GNPDA1 is widely expressed across tissues, with elevated levels observed in:
Antibody validation data (12312-1-AP):
Application | Dilution Range | Detected Tissues |
---|---|---|
Western Blot (WB) | 1:500–1:2000 | Human brain, rat kidney |
Immunohistochemistry (IHC) | 1:20–1:200 | Pancreatic cancer tissue |
Proteintech’s GNPDA1 antibody (RRID: AB_2110553) shows reactivity with human, mouse, and rat samples .
GNPDA1 is overexpressed in HCC tissues compared to normal liver () and correlates with advanced tumor stage and poor survival .
Clinical associations (TCGA data):
Mechanistically, GNPDA1 knockdown in HCC cell lines (SMMC-7721, Huh7) reduces proliferation by 40% and inhibits migration/invasion by 60–75% . Its activity fuels glycolysis via Fru6P production and promotes lipogenesis through ammonium-induced SREBP-1 activation .
GNPDA1 interacts with metabolic enzymes:
Partner Protein | Function | Interaction Score |
---|---|---|
GNPNAT1 | GlcN6P acetylation | 0.971 |
GFPT1/2 | HBP rate-limiting step | 0.969 |
AMDHD2 | GlcNAc6P deacetylation | 0.994 |
These interactions position GNPDA1 as a nodal enzyme connecting HBP with nucleotide synthesis and protein glycosylation .
Targeting GNPDA1 may disrupt cancer metabolic reprogramming. Preclinical studies show that:
Ammonium accumulation from GNPDA1 activity enhances autophagy and chemoresistance .
O-GlcNAcylation, a downstream process, stabilizes oncogenic proteins like MEK2 .
Current research focuses on small-molecule inhibitors of its allosteric site to modulate GlcNAc6P binding .
MGSSHHHHHH SSGLVPRGSH MKLIILEHYS QASEWAAKYI RNRIIQFNPG PEKYFTLGLP TGSTPLGCYK KLIEYYKNGD LSFKYVKTFN MDEYVGLPRD HPESYHSFMW NNFFKHIDIH PENTHILDGN AVDLQAECDA FEEKIKAAGG IELFVGGIGP DGHIAFNEPG SSLVSRTRVK TLAMDTILAN ARFFDGELTK VPTMALTVGV GTVMDAREVM ILITGAHKAF ALYKAIEEGV NHMWTVSAFQ QHPRTVFVCD EDATLELKVK TVKYFKGLML VHNKLVDPLY SIKEKETEKS QSSKKPYSD.
GNPDA1 (Glucosamine-6-Phosphate Deaminase 1) belongs to the glucosamine 6-phosphate deaminase family and catalyzes the conversion of glucosamine 6-phosphate to fructose 6-phosphate . This enzyme serves as a critical link between the hexosamine system and the glycolytic pathway, enabling the catabolism of hexosamines derived from glycoproteins, glycolipids, and sialic acids into phosphate sugars to provide energy sources . Through this conversion process, GNPDA1 effectively increases the availability of raw materials for glycolysis, which is particularly significant in understanding cellular energy metabolism. The enzyme's central role in connecting these metabolic pathways makes it an important focus for research into metabolic reprogramming in both normal and disease states.
In normal cellular metabolism, GNPDA1 facilitates the conversion of the hexosamine system to the glycolytic pathway, affecting energy metabolism through the conversion of glucosamine 6-phosphate to fructose 6-phosphate . Under physiological conditions, this enzyme helps maintain the balance between different metabolic pathways and ensures the efficient utilization of hexosamines derived from various cellular components.
In pathological conditions, particularly in cancer, GNPDA1 contributes to metabolic reprogramming—a hallmark of cancer cells . In hepatocellular carcinoma (HCC), for instance, GNPDA1 is highly expressed compared to normal liver tissues . This aberrant expression promotes the "Warburg effect," where tumor cells exhibit an abnormally high rate of glycolysis accompanied by weakened mitochondrial aerobic metabolism . By increasing the conversion of glucosamine to fructose 6-phosphate, GNPDA1 increases the raw materials available for glycolysis, thereby supporting the high energy demands of rapidly proliferating cancer cells.
For measuring GNPDA1 expression in human tissues, researchers commonly employ a multi-modal approach combining RNA and protein detection methods. RNA-sequencing (RNA-seq) has proven particularly valuable for quantifying GNPDA1 transcript levels, as demonstrated in studies utilizing datasets like GSE98622 and GSE134386 . For protein-level detection, Western blot analysis using anti-GNPDA1 antibodies (such as Catalog No.: 12,312-1-AP, 1:1000, Proteintech) provides reliable quantification when paired with loading controls like GAPDH .
