ALDOC Human, Active

Aldolase C Fructose-Bisphosphate Human Recombinant, Active
Shipped with Ice Packs
In Stock

Description

Cancer Biology

  • Non-Small Cell Lung Cancer (NSCLC):

    • ALDOC overexpression correlates with advanced tumor stage, lymph node metastasis, and poor survival .

    • Mechanistically, ALDOC upregulates UBE2N via MYC transcription, enhancing Wnt/β-catenin signaling and promoting tumor proliferation .

    • In 3D tumor spheroids, ALDOC sustains glucose metabolism under nutrient-restricted conditions, enabling tumor growth .

  • Therapeutic Target:

    • ALDOC-driven glycolysis supports tumor survival; inhibitors targeting aldolase-protein interactions (e.g., raltegravir analogs) show anti-tumor potential .

Diabetic Retinopathy (DR)

  • ALDOC is causally linked to proliferative DR (PDR), with elevated levels associated with insulin dysregulation and Wnt signaling activation .

Neurological Roles

  • Neuroprotection: ALDOC mitigates excitotoxic damage in cerebellar Purkinje cells .

  • Neurodegeneration: Oxidative inactivation of ALDOC in Alzheimer’s disease disrupts ATP production, contributing to metabolic dysfunction .

Key Research Findings

Recent studies highlight ALDOC’s multifunctional roles beyond glycolysis:

Study FocusKey FindingsSource
NSCLC MetastasisALDOC knockdown reduces UBE2N expression and suppresses tumor growth .
Diabetic RetinopathyALDOC levels correlate with plasma free fatty acids and PDR risk .
Tumor MetabolismALDOC and ENO2 drive glycolysis in 3D spheroids, independent of nutrient supply .
Therapeutic TargetingAldolase inhibitors disrupt cancer cell cytoskeleton, inducing apoptosis .

Research Applications

  • In Vitro Studies: Used to investigate ALDOC’s role in glycolysis, cancer cell migration, and apoptosis .

  • Biomarker Development: Explored in NSCLC prognosis and diabetic retinopathy screening .

  • Drug Discovery: Screened for inhibitors targeting ALDOC-protein interactions in cancer .

Product Specs

Introduction
Aldolase C Fructose-Bisphosphate (ALDOC), a member of the class I fructose-bisphosphate aldolase family, is a glycolytic enzyme. It plays a crucial role in the reversible aldol cleavage of fructose-1,6-biphosphate and fructose 1-phosphate into dihydroxyacetone phosphate and either glyceraldehyde-3-phosphate or glyceraldehydes, respectively. ALDOC expression is highly specific to the brain, particularly in the hippocampus and Purkinje cells.
Description
Recombinant human ALDOC, produced in E. coli, is a single, non-glycosylated polypeptide chain. It comprises 364 amino acids (1-364 a.a.) and exhibits a molecular weight of 39.4kDa. The purification process of ALDOC involves proprietary chromatographic techniques.
Physical Appearance
A clear solution that has been sterilized through filtration.
Formulation
The ALDOC solution, with a concentration of 1mg/ml, is formulated in a buffer containing 20% glycerol, 20mM Tris-HCl (pH 8.0), 2mM DTT, and 0.1M NaCl.
Stability
For short-term storage (2-4 weeks), the ALDOC vial should be kept at 4°C. For extended storage, it is recommended to store the vial frozen at -20°C. To ensure optimal long-term stability, adding a carrier protein (0.1% HSA or BSA) is advisable. It is important to avoid repeated freeze-thaw cycles.
Purity
The purity of ALDOC is determined to be greater than 90.0% using SDS-PAGE analysis.
Biological Activity
The specific activity of ALDOC is measured to be greater than 6 units/mg. One unit of activity is defined as the amount of enzyme required to convert 1.0 µmol of fructose 1,6-diphosphate to dihydroxyacetone phosphate and glyceraldehydes 3-phosphate per minute at a pH of 7.5 and a temperature of 37°C.
Synonyms

Aldolase, Fructose-Bisphosphate C, Aldolase C, Fructose-Bisphosphate, Brain-Type Aldolase, EC 4.1.2.13, ALDC, Fructose-1,6-Biphosphate Triosephosphate Lyase, Fructose-Bisphosphate Aldolase C, Fructoaldolase C, Aldolase 3, ALDOC .

