GPD2 Human

Glycerol-3-Phosphate Dehydrogenase 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

GPD2 Human Recombinant produced in E.Coli is single, a non-glycosylated, Polypeptide chain containing 558 amino acids fragment (43-600) corresponding to the GlpA domain fragment of the mature protein, having a total molecular mass of 66.26kDa and fused with a 4.5kDa amino-terminal hexahistidine tag.
The GPD2 is purified by proprietary chromatographic techniques.

Product Specs

Introduction

Mitochondrial glycerol-3-phosphate dehydrogenase (GPD2) is a calcium-sensitive enzyme that binds to flavin adenine dinucleotide (FAD) and resides on the outer surface of the inner mitochondrial membrane. It exists in two isoforms: Isoform 1, containing 727 amino acids, and Isoform 2, lacking 126 amino acids at the N-terminus. GPD2 catalyzes the oxidation of glycerol-3-phosphate to dihydroxyacetone phosphate (DHAP) while simultaneously reducing the FAD molecule bound to the enzyme. As a testis-specific promoter of mitochondrial GPDH, GPD2, along with a cytosolic NAD-linked GPD, forms the glycerol phosphate shuttle. This shuttle utilizes the interconversion of glycerol-3-phosphate and DHAP to transport reducing equivalents into mitochondria, ultimately leading to the reoxidation of NADH generated during glycolysis. Deficiencies in GPD2 contribute to impaired glucose-stimulated insulin secretion in various animal models of non-insulin-dependent diabetes mellitus. Conversely, in a highly glycolytic environment, GPD2 upregulation contributes to the overall increase in reactive oxygen species (ROS) generation, potentially leading to the progression of prostate cancer.

Description
Recombinant human GPD2, expressed in E. coli, is a non-glycosylated polypeptide chain containing a 558 amino acid fragment (residues 43-600). This fragment corresponds to the GlpA domain of the mature protein and has a molecular mass of 66.26 kDa, including a 4.5 kDa amino-terminal hexahistidine tag. GPD2 is purified using proprietary chromatographic techniques.
Physical Appearance
A clear solution that has been sterilized by filtration.
Formulation
The GPD2 protein solution is supplied in a buffer containing 20mM Tris-HCl (pH 8), 1mM EDTA, and 50% glycerol.
Stability
For best results, store the vial at 4°C if it will be used within 2-4 weeks. For longer storage periods, freeze the solution at -20°C. Repeated freezing and thawing of the solution should be avoided.
Purity
The purity of the GPD2 protein is greater than 95.0% as determined by SDS-PAGE analysis.
Synonyms
Glycerol-3-phosphate dehydrogenase mitochondrial, glycerol-3-phosphate dehydrogenase 2 (mitochondrial), GPDH-M, GPD-M, mtGPD, GPD2, GDH2, GPDM, mGPDH.
Source
Escherichia Coli.

Q&A

What is the molecular structure and localization of human GPD2?

GPD2, also known as glycerol-3-phosphate dehydrogenase 2 (mitochondrial) or GPDM, is a 727 amino acid protein belonging to the FAD-dependent glycerol-3-phosphate dehydrogenase family. The protein contains two EF-hand domains that are critical for its enzymatic activity and regulation. GPD2 localizes specifically to the outer surface of the inner mitochondrial membrane, positioning it strategically for its involvement in electron transport processes .

The GPD2 gene is located on human chromosome 2q24.1, and the homologous mouse gene maps to chromosome 2 C1.1. The protein exists as two alternatively spliced isoforms, allowing for potential tissue-specific functions. For experimental detection, antibodies targeting epitopes between amino acids 463-492 within the internal region of human GPD2 have been successfully employed in various research applications including Western blotting, immunoprecipitation, and immunofluorescence .

What are the primary biochemical functions of GPD2 in normal cellular metabolism?

GPD2 catalyzes the conversion of glycerol-3-phosphate (G3P) to dihydroxyacetone phosphate (DHAP) while simultaneously reducing enzyme-bound FAD. This reaction is a critical component of the glycerol phosphate shuttle, which functions in conjunction with cytosolic GPD1 (a NAD-linked glycerol-3-phosphate dehydrogenase) to facilitate the reoxidation of NADH formed during glycolysis .

