GPD1L Human

Glycerol-3-Phosphate Dehydrogenase 1 Like Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to GPD1L Human

GPD1L (glycerol-3-phosphate dehydrogenase 1-like) is a human gene located on chromosome 3p22.3, encoding a cytoplasmic protein associated with the plasma membrane. It shares 72% sequence identity with GPD1, a key enzyme in glycerolipid metabolism and energy production . GPD1L converts sn-glycerol-3-phosphate to glycerone phosphate, influencing metabolic pathways and ion channel regulation . Mutations in GPD1L are linked to Brugada syndrome (a cardiac arrhythmia disorder) and sudden infant death syndrome (SIDS) .

Enzymatic Activity and Localization

GPD1L catalyzes the oxidation of sn-glycerol-3-phosphate to glycerone phosphate, a step critical for glycerolipid biosynthesis and NAD+/NADH balance . It localizes to the cytoplasm and plasma membrane, interacting with voltage-gated sodium channel SCN5A to modulate cardiac sodium currents .

PropertyValue/Description
Molecular Weight40.6 kDa (recombinant protein) ; 38–40 kDa (observed via Western blot)
Subcellular LocalizationCytoplasm, plasma membrane association
Key DomainsN-terminal NAD+ binding site, C-terminal lysine-206 residue
Recombinant ProductionExpressed in E. coli, fused with N-terminal His-tag (20 aa), purified via chromatography

Functional Interactions

GPD1L interacts with enzymes involved in lipid metabolism and energy regulation:

Interaction PartnerFunctional RoleSTRING Score
GPAT2Glycerol-3-phosphate acylation (lipid biosynthesis) 0.959
GPAMMitochondrial glycerol-3-phosphate acylation 0.957
GPD1Cytosolic glycerol-3-phosphate dehydrogenase (NAD+ shuttle) 0.942
ADPRMADP-ribose/CDP-alcohol diphosphatase (immune signaling) 0.902

These interactions highlight GPD1L’s role in lipid synthesis and redox balance .

Cardiac Disorders

Mutations in GPD1L disrupt sodium channel regulation, causing arrhythmias. The A280V mutation is implicated in Brugada syndrome, characterized by reduced sodium current and increased risk of ventricular fibrillation .

Cancer (Hepatocellular Carcinoma, HCC)

High GPD1L expression in HCC correlates with:

  • Poor prognosis: Advanced tumor stage, larger size, microvascular invasion

  • Metabolic dysregulation: Enrichment of cell cycle, EMT, and E2F target pathways

  • Predictive biomarker: Inverse correlation with therapeutic response to FAK inhibitors (e.g., PF-562271) and other agents

Genetic Variants and Diseases

The LOVD database reports 66 public variants in GPD1L, including missense, nonsense, and frameshift mutations. Key associations include:

DiseaseVariantMechanism
Brugada Syndrome (Type 2)A280V, R164H, GPD1L truncationsAltered SCN5A phosphorylation, reduced sodium current
SIDSGPD1L loss-of-functionDisrupted glycerol-3-phosphate metabolism, arrhythmia risk

Detection Methods

GPD1L is analyzed via:

MethodDetails
Western BlotRabbit polyclonal antibody (1:500–1:2000 dilution), 38–40 kDa band
Recombinant ProteinE. coli-derived, His-tagged, >95% purity (SDS-PAGE)
Single-Cell TranscriptomicsElevated expression in HCC tumor vs. adjacent normal tissue

Metabolic Regulation

GPD1L regulates HIF-1α stability via proline hydroxylation, modulating hypoxia responses . In obesity, GPD1L downregulation (via miR-210) promotes HIF-1α activation, exacerbating insulin resistance and adiposity .