Source
Escherichia Coli.
Amino Acid Sequence

MPHSYPALSA EQKKELSDIA LRIVAPGKGI LAADESVGSM AKRLSQIGVE NTEENRRLYR QVLFSADDRV KKCIGGVIFF HETLYQKDDN GVPFVRTIQD KGIVVGIKVD KGVVPLAGTD GETTTQGLDG LSERCAQYKK DGADFAKWRC VLKISERTPS ALAILENANV LARYASICQQ NGIVPIVEPE ILPDGDHDLK RCQYVTEKVL AAVYKALSDH HVYLEGTLLK PNMVTPGHAC PIKYTPEEIA MATVTALRRT VPPAVPGVTF LSGGQSEEEA SFNLNAINRC PLPRPWALTF SYGRALQASA LNAWRGQRDN AGAATEEFIK RAEVNGLAAQ GKYEGSGEDG GAAAQSLYIA NHAY

Q&A

What is ALDOC and what is its primary function in human cells?

ALDOC is a metabolic enzyme that functions primarily in glycolysis, converting fructose 1,6-bisphosphate to glyceraldehyde 3-phosphate and dihydroxyacetone phosphate. Beyond this canonical role, ALDOC interacts with various signaling molecules and transcription factors, suggesting its involvement in cellular processes beyond metabolism. Recent research indicates ALDOC can influence gene expression patterns and regulate signaling pathways, particularly in pathological contexts such as cancer. The enzyme has been implicated in regulating the Wnt/β-catenin pathway, which plays a crucial role in cell proliferation and differentiation .

How is ALDOC activity measured in laboratory settings?

ALDOC enzymatic activity is typically measured through spectrophotometric assays that monitor the formation or consumption of NADH at 340 nm. The standard approach involves coupling the ALDOC reaction with glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and following the reduction of NAD+ to NADH. For research focusing on non-metabolic functions, activity can be assessed indirectly through downstream effects on target pathways, such as changes in Wnt/β-catenin signaling activity using reporter assays or changes in the expression of target genes like UBE2N . When evaluating ALDOC's functional effects, researchers typically measure cell proliferation, migration, colony formation, and apoptosis after manipulating ALDOC expression through knockdown or overexpression approaches .

What are the expression patterns of ALDOC in normal versus pathological tissues?

ALDOC expression varies significantly between normal and pathological tissues. In NSCLC, ALDOC shows significantly elevated expression compared to normal lung tissue. Analysis of clinical samples has revealed that ALDOC expression positively correlates with lymph node metastasis, lymphatic metastasis, and pathological stage in NSCLC patients . Laboratory studies confirm this pattern, with elevated ALDOC levels in both NSCLC cell lines (A549 and NCI-H1299) compared to normal bronchial epithelial cells (BEAS-2B) . The clinical significance of this overexpression is substantial, as patients with high ALDOC levels exhibit decreased survival probabilities, indicating poor prognosis . This differential expression pattern suggests that ALDOC may serve as a potential biomarker for cancer diagnosis and prognosis.

What methods are effective for modulating ALDOC expression in experimental settings?

Several methodologies have proven effective for modulating ALDOC expression in experimental settings:

  • RNA interference: shRNA targeting ALDOC can achieve significant reduction in both mRNA and protein expression. The research demonstrates successful ALDOC knockdown in A549 and NCI-H1299 cells using this approach .

  • Overexpression systems: Forced expression of ALDOC can be achieved through transfection or viral transduction of expression vectors containing the ALDOC cDNA.

  • CRISPR-Cas9 genome editing: While not explicitly mentioned in the search results, this technology allows for precise genetic manipulation to create knockout models or introduce specific mutations.

  • Validation approaches: Effective modulation should be verified through Western blotting and qRT-PCR to confirm changes in expression levels, as demonstrated in the referenced studies .

  • Functional assessment: Following modulation, changes in cellular phenotypes should be measured through proliferation assays, migration assays (transwell and wound-healing), and apoptosis assessment (flow cytometry) .

What cell culture systems are optimal for studying ALDOC function?

