functional studies presents several methodological challenges that require strategic approaches to overcome. The process begins with optimizing expression conditions, as high-level expression can lead to inclusion body formation; researchers should test multiple temperatures (16-30°C), inducer concentrations, and expression durations to maximize soluble protein yield. The bacterial strain selection is critical, with specialized strains like Rosetta or Arctic Express offering advantages for eukaryotic protein expression through provision of rare codons or enhanced folding at low temperatures. During purification, maintaining protein stability requires careful buffer optimization, typically including glycerol (10-20%), reducing agents (DTT or β-mercaptoethanol), and protease inhibitor cocktails. To enhance solubility while preserving function, researchers should evaluate different detergents (Triton X-100, CHAPS) and stabilizing agents (arginine, trehalose) at various concentrations. The GST tag itself presents challenges, as its dimerization tendency can affect APOD function; therefore, on-column cleavage using site-specific proteases (PreScission, thrombin) should be optimized to remove the tag while maintaining protein on the resin for final purification. Quality control is essential and should include size-exclusion chromatography to confirm monodispersity, circular dichroism to verify secondary structure, and thermal shift assays to assess stability. Functional validation requires comparison of purified GST-APOD with native APOD in cellular assays, ensuring that the fusion protein retains biological activity before use in detailed mechanistic studies.

How should researchers analyze potential synergistic effects between APOD and growth factors in experimental datasets?

Analyzing potential synergistic effects between APOD and growth factors requires rigorous statistical approaches that can distinguish between additive and true synergistic interactions. Researchers should implement factorial experimental designs where APOD levels (normal, overexpressed, knockdown) and growth factor concentrations (multiple doses including subthreshold levels) are systematically varied . For statistical analysis, two-way ANOVA with interaction terms provides the foundation for detecting synergy, with a significant interaction term indicating that the combined effect differs from what would be predicted by simple addition of individual effects . Researchers should calculate theoretical additive effects (Effect A + Effect B) and compare these to observed combined effects (Effect AB), with synergy defined as cases where observed effects significantly exceed predicted additive effects. Dose-response curve analysis is particularly informative, comparing EC50 values and Hill slopes between control and APOD-manipulated conditions to detect sensitization effects . Response surface methodology (RSM) provides a more comprehensive approach for complex interactions, generating three-dimensional models of how APOD and growth factor levels jointly influence cellular responses. For time-course data, researchers should analyze area-under-curve (AUC) measurements and employ repeated measures statistical approaches to capture temporal aspects of synergy. Importantly, validation should include multiple readouts (migration distance, Rac1 activation, proliferation) to confirm that synergistic effects extend across different cellular processes rather than reflecting measurement artifacts in a single assay .

How can researchers effectively compare APOD function across different cell types and experimental systems?

Comparing APOD function across different cell types and experimental systems requires standardized methodologies combined with context-specific interpretations to account for biological variation. Researchers should implement a systematic approach beginning with baseline characterization of endogenous APOD expression profiles in each cell type using both RT-qPCR and western blotting, as post-transcriptional regulation may create discrepancies between mRNA and protein levels . Functional assays should employ consistent protocols across cell types, with migration studies utilizing identical substrate coatings, cell densities, and quantification methods to enable direct comparisons . When manipulating APOD expression, researchers should calibrate overexpression or knockdown efficiencies to achieve comparable levels across different cell types, using quantitative western blotting with recombinant protein standards for absolute quantification. Response normalization represents a crucial analytical approach, where each cell type's response to APOD manipulation is calculated as a percentage change from its own baseline, allowing comparison of relative effects rather than absolute values. For mechanistic studies, researchers should systematically assess the same downstream effectors (particularly Rac1 activation) across cell types to determine whether APOD utilizes conserved or context-specific signaling pathways . Meta-analysis techniques can integrate results across multiple studies and cell types, identifying consistent patterns while accounting for heterogeneity. When interpreting differences, researchers should consider cell type-specific factors including receptor expression profiles, baseline migration capacities, and growth factor responsiveness as potential explanatory variables that may influence APOD function in different cellular contexts .

What approaches should be used to validate potential interactions between APOD and GST fusion proteins in biochemical studies?

Validating potential interactions between APOD and GST fusion proteins in biochemical studies requires a multi-faceted approach that addresses both technical artifacts and biological significance. Researchers should implement reciprocal co-immunoprecipitation assays where anti-APOD antibodies pull down the GST fusion protein partner and vice versa, with detection by western blotting using antibodies against both interaction partners. Control experiments are critical and must include GST-only pulls downs to identify non-specific binding to the GST tag itself rather than the fusion partner. Competitive binding assays provide further validation, where increasing concentrations of untagged proteins are added to disrupt interactions in a dose-dependent manner, confirming specificity. Surface plasmon resonance (SPR) or isothermal titration calorimetry (ITC) offers quantitative analysis of binding kinetics and thermodynamics, determining association/dissociation constants and stoichiometry. For structural validation, hydrogen-deuterium exchange mass spectrometry (HDX-MS) can map interaction interfaces by identifying protected regions upon complex formation. In cellular contexts, proximity ligation assays (PLA) visualize interactions within intact cells, while FRET/BRET approaches using fluorescently tagged proteins provide real-time interaction monitoring. Functional validation is essential, testing whether mutations in putative interaction interfaces disrupt both biochemical binding and downstream cellular effects. Researchers should also perform domain mapping experiments using truncated protein variants to identify specific regions responsible for interactions, and cross-linking mass spectrometry to identify precise contact residues between APOD and GST fusion protein partners.