The glycerol phosphate shuttle serves as an important metabolic link between several key pathways:

  • It connects glycolysis to oxidative phosphorylation by transferring reducing equivalents from the cytosol to mitochondria

  • It serves as an interface between glucose and lipid metabolism through the interconversion of G3P and DHAP

  • It contributes to the electron transport chain (ETC) by transferring electrons to coenzyme Q (CoQ), thereby supporting ATP generation

While the enzymatic reaction is theoretically reversible, under physiological conditions, GPD2 predominantly operates in the direction that generates DHAP. The transport of G3P and DHAP between the mitochondrial intermembrane space and cytosol is facilitated by voltage-dependent anion channels (VDACs) on the mitochondrial outer membrane .

What are the most effective techniques for studying GPD2 expression and activity in cellular models?

For comprehensive investigation of GPD2 in cellular models, researchers should employ a multi-modal approach:

Gene Expression Analysis:

  • RT-qPCR for mRNA quantification

  • RNA-seq for transcriptomic profiling and splicing variant detection

  • Western blotting using specific antibodies (such as mouse monoclonal antibodies targeting epitopes between amino acids 463-492) at dilutions ranging from 1:100 to 1:1000

  • Immunofluorescence (starting at 1:50 dilution) for subcellular localization studies

Enzymatic Activity Assays:

  • Spectrophotometric measurement of GPD2 activity by monitoring the reduction of artificial electron acceptors

  • Oxygen consumption rate (OCR) measurement using a Seahorse XF analyzer to assess contribution to mitochondrial respiration

  • Isotope tracing experiments using 13C-labeled substrates to track flux through the glycerol phosphate shuttle

Genetic Manipulation:

  • CRISPR-Cas9 system for GPD2 knockout, as demonstrated in the 4T1 cell line using guide RNA sequences:

    • 5'-TCAGGTGAGCCTGGCATATGTGG-3'

    • 5'-GCACTAGATGCCGTCACCAGAGG-3'

  • Overexpression systems using vectors like Gpd2 (BC021359) Mouse Untagged Clone with subsequent selection using 400 μg/mL Geneticin

  • siRNA or shRNA for transient knockdown studies

Pharmacological Inhibition:

  • Chemical inhibitors such as KM04416 (at concentrations of 5-20 μM) for acute inhibition of GPD2 activity

When selecting cellular models for GPD2 research, consider tissue relevance - GPD2 is widely expressed but shows highest expression in pancreatic islets, which is particularly important when studying its role in glucose metabolism and diabetes .

How can researchers effectively generate and validate GPD2 knockout models?

Generating reliable GPD2 knockout models requires careful consideration of experimental design and thorough validation:

CRISPR-Cas9 Knockout Generation:

  • Design guide RNAs targeting exons with essential functional domains. Example sequences that have been successfully used include:

    • 5'-TCAGGTGAGCCTGGCATATGTGG-3'

    • 5'-GCACTAGATGCCGTCACCAGAGG-3'

  • Transfect cells with CRISPR-Cas9 vectors using an appropriate transfection reagent like Lipofectamine 3000 for 24 hours

  • Select transfected cells using an appropriate antibiotic (e.g., 5 μg/mL puromycin for over 72 hours)

  • Establish single-cell-derived clones through limiting dilution in 96-well plates

Comprehensive Validation Approaches:

  • Protein Expression Verification:

    • Western blot analysis to confirm complete absence of GPD2 protein

    • Immunofluorescence microscopy as a complementary approach to verify loss of GPD2 in cellular compartments

  • Genomic Validation:

    • PCR amplification and sequencing of the targeted genomic region to confirm mutations

    • Next-generation sequencing to rule out off-target effects

  • Functional Validation:

    • Enzymatic activity assays to confirm loss of GPD2 function

    • Metabolite analysis to detect expected changes in G3P and DHAP levels

    • Rescue experiments by reintroducing wild-type GPD2 to confirm phenotype specificity

  • Phenotypic Characterization:

    • Growth curves to assess proliferation defects

    • Metabolomic profiling to identify broader metabolic perturbations

    • In vivo tumor progression studies for cancer models

For rescue experiments specifically, researchers have successfully used:

  • Supplementation with 2 mM DHA (dihydroxyacetone) as a DHAP precursor

  • Addition of 100 μM plasmalogen PC (18:0p/18:1) prepared in liposome form

These approaches confirm the specificity of observed phenotypes and help rule out potential adaptation or compensation mechanisms that might arise during knockout generation.