The optimal cell culture systems for studying ALDOC function depend on the specific research question:

  • NSCLC cell lines: A549 and NCI-H1299 cells have been successfully used to study ALDOC's role in cancer progression, as they express elevated levels of ALDOC compared to normal cells .

  • Normal counterparts: BEAS-2B bronchial epithelial cells serve as appropriate controls when studying ALDOC in the context of lung cancer .

  • Primary cultures: While not specifically mentioned for ALDOC research, techniques similar to those described for primary glial cultures could be adapted for relevant tissues . This involves tissue digestion protocols, specialized culture conditions, and purity verification through immunocytochemistry.

  • Genetic manipulation models: Creating stable cell lines with ALDOC knockdown or overexpression provides valuable models for studying ALDOC function .

What are the non-canonical functions of ALDOC beyond glycolysis?

ALDOC exhibits several non-canonical functions extending beyond its traditional role in glycolysis:

  • Transcriptional regulation: ALDOC influences gene expression by affecting transcription factors. Research demonstrates that ALDOC affects MYC-mediated transcription of UBE2N, suggesting a role in regulating gene expression programs .

  • Signaling pathway modulation: ALDOC interacts with and regulates key signaling pathways, particularly the Wnt/β-catenin pathway. This interaction has significant implications for cell proliferation, differentiation, and oncogenesis .

  • Protein-protein interactions: ALDOC forms complexes with various cellular proteins beyond metabolic enzymes, serving as a scaffold or regulator for multi-protein complexes.

  • Cancer progression promotion: ALDOC promotes NSCLC progression through multiple mechanisms including enhanced cell proliferation, migration, and resistance to apoptosis .

To investigate these non-canonical functions, researchers employ techniques such as gene expression analysis, which has identified thousands of differentially expressed genes (4,182) upon ALDOC knockdown, with 2,398 upregulated and 1,884 downregulated genes .

How does ALDOC contribute to cancer progression in NSCLC?

ALDOC contributes to NSCLC progression through multiple mechanisms:

  • Promotion of cell proliferation: ALDOC knockdown substantially decreases cell proliferation in A549 and NCI-H1299 cells, while overexpression enhances proliferation .

  • Enhancement of cell migration: ALDOC depletion leads to a significant reduction in cell migration in transwell chamber assays and wound-healing assays, suggesting a role in promoting metastasis .

  • Inhibition of apoptosis: Flow cytometry analysis reveals that silencing ALDOC promotes apoptosis in NSCLC cells, indicating that ALDOC normally suppresses cell death pathways .

  • Regulation of UBE2N expression: ALDOC affects MYC-mediated transcription of UBE2N, a gene involved in DNA damage tolerance and cellular signaling. UBE2N knockdown inhibits the proliferation and migration of ALDOC-overexpressing cells, indicating that UBE2N is a key mediator of ALDOC's oncogenic effects .

  • Modulation of the Wnt/β-catenin pathway: ALDOC regulates this pathway, which is crucial for cancer development and progression .

These findings are supported by clinical data showing that high ALDOC expression correlates with lymph node metastasis, advanced disease stage, and poor survival outcomes in NSCLC patients .

What transcriptomic approaches reveal ALDOC-regulated pathways?

Various transcriptomic approaches have been valuable for uncovering ALDOC-regulated pathways:

  • Microarray analysis: The research employed genechip primeview human patharrayTM in A549 cells after shALDOC and shCtrl infection, identifying 4,182 differentially expressed genes (DEGs) .

  • Statistical filtering: Researchers applied specific criteria to identify the most relevant targets: (i) downregulation by shALDOC, (ii) logFC ≥ 2, and (iii) relation to NSCLC patients' prognosis. This stringent filtering identified three genes: RAD51AP1, UBE2N, and KIAA0101 .

  • Validation of key targets: qRT-PCR and Western blotting confirmed a clear reduction in UBE2N mRNA and protein levels upon ALDOC knockdown .

  • RNAseq methodology: While not directly applied to ALDOC in the search results, comprehensive RNAseq approaches could be adapted for ALDOC studies, including mapping raw data to reference genomes, building indices with Bowtie2, trimming adaptors with TrimGalore, mapping reads with Tophat2, and normalizing datasets with DESeq2 .

  • Pathway analysis: Functional annotation of differentially expressed genes can reveal biological processes and signaling pathways affected by ALDOC modulation.