What emerging technologies could advance our understanding of APOD function and regulation?

Emerging technologies offer unprecedented opportunities to advance our understanding of APOD function and regulation beyond current methodological limitations. Single-cell multi-omics approaches represent a frontier technology, enabling simultaneous analysis of APOD at genomic, transcriptomic, and proteomic levels within individual cells to reveal heterogeneity in expression patterns and regulatory mechanisms. Spatial transcriptomics and proteomics techniques can map APOD distribution within tissues with subcellular resolution, providing insights into localization patterns observed in migration studies . For real-time tracking of APOD dynamics, researchers should implement optogenetic tools that allow light-controlled activation or inhibition of APOD expression or function with precise temporal and spatial control. CRISPR-based epigenome editing can manipulate APOD's chromatin environment to investigate epigenetic regulatory mechanisms, while RNA-targeting CRISPR systems enable precise manipulation of APOD mRNA to study post-transcriptional regulation . Protein structure determination through cryo-EM or AlphaFold2 computational prediction can reveal APOD's three-dimensional conformation and potential interaction interfaces. Microfluidic organ-on-chip technologies provide physiologically relevant models for studying APOD function in complex tissue microenvironments, while advanced intravital imaging techniques allow visualization of APOD dynamics in living organisms. Biomaterial approaches using engineered extracellular matrices with controlled stiffness and composition can investigate how mechanical cues influence APOD expression and function in migration contexts . These emerging technologies collectively promise to illuminate APOD's multifaceted roles across cellular contexts and physiological states.

How might comparative studies of APOD and APOE contribute to understanding their distinct functional roles?

Comparative studies of APOD and APOE offer valuable opportunities to differentiate their distinct functional roles despite their shared classification as apolipoproteins. Although the search results primarily focus on these proteins separately, a strategic comparative research program would leverage methodological approaches from both domains . Researchers should implement parallel expression analysis across tissues and developmental stages using RNA-seq and proteomics to identify contexts where APOD and APOE expression patterns diverge or overlap. Functional comparisons require systematically testing both proteins in identical experimental systems, particularly focusing on smooth muscle cell migration where APOD has demonstrated effects , while also examining contexts where APOE variants influence physiological processes like fertility and development . Structural biology approaches comparing the lipid-binding domains of both proteins can reveal differences in lipid preferences and binding mechanisms. For mechanistic insights, researchers should conduct comprehensive signaling pathway analysis, particularly examining whether APOE influences Rac1 activation similar to APOD or operates through distinct signaling cascades. Evolutionary analyses comparing sequence conservation across species can identify functionally important domains unique to each protein. Within human populations, researchers should investigate potential functional interactions between APOD expression levels and APOE genotypes, particularly examining whether the fertility effects associated with APOE-ε4 correlate with altered APOD expression or function. CRISPR-based genome editing enabling protein domain swapping between APOD and APOE would provide definitive evidence regarding which structural elements confer their distinct functional properties, potentially revealing how these related proteins have evolved specialized roles in human physiology.

Product Science Overview

Structure and Function

Apo-D binds to various ligands, including cholesterol, progesterone, pregnenolone, bilirubin, and arachidonic acid . This binding capability makes Apo-D a multi-ligand, multi-functional transporter. It can transport ligands from one cell to another within an organ, scavenge ligands within an organ for transport to the blood, or transport ligands from the circulation to specific cells within a tissue .

Expression and Localization

Apo-D is expressed in numerous tissues, with high levels of expression in the spleen, testes, and brain . It is also present at high concentrations in the cyst fluid of women with gross cystic disease of the breast, a condition associated with an increased risk of breast cancer . Additionally, Apo-D accumulates in regenerating peripheral nerves and in the cerebrospinal fluid of patients with neurodegenerative conditions such as Alzheimer’s disease .

Recombinant Apo-D with GST Tag

The recombinant form of Apo-D, tagged with Glutathione S-transferase (GST), is produced in Escherichia coli (E. coli) and purified using proprietary chromatographic techniques . The GST tag aids in the purification process and enhances the solubility of the protein. The recombinant Apo-D with GST tag shows a 48 kDa band on SDS-PAGE .

Applications and Stability

Recombinant Apo-D is used in various laboratory research applications, including studies on lipid metabolism, neuroprotection, and protein-ligand interactions. The protein is typically formulated in a solution containing Tris-Acetate, EDTA, and glycerol, and it is recommended to store the vial at -20°C to -80°C for long-term stability .

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.