What are the mechanisms linking GPD2 to cancer progression?

GPD2 contributes to cancer progression through both bioenergetic and non-bioenergetic mechanisms, with recent evidence highlighting the latter as particularly significant:

The GPD2-Ether Lipid-Akt Axis:
Studies using mtDNA-deficient (ρ0) cancer cells and GPD2 knockout models have revealed a novel non-bioenergetic pathway by which GPD2 promotes tumor growth. This mechanism involves:

  • Production of dihydroxyacetone phosphate (DHAP) by GPD2, which serves as a critical precursor for ether lipid biosynthesis

  • Biosynthesis of ether lipids (including plasmalogens) through a peroxisomal pathway involving:

    • Glyceronephosphate O-acyltransferase (GNPAT)

    • Alkylglycerone phosphate synthase (AGPS)

    • Fatty acyl-CoA reductase 1 (FAR1)

  • Ether lipid-mediated activation of the Akt/mTORC1 signaling pathway, which promotes cancer cell growth and survival

  • Rescue of growth defects in GPD2 knockout cells through supplementation with either:

    • DHAP precursors (such as dihydroxyacetone)

    • Exogenous ether lipids (like plasmalogen PC)

Tissue-Specific Oncogenic Mechanisms:
GPD2 exhibits varying contributions to cancer progression depending on tumor type:

  • In thyroid cancer, upregulated GPD2 enhances oxidative phosphorylation rates

  • In glioma, GPD2 activation increases glycolytic flux

  • In liver cancer, GPD2 supports anchorage-independent growth through energy metabolism regulation

This metabolic function is particularly relevant as ether lipids are elevated in various cancer cell lines and tumors, with increased levels reported in breast and colon cancer patients . The connection between GPD2 activity, ether lipid metabolism, and the Akt signaling pathway represents a novel therapeutic target for cancer intervention.

How does GPD2 function as a host restriction factor in HBV replication?

GPD2 has been identified as a significant host restriction factor in hepatitis B virus (HBV) replication, representing a novel antiviral mechanism distinct from its metabolic functions. Research has elucidated the following mechanisms:

  • Direct Interaction with Viral Components:
    GPD2 inhibits HBV replication through the recruitment of TRIM28 (tripartite motif-containing protein 28), a multifunctional protein with known antiviral activities. This interaction leads to the degradation of HBx, a viral regulatory protein essential for efficient HBV replication .

  • HBx-GPD2 Interaction as a Therapeutic Target:
    The specific interaction between GPD2 and the viral protein HBx represents a potential target for anti-HBV drug development. Enhancing this interaction or preventing HBx from neutralizing GPD2's restrictive effects could yield novel therapeutic approaches .

  • Metabolic Linkage to Antiviral Activity:
    As a key enzyme in the glycerol-3-phosphate shuttle, GPD2 plays a role in cellular energy metabolism that may create an intracellular environment less conducive to viral replication. This metabolic component of GPD2's antiviral activity warrants further investigation .

This finding adds to our understanding of host-virus interactions and identifies GPD2 as part of the innate cellular defense against HBV infection. Exploiting this natural restriction mechanism could provide alternative approaches to developing antiviral therapeutics with potentially fewer side effects than direct-acting antivirals.

What is the role of GPD2 in glucose metabolism and diabetes?

GPD2 plays a critical role in glucose homeostasis and pancreatic β-cell function, with significant implications for diabetes:

Pancreatic β-cell Function:
GPD2 is expressed at particularly high levels in human pancreatic islets, where it serves an essential function in glucose-sensing mechanisms. The enzyme contributes to the generation of ATP through the glycerol phosphate shuttle, which is crucial for glucose-stimulated insulin secretion (GSIS) .