Immunohistochemical staining represents another crucial technique for visualizing GNPDA1 expression patterns within tissue architecture, similar to methods used for other proteins like Bsnd and Ranbp3l . For large-scale analyses, bioinformatic approaches leveraging databases such as The Cancer Genome Atlas (TCGA) enable researchers to extract GNPDA1 expression data using platforms like the limma package (version 3.8) in R software (version 3.5.1) . This combination of experimental and computational methods provides comprehensive insights into GNPDA1 expression across different tissues and disease states.
Designing effective GNPDA1 knockdown experiments requires careful consideration of several methodological aspects. Based on established protocols, researchers should begin by selecting appropriate cell lines that express GNPDA1, such as SMMC-7721 and Huh7 for liver-related studies . For RNA interference, the construction of lentiviral vectors carrying short hairpin RNAs (shRNAs) targeting GNPDA1 has demonstrated high efficiency.
The experimental design should include:
Vector selection: The pLV-sh-puro vector has been successfully used for GNPDA1 knockdown
shRNA design: Multiple shRNA sequences (e.g., shGNPDA1-1 and shGNPDA1-2) should be designed based on the human GNPDA1 gene to identify the most effective construct
Lentiviral particle preparation: Co-transfection of the shGNPDA1 vector with packaging plasmids (pMD2G and pspax2) into 293T cells using Lipofectamine 2000
Target cell transduction: Culture target cells with lentivirus-containing medium for 48 hours, followed by selection with puromycin (1.0 μg/mL) for 72 hours
Validation: Confirm knockdown efficiency at both mRNA level (qRT-PCR) and protein level (Western blot)
Control setup: Include proper controls using non-targeting shRNA (shctrl) for comparative analysis
This systematic approach ensures reliable GNPDA1 knockdown for subsequent functional studies.
For GNPDA1-related transcriptomic data analysis, a robust analytical pipeline should incorporate quality control, differential expression analysis, and functional interpretation. Based on established methodologies, researchers should:
Data normalization: Apply appropriate normalization methods as demonstrated in studies utilizing boxplot and PCA for quality assessment
Differential expression analysis: Implement packages like limma (version 3.8) for RNA-seq data analysis to identify differentially expressed genes (DEGs)
Integration of multiple datasets: Consider using Robust Rank Aggregation (RRA) for integrating results from multiple studies as exemplified in analyses combining GSE39548, GSE52004, GSE71647, GSE87025, and GSE131288
Validation with independent datasets: Verify findings using independent RNA-seq datasets (e.g., GSE98622, GSE134386) to ensure reliability
Network analysis: Construct regulatory networks connecting GNPDA1 with transcription factors and miRNAs to elucidate molecular mechanisms
Survival analysis: Conduct Kaplan-Meier analysis for correlating GNPDA1 expression with clinical outcomes, stratifying patients into high and low expression groups
For proteomic data, complement transcriptomic findings with protein expression validation through Western blotting and immunohistochemistry, correlating results with databases like The Human Protein Atlas . This integrated approach enables comprehensive understanding of GNPDA1's biological significance across multiple molecular levels.
The prognostic value of GNPDA1 is further supported by ROC analysis, confirming its potential as a diagnostic biomarker in HCC . Notably, while GNPDA1 expression correlates with several clinicopathological parameters, no significant association has been found with gender, indicating that its prognostic value transcends this demographic factor . These findings collectively establish GNPDA1 as a potential novel prognostic biomarker for HCC, with high expression consistently predicting poor clinical outcomes.
GNPDA1 promotes tumor progression through several interconnected molecular mechanisms centered around metabolic reprogramming and cellular behavior modulation. At the metabolic level, GNPDA1 catalyzes the conversion of glucosamine 6-phosphate to fructose 6-phosphate, which feeds into the glycolytic pathway . This conversion increases the availability of raw materials for glycolysis, supporting the "Warburg effect" characteristic of cancer cells, where glycolysis is abnormally active even in the presence of oxygen .
Functionally, experimental studies using GNPDA1 knockdown in HCC cell lines (SMMC-7721 and Huh7) have revealed that GNPDA1 significantly enhances cellular proliferation, migration, and invasion while inhibiting apoptosis . These effects were demonstrated through multiple methodologies including MTT assay, EdU incorporation, cell cycle analysis, transwell migration/invasion assays, and wound healing assays . The molecular basis for these functional effects likely involves GNPDA1's impact on energy metabolism, providing cancer cells with the metabolic flexibility needed to sustain rapid proliferation and metastatic potential.