Therapeutic Implications

  • HCC: GPD1L expression predicts resistance to kinase inhibitors, guiding precision therapy

  • Cardiac Diseases: Targeting GPD1L-SCN5A interaction may restore sodium current in Brugada syndrome

Product Specs

Introduction
GPD1L, or Glycerol-3-phosphate dehydrogenase 1-like protein, plays a crucial role in converting sn-glycerol 3-phosphate to glycerone phosphate. This protein is primarily located in the cytoplasm and is associated with the plasma membrane. At the plasma membrane, GPD1L binds to SCN5A, the alpha subunit of the voltage-gated sodium channel, type V. Genetic mutations affecting the GPD1L gene have been linked to serious medical conditions like SIDS (sudden infant death syndrome) and Brugada syndrome type 2, a type of inherited cardiac arrhythmia.
Description
Recombinant GPD1L Human, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 371 amino acids (specifically, amino acids 1 through 351). With a molecular weight of 40.6 kDa, this protein is fused to a 20 amino acid His-tag at its N-terminus. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution that has been sterilized through filtration.
Formulation
The GPD1L protein solution is provided at a concentration of 1 mg/ml. It is formulated in a buffer containing 20mM Tris-HCl (pH 8.0), 20% glycerol, and 1mM DTT.
Stability
For short-term storage (up to 2-4 weeks), the product can be kept at 4°C. For extended storage, it is recommended to freeze the product at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is advisable for long-term storage. Repeated freezing and thawing should be avoided.
Purity
Purity of the GPD1L protein is greater than 95%, as determined by SDS-PAGE analysis.
Synonyms
Glycerol-3-phosphate dehydrogenase 1-like protein, GPD1-L, GPD1L, KIAA0089.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MAAAPLKVCI VGSGNWGSAV AKIIGNNVKK LQKFASTVKM WVFEETVNGR KLTDIINNDH ENVKYLPGHK LPENVVAMSN LSEAVQDADL LVFVIPHQFI HRICDEITGR VPKKALGITL IKGIDEGPEG LKLISDIIRE KMGIDISVLM GANIANEVAA EKFCETTIGS KVMENGLLFK ELLQTPNFRI TVVDDADTVE LCGALKNIVA VGAGFCDGLR CGDNTKAAVI RLGLMEMIAF ARIFCKGQVS TATFLESCGV ADLITTCYGG RNRRVAEAFA RTGKTIEELE KEMLNGQKLQ GPQTSAEVYR ILKQKGLLDK FPLFTAVYQI CYESRPVQEM LSCLQSHPEH T.

Q&A

What is GPD1L and what is its primary function in human physiology?

GPD1L is a member of the glycerol-3-phosphate dehydrogenase family, a protein that shares homology with the enzymes involved in the glycerol 3-phosphate shuttle . The gene was first discovered in 2002 . Its primary physiological function involves interaction with sodium channels, particularly Nav1.5 (encoded by SCN5A), where it regulates channel membrane expression and current . GPD1L appears to play a critical role in cardiac electrophysiology, as evidenced by its association with cardiac arrhythmias when mutated . Studies have shown that functional GPD1L is necessary for normal sodium channel trafficking and function, which is essential for proper cardiac conduction .

How can researchers identify potentially pathogenic GPD1L variants?

Researchers can employ several complementary approaches to identify potentially pathogenic GPD1L variants:

  • Genetic linkage analysis: Using microsatellite or SNP markers to identify co-segregation of variants with disease phenotypes in families. This approach helped identify the original GPD1L-A280V variant with a LOD score >4 in a multigenerational family with Brugada Syndrome .

  • Whole exome sequencing: High-depth sequencing of all coding regions can identify rare variants, which can then be filtered based on population frequency databases like gnomAD .

  • In silico prediction tools: Structural modeling using tools like PyMol can predict the impact of amino acid substitutions on protein structure and function. For example, mutagenesis functions within PyMol can be used to visualize the impact of predicted rotamers and calculate strain scores .

  • Functional validation: Heterologous expression systems (like HEK293 cells) co-transfected with Nav1.5 and wild-type or mutant GPD1L can be used to measure peak sodium current and channel membrane expression .

  • Clinical correlation: Careful phenotyping of patients, including ECG patterns and response to provocative testing (e.g., with procainamide), helps establish genotype-phenotype relationships .

What experimental models are most effective for studying GPD1L function?

Several experimental models have proven valuable for studying GPD1L function:

  • Heterologous expression systems: HEK293 cells transfected with Nav1.5 and GPD1L constructs allow for controlled assessment of electrophysiological parameters. This system demonstrated that GPD1L-A280V decreases peak sodium current and reduces Nav1.5 membrane expression .

  • Primary cardiac myocytes: Neonatal cardiac myocytes have been used to study the effects of GPD1L mutations on sodium current, providing a more physiologically relevant cellular context .

