Glioblastoma (GBM):
Non-Small Cell Lung Cancer (NSCLC):
Melanoma Brain Metastasis (MBM):
Esophageal Cancer (EC):
Schizophrenia (SCZ): Upregulated in male SCZ patients’ anterior cingulate cortex, suggesting sex-specific glycolytic dysregulation .
Alzheimer’s Disease (AD): Oxidative inactivation of ALDOC disrupts glycolysis, favoring gluconeogenesis and ATP depletion .
ALDOC Human is utilized in:
Glycolysis Studies: Investigating metabolic rewiring in cancer .
Drug Development: Screening PPAR-γ agonists for GBM therapy .
Molecular Diagnostics: Prognostic biomarker for NSCLC and EC .
Quantitative assessment of ALDOC requires a multi-analytical approach. For mRNA quantification, RT-qPCR using validated primers targeting conserved ALDOC regions provides high sensitivity. For protein detection, western blotting with monoclonal antibodies (clone specifications based on application) and immunohistochemistry (IHC) offer complementary insights. IHC scoring should follow standardized protocols (0-3+ scale) with blinded pathologist review. For large-scale analysis, mining TCGA and GEO datasets provides comparative expression data across tissues, though validation with fresh samples is essential due to potential RNA degradation effects in archival specimens .
ALDOC expression demonstrates significant correlation with several clinical parameters as shown in the table below:
Features | No. of patients | ALDOC expression | P value |
---|---|---|---|
All patients | 79 | 41 (Low) | 38 (High) |
Lymph node metastasis | |||
No | 37 | 24 | 13 |
Yes | 42 | 17 | 25 |
Stage | |||
I | 28 | 21 | 7 |
II | 32 | 12 | 20 |
III | 13 | 7 | 6 |
IV | 6 | 1 | 5 |
This data reveals that high ALDOC expression significantly correlates with lymph node metastasis (p=0.031) and advanced pathological stage (p=0.008), suggesting its potential value as a prognostic biomarker. Further analysis should employ multivariate Cox regression to control for confounding variables when assessing survival implications .
ALDOC knockdown experiments in NSCLC cell lines produce consistent phenotypic changes that can be quantified using multiple methodological approaches:
Proliferation: MTT or BrdU incorporation assays show significant reduction in proliferation following ALDOC knockdown (p<0.001)
Colony formation: Crystal violet staining reveals decreased colony-forming ability after ALDOC silencing (p<0.001)
Migration: Transwell chamber assays and wound-healing experiments demonstrate reduced migration capacity
Apoptosis: Flow cytometry with Annexin V/PI staining shows increased apoptotic rates after ALDOC depletion (p<0.001)
These effects are consistent across multiple NSCLC cell lines (A549 and NCI-H1299), suggesting a conserved mechanism rather than cell line-specific artifacts .
A comprehensive approach to identifying ALDOC transcriptional targets requires a sequential experimental design:
Differential expression screening: Employ high-throughput transcriptomics (RNA-seq or microarray) comparing ALDOC-knockdown versus control cells
Candidate filtering: Apply stringent statistical thresholds (adjusted p<0.05, logFC≥2) and biological relevance criteria
Validation strategy: Confirm top candidates via RT-qPCR and western blotting across multiple cell lines
Mechanistic exploration: For each validated target, perform:
Promoter analysis using bioinformatics tools to identify potential transcription factor binding sites
Chromatin immunoprecipitation (ChIP) to assess transcription factor binding at candidate gene promoters
Luciferase reporter assays with wild-type and mutated promoter constructs
Functional significance: Rescue experiments where ALDOC-knockdown phenotypes are assessed after overexpressing the candidate target gene
This approach successfully identified UBE2N as a key downstream mediator of ALDOC's oncogenic effects in NSCLC, with potential therapeutic implications .