Diabetes Pathophysiology:
Decreased levels or activity of GPD2 leads to impaired glucose-stimulated insulin release, a hallmark of non-insulin-dependent diabetes mellitus (Type 2 diabetes). This establishes GPD2 as a key molecular link in the pathophysiology of diabetes .

Metabolic Regulatory Roles:
GPD2 has several additional metabolic functions relevant to diabetes:

  • It is a target for metformin, a common diabetes medication, which suppresses gluconeogenesis through GPD2 inhibition

  • It regulates hepatic gluconeogenesis from glycerol, an important metabolic pathway in diabetes

  • It participates in retrograde electron transport, which can influence inflammatory responses associated with metabolic diseases

Potential Therapeutic Applications:
The specific localization and function of GPD2 in pancreatic β-cells make it a potential target for diabetes intervention. Modulating GPD2 activity could enhance insulin secretion in response to glucose, providing a novel approach to diabetes management. Conversely, the inhibition of GPD2 by metformin highlights the complex balance required when targeting this enzyme therapeutically.

Understanding the tissue-specific roles of GPD2 in glucose metabolism offers insights into both the pathogenesis of diabetes and potential treatment strategies aimed at restoring normal glucose homeostasis.

How do mitochondrial dynamics affect GPD2 function and regulation?

The relationship between mitochondrial dynamics and GPD2 function represents an emerging research area with significant implications:

Mitochondrial Localization and Activity:
GPD2 is strategically positioned at the outer surface of the inner mitochondrial membrane, placing it at a critical interface for electron transport and metabolite exchange. This localization enables GPD2 to:

  • Transfer electrons to coenzyme Q in the respiratory chain

  • Interact with voltage-dependent anion channels (VDACs) that facilitate transport of G3P and DHAP across the outer membrane

Effects of Mitochondrial Architecture:
Mitochondrial dynamics (fission, fusion, and structural remodeling) likely influence GPD2 activity through:

  • Changes in membrane curvature affecting enzyme conformation and activity

  • Redistribution of GPD2 within the mitochondrial network during dynamic events

  • Alterations in the proximity of GPD2 to other components of metabolic pathways

mtDNA Depletion Studies:
Research using mtDNA-deficient (ρ0) cancer cells has revealed that despite extensive metabolic downregulation, these cells exhibit high glycerol-3-phosphate levels due to decreased GPD2 activity. This suggests a complex relationship between mitochondrial genetics, respiratory function, and GPD2 regulation .

Research Methodology for Investigating Mitochondrial-GPD2 Interactions:

  • Super-resolution microscopy to visualize GPD2 distribution within dynamic mitochondrial networks

  • Proximity labeling techniques (BioID, APEX) to identify proteins interacting with GPD2 in different mitochondrial states

  • Live-cell imaging with fluorescent GPD2 constructs during induced mitochondrial fission or fusion events

  • Mitochondrial isolation and subfractionation to assess GPD2 distribution and activity in different mitochondrial compartments

Understanding these relationships could provide insights into mitochondrial biology and identify new approaches to modulating GPD2 activity in pathological conditions where mitochondrial dynamics are altered, such as neurodegenerative diseases, cancer, and diabetes.

What are the technical challenges in targeting GPD2 for therapeutic development?

Developing therapeutics targeting GPD2 presents several significant technical challenges that researchers must address:

Specificity Considerations:

  • Isoform Selectivity: Distinguishing between GPD2 and its cytosolic counterpart GPD1 is essential, particularly when both enzymes catalyze similar reactions with opposite directionality

  • Mitochondrial Targeting: Delivering compounds specifically to mitochondria where GPD2 resides requires specialized approaches such as:

    • Lipophilic cation-based carriers (e.g., triphenylphosphonium)

    • Mitochondrial targeting sequences

    • Nanoparticle formulations with mitochondrial affinity

Tissue-Specific Considerations:

  • Differential Expression: GPD2 exhibits varying expression levels across tissues, with highest expression in pancreatic islets

  • Context-Dependent Functions: GPD2 inhibition may be beneficial in cancer contexts but potentially detrimental in pancreatic β-cells where it supports insulin secretion

  • Development of Tissue-Selective Delivery Systems: Methods such as:

    • Antibody-drug conjugates targeting tissue-specific surface markers

    • Nanoparticles with tissue-tropic properties

    • Prodrug approaches activated by tissue-specific enzymes

Pharmacological Approaches:

  • Chemical Inhibitors: Compounds like KM04416 have shown efficacy in inhibiting GPD2 at concentrations of 5-20 μM in cellular models , but further optimization for:

    • Increased potency

    • Improved pharmacokinetics

    • Reduced off-target effects
      is required for clinical development

  • Biologic Approaches: Alternative strategies including:

    • Antisense oligonucleotides targeting GPD2 mRNA

    • siRNA delivery systems for transient knockdown

    • Protein-protein interaction disruptors for pathology-specific functions

Biomarker Development:
Establishing reliable biomarkers to monitor GPD2 inhibition in vivo through:

  • Measurement of substrate/product ratios (G3P/DHAP) in accessible biofluids

  • Development of positron emission tomography (PET) tracers targeting GPD2

  • Identification of downstream metabolic signatures reflective of GPD2 modulation

Addressing these challenges requires interdisciplinary approaches combining structural biology, medicinal chemistry, advanced drug delivery systems, and precision medicine strategies to achieve therapeutic efficacy while minimizing adverse effects.

How can multi-omics approaches advance our understanding of GPD2 regulatory networks?

Multi-omics integration offers powerful approaches to comprehensively map GPD2 regulatory networks and functional impacts:

Integrated Omics Strategies:

  • Metabolomics:

    • Targeted analysis of glycerol-3-phosphate, DHAP, and ether lipid species to directly track GPD2 activity

    • Global untargeted metabolomics to identify broader metabolic consequences of GPD2 modulation

    • Stable isotope tracing (e.g., using 13C-labeled substrates) to quantify metabolic flux through GPD2-dependent pathways

  • Lipidomics:

    • Comprehensive profiling of ether lipids, particularly plasmalogens, which are downstream products of GPD2 activity

    • Analysis of membrane lipid composition changes following GPD2 manipulation

    • Spatial lipidomics to map subcellular distribution of GPD2-dependent lipid species

  • Transcriptomics:

    • RNA-seq analysis to identify genes co-regulated with GPD2 across different conditions

    • Single-cell transcriptomics to detect cell-type specific GPD2 regulatory networks

    • Alternative splicing analysis to characterize GPD2 isoform expression patterns

  • Proteomics:

    • Proximity labeling approaches to identify GPD2 protein interaction networks

    • Phosphoproteomics to map signaling changes downstream of GPD2, particularly in the Akt/mTORC1 pathway

    • Thermal proteome profiling to identify proteins stabilized or destabilized by GPD2 activity

Computational Integration Methods:

  • Network analysis algorithms to identify hub proteins connecting GPD2 to broader cellular processes

  • Machine learning approaches to predict functional consequences of GPD2 perturbation

  • Pathway enrichment analysis to contextualize GPD2 within known biological processes

  • Causal reasoning algorithms to infer directionality in regulatory relationships

Example Research Applications:

  • Cancer Metabolism: Multi-omics has revealed the previously unrecognized GPD2-ether lipid-Akt axis in tumor progression, demonstrating how GPD2 influences cancer growth through non-bioenergetic mechanisms

  • Antiviral Mechanisms: Integrating proteomics and functional genomics has identified GPD2 as a host restriction factor in HBV replication, suggesting novel therapeutic approaches

  • Metabolic Disease: Comprehensive metabolic profiling in diabetes models can elucidate how GPD2 dysfunction contributes to impaired glucose-stimulated insulin secretion

The implementation of multi-omics approaches provides a systems-level view of GPD2 function, moving beyond isolated pathways to understand its role in broader cellular networks and identifying novel therapeutic opportunities across multiple disease contexts.