  • Cancer cell lines: For studying GPD1L's role in cancer, researchers have used various cell lines with different GPD1L expression levels. For instance, PLC/PRF/5, HepG2, and Hep3B cell lines with varying GPD1L expression have been used to study drug sensitivity in hepatocellular carcinoma .

  • siRNA knockdown approaches: Targeted suppression of GPD1L expression using siRNA in cell lines like Hep3B has helped establish the relationship between GPD1L expression and sensitivity to specific therapeutic agents .

  • Patient-derived samples: Analysis of clinical samples with appropriate IRB approval provides the most direct evidence of GPD1L's role in human disease .

How does GPD1L expression vary across human tissues?

While the search results don't provide comprehensive information about GPD1L expression across all human tissues, they do offer insights into expression patterns in specific contexts:

  • Cardiac tissue: GPD1L is expressed in cardiac tissue, where it regulates sodium channel function. Mutations in GPD1L in this tissue have been associated with Brugada Syndrome, ventricular tachycardia, and sudden cardiac death .

  • Liver tissue and hepatocellular carcinoma: Spatial and single-cell transcriptome datasets have revealed elevated GPD1L expression in hepatocellular carcinoma (HCC) tumor tissue compared to adjacent normal liver tissue. Expression increases with advancing tumor stage, suggesting positive selection during tumorigenesis .

  • Expression regulation: In HCC, GPD1L exhibits promoter demethylation with advancing tumor stage, indicating epigenetic regulation of its expression .

Researchers studying tissue-specific expression patterns of GPD1L should consider using single-cell RNA sequencing, spatial transcriptomics, and tissue microarrays to obtain comprehensive expression profiles across normal and disease states.

What molecular mechanisms underlie GPD1L's effects on sodium channel function?

The molecular mechanisms by which GPD1L influences sodium channel function appear to be multifaceted:

  • Membrane trafficking regulation: Studies suggest that wild-type GPD1L facilitates the proper trafficking of Nav1.5 to the cell membrane, while mutant forms like GPD1L-A280V decrease Nav1.5 membrane expression .

  • Current modulation: When co-expressed with Nav1.5 in heterologous systems, wild-type GPD1L supports normal peak sodium current, whereas mutant GPD1L (such as A280V, E83K, I124V, and R273C) significantly decreases this current .

  • Interaction with common variants: Research suggests that GPD1L mutations may interact with common variants in sodium channel genes (SCN5A and SCN10A). In one multigenerational family, two common variants associated with Brugada Syndrome were linked to the mutant GPD1L allele in 12 of 14 affected individuals. This genetic interaction may explain why the relatively common A280V-GPD1L variant becomes pathogenic in specific genetic backgrounds .

  • Structural impacts: Structural modeling of GPD1L variants using tools like AlphaFold and PyMol has provided insights into how mutations might alter protein conformation and function. For example, the A280V mutation can be analyzed for rotamer strain scores to predict structural perturbations .

Researchers investigating these mechanisms should consider combining structural biology approaches with electrophysiological studies and protein-protein interaction analyses to fully elucidate how GPD1L modulates sodium channel function.

How can researchers reconcile the variable penetrance and expressivity of GPD1L mutations in Brugada Syndrome?

The variable penetrance and expressivity of GPD1L mutations in Brugada Syndrome present significant research challenges. Several approaches can help address this complexity:

  • Comprehensive family studies: Detailed phenotyping of all family members carrying GPD1L mutations, including those with subtle or no clinical manifestations. The case of patient II-7, an obligate carrier with Type III BrS ECG pattern whose male children displayed Type I and Type II patterns, exemplifies incomplete penetrance that is more common in women .

  • Genetic modifier analysis: Identifying additional genetic variants that modify the phenotypic expression of GPD1L mutations. For instance, the co-inheritance of common variants in SCN5A and SCN10A with GPD1L-A280V may exacerbate sodium current loss and explain variable penetrance .

  • Sex-specific factors: Investigating sex-specific modifiers, given the observation that incomplete penetrance is more common in women .

  • Environmental triggers: Studying how environmental factors (medications, electrolyte disturbances, fever) may trigger clinical manifestations in mutation carriers.

