C12ORF5/TIGAR regulates glycolysis and oxidative stress through enzymatic activity:
Fructose-2,6-bisphosphatase Activity: Hydrolyzes fructose-2,6-bisphosphate (Fru-2,6-P₂), a key glycolytic activator, thereby inhibiting glycolysis and redirecting glucose flux to the pentose phosphate pathway (PPP) .
ROS Reduction: Enhances PPP flux to produce NADPH, which regenerates glutathione and reduces intracellular reactive oxygen species (ROS) .
p53-Dependent Regulation: Induced by low oxidative stress via p53, promoting cell survival during mild stress .
C12ORF5 amplification is observed in ovarian, colorectal, and other cancers, correlating with poor prognosis . Knockdown of TIGAR enhances sensitivity to PARP inhibitors (e.g., olaparib) by:
Reducing BRCA1 and Fanconi Anemia Pathway Activity: Impairing DNA repair mechanisms .
Increasing Senescence and Cytotoxicity: Elevating cellular ROS and DNA damage .
Cancer Type | TIGAR Expression | Clinical Impact |
---|---|---|
Ovarian | Amplified | Poor survival, PARP inhibitor resistance |
Colorectal | Overexpressed | Promotes metastasis and autophagy inhibition |
Ischemic Brain Injury: Enhances PPP flux to preserve mitochondrial function .
Cardiac Stress: Reduces mitochondrial ROS via HK2 interaction, though excessive TIGAR may exacerbate apoptosis .
C12ORF5 Human, His is produced via bacterial expression and purified using chromatography:
Parameter | Details | Source |
---|---|---|
Host System | E. coli | |
Formulation | 20 mM Tris-HCl (pH 8.0), 0.2 M NaCl, 2 mM DTT, 10% glycerol | |
Storage | -20°C (long-term) or 4°C (short-term); avoid freeze-thaw cycles |
C12ORF5’s enzymatic activity is quantified using phosphatase assays:
SDS-PAGE Verification: Bands at 27–30 kDa under reducing/non-reducing conditions confirm integrity .
TIGAR inhibition is explored for:
MGSSHHHHHH SSGLVPRGSH MGSHMARFAL TVVRHGETRF NKEKIIQGQG VDEPLSETGF KQAAAAGIFL NNVKFTHAFS SDLMRTKQTM HGILERSKFC KDMTVKYDSR LRERKYGVVE GKALSELRAM AKAAREECPV FTPPGGETLD QVKMRGIDFF EFLCQLILKE ADQKEQFSQG SPSNCLETSL AEIFPLGKNH SSKVNSDSGI PGLAASVLVV SHGAYMRSLF DYFLTDLKCS LPATLSRSEL MSVTPNTGMS LFIINFEEGR EVKPTVQCIC MNLQDHLNGL TETR.
C12ORF5/TIGAR is a 270-amino acid protein encoded by the TIGAR gene (previously known as C12orf5) located on chromosome 12p13-3 in humans. It was discovered in 2005 by Kuang-Yu Jen and Vivian G. Cheung as a p53-regulated gene activated in response to ionizing radiation . The protein functions primarily as a fructose-bisphosphatase that hydrolyzes fructose-2,6-bisphosphate and fructose-1,6-bisphosphate.
TIGAR's primary biological functions include:
Negative regulation of glycolysis by decreasing intracellular levels of fructose-2,6-bisphosphate
Activation of the pentose phosphate pathway (PPP), leading to increased NADPH production
Decreasing reactive oxygen species (ROS) levels and protecting cells from oxidative stress
Providing protection against DNA damage-induced apoptosis
Recombinant Human TIGAR/C12ORF5 His-tag protein typically consists of the human TIGAR sequence (Ala2-Arg270) with a C-terminal 6-His tag . The protein has the following structural characteristics:
Molecular mass of approximately 32.6 kDa (including the His-tag)
Single, non-glycosylated polypeptide chain containing 294 amino acids (270 amino acids from TIGAR sequence plus 24 amino acids from the His-tag)
Histidine phosphatase fold with a phosphate coordinated to a catalytic histidine in the active site
The recombinant protein is typically produced in E. coli expression systems and purified using chromatographic techniques to achieve >90-95% purity as determined by SDS-PAGE .