What are the most reliable antibodies and detection methods for GPD2 research?

Selecting appropriate detection tools is critical for reliable GPD2 research. Based on validated resources from the literature:

Recommended Antibodies:

  • Mouse Monoclonal Antibodies:

    • Epitope mapping between amino acids 463-492 within the internal region of human GPD2

    • Recommended dilutions:

      • Western blotting: 1:100-1:1000

      • Immunoprecipitation: 1-2 μg per 100-500 μg total protein

      • Immunofluorescence: 1:50-1:500

  • Blocking Peptides:

    • Available for competition studies to validate antibody specificity

    • Typically used at 100 μg peptide in 0.5 ml PBS containing <0.1% sodium azide

Western Blotting Protocols:

  • Sample Preparation:

    • Lysate cells in RIPA buffer supplemented with protease inhibitors

    • For mitochondrial enrichment, use differential centrifugation protocols

    • Load 20-50 μg of total protein per lane

  • Detection Conditions:

    • Use 8% SDS-PAGE gels for optimal separation

    • Transfer to PVDF membranes at 100V for 90 minutes

    • Block with 5% non-fat milk in TBST

    • Incubate with primary antibody overnight at 4°C

    • Use appropriate HRP-conjugated secondary antibodies (anti-mouse IgM)

    • Develop using enhanced chemiluminescence

Alternative Detection Methods:

  • Activity Assays:

    • Spectrophotometric measurement of GPD2 activity using artificial electron acceptors

    • Enzyme-coupled assays monitoring DHAP production

  • Mass Spectrometry:

    • Targeted proteomics approaches using specific peptide signatures

    • Multiple reaction monitoring (MRM) for quantitative analysis

  • Genetic Reporters:

    • CRISPR-based tagging with fluorescent proteins

    • Promoter-reporter constructs for transcriptional regulation studies

When selecting detection methods, consider the specific research question, required sensitivity, and potential for cross-reactivity with other dehydrogenases. Validation using multiple detection methods and appropriate controls (such as GPD2 knockout samples) is strongly recommended for conclusive results.

What methodological approaches are most effective for studying GPD2-dependent ether lipid metabolism?

Investigating GPD2-dependent ether lipid metabolism requires specialized methodological approaches spanning analytical chemistry, cell biology, and genetic manipulation:

Analytical Methods for Ether Lipid Detection:

  • Liquid Chromatography-Mass Spectrometry (LC-MS):

    • For comprehensive ether lipid profiling, particularly plasmalogen species

    • Sample preparation involves lipid extraction protocols optimized for phospholipids

    • Multiple reaction monitoring (MRM) for targeted analysis of specific ether lipid species

  • Nuclear Magnetic Resonance (NMR) Spectroscopy:

    • 1H-13C Heteronuclear Single Quantum Coherence (HSQC) for structural characterization

    • Sample preparation using appropriate solvents (e.g., chloroform-d or deuterated methanol)

    • Spectral analysis using software such as Topspin 3.6.2

  • Thin-Layer Chromatography (TLC):

    • For rapid screening of ether lipid classes

    • Visualization using specific stains for plasmalogen detection

Functional Analysis Methods:

  • Metabolic Labeling:

    • Incorporation of isotope-labeled precursors (13C-glucose, 13C-glycerol) to track carbon flux into ether lipids

    • Pulse-chase experiments to determine synthesis and turnover rates

  • Enzyme Activity Assays:

    • Measuring activities of peroxisomal enzymes involved in ether lipid synthesis (GNPAT, AGPS)

    • Assessing GPD2 activity in relation to DHAP production for ether lipid synthesis

  • Rescue Experiments:

    • Supplementation with DHAP precursors (2 mM dihydroxyacetone)

    • Addition of preformed ether lipids (100 μM plasmalogen PC) prepared as liposomes

    • Preparation protocol: evaporation of chloroform-dissolved lipids under nitrogen, hydration with growth medium, agitation for 1 hour at room temperature, and sonication for 10 minutes

Genetic and Pharmacological Manipulation:

  • GPD2 Knockout Models:

    • CRISPR-Cas9 system with specific guide RNAs targeting GPD2

    • shRNA knockdown for transient suppression

  • GPD2 Inhibition:

    • Chemical inhibitors such as KM04416 at concentrations of 5-20 μM

    • Dose-response and time-course analyses to establish optimal inhibition conditions

  • Peroxisomal Enzyme Manipulation:

    • AGPS or GNPAT knockdown to complement GPD2 studies

    • Ether lipid pathway-specific inhibitors to distinguish from other GPD2 functions

Imaging Approaches:

  • Fluorescent Ether Lipid Analogs:

    • Tracking subcellular distribution and dynamics of ether lipids

    • Live-cell imaging to monitor trafficking between organelles

  • Correlative Light and Electron Microscopy:

    • Visualization of peroxisome-mitochondria contacts relevant for ether lipid metabolism

    • Immunogold labeling for GPD2 and ether lipid biosynthetic enzymes

These methodological approaches enable researchers to comprehensively investigate the newly described GPD2-ether lipid-Akt axis and its implications in cancer progression and other cellular processes.

What are the most promising therapeutic applications for GPD2 modulation?

Based on current understanding of GPD2 functions, several promising therapeutic applications warrant further investigation:

Cancer Therapeutics:

  • Small Molecule Inhibitors:

    • Development of selective GPD2 inhibitors to disrupt the GPD2-ether lipid-Akt axis in tumor cells

    • Potential for combination therapy with existing Akt/mTOR pathway inhibitors for synergistic effects

    • Targeted delivery strategies to limit effects on normal tissues

  • Metabolic Vulnerability Exploitation:

    • Identification of cancer subtypes particularly dependent on GPD2 function

    • Synthetic lethality approaches combining GPD2 inhibition with other metabolic interventions

    • Biomarker development to predict sensitivity to GPD2-targeted therapies

Antiviral Strategies:

  • HBV Treatment Approaches:

    • Enhancement of GPD2's natural restriction factor activity against HBV

    • Development of small molecules that stabilize the GPD2-TRIM28 interaction for degrading HBx

    • Screening for compounds that prevent viral evasion of GPD2-mediated restriction

  • Broad-Spectrum Antiviral Potential:

    • Investigation of GPD2's role against other viral pathogens

    • Determining whether the antiviral mechanism extends beyond HBV

Metabolic Disease Interventions:

  • Diabetes Management:

    • Selective GPD2 activators to enhance glucose-stimulated insulin secretion in pancreatic β-cells

    • Tissue-specific modulation approaches to avoid potentially contradictory effects in different tissues

    • Personalized medicine strategies based on genetic variants affecting GPD2 function

  • Obesity-Related Conditions:

    • Exploration of GPD2's role in adipose tissue metabolism

    • Strategic modulation to influence lipid storage vs. utilization

    • Potential impact on thermogenesis and energy expenditure

Neurological Applications:

  • Neuroprotective Strategies:

    • Given the high ether lipid content in brain tissue, investigation of GPD2 modulation for neuroprotection

    • Potential applications in neurodegenerative diseases where ether lipid metabolism is disrupted

    • Mitochondrial enhancement approaches leveraging GPD2's role in bioenergetics

For effective therapeutic development, researchers must address the tissue-specific functions of GPD2 and develop methods for targeted modulation to maximize efficacy while minimizing off-target effects. The dual role of GPD2 in both bioenergetic and non-bioenergetic processes suggests that context-specific approaches will be necessary for successful therapeutic applications.

How might single-cell technologies advance our understanding of GPD2 heterogeneity in tissues?

