Resource

Submit Your Request Now

Submit Your Request Now

×

Metabolic Analysis Method of Glycolysis

Glycolysis, the central biochemical pathway converting glucose to pyruvate, underpins cellular energy production, biosynthetic processes, and pathological mechanisms in conditions such as cancer and metabolic disorders. Elucidating its dynamic flux, intermediate metabolite kinetics, and enzymatic regulation demands a synergistic integration of analytical approaches. This review delineates the evolution of glycolysis research methodologies—from foundational enzymatic assays and isotopic tracer studies to cutting-edge metabolomic platforms and multi-omics frameworks—highlighting their transformative roles in mechanistic discovery and clinical applications.

Glycolysis: A Central Metabolic Pathway in Cellular Energy Production

Glycolysis represents a fundamental biochemical process in cellular metabolism, wherein glucose is enzymatically converted into pyruvate, yielding energy (adenosine triphosphate, ATP) and reducing equivalents (nicotinamide adenine dinucleotide, NADH). This pathway is systematically divided into two phases: a preparatory phase (ATP investment) and an energy-yielding phase (ATP generation).

1. Preparatory Phase (ATP Utilization)

During this initial stage, glucose undergoes activation via ATP-dependent phosphorylation:

  • Hexokinase Catalysis: Glucose reacts with ATP to form glucose-6-phosphate, a critical regulatory step preventing glucose efflux from cells.
  • Phosphofructokinase-1 (PFK-1) Activity: Glucose-6-phosphate is isomerized to fructose-6-phosphate, then phosphorylated by ATP to yield fructose-1,6-bisphosphate—a pivotal intermediate and major regulatory node in glycolysis.

This phase consumes 2 ATP molecules to prime glucose for subsequent cleavage.

2. Energy-Yielding Phase (ATP and NADH Synthesis)

  • The second phase harvests energy through substrate-level phosphorylation:
    • Aldolase Cleavage: Fructose-1,6-bisphosphate splits into two triose phosphates (glyceraldehyde-3-phosphate and dihydroxyacetone phosphate).
    • Oxidative Phosphorylation: Glyceraldehyde-3-phosphate is oxidized to 1,3-bisphosphoglycerate, generating NADH.
    • ATP Synthesis: Enzymatic steps catalyzed by phosphoglycerate kinase and pyruvate kinase produce 4 ATP molecules (net gain: 2 ATP), culminating in pyruvate formation.
  • Key Regulatory Enzyme: Pyruvate kinase catalyzes the final ATP-generating step, ensuring pathway flux toward pyruvate.

Critical Metabolites and Their Roles

  • Glucose-6-Phosphate: Serves as a metabolic hub, entering glycolysis, the pentose phosphate pathway, or glycogen synthesis.
  • Fructose-1,6-Bisphosphate: A regulatory checkpoint amplifying pathway commitment via PFK-1 allosteric control.
  • 1,3-Bisphosphoglycerate/3-Phosphoglycerate: Direct precursors for ATP synthesis during energy-yielding reactions.
  • Pyruvate: The end-product enters mitochondria for oxidative decarboxylation to acetyl-CoA, linking glycolysis to the citric acid cycle.

Functional Significance

Glycolysis not only supplies immediate ATP but also provides intermediates for biosynthesis (e.g., amino acids, lipids) and redox balance via NADH production. Its dysregulation is implicated in pathologies such as cancer (Warburg effect) and metabolic syndromes, underscoring its therapeutic relevance.

Glycolytic metabolism.Glycolytic metabolism (Cordani M et al., 2024).

Analytical Strategies for Glycolytic Metabolism Profiling

1. Metabolite Quantification in Glycolytic Flux Assessment

Dynamic shifts in glycolytic intermediates (e.g., glucose-6-phosphate, lactate, pyruvate) provide critical insights into cellular energy dynamics, pathway regulation, and metabolic adaptations. Advanced quantification techniques enable precise evaluation of these biomarkers.

2. Enzymatic Assays for Targeted Metabolite Analysis

Spectrophotometric Methods

Enzymatic colorimetry employs substrate-specific enzymes to catalyze reactions, with metabolite levels quantified via absorbance changes.

Lactate Quantification

  • Principle: Lactate dehydrogenase (LDH) oxidizes lactate to pyruvate, reducing NAD⁺ to NADH, detectable at 340 nm.
  • Strengths: Cost-effective, high-throughput, and suitable for single-analyte detection.
  • Constraints: Limited to individual metabolites; incompatible with multiplexed profiling.

Glucose-6-Phosphate (G6P) Assay

  • Principle: G6P dehydrogenase (G6PDH) oxidizes G6P, generating NADPH, measured spectrophotometrically.
  • Utility: Robust for routine monitoring but lacks sensitivity for trace-level analytes.

3. Metabolomics-Driven Approaches

LC-MS

LC-MS combines chromatographic separation with mass detection, excelling in polar metabolite analysis (e.g., phosphorylated sugars).