  • Functional gradients: Assessing the functional impact of different GPD1L mutations through standardized in vitro assays may help establish genotype-phenotype correlations.

Researchers should implement an integrated approach combining clinical, genetic, and functional data to develop predictive models for disease penetrance and expressivity, which could improve risk stratification and management strategies for affected families.

What is the biological basis for GPD1L's paradoxical role in cancer progression?

The seemingly paradoxical role of GPD1L in cancer, particularly its association with poor prognosis in hepatocellular carcinoma (HCC), presents an intriguing research question:

  • Metabolic reprogramming: High GPD1L expression in HCC is associated with metabolic dysregulation, suggesting that GPD1L may contribute to the altered metabolic state that supports cancer cell survival and proliferation .

  • Cell cycle regulation: GPD1L overexpression correlates with enrichment of gene sets related to cell cycle control, epithelial-mesenchymal transition (EMT), and E2F targets, indicating its involvement in key oncogenic pathways .

  • Epigenetic regulation: GPD1L exhibits promoter demethylation with advancing tumor stage in HCC, suggesting that epigenetic mechanisms drive its increased expression during cancer progression .

  • Therapeutic resistance mechanisms: The inverse correlation between GPD1L expression and sensitivity to certain therapeutic agents (PF-562271, Linsitinib, and BMS-754807) suggests that GPD1L may modulate signaling pathways involved in drug response, particularly those targeting focal adhesion kinase (FAK) and insulin-like growth factor 1 receptor (IGF1R) .

  • Context-dependent functions: GPD1L may function differently depending on the cellular context and tissue type, potentially explaining why it appears to be a tumor suppressor in some cancers but is associated with poor prognosis in HCC.

Researchers investigating this paradox should consider multi-omics approaches that integrate transcriptomic, proteomic, and metabolomic data, along with functional studies that directly manipulate GPD1L expression in different cancer models to elucidate its context-specific roles.

How can GPD1L expression be leveraged as a predictive biomarker for therapeutic response?

Research suggests GPD1L has potential as a predictive biomarker for treatment response, particularly in hepatocellular carcinoma. Multiple approaches can help develop and validate this potential:

  • Correlation analysis with drug sensitivity databases: Analysis of GPD1L mRNA levels in HCC cell lines from databases like GDSC1 has revealed inverse correlations with response to specific therapeutic agents. For example, HCC cell lines with higher GPD1L expression showed reduced sensitivity to PF-562271 (FAK inhibitor), Linsitinib, and BMS-754807 (both IGF1R inhibitors) .

  • In silico validation: Computational approaches like the oncoPredict algorithm can be used to impute IC50 values in patient datasets like TCGA LIHC, predicting differential responses to agents such as BMS-754807 and PF-562271 based on GPD1L expression .

  • In vitro validation: Testing drug sensitivity in cell lines with varying natural GPD1L expression levels (e.g., PLC/PRF/5, HepG2, and Hep3B) provides functional validation of predicted associations .

  • Genetic manipulation studies: Knockdown experiments using siRNA to suppress GPD1L expression in cell lines can directly test the causal relationship between GPD1L expression and drug sensitivity. For instance, GPD1L knockdown in Hep3B cells reduced sensitivity specifically to PF-562271 .

  • Mechanistic studies: Investigating the signaling pathways linking GPD1L to drug response mechanisms, such as those involving FAK and IGF1R, can provide biological rationale for observed correlations.

Researchers developing GPD1L as a predictive biomarker should integrate these approaches with prospective clinical validation studies to establish robust evidence for its clinical utility in treatment selection, particularly for targeted therapies like FAK inhibitors in HCC patients.

What experimental approaches can best determine the pathogenicity of novel GPD1L variants?

Determining the pathogenicity of novel GPD1L variants requires a multi-faceted approach:

Researchers should integrate these approaches to build comprehensive evidence portfolios for novel GPD1L variants, recognizing that pathogenicity may be influenced by genetic background and environmental factors.

What are the major challenges in translating GPD1L research into clinical applications?

Several significant challenges hinder the translation of GPD1L research into clinical applications:

  • Variant interpretation uncertainties: The explosion of genetic data has led to a "plethora of putative mutations but a dearth of scientific evidence supporting the pathogenicity of these variants" . This uncertainty complicates clinical genetic testing and counseling.