For optimal stability and activity of recombinant TIGAR/C12ORF5 His-tag protein, researchers should follow these methodological guidelines:
Short-term storage (2-4 weeks): Store at 4°C in the recommended buffer solution
Long-term storage: Store at -20°C with proper aliquoting to avoid freeze-thaw cycles
For extended storage periods, addition of a carrier protein (0.1% HSA or BSA) is recommended to enhance stability unless working with carrier-free preparations
Typical formulation includes:
For carrier-free versions, researchers should be aware that the absence of stabilizing BSA may affect shelf-life but is beneficial for applications where BSA might interfere with experimental outcomes .
Researchers can employ several methodological approaches to measure TIGAR enzymatic activity:
Phosphatase Activity Assay:
Fructose-2,6-bisphosphate Hydrolysis Assay:
Direct measurement of F2,6BP degradation
Detection: Coupled enzymatic assays measuring phosphofructokinase activity
Indirect Assessment via Metabolic Flux Analysis:
Measurement of glycolytic intermediates
Analysis of pentose phosphate pathway activation
NADPH production quantification
A standardized protocol for phosphatase activity includes:
Incubation of TIGAR (typically 2 μg per reaction) with 1.25 mM substrate at 37°C for 3 hours
Reaction termination with 0.2 M NaOH
Spectrophotometric analysis and calculation of specific activity using appropriate conversion factors and controls
TIGAR demonstrates complex roles in cancer biology, exhibiting both tumor-suppressive and tumor-promoting functions depending on cellular context. Research methodologies to investigate TIGAR's role in cancer include:
Metabolic Profiling Approaches:
Isotope tracing using 13C-labeled glucose to track metabolic flux through glycolysis versus pentose phosphate pathway
Measurement of NADPH/NADP+ ratios to assess redox status
Quantification of ribose-5-phosphate production for nucleotide synthesis
In Vitro Cancer Models:
TIGAR knockdown/overexpression studies in cancer cell lines
Assessment of proliferation, migration, invasion capacity
Evaluation of sensitivity to chemotherapeutic agents and radiation
In Vivo Models:
Xenograft studies with TIGAR-modulated cancer cells
Conditional knockout mouse models
Analysis of metastatic potential and tumor microenvironment interactions
Research findings indicate that TIGAR can promote cancer metastasis through:
Enzymatic functions that redirect glucose metabolism toward the pentose phosphate pathway
Interactions with signaling pathways that inhibit autophagy and apoptosis
Nuclear translocation during chemotherapy and hypoxia, promoting cell survival
Mitochondrial translocation during hypoxia, enhancing hexokinase 2 (HK2) activity
TIGAR is highly expressed in various tumor types and represents a potential therapeutic target for cancer treatment strategies .
Distinguishing between TIGAR's metabolic and non-metabolic functions requires sophisticated experimental designs:
Enzymatic Activity Mutations:
Generation of catalytically inactive TIGAR mutants that retain protein-protein interaction capabilities
Compare phenotypes between wild-type and mutant TIGAR expression
Subcellular Localization Studies:
Creation of localization-restricted TIGAR variants (nuclear, cytoplasmic, mitochondrial)
Fluorescent tagging and live cell imaging to track translocation events
Correlation of localization with specific functions
Protein-Protein Interaction Analysis:
Co-immunoprecipitation studies to identify interaction partners in different cellular compartments
Proximity ligation assays to confirm interactions in situ
Mass spectrometry-based interactome analysis
Metabolic Rescue Experiments:
Provision of metabolic intermediates to bypass TIGAR's enzymatic functions
Administration of antioxidants to neutralize ROS-related effects
Direct NADPH supplementation to distinguish between redox and other functions
Research has revealed that TIGAR's protective function during hypoxia occurs through a mechanism independent of its fructose-bisphosphatase activity, involving interactions with HK2 . Similarly, TIGAR's interaction with ATP5A1 reduces oxidative stress through protein-protein interactions rather than enzymatic activity .