These approaches have successfully identified UBE2N as a key mediator of ALDOC's effects in NSCLC, demonstrating the value of transcriptomic analysis in understanding ALDOC-regulated pathways .

How do experimental conditions affect ALDOC activity and function?

Experimental conditions significantly impact ALDOC activity and function, requiring careful consideration in research design:

Standardizing these conditions across experiments is essential for obtaining reproducible results when studying ALDOC function.

What are the methodological considerations for validating ALDOC-regulated targets?

Validating ALDOC-regulated targets requires rigorous methodological approaches:

  • Hierarchical validation strategy: Initially identified targets from high-throughput approaches should be validated through multiple complementary methods. For UBE2N, researchers confirmed its downregulation at both mRNA and protein levels after ALDOC knockdown .

  • Functional validation: Demonstrating the biological relevance of potential targets through gain and loss of function experiments. The research showed that UBE2N knockdown inhibited the proliferation and migration of ALDOC-overexpressing cells, confirming UBE2N as a functional mediator of ALDOC's effects .

  • Rescue experiments: Restoring the expression of a putative target in ALDOC-knockdown cells should rescue the observed phenotypes if the target is indeed mediating ALDOC's effects.

  • Direct versus indirect regulation: Determining whether ALDOC directly or indirectly regulates the target. The research indicated that ALDOC affects MYC-mediated transcription of UBE2N, suggesting a mechanistic link .

  • Clinical correlation: Validating the relationship between ALDOC and its targets in patient samples. High ALDOC expression was associated with poor prognosis in NSCLC patients, supporting the clinical relevance of identified pathways .

  • Pathway context: Placing validated targets within broader signaling networks. UBE2N was positioned within the context of the Wnt/β-catenin pathway, providing a framework for understanding ALDOC's effects .

These methodological considerations ensure that identified ALDOC-regulated targets represent genuine biological relationships rather than experimental artifacts.

What are the optimal protocols for isolating active human ALDOC?

While the search results don't provide specific protocols for ALDOC isolation, we can adapt general protein purification principles and similar approaches used for other enzymes:

  • Cell lysis optimization: Determining the optimal lysis buffer composition that preserves ALDOC activity is critical. Typically, a buffer containing mild detergents (0.1-1% Triton X-100), protease inhibitors, and stabilizing agents at physiological pH would be suitable.

  • Chromatographic techniques:

    • Affinity chromatography: Using substrate analogs or specific antibodies against ALDOC

    • Ion-exchange chromatography: Based on ALDOC's charge properties

    • Size-exclusion chromatography: To separate ALDOC tetramers from other cellular components

  • Activity-guided fractionation: Tracking ALDOC enzymatic activity throughout the purification process to identify fractions containing active enzyme.

  • Recombinant expression systems: For large-scale production, expressing human ALDOC with appropriate tags (His, GST) in bacterial, insect, or mammalian expression systems.

  • Quality control: Verifying purity through SDS-PAGE, Western blotting, and mass spectrometry, while confirming activity through enzymatic assays.

  • Stability considerations: Adding glycerol (10-20%) and reducing agents to maintain long-term stability of purified ALDOC.

For research focused on studying ALDOC in specific cell contexts, genetic approaches may be more practical than protein purification. The research successfully employed shRNA for knockdown and expression vectors for overexpression to manipulate ALDOC levels in NSCLC cell lines .

What experimental designs best elucidate ALDOC's role in signaling pathways?

Comprehensive experimental designs for elucidating ALDOC's role in signaling pathways include:

  • Genetic manipulation coupled with pathway analysis:

    • Generate ALDOC knockdown and overexpression models as demonstrated in the research

    • Assess effects on key signaling pathways (Wnt/β-catenin, MYC) using reporter assays, Western blotting for pathway components, and transcriptomic analysis

  • Sequential validation approach:

    • Large-scale screening (e.g., transcriptomics) to identify affected pathways and genes

    • Targeted validation of key components through qRT-PCR and Western blotting

    • Functional validation through loss/gain of function of pathway components

  • Time-course experiments:

    • Analyze temporal dynamics of pathway activation after ALDOC modulation

    • Differentiate between early (direct) and late (indirect) effects

  • Combinatorial perturbations:

    • Simultaneously manipulate ALDOC and key pathway components

    • The research demonstrated this approach by silencing UBE2N in ALDOC-overexpressing cells

  • Protein interaction studies:

    • Co-immunoprecipitation to identify direct ALDOC-interacting proteins

    • Proximity ligation assays to verify interactions in situ

  • Systems biology approach:

    • Integrate transcriptomic, proteomic, and functional data

    • Construct network models of ALDOC-regulated pathways

  • In vivo validation:

    • Test findings from cell culture in appropriate animal models

    • Correlate with patient data from clinical samples

This multi-faceted approach provides robust evidence for ALDOC's role in specific signaling pathways and helps distinguish direct from indirect effects.

How can RNAseq approaches be optimized for studying ALDOC-related gene expression?

Optimizing RNAseq approaches for studying ALDOC-related gene expression requires careful consideration of several technical aspects:

  • Experimental design considerations:

    • Include biological replicates (minimum n=3 per condition) as demonstrated in the RNAseq approach described

    • Design appropriate controls (e.g., shCtrl for shALDOC experiments)

    • Consider time-course experiments to capture dynamic changes

  • Sample preparation:

    • Ensure high-quality RNA extraction (RIN > 8)

    • Select appropriate library preparation methods based on research questions

    • Consider strand-specific protocols to distinguish sense and antisense transcription

  • Sequencing parameters:

    • Determine appropriate sequencing depth (typically 30-50 million reads per sample for differential expression analysis)

    • Select optimal read length (paired-end sequencing provides better transcript reconstruction)

  • Bioinformatic analysis pipeline:

    • Build the index of reference genomes with Bowtie2

    • Trim adaptors with TrimGalore

    • Map reads to reference genome with Tophat2

    • Count reads per gene with HTseq

    • Normalize data with DESeq2

  • Statistical analysis:

    • Set appropriate significance thresholds (the research used padj < 0.1, equivalent to a false discovery rate of 10%)

    • Apply additional filtering criteria based on biological relevance (e.g., logFC ≥ 2 and relevance to disease prognosis as used in the research)

  • Validation strategies:

    • Confirm key findings with qRT-PCR and protein-level analysis

    • Perform functional validation of identified targets

  • Integration with other data types:

    • Combine RNAseq data with clinical information, proteomics, or metabolomics for comprehensive understanding

The optimized approach used in the research successfully identified UBE2N as a key mediator of ALDOC's effects, demonstrating the value of carefully designed RNAseq experiments .

What techniques are most effective for studying ALDOC protein-protein interactions?

Several complementary techniques can effectively investigate ALDOC protein-protein interactions:

  • Co-immunoprecipitation (Co-IP):

    • Use antibodies against ALDOC to pull down protein complexes

    • Identify interacting partners through Western blotting or mass spectrometry

    • Perform reciprocal Co-IP with antibodies against suspected interaction partners

  • Proximity-based approaches:

    • BioID: Fuse ALDOC to a biotin ligase to biotinylate proximal proteins

    • APEX: Fuse ALDOC to an engineered peroxidase to label neighboring proteins

    • These methods capture transient and weak interactions that may be missed by Co-IP

  • Yeast two-hybrid screening:

    • Use ALDOC as bait to screen for potential interacting proteins

    • Confirm positive hits with orthogonal methods

  • Protein fragment complementation assays:

    • Split reporter proteins (luciferase, GFP) fused to ALDOC and potential partners

    • Signal generated only when proteins interact

  • FRET/BRET analysis:

    • Tag ALDOC and potential partners with appropriate fluorophores

    • Measure energy transfer as indication of protein proximity

  • Crosslinking mass spectrometry:

    • Chemically crosslink protein complexes in intact cells

    • Identify interaction sites through mass spectrometry

  • Functional validation:

    • Validate biological relevance of identified interactions through mutagenesis

    • Perform functional assays after disrupting specific interactions

  • Structural analysis:

    • Determine crystal structures of ALDOC-protein complexes

    • Use computational docking to predict interaction interfaces

When investigating ALDOC interactions with the MYC transcription factor or components of the Wnt pathway (as suggested by the research) , chromatin immunoprecipitation (ChIP) assays would be particularly valuable to detect interactions at specific genomic loci.

How should researchers analyze transcriptomic data after ALDOC modulation?

Analyzing transcriptomic data after ALDOC modulation requires a systematic approach:

  • Quality control and normalization:

    • Assess RNA quality metrics and sequencing quality scores

    • Normalize count data appropriately (e.g., using DESeq2 as mentioned in the research)

    • Perform batch correction if necessary

  • Differential expression analysis:

    • Compare ALDOC-modulated samples with controls to identify differentially expressed genes (DEGs)

    • Apply appropriate statistical thresholds (e.g., adjusted p-value < 0.1 as used in the research)

    • Consider both statistical significance and fold-change magnitude

  • Strategic filtering of DEGs:

    • Apply biologically relevant filters as demonstrated in the research:

      • Direction of regulation (e.g., downregulated by shALDOC)

      • Magnitude of change (e.g., logFC ≥ 2)

      • Clinical relevance (e.g., relation to patient prognosis)

  • Pathway and functional enrichment analysis:

    • Identify enriched biological processes, molecular functions, and pathways

    • Focus on pathways relevant to the phenotype (e.g., Wnt signaling for NSCLC progression)

  • Network analysis:

    • Construct gene co-expression networks

    • Identify hub genes and modules related to ALDOC function

  • Integration with protein data:

    • Validate key transcriptional changes at the protein level

    • The research confirmed UBE2N downregulation at both mRNA and protein levels

  • Visualization techniques:

    • Generate heat maps of top DEGs as demonstrated in the research (Figure 3A)

    • Create volcano plots to visualize both significance and magnitude of changes

    • Develop pathway maps highlighting affected components

  • Functional validation:

    • Design experiments to test the biological relevance of identified genes

    • The research validated UBE2N's role through knockdown experiments in ALDOC-overexpressing cells

This comprehensive analytical approach successfully identified UBE2N as a key mediator of ALDOC's effects in NSCLC, demonstrating its effectiveness in uncovering biologically meaningful insights from transcriptomic data .

What statistical methods are appropriate for analyzing ALDOC's association with clinical outcomes?

When analyzing ALDOC's association with clinical outcomes, several statistical methods are appropriate:

  • Survival analysis:

    • Kaplan-Meier survival curves: The research used this approach to demonstrate that patients with high ALDOC levels exhibited decreased survival probabilities, implying poor prognosis

    • Log-rank tests: To assess statistical significance between survival curves of high versus low ALDOC expression groups

    • Cox proportional hazards regression: For multivariate analysis accounting for other clinical factors while assessing ALDOC's independent prognostic value

  • Correlation with clinicopathological parameters:

    • Chi-square or Fisher's exact tests: The research established significant correlations between ALDOC expression and lymph node metastasis, lymphatic metastasis, and pathological stage

    • Spearman or Pearson correlation: For continuous variables and expression levels

    • Logistic regression: To determine odds ratios for specific clinical outcomes based on ALDOC expression

  • Cohort comparison methods:

    • T-tests or Mann-Whitney tests: To compare ALDOC expression between different patient groups

    • ANOVA or Kruskal-Wallis tests: For comparing multiple groups (e.g., different cancer stages)

  • Predictive modeling:

    • ROC curve analysis: To assess ALDOC's potential as a diagnostic or prognostic biomarker

    • Decision tree or random forest algorithms: To integrate ALDOC with other markers for improved prediction

  • Meta-analysis approaches:

    • Fixed or random effects models: When combining data from multiple cohorts, as the research did by expanding analysis to larger cohorts like TCGA and GEO databases

    • Forest plots: To visualize effect sizes across different studies or cohorts

  • Multiple testing correction:

    • Benjamini-Hochberg procedure: To control false discovery rate when testing associations with multiple clinical parameters

    • Bonferroni correction: For more stringent control of family-wise error rate

  • Power analysis:

    • Sample size calculation: To ensure adequate statistical power for detecting clinically meaningful associations

The research successfully employed several of these methods to establish ALDOC as a clinically relevant marker in NSCLC, demonstrating significant associations with pathological parameters and survival outcomes .

How can researchers integrate ALDOC functional data with clinical information?

Integrating ALDOC functional data with clinical information requires multifaceted approaches:

  • Translational validation strategy:

    • Validate mechanisms identified in cell models using patient samples

    • The research demonstrated this by correlating ALDOC expression in clinical samples with lymph node metastasis and survival outcomes

    • Further validate key downstream targets (e.g., UBE2N) in patient tissues

  • Multi-level data integration:

    • Correlate ALDOC expression with:

      • Clinical parameters (stage, metastasis status)

      • Molecular subtypes

      • Treatment response

      • Patient outcomes (survival, recurrence)

  • Biomarker development pipeline:

    • Use functional insights to develop clinically relevant biomarkers

    • Design practical assays for measuring ALDOC or its downstream effectors

    • Validate in retrospective and prospective clinical cohorts

  • Mechanistic classification of patients:

    • Stratify patients based on ALDOC-related molecular signatures

    • Correlate with differential treatment responses or outcomes

    • Develop ALDOC pathway activity scores from expression data

  • Therapeutic targeting opportunities:

    • Identify druggable nodes in ALDOC-regulated pathways

    • Design rational combination strategies based on functional insights

    • Predict patient subgroups most likely to benefit from pathway-targeted therapies

  • Data visualization and communication tools:

    • Develop clinician-friendly visualization of complex molecular data

    • Create nomograms or prediction tools incorporating ALDOC-related parameters

    • Design interactive databases linking functional and clinical information

  • Collaborative research frameworks:

    • Establish biobanking with linked clinical data

    • Develop standardized protocols for functional studies

    • Create shared platforms for data integration and analysis

The research successfully integrated functional findings with clinical data by demonstrating that the same pathways affected by ALDOC in cell models (proliferation, migration) correlate with metastasis and survival in patient cohorts, providing a model for effective translational research .

What are the promising areas for future ALDOC research in cancer biology?

Several promising areas for future ALDOC research in cancer biology emerge from the current findings:

  • Expanded cancer type exploration: While the research focused on NSCLC, investigating ALDOC's role in other cancer types could reveal common mechanisms or tissue-specific differences.

  • Therapeutic targeting strategies: Developing specific inhibitors of ALDOC or its downstream effectors like UBE2N could provide new treatment options for cancers with high ALDOC expression .

  • Combination therapy approaches: Exploring synergistic effects between ALDOC pathway inhibition and standard treatments (chemotherapy, immunotherapy, targeted therapies).

  • Biomarker development: Validating ALDOC as a prognostic or predictive biomarker in larger clinical cohorts, potentially in combination with other markers.

  • Resistance mechanisms: Investigating how ALDOC contributes to treatment resistance and tumor recurrence.

  • Metabolic-signaling crosstalk: Further elucidating how ALDOC connects metabolic changes with signaling pathways like Wnt/β-catenin .

  • Structural biology approaches: Determining the three-dimensional structure of ALDOC in complex with key interacting partners to enable structure-based drug design.

  • Single-cell analysis: Applying single-cell transcriptomic approaches similar to those described for fibroblasts to understand heterogeneity in ALDOC expression and function within tumors.

  • In vivo models: Developing sophisticated animal models to study ALDOC's role in tumor initiation, progression, and metastasis.

  • Clinical trials: Ultimately, testing ALDOC-targeting approaches in clinical settings based on preclinical findings.

The groundwork laid by current research, particularly the identification of the ALDOC-MYC-UBE2N-Wnt axis in NSCLC, provides a solid foundation for these future research directions .

How can emerging technologies advance our understanding of ALDOC function?

Emerging technologies can significantly advance our understanding of ALDOC function:

  • Single-cell multiomics: Combining single-cell RNA sequencing with proteomics or epigenomics to provide comprehensive insights into ALDOC's cell-type specific functions and heterogeneity, similar to approaches described for fibroblast research .

  • CRISPR screening technologies: Genome-wide or targeted CRISPR screens to identify synthetic lethal interactions with ALDOC or its downstream pathways.

  • Spatial transcriptomics: Mapping ALDOC expression and its correlation with other genes in spatial context within tumors to understand microenvironmental influences.

  • Protein structure prediction algorithms: Using AI-based approaches like AlphaFold to predict ALDOC structure and interactions with higher accuracy.

  • Live-cell imaging technologies: Advanced microscopy techniques to visualize ALDOC localization and dynamics in real-time during various cellular processes.

  • Organoid and patient-derived xenograft models: More physiologically relevant systems to study ALDOC function in three-dimensional contexts.

  • Metabolic flux analysis: Advanced techniques to trace how ALDOC affects metabolic pathways beyond glycolysis.

  • Proteomics approaches: Mass spectrometry-based techniques to comprehensively identify ALDOC-interacting proteins and post-translational modifications.

  • Systems biology modeling: Computational approaches to model the complex interplay between ALDOC, metabolism, and signaling pathways.

  • Liquid biopsy technologies: Non-invasive methods to monitor ALDOC or its downstream effectors in patient samples over time.

These technologies could help resolve current knowledge gaps regarding ALDOC's complex roles in normal physiology and disease, potentially leading to novel therapeutic approaches.

What methodological advances could improve ALDOC-targeted therapy development?

Several methodological advances could improve the development of ALDOC-targeted therapies:

  • Structure-based drug design: Leveraging crystal structures or computational models of ALDOC to design small molecules that specifically inhibit its enzymatic activity or protein-protein interactions.

  • Allosteric modulator development: Identifying allosteric sites on ALDOC that could be targeted to modify its function without completely inhibiting its essential metabolic roles.

  • Targeted protein degradation: Employing PROTAC (Proteolysis-Targeting Chimera) technology to selectively degrade ALDOC protein in cancer cells.

  • RNA therapeutics: Developing siRNA, antisense oligonucleotides, or mRNA-targeting approaches to modulate ALDOC expression, similar to the shRNA approach used in the research .

  • Downstream target inhibition: Developing inhibitors of key downstream effectors like UBE2N or components of the Wnt pathway, which may provide more specific therapeutic effects .

  • Combination therapy rational design: Using functional genomics and computational approaches to identify synergistic drug combinations targeting ALDOC-related pathways.

  • Biomarker-guided trial design: Incorporating ALDOC expression or pathway activity measurements to select patients most likely to benefit from targeted therapies.

  • Drug delivery innovations: Developing cancer-specific delivery systems for ALDOC-targeting agents to minimize off-target effects.

  • Predictive preclinical models: Establishing patient-derived organoids or xenografts with varying ALDOC levels to test therapeutic approaches before clinical trials.

  • Adaptive trial designs: Implementing flexible clinical trial protocols that can adapt based on emerging biomarker data related to ALDOC pathways.

The research findings on ALDOC's role in regulating UBE2N expression and the Wnt pathway provide specific molecular targets for these therapeutic development approaches .

Product Science Overview

Introduction

Aldolase C, also known as fructose-bisphosphate aldolase C (ALDOC), is a key enzyme involved in glycolysis and gluconeogenesis. It catalyzes the reversible conversion of fructose-1,6-bisphosphate into glyceraldehyde-3-phosphate (G3P) and dihydroxyacetone phosphate (DHAP) through aldol cleavage .

Structure and Expression

Aldolase C is a member of the class I fructose-bisphosphate aldolase family. The human recombinant form of Aldolase C is expressed in Escherichia coli and is typically purified to a high degree of purity (>90%) using conventional chromatography techniques . The protein consists of 364 amino acids and has a molecular weight of approximately 39.4 kDa .

Biological Function

Aldolase C plays a crucial role in the fourth step of glycolysis, a metabolic pathway that breaks down glucose to produce energy. It is also involved in gluconeogenesis, the reverse pathway that generates glucose from non-carbohydrate precursors . The enzyme is specifically expressed in the hippocampus and Purkinje cells of the brain, indicating its importance in brain metabolism .

Enzymatic Activity

The specific activity of recombinant human Aldolase C is greater than 6 units/mg. One unit of the enzyme will convert 1.0 μmol of fructose-1,6-diphosphate to dihydroxyacetone phosphate and glyceraldehyde-3-phosphate per minute at pH 7.5 and 37°C . This high level of activity makes it suitable for various biochemical and physiological studies.

Applications

Recombinant human Aldolase C is used in research to study its role in metabolic pathways, particularly in glycolysis and gluconeogenesis. It is also utilized in structural biology to understand the enzyme’s mechanism of action and its interactions with other molecules. Additionally, it serves as a valuable tool in the development of therapeutic strategies for metabolic disorders and neurological diseases.

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 2025 TheBiotek. All Rights Reserved.