GNPDA1 participates in complex molecular networks that connect metabolic pathways with transcriptional regulation in HCC and potentially other cancers. Research has identified interactions between GNPDA1 and transcription factors (TFs) including Junb, Fos, Fosl1, Fosl2, Egr2, and Cebpb, which have been verified in RNA-seq analysis to be up-regulated in renal ischemia-reperfusion injury (IRI) . These TFs may regulate GNPDA1 expression or work in concert with GNPDA1 to influence downstream pathways.
The table below summarizes key molecular interactions identified in recent studies:
Molecular Partner | Relationship with GNPDA1 | Verified In | Functional Implication |
---|---|---|---|
Fosl1, Junb, Fosl2, Egr2, Cebpb | Negative regulation with target genes | GSE52004, GSE98622 | Transcriptional control of metabolic pathways |
Fos | Correlation with Ass1 and Crebl2 | GSE98622 | Inconsistent with GSE52004, suggesting context-specific regulation |
Atf4 | No significant correlation with target genes | GSE98622 | Independent regulatory mechanism |
SPP1, TIMP1, TNC | Co-expression with GNPDA1 | Human HCC samples | Potential cooperative effect in tumor progression |
These interactions suggest that GNPDA1 functions within a broader regulatory network that coordinates metabolic reprogramming with other cancer hallmarks, highlighting its potential importance as a therapeutic target in HCC .
For comprehensive investigation of GNPDA1 function, researchers should employ a multi-faceted approach combining enzymatic, cellular, and molecular assays. Based on established methodologies, the following assays have proven particularly valuable:
Proliferation Assays:
Migration and Invasion Assays:
Apoptosis Assays:
Enzymatic Activity Assays:
Specific assays to measure the conversion of glucosamine 6-phosphate to fructose 6-phosphate
Glycolytic rate measurements to assess downstream metabolic effects
Molecular Interaction Studies:
Co-immunoprecipitation to identify protein-protein interactions
Chromatin immunoprecipitation (ChIP) for studying transcription factor binding
These assays, when applied in combination following GNPDA1 manipulation (overexpression or knockdown), provide comprehensive insights into its functional roles in cellular processes relevant to both normal physiology and disease states.
Establishing causal relationships between GNPDA1 and phenotypic changes requires a systematic experimental approach with appropriate controls and rescue experiments. Based on published methodologies, researchers should implement the following strategy:
Gene Manipulation Techniques:
Validation of Manipulation:
Rescue Experiments:
Re-introduce wild-type GNPDA1 in knockdown/knockout cells
Utilize enzymatically inactive GNPDA1 mutants to distinguish between enzymatic and structural roles
Comprehensive Phenotypic Analysis:
Pathway Validation:
Examine downstream metabolic changes through metabolomics
Analyze expression of pathway components through transcriptomics/proteomics
This multi-layered approach with appropriate controls enables researchers to establish direct causal relationships between GNPDA1 and observed phenotypic changes, distinguishing primary effects from secondary consequences.
For investigating GNPDA1 function in human disease contexts, several in vivo models offer complementary advantages for translational research. Based on current methodologies and disease relevance, researchers should consider:
Xenograft Mouse Models:
Genetically Engineered Mouse Models (GEMMs):
Conditional GNPDA1 knockout or overexpression models using tissue-specific promoters (e.g., Alb-Cre for liver-specific manipulation)
Inducible systems to study temporal aspects of GNPDA1 function
Patient-Derived Xenografts (PDXs):
Implantation of tumor tissues from patients with varying GNPDA1 expression levels to maintain tumor heterogeneity
Correlation of GNPDA1 expression with growth characteristics and treatment responses
Metabolism-Focused Models:
High-fat diet or metabolic stress models to investigate GNPDA1's role in metabolic reprogramming
Combination with genetic backgrounds predisposed to metabolic disorders
Ischemia-Reperfusion Injury Models:
These models should be selected based on specific research questions, with consideration of ethical standards and translational relevance. Combining multiple models provides the most comprehensive understanding of GNPDA1 function in human disease contexts.