When designing experiments with recombinant TIGAR protein, researchers should implement these essential controls:
Control Type | Purpose | Implementation |
---|---|---|
Enzyme-free control | Account for spontaneous substrate degradation | Reaction mixture without TIGAR |
Heat-inactivated TIGAR | Verify enzymatic versus non-enzymatic effects | TIGAR protein denatured at 95°C for 10 minutes |
Catalytically inactive mutant | Distinguish enzymatic from structural roles | H11A or equivalent mutation in catalytic site |
Concentration gradient | Establish dose-response relationship | Serial dilutions of TIGAR protein |
Time course | Determine reaction kinetics | Sampling at multiple time points |
Buffer composition controls | Evaluate buffer component effects | Variations in pH, salt concentration, reducing agents |
Substrate specificity | Confirm enzymatic selectivity | Multiple potential substrates (F2,6BP, F1,6BP, pNPP) |
For phosphatase activity assays specifically, an enzyme control prepared by combining TIGAR with NaOH before substrate addition is critical to account for background signal and ensure accurate activity calculations .
Contradictory findings regarding TIGAR's role in autophagy regulation reflect its context-dependent functions. Methodological approaches to resolve these contradictions include:
Cell Type-Specific Analysis:
Systematic evaluation across multiple cell types (primary cells, cancer cell lines, non-transformed cells)
Correlation with baseline metabolic state (glycolytic vs. oxidative phosphorylation-dependent)
Analysis of p53 status and correlation with autophagy outcomes
Stress Condition Stratification:
Precise categorization of stress stimuli (nutrient deprivation, hypoxia, DNA damage)
Temporal analysis of TIGAR expression and autophagy markers
Quantification of autophagic flux rather than static autophagosome numbers
Molecular Mechanism Dissection:
Evaluation of mTOR signaling pathway components
Assessment of AMPK activation status
Analysis of Beclin1 complexes and interacting partners
In Vivo Validation:
Tissue-specific conditional knockout models
Physiological versus pathological stress conditions
Correlation with clinical samples and patient outcomes
Research has demonstrated that TIGAR mediates p53-induced inhibition of myocyte mitophagy through ROS reduction and subsequent inactivation of BNIP3, which can exacerbate cardiac damage . Conversely, in other contexts, TIGAR may promote autophagy to maintain cellular homeostasis, highlighting the importance of experimental context in interpreting results.
The production of high-quality recombinant TIGAR protein involves several methodological considerations:
Expression System Selection:
Expression Vector Design:
Incorporate appropriate promoter (T7, tac)
Include affinity tag (6xHis) for purification
Consider fusion partners (GST, MBP) for enhanced solubility
Include precision protease cleavage sites if tag removal is required
Expression Conditions Optimization:
Test multiple induction temperatures (16°C, 25°C, 37°C)
Vary IPTG concentrations (0.1-1.0 mM)
Optimize expression duration (3-24 hours)
Consider auto-induction media for higher yields
Purification Protocol:
Initial capture: Immobilized metal affinity chromatography (IMAC)
Secondary purification: Size exclusion chromatography
Quality control: SDS-PAGE, Western blotting, mass spectrometry
Activity verification: Enzymatic assays
Buffer Optimization:
For optimal results, researchers should verify protein purity (>90% by SDS-PAGE) and confirm enzymatic activity through phosphatase assays before using in experimental procedures .
Studying TIGAR's impact on the pentose phosphate pathway (PPP) and redox regulation requires integrated methodological approaches:
Metabolic Flux Analysis:
Use of 13C-labeled glucose to track carbon flow through glycolysis versus PPP
GC-MS or LC-MS/MS analysis of labeled metabolites
Computational modeling of flux distributions
Redox Status Assessment:
Quantification of NADPH/NADP+ ratio using enzymatic assays or targeted metabolomics
Measurement of reduced/oxidized glutathione levels (GSH/GSSG)
ROS detection using fluorescent probes (CM-H2DCFDA, MitoSOX)
Protein oxidation assessment (protein carbonylation, thiol oxidation)
Gene Expression and Protein Analysis:
Quantification of PPP enzyme expression (G6PD, 6PGD, TKT)
Assessment of antioxidant defense systems (glutathione peroxidase, peroxiredoxins)
Analysis of redox-sensitive transcription factors (Nrf2, NF-κB)
Functional Outcomes Measurement:
Cell viability under oxidative stress conditions
DNA damage assessment (comet assay, γH2AX foci)
Mitochondrial function analysis (oxygen consumption rate, membrane potential)
Integration with Disease Models:
Cancer cells with varying metabolic dependencies
Ischemia-reperfusion injury models
Neurodegenerative disease models sensitive to oxidative stress
Research has established that TIGAR contributes to the generation of reduced glutathione to decrease intracellular ROS, correlating with its ability to protect cells from oxidative stress-induced death . Additionally, TIGAR plays a neuroprotective role against ischemic brain damage by enhancing PPP flux and preserving mitochondrial functions .