  • Inconsistent classification systems: Frameworks like ClinGen have downgraded GPD1L as a potential cause of Brugada Syndrome despite evidence from family studies, highlighting difficulties in standardizing variant classification .

  • Low genetic testing yield: Current genetic testing in Brugada Syndrome identifies causative mutations in only approximately 25% of cases, limiting the clinical utility of testing .

  • Limited understanding of modifier effects: The variable penetrance and expressivity of GPD1L mutations suggest the influence of genetic modifiers and environmental factors that remain incompletely characterized .

  • Contradictory roles in different diseases: GPD1L appears to have context-specific functions, acting as a contributor to arrhythmogenic disease in some contexts while being associated with poor cancer prognosis in others .

  • Therapeutic targeting complexities: GPD1L's involvement in fundamental cellular processes makes it challenging to develop targeted interventions without unintended consequences.

Researchers addressing these challenges should focus on developing standardized, evidence-based frameworks for variant interpretation, expanding the investigation of genetic modifiers, and exploring tissue-specific and context-dependent functions of GPD1L to inform precision medicine approaches.

How might single-cell and spatial transcriptomic technologies advance our understanding of GPD1L function?

Emerging single-cell and spatial technologies offer powerful approaches to elucidate GPD1L's context-specific functions:

  • Cellular heterogeneity analysis: Single-cell RNA sequencing can reveal cell type-specific expression patterns of GPD1L in complex tissues like heart and liver, potentially identifying specific cellular populations where GPD1L plays critical roles .

  • Spatial context definition: Spatial transcriptomics can map GPD1L expression within tissue microenvironments, providing insights into its function in relation to anatomical features such as nodal tissue in the heart or tumor margins in cancer .

  • Co-expression network identification: Single-cell approaches enable the identification of genes co-expressed with GPD1L in specific cell types, revealing potential functional partners and pathways.

  • Temporal dynamics assessment: Single-cell technologies applied across developmental timepoints or disease progression stages can track changes in GPD1L expression and function over time.

  • Response to perturbation: Combining single-cell approaches with genetic or pharmacological perturbations can elucidate how manipulation of GPD1L affects cellular states and intercellular communication.

  • Integration with multi-omics data: Combining single-cell transcriptomics with proteomics, metabolomics, and epigenomics can provide a comprehensive view of how GPD1L functions within cellular regulatory networks.

Researchers utilizing these technologies should focus on integrative analyses that connect transcriptional patterns to functional outcomes and disease phenotypes, potentially revealing new therapeutic targets or biomarkers related to GPD1L function.

Product Science Overview

Structure and Expression

The GPD1L protein is a member of the NAD-dependent glycerol-3-phosphate dehydrogenase family. It is primarily found in the cytoplasm and is associated with the plasma membrane. The protein is highly expressed in heart tissue, with lower levels in skeletal muscle, kidney, lung, and other organs .

Function

GPD1L plays a significant role in regulating cardiac sodium current. It binds to the sodium channel, voltage-gated, type V, alpha subunit (SCN5A). Decreased enzymatic activity of GPD1L leads to increased levels of glycerol-3-phosphate, which activates the DPD1L-dependent SCN5A phosphorylation pathway. This may ultimately result in decreased sodium current. Additionally, alterations in the NAD(H) balance induced by GPD1L can also reduce cardiac sodium current .

Clinical Significance

Mutations or defects in the GPD1L gene are associated with several medical conditions:

  • Brugada Syndrome Type 2 (BRS2): This is an autosomal dominant tachyarrhythmia characterized by right bundle branch block and ST segment elevation on an electrocardiogram (ECG). It can cause the ventricles to beat so fast that the blood is prevented from circulating efficiently in the body, leading to fainting or sudden death if the heart is not reset .
  • Sudden Infant Death Syndrome (SIDS): GPD1L defects are also linked to SIDS, which is the sudden death of an infant younger than one year that remains unexplained after a thorough case investigation, including a complete autopsy, examination of the death scene, and review of clinical history .
Recombinant GPD1L

Recombinant human GPD1L is produced using an expression system, typically in Escherichia coli. The recombinant protein is purified to a high degree of purity, often greater than 95%, and is used in various research applications, including SDS-PAGE and mass spectrometry .

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.