Single-cell technologies offer unprecedented opportunities to unravel the heterogeneous expression and function of GPD2 across different cell types and disease states:

Single-Cell Transcriptomics:

  • Cell Type-Specific Expression Patterns:

    • Single-cell RNA sequencing (scRNA-seq) to map GPD2 expression across diverse cell populations

    • Identification of cell types with particularly high or regulated GPD2 expression

    • Discovery of co-expression patterns that might indicate functional modules

  • Disease-Associated Transcriptional Regulation:

    • Comparison of GPD2 expression in normal versus diseased tissues at single-cell resolution

    • Identification of specific cell populations where GPD2 is dysregulated in cancer or metabolic disorders

    • Correlation with other metabolic genes to infer pathway alterations

Spatial Transcriptomics and Proteomics:

  • Tissue Architecture Influence:

    • Spatial mapping of GPD2 expression relative to tissue microenvironment features

    • Correlation with oxygen gradients, nutrient availability, and cellular interactions

    • Integration with histopathological markers in disease contexts

  • Subcellular Localization Studies:

    • Super-resolution imaging of GPD2 protein distribution within single cells

    • Co-localization with mitochondrial markers to assess potential functional heterogeneity

    • Assessment of mitochondrial morphology in relation to GPD2 expression

Single-Cell Metabolomics:

  • Metabolic Phenotyping:

    • Emerging mass spectrometry approaches for single-cell metabolomic analysis

    • Correlation of GPD2 expression with G3P/DHAP ratios and ether lipid profiles

    • Identification of metabolically distinct cell populations within tissues

  • Functional Consequences:

    • Live-cell metabolic imaging using fluorescent sensors for metabolites

    • Single-cell respirometry to assess contribution to cellular bioenergetics

    • Correlation of metabolic states with cell fate decisions or disease progression

Integrative Multi-modal Analysis:

  • Computational Integration:

    • Machine learning approaches to integrate transcriptomic, proteomic, and metabolomic data

    • Trajectory inference to map temporal dynamics of GPD2 regulation

    • Network analysis to position GPD2 within cell type-specific regulatory frameworks

  • Clinical Applications:

    • Patient-derived samples analyzed at single-cell resolution to identify disease-specific patterns

    • Correlation with treatment response and clinical outcomes

    • Development of precision medicine approaches based on cellular heterogeneity

These technologies would be particularly valuable for understanding:

  • The role of GPD2 in specific pancreatic β-cell subpopulations relevant to diabetes

  • Heterogeneity of GPD2 expression and function within tumors

  • Cell type-specific responses to GPD2-targeting therapeutics

  • Developmental regulation of GPD2 in tissue differentiation and maturation

Product Science Overview

Introduction

Glycerol-3-Phosphate Dehydrogenase 2 (GPD2) is a mitochondrial enzyme that plays a crucial role in cellular metabolism. It is part of the glycerol-3-phosphate shuttle, which is essential for transferring reducing equivalents from cytosolic NADH into the mitochondria for oxidative phosphorylation. This enzyme is encoded by the GPD2 gene located on chromosome 3p22.3 in humans .

Structure and Function

GPD2 is a flavoprotein that catalyzes the oxidation of glycerol-3-phosphate to dihydroxyacetone phosphate (DHAP) with the concomitant reduction of flavin adenine dinucleotide (FAD) to FADH2. This reaction is a key step in the glycerol-3-phosphate shuttle, which is one of the two main mechanisms for transferring electrons from NADH in the cytosol to the mitochondrial electron transport chain .

Biological Significance

The glycerol-3-phosphate shuttle, involving GPD2, is particularly important in tissues with high metabolic rates, such as brown adipose tissue and muscle. It plays a significant role in thermogenesis and energy metabolism. Additionally, GPD2 has been implicated in various metabolic diseases and cancers due to its role in cellular energy homeostasis .

Recombinant GPD2

Recombinant GPD2 refers to the enzyme produced through recombinant DNA technology, which allows for the expression of the human GPD2 gene in a host organism, such as bacteria or yeast. This method enables the production of large quantities of the enzyme for research and therapeutic purposes. Recombinant GPD2 retains the same functional properties as the naturally occurring enzyme and is used in various biochemical assays and studies .

Applications in Research

Recombinant GPD2 is widely used in research to study its role in metabolism and disease. It is utilized in enzymatic assays to understand its kinetic properties and interactions with other metabolic pathways. Additionally, recombinant GPD2 is used in structural biology to determine its three-dimensional structure, which can provide insights into its function and regulation .

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.