Robust xenograft studies investigating ALDOC function should incorporate these methodological considerations:
Cell preparation: Establish stable ALDOC-modulated cell lines (knockdown, overexpression, and controls) and validate expression before injection
Experimental groups: Include multiple conditions (minimum 4 groups):
Control (shCtrl)
ALDOC knockdown (shALDOC)
ALDOC overexpression (ALDOC-OE)
Combined manipulation (e.g., ALDOC-OE + shUBE2N to test pathway dependencies)
Randomization: Randomly assign animals to treatment groups to minimize selection bias
Power analysis: Calculate appropriate sample size based on expected effect size (typically n≥5 per group)
Tumor measurements: Monitor tumor dimensions at regular intervals (2-3 times weekly) using digital calipers
Endpoint analyses:
Tumor volume calculation using the formula V = (length × width²)/2
Tumor weight measurement after excision
IHC staining for proliferation markers (Ki67) and pathway components
RT-qPCR and western blotting of tumor tissue to confirm sustained ALDOC modulation
This comprehensive design enables detection of both macroscopic tumor growth effects and molecular mechanisms in vivo .
Resolving contradictory findings (such as favorable prognostic association in glioblastoma versus unfavorable in NSCLC) requires systematic analytical approaches:
Data harmonization: Standardize expression measurements across studies using normalized units
Meta-analysis methodology:
Apply random-effects models to account for between-study heterogeneity
Calculate pooled effect sizes with confidence intervals
Perform subgroup analyses by cancer type, stage, and molecular subtype
Molecular context exploration:
Compare co-expression networks across cancer types to identify differential partner proteins
Analyze mutational landscape and genomic alterations that might modify ALDOC function
Perform pathway enrichment analysis to identify cancer-specific signaling contexts
Experimental validation:
Design paired experiments using cell lines from different cancer types
Apply identical ALDOC modulation techniques across all models
Measure identical endpoints using standardized protocols
Single-cell resolution: Analyze scRNA-seq data to determine if apparent contradictions reflect cellular heterogeneity within tumors
This approach can transform seemingly contradictory findings into mechanistic insights about context-dependent functions .
Investigating ALDOC's regulation of Wnt signaling requires a multi-modal strategy:
Pathway activity measurement:
TOPFlash/FOPFlash luciferase reporter assays to quantify β-catenin-mediated transcription
Immunoblotting for active (non-phosphorylated) β-catenin levels
Nuclear/cytoplasmic fractionation to assess β-catenin nuclear translocation
Gene expression analysis:
qPCR panel of canonical Wnt target genes (AXIN2, CCND1, MYC)
RNA-seq with gene set enrichment analysis (GSEA) for Wnt pathway signatures
Protein interaction studies:
Co-immunoprecipitation to detect physical interactions between ALDOC and Wnt pathway components
Proximity ligation assay to visualize protein interactions in situ
Functional validation:
Combined manipulation experiments where ALDOC overexpression is performed with and without Wnt pathway inhibitors
Rescue experiments assessing whether constitutively active β-catenin can overcome ALDOC knockdown phenotypes
These approaches have revealed that ALDOC influences Wnt signaling in NSCLC, though the complete molecular mechanism requires further elucidation .
A comprehensive investigation of the ALDOC-UBE2N axis requires a structured approach:
Expression correlation analysis:
Assess correlation between ALDOC and UBE2N expression in patient samples
Quantify UBE2N mRNA and protein levels after ALDOC modulation
Transcriptional regulation assessment:
Chromatin immunoprecipitation (ChIP) for transcription factors at the UBE2N promoter
UBE2N promoter-reporter constructs to measure transcriptional activity
DNA-protein interaction studies (EMSA) to confirm direct binding
Functional relationship characterization:
Phenotypic analysis of combined manipulation models:
ALDOC-overexpression with UBE2N-knockdown
ALDOC-knockdown with UBE2N-overexpression
Epistasis testing through sequential gene manipulation
Pathway integration:
Analyze downstream effectors shared between ALDOC and UBE2N
Investigate convergence on Wnt/β-catenin pathway components
This methodology successfully demonstrated that UBE2N knockdown inhibited proliferation and migration while enhancing apoptosis in ALDOC-overexpressing NSCLC cells, establishing UBE2N as a critical downstream effector of ALDOC's oncogenic function .
Separating ALDOC's glycolytic role from its non-metabolic functions requires specialized experimental design:
Structure-function analysis:
Generate catalytically inactive ALDOC mutants (targeting the active site)
Create domain-specific mutants that selectively disrupt protein-protein interactions
Express these variants in ALDOC-knockout backgrounds to assess functional complementation
Metabolic uncoupling experiments:
Perform ALDOC manipulation under conditions where glycolysis is either enhanced or inhibited
Use alternative carbon sources (glutamine, fatty acids) to bypass glycolytic dependence
Employ metabolic inhibitors to isolate pathway contributions
Cellular compartmentalization studies:
Create subcellular-targeted ALDOC variants (nuclear, mitochondrial, membrane-bound)
Analyze compartment-specific functions using fractionation and imaging techniques
Temporal dynamics:
Employ rapidly inducible ALDOC modulation systems to distinguish immediate (likely metabolic) from delayed (likely signaling) effects
Perform time-course analyses of metabolic parameters and signaling events after ALDOC perturbation
These approaches can disentangle ALDOC's dual roles and potentially identify targetable non-metabolic functions while preserving essential metabolic activities .
Evaluating ALDOC's biomarker potential requires rigorous clinical validation methodology:
Patient cohort selection:
Prospective collection with defined inclusion/exclusion criteria
Stratification by disease stage, treatment history, and molecular subtypes
Adequate sample size determined by power analysis (typically n>100)
Specimen processing standardization:
Consistent collection, preservation, and storage protocols
Quality control metrics for tissue integrity and RNA/protein quality
Expression analysis techniques:
IHC with standardized scoring system (H-score or 0-3+ scale)
RT-qPCR with validated reference genes for normalization
Digital spatial profiling for heterogeneity assessment
Statistical methodology:
ROC curve analysis to determine optimal cutoff values
Kaplan-Meier survival analysis with log-rank test
Multivariate Cox regression to control for confounding variables
Concordance index (C-index) to assess predictive accuracy
Internal and external validation:
Training/validation set approach or cross-validation
Independent cohort validation
This approach can establish whether ALDOC expression levels provide clinically meaningful prognostic or predictive information beyond standard clinicopathological parameters .
Establishing ALDOC as a viable therapeutic target requires a systematic validation approach:
Target dependency confirmation:
CRISPR-Cas9 knockout or inducible shRNA systems in multiple cell lines
Colony formation and 3D spheroid assays for long-term viability assessment
Xenograft studies with inducible knockdown systems for in vivo validation
Therapeutic window evaluation:
Parallel manipulation in cancer and normal cell lines
Primary cell cultures from patient and normal adjacent tissue
Ex vivo tissue slice cultures to maintain tissue architecture
Resistance mechanism investigation:
Development of resistant models through prolonged partial inhibition
RNA-seq and phosphoproteomics to identify adaptation pathways
Combination strategy testing based on resistance mechanisms
Pharmacological approach assessment:
In silico modeling of ALDOC structure for druggable pocket identification
Fragment-based screening or virtual screening approaches
Development of tool compounds with documented specificity
Predictive biomarker identification:
Correlation of ALDOC dependency with molecular features
Multi-omics integration to identify sensitive patient subpopulations
This comprehensive approach can establish whether ALDOC represents a therapeutically viable target and identify which patient populations would most likely benefit from ALDOC-targeted therapies .
Patient-derived models require specialized methodology to maintain fidelity to the original tumor:
PDX establishment protocol:
Direct implantation of fresh tumor fragments (rather than dissociated cells)
Low-passage utilization to prevent drift from original tumor characteristics
Molecular characterization at each passage to monitor stability
Implantation in corresponding anatomical locations (orthotopic)
Organoid development methodology:
Optimization of extracellular matrix composition for each tumor type
Defined medium formulation with documented rationale for supplements
Single-cell versus fragment seeding comparison
Growth pattern documentation and quantification
Experimental manipulation approaches:
Lentiviral transduction protocols optimized for primary tissue
Inducible systems to control timing of ALDOC modulation
Ex vivo electroporation for transient manipulation
Analytical methods:
3D imaging techniques (confocal, light-sheet microscopy)
Live-cell imaging for dynamic phenotypes
Single-cell analysis from dissociated structures
Histological analysis with spatial preservation
These specialized patient-derived models offer superior physiological relevance compared to cell lines and can more accurately predict clinical efficacy of ALDOC-targeted strategies .
Comprehensive interactome mapping requires advanced proteomics approaches:
Proximity-based labeling methods:
BioID or TurboID fusion proteins to identify proximal interactors
APEX2-based proximity labeling for temporal resolution
Split-BioID for interaction-specific labeling
Cross-linking mass spectrometry (XL-MS):
Chemical cross-linking of protein complexes
MS/MS analysis to identify cross-linked peptides
Structural modeling based on distance constraints
Native complex isolation:
Size-exclusion chromatography with native conditions
Blue native PAGE for complex integrity
Protein correlation profiling across fractions
Interactome dynamics:
SILAC or TMT labeling to quantify interaction changes
Pulse-chase approaches for temporal dynamics
Stimulus-dependent interaction changes
Computational integration:
Network analysis with existing interactome databases
Structural modeling of key interactions
Pathway enrichment of interacting proteins
These approaches would significantly expand our understanding of ALDOC's functional network beyond the currently identified interactions with MYC and components of the Wnt pathway .
Metabolic reprogramming studies require specialized metabolic analysis techniques:
Flux analysis methodology:
13C-glucose or 13C-glutamine tracing with mass spectrometry
Seahorse extracellular flux analysis for glycolytic and mitochondrial function
Isotopomer distribution analysis for pathway utilization
Metabolite profiling:
Targeted metabolomics of glycolytic intermediates
Untargeted metabolomics for novel metabolite identification
In situ metabolite imaging using mass spectrometry imaging
Enzyme activity assays:
Direct measurement of ALDOC catalytic activity
Analysis of connected enzyme activities
Assessment of enzyme complex formation
Metabolic inhibitor studies:
Strategic pathway inhibition to assess metabolic dependencies
Synthetic lethality screening with ALDOC modulation
Nutrient limitation studies combined with ALDOC manipulation
In vivo metabolism:
Hyperpolarized 13C-MRI for real-time metabolic imaging
PET imaging with metabolic tracers
Ex vivo analysis of tumor metabolites
This multi-faceted approach can determine how ALDOC influences metabolic reprogramming in cancer and identify potential vulnerabilities for therapeutic exploitation .
Isoform-specific studies require specialized methodology:
Isoform expression profiling:
RNA-seq with specific attention to splice junction reads
Isoform-specific qPCR with junction-spanning primers
Proteomics with isoform-distinguishing peptide detection
Isoform-specific manipulation:
CRISPR-Cas13 for isoform-selective RNA targeting
Antisense oligonucleotides targeting unique exon junctions
Overexpression of individual isoforms in knockout backgrounds
Structural and functional characterization:
In vitro enzymatic activity comparisons
Protein stability and half-life assessment
Subcellular localization studies
Isoform-specific interactome:
Selective immunoprecipitation with isoform-specific antibodies
Domain-specific protein interaction studies
Comparative BioID experiments with different isoforms
Clinical correlation:
Isoform ratio analysis in patient samples
Survival correlation with specific isoform expression
Treatment response prediction based on isoform patterns
This approach can identify potentially targetable isoform-specific functions of ALDOC that may have distinct roles in cancer development and progression .
Aldolase C is primarily expressed in the brain, specifically in the hippocampus and Purkinje cells . It plays a crucial role in glycolysis, the metabolic pathway that converts glucose into pyruvate, releasing energy and producing intermediates for other metabolic processes . The enzyme catalyzes the reversible aldol cleavage of fructose-1,6-bisphosphate and fructose 1-phosphate into dihydroxyacetone phosphate and either glyceraldehyde-3-phosphate or glyceraldehyde, respectively .
The complete amino acid sequence of aldolase C was determined from recombinant genomic clones, revealing a polypeptide of 363 amino acids . Aldolase C shares 81% amino acid identity with aldolase A and 70% identity with aldolase B . The mRNA expression of ALDOC has been detected specifically in the brain .
Recombinant human aldolase C is produced using recombinant DNA technology, which involves inserting the ALDOC gene into a suitable expression system, such as bacteria or yeast, to produce the enzyme in large quantities . This recombinant enzyme is used in various research applications, including studies on glycolysis, brain metabolism, and related disorders .
Mutations or dysregulation of the ALDOC gene can be associated with certain metabolic disorders. For example, congenital disorders of glycosylation, type In, have been linked to abnormalities in aldolase C . Understanding the function and regulation of aldolase C is essential for developing potential therapeutic strategies for these conditions.