  • Workflow:
    • Separation: Hydrophilic interaction liquid chromatography (HILIC; e.g., Waters XBridge BEH Amide).
    • Detection: Multiple reaction monitoring (MRM) for targeted quantification:
      • G6P: m/z 259.0 → 97.0
      • Fructose-1,6-bisphosphate: m/z 339.0 → 79.0
    • Advantages: Multiplexed detection, sub-nanomolar sensitivity, and matrix tolerance.
    • Challenges: Demands technical expertise; limited for low-abundance analytes (e.g., ATP).

GC-MS

GC-MS profiles volatile or derivatized metabolites (e.g., pyruvate via methoxyamine derivatization).

  • Protocols: Chemical modification (e.g., silylation) enhances volatility for chromatographic resolution.
  • Strengths: Unmatched resolution for ketones, fatty acids, and alcohols.
  • Limitations: Thermally labile metabolites require rapid quenching; derivatization complicates workflows.

4. Methodological Trade-offs and Innovations

TechniqueStrengthsLimitations
EnzymaticLow cost, simplicitySingle-analyte focus
LC-MSHigh multiplexing capabilityEquipment-intensive
GC-MSSuperior volatility resolutionDerivatization dependency

Emerging Trends

  • Hybrid Platforms: Coupling LC-MS with ion mobility for enhanced isomer separation.
  • Microsampling: Minimizing pre-analytical degradation via rapid freezing techniques.

Metabolic Flux Analysis in Glycolysis: Techniques and Innovations

Introduction to Metabolic Flux Analysis

Metabolic flux analysis (MFA) is a pivotal approach for investigating the dynamic rates of metabolites within cellular pathways. By quantifying metabolic flow, researchers gain insights into regulatory mechanisms, cellular energy requirements, and shifts in metabolic equilibrium. Advanced methodologies, including isotopic tracer techniques and computational MFA, integrate experimental data with mathematical modeling to map pathway dynamics accurately.

Isotopic Tracer Techniques

Carbon-13 Labeling Strategies

Stable isotope labeling, particularly with carbon-13 (¹³C), enables precise tracking of metabolic pathways. Substrates such as uniformly labeled [U-¹³C] glucose or position-specific [1,2-¹³C] glucose are utilized to trace carbon redistribution, offering insights into pathway contributions and branching points.

  • [U-¹³C] Glucose: Provides a uniformly labeled backbone for studying glycolysis, the tricarboxylic acid (TCA) cycle, and interconnected pathways.
  • [1,2-¹³C] Glucose: Distinguishes glycolysis from the pentose phosphate pathway (PPP) by analyzing differential carbon labeling patterns.

Applications

  • Pathway Contribution Analysis:
    • Glycolysis vs. PPP: Position-specific ¹³C labeling reveals distinct carbon fates, allowing calculation of flux ratios between glycolysis and PPP.
    • Pyruvate Utilization: Tracking ¹³C in pyruvate under varying oxygen conditions clarifies its conversion to lactate (anaerobic) or acetyl-CoA (aerobic).

[2,3-13C2]glucose for the assessment of the PPP and glycolysis.[2,3-13C2]glucose for the assessment of the PPP and glycolysis (Lee MH et al., 2019).

Instrumentation

  • LC-MS/MS: High-sensitivity separation and quantification of labeled metabolites in complex matrices, ideal for targeted flux studies.
  • NMR Spectroscopy: Non-destructive analysis offering real-time metabolite concentration and labeling data, albeit with lower resolution.

Metabolic Flux Analysis (MFA)

Principles and Workflow

MFA combines isotopic data with stoichiometric models to quantify reaction rates within metabolic networks.

  • Network Construction: Maps include glycolysis, TCA cycle, and ancillary pathways, detailing metabolite interconversions.
  • Data Acquisition: Isotope labeling and metabolite concentration data are collected via MS or NMR.
  • Computational Optimization: Algorithms minimize discrepancies between experimental data and model predictions to estimate flux distributions.

Software Tools

  • INCA: Facilitates flux estimation in complex networks using isotopic labeling data.
  • ¹³CFLUX2: Specialized for ¹³C tracer studies, optimizing flux calculations through iterative modeling.

Challenges in Flux Analysis

  • Data Variability: Experimental noise from sample handling or instrumentation affects accuracy.
  • Network Complexity: High-dimensional models with numerous reactions demand robust computational frameworks.
  • Resource Intensity: Large-scale networks require optimized algorithms to reduce computational overhead.

Future Directions

  • Temporal Flux Profiling: Transition from steady-state to dynamic models capturing real-time metabolic adaptations.
  • Integrative Omics: Coupling flux data with genomic, transcriptomic, and proteomic insights for holistic metabolic mapping.
  • Single-Cell Resolution: Advancing techniques to explore metabolic heterogeneity at the individual cell level, crucial in cancer and immunology.

Enzyme Activity and Regulatory Mechanisms

Objective

This investigation aims to elucidate the molecular mechanisms governing the catalytic function of key glycolytic enzymes, such as hexokinase (HK2) and pyruvate kinase M2 (PKM2), with a focus on post-translational modifications (PTMs). By exploring these regulatory