Advanced imaging techniques provide crucial insights into TIGAR's subcellular localization and dynamics:
Fluorescent Fusion Protein Approaches:
Generation of TIGAR-GFP/mCherry fusion constructs
Lentiviral transduction for stable expression
Live-cell confocal microscopy for dynamic tracking
Photoactivatable or photoconvertible tags for pulse-chase localization studies
Super-Resolution Microscopy:
Stimulated emission depletion (STED) microscopy for nanoscale resolution
Stochastic optical reconstruction microscopy (STORM) for single-molecule localization
Structured illumination microscopy (SIM) for enhanced resolution of subcellular structures
Multi-Channel Co-Localization Analysis:
Simultaneous imaging of TIGAR with organelle markers:
Mitochondria: MitoTracker, TOM20 antibodies
Nucleus: DAPI, Hoechst
ER: Calnexin, PDI
Lysosomes: LAMP1, LysoTracker
Quantitative co-localization metrics (Pearson's coefficient, Manders' overlap)
Förster Resonance Energy Transfer (FRET):
Detection of protein-protein interactions in live cells
Measurement of conformational changes in TIGAR protein
Biosensor development for TIGAR activity
Fluorescence Recovery After Photobleaching (FRAP):
Assessment of TIGAR mobility within cellular compartments
Determination of binding dynamics to subcellular structures
Research has demonstrated that TIGAR translocates to different subcellular compartments under specific conditions:
Nuclear translocation promotes cell survival during chemotherapy and hypoxia
Mitochondrial translocation under hypoxia enhances HK2 activity and maintains cell survival
Interaction with ATP5A1 in mitochondria reduces oxidative stress
These dynamic localizations highlight the importance of advanced imaging techniques in understanding TIGAR's multifaceted functions.
Variability in TIGAR phosphatase activity assays presents a common challenge. Methodological solutions include:
Standardization of Protein Quality:
Implement batch-to-batch quality control testing
Verify protein purity by SDS-PAGE and Western blotting
Perform thermal shift assays to confirm proper folding
Use freshly thawed aliquots for each experiment
Reaction Condition Optimization:
Systematically test buffer components:
pH range (6.5-8.5)
Salt concentration (50-300 mM NaCl)
Divalent cations (Mg2+, Mn2+)
Reducing agents (DTT, β-mercaptoethanol)
Determine optimal enzyme:substrate ratio
Establish temperature dependence (25°C, 30°C, 37°C)
Substrate Considerations:
Use freshly prepared substrate solutions
Verify substrate purity by analytical methods
Consider competing reactions or substrate instability
Include substrate-only controls in every experiment
Advanced Data Analysis:
Apply baseline correction algorithms
Calculate initial velocities from linear portions of progress curves
Use internal standards for assay normalization
Consider mathematical modeling of reaction kinetics
Technical Replicates and Controls:
Following the standardized protocol described in source , which includes appropriate enzyme controls and specific activity calculations, can significantly reduce assay variability and improve reproducibility.
When investigating TIGAR's role in cancer metabolism, researchers encounter several methodological challenges:
Cell Line Heterogeneity:
Pitfall: Varying baseline TIGAR expression and p53 status across cancer cell lines
Solution: Systematically characterize multiple cell lines before selection; create isogenic cell line panels; document p53 status
Metabolic Adaptation:
Pitfall: Compensatory metabolic pathways activated upon TIGAR modulation
Solution: Employ acute (inducible) rather than chronic TIGAR modulation; perform time-course studies; analyze multiple metabolic pathways simultaneously
In Vitro vs. In Vivo Discrepancies:
Pitfall: Cell culture conditions poorly recapitulate tumor microenvironment
Solution: Use 3D culture systems; implement co-culture with stromal cells; validate findings in patient-derived xenografts
Technical Limitations in Metabolite Analysis:
Pitfall: Rapid turnover and instability of key metabolites
Solution: Optimize rapid quenching techniques; employ stable isotope tracing; integrate computational modeling with experimental data
Contextual Dependency:
Pitfall: TIGAR functions vary with nutrient availability and oxygen tension
Solution: Precisely control and document culture conditions; systematically test nutrient limitation and hypoxia; correlate with in vivo tumor regions
Research has shown that TIGAR protects glioma cells from hypoxia and ROS-induced cell death by inhibiting glycolysis and activating mitochondrial energy metabolism in a TKTL1-dependent manner . Additionally, TIGAR plays a role in cancer cell survival by promoting DNA repair through activating PPP flux in a CDK5-ATM-dependent signaling pathway during hypoxia and/or genome stress-induced DNA damage responses .
Distinguishing specific TIGAR effects from general metabolic perturbations requires methodological rigor:
Genetic Approach Considerations:
Complementary Techniques: Employ both knockdown (siRNA, shRNA) and knockout (CRISPR-Cas9) approaches
Rescue Experiments: Re-express TIGAR wild-type or mutant variants to confirm specificity
Dose-Response Analysis: Establish graded expression levels to identify threshold effects
Metabolic Intervention Design:
Targeted Metabolite Supplementation: Provide specific metabolites (ribose-5-phosphate, NADPH) to bypass TIGAR effects
Parallel Pathway Modulation: Simultaneously manipulate alternative metabolic enzymes (G6PD, PKM2)
Metabolic Inhibitors: Use specific inhibitors of glycolysis or PPP to dissect pathway contributions
Temporal Analysis:
Early vs. Late Responses: Distinguish immediate TIGAR effects from adaptive responses
Pulse-Chase Experiments: Track metabolic flux changes over time using labeled precursors
Inducible Systems: Employ temporal control of TIGAR expression or activity
Multi-Omics Integration:
Correlative Analysis: Integrate transcriptomics, proteomics, and metabolomics data
Network Analysis: Apply pathway enrichment and network modeling to identify specific nodes
Single-Cell Technologies: Assess heterogeneity of responses within cell populations
Direct Target Validation:
In Vitro Enzyme Assays: Confirm direct enzymatic activities with purified components
Target Engagement Studies: Assess physical interactions in cellular contexts
Structure-Function Analysis: Utilize domain-specific mutations to dissect activities
Research indicates that TIGAR has both enzymatic and non-enzymatic functions, including interactions with mitochondrial proteins that occur independently of its fructose-bisphosphatase activity . Careful experimental design is essential to distinguish these diverse mechanistic effects.
Current evidence points to several promising research directions for TIGAR in disease contexts:
Cancer Therapeutics Development:
Cardiovascular Disease Interventions:
Neurological Disease Applications:
Metabolic Disorder Strategies:
Exploration of TIGAR's role in insulin resistance and diabetes
Assessment of potential in non-alcoholic fatty liver disease
Investigation of interactions with other metabolic regulators
Aging and Cellular Senescence:
Examination of TIGAR in longevity pathways
Connection to cellular senescence mechanisms
Potential for rejuvenation strategies through metabolic reprogramming
Research has established TIGAR as a multifunctional protein with diverse roles in cellular homeostasis and stress responses. The development of specific modulators of TIGAR activity, combined with precise understanding of context-dependent functions, holds significant promise for therapeutic applications across multiple disease states .
The C12orf5 gene is located on chromosome 12 and encodes a protein that is involved in the regulation of glycolysis and apoptosis. The protein is composed of 270 amino acids and has a molecular mass of approximately 31 kDa . The recombinant form of this protein is often produced in Escherichia coli (E. coli) and includes a C-terminal 6-His tag for purification purposes .
TIGAR functions as a fructose-2,6-bisphosphatase, which means it can lower the levels of fructose-2,6-bisphosphate in cells. By doing so, it diverts glucose metabolism away from glycolysis and towards the pentose phosphate pathway. This shift helps in reducing reactive oxygen species (ROS) and protecting cells from oxidative stress .
Additionally, TIGAR is involved in the regulation of apoptosis. By modulating the levels of ROS, TIGAR can influence cell survival and death, making it a critical player in cellular responses to stress and damage .
The recombinant form of C12orf5 (Human Recombinant, His Tag) is widely used in research to study its biochemical properties and its role in cellular metabolism and apoptosis. The His tag allows for easy purification of the protein using affinity chromatography, which is essential for obtaining high-purity samples for experimental purposes .
Researchers use this recombinant protein to investigate its enzymatic activity, interaction with other cellular components, and its overall impact on cellular physiology. Studies involving TIGAR have implications in understanding cancer metabolism, neurodegenerative diseases, and other conditions where cellular metabolism and oxidative stress play a significant role .