Developing GNPDA1 as a prognostic biomarker for hepatocellular carcinoma (HCC) requires systematic validation through multiple clinical datasets and methodological approaches. Based on existing research, the development pathway should include:
Expression Analysis in Large Cohorts:
Analysis of GNPDA1 expression in extensive HCC datasets such as TCGA, which has already demonstrated significant association between high GNPDA1 expression and advanced tumor stage, TNM stage, and grade
Validation in independent cohorts across diverse populations to establish universality of the biomarker
Survival Correlation Studies:
Diagnostic Performance Assessment:
Clinically Applicable Detection Methods:
Development of standardized immunohistochemistry protocols for GNPDA1 detection in routine pathology specimens
Exploration of less invasive detection methods in serum or circulating tumor cells
Integration with Existing Biomarkers:
Combination with established HCC biomarkers to improve prognostic accuracy
Development of integrated scoring systems incorporating GNPDA1 expression
This systematic approach would establish GNPDA1 as a clinically valuable prognostic biomarker for HCC, potentially guiding treatment decisions and patient stratification in clinical trials.
Given GNPDA1's role in metabolic reprogramming and cancer progression, several therapeutic strategies could be developed to target this enzyme or its associated pathways:
Direct Enzymatic Inhibition:
Development of small molecule inhibitors specifically targeting GNPDA1's catalytic domain to block the conversion of glucosamine 6-phosphate to fructose 6-phosphate
Structure-based drug design utilizing crystallographic data of GNPDA1
RNA Interference Therapeutics:
Metabolic Pathway Modulation:
Targeting the hexosamine biosynthetic pathway upstream of GNPDA1
Exploiting synthetic lethality by inhibiting glycolysis in GNPDA1-overexpressing tumors
Transcriptional Regulation:
Combination Therapies:
Combining GNPDA1 inhibition with conventional chemotherapeutics to enhance efficacy
Pairing with immune checkpoint inhibitors to potentially improve immunotherapy responses
These strategies represent promising avenues for therapeutic development, with the potential to exploit cancer cells' dependence on altered metabolism while minimizing effects on normal tissues where GNPDA1 expression is typically lower .
GNPDA1 research has significant potential to contribute to precision medicine approaches for cancer patients, particularly those with hepatocellular carcinoma. This contribution would manifest through several pathways:
Patient Stratification:
GNPDA1 expression profiling could identify patient subgroups with differential prognosis, as evidenced by survival analyses showing poorer outcomes in HCC patients with high GNPDA1 expression
Integration of GNPDA1 status into clinical decision algorithms to guide treatment intensity and monitoring frequency
Treatment Selection:
Development of companion diagnostics to identify patients likely to respond to GNPDA1-targeting therapies
Correlation of GNPDA1 expression with response to existing therapies to optimize treatment selection
Resistance Mechanism Identification:
Investigation of GNPDA1's role in therapy resistance through metabolic adaptation
Development of strategies to overcome resistance by targeting GNPDA1-dependent metabolic pathways
Monitoring Disease Progression:
Serial assessment of GNPDA1 expression or activity as a biomarker for treatment response and disease progression
Development of liquid biopsy approaches to detect GNPDA1-expressing circulating tumor cells or cell-free DNA
Metabolic Profiling Integration:
Combination of GNPDA1 status with broader metabolic profiles to create comprehensive metabolic signatures for individual tumors
Tailoring of dietary or metabolic interventions based on GNPDA1-dependent metabolic vulnerabilities
These applications could substantially advance precision medicine approaches by moving beyond genetic mutation-based stratification to include metabolic reprogramming as a targetable cancer vulnerability, potentially expanding therapeutic options for patients with limited treatment choices.
GNPDA1 catalyzes the deamination of D-glucosamine-6-phosphate, a key step in the hexosamine biosynthetic pathway. This reaction is essential for the production of uridine diphosphate-N-acetylglucosamine (UDP-GlcNAc), which is a critical substrate for glycosylation processes in cells . The enzyme’s activity is allosterically regulated, meaning its function can be modulated by the binding of effector molecules .
The enzyme belongs to the family of hydrolases, specifically those acting on carbon-nitrogen bonds other than peptide bonds . Structural studies have revealed that GNPDA1 functions as a hexamer, with each subunit contributing to the overall catalytic activity . The enzyme’s structure has been extensively studied, with several crystallographic structures available in protein databases .
GNPDA1 is expressed in various tissues and is particularly important in metabolic pathways related to glycolysis and glycosaminoglycan metabolism . It has been implicated in the regulation of cytosolic UDP-GlcNAc levels, which in turn affects hyaluronan synthesis during tissue remodeling . Additionally, GNPDA1 has a role in triggering calcium oscillations in mammalian eggs, which are essential for egg activation and early embryonic development .
Human recombinant GNPDA1 is produced using recombinant DNA technology, which involves cloning the GNPDA1 gene into an expression vector and introducing it into a suitable host organism, such as Escherichia coli. The host cells then express the enzyme, which can be purified and used for various research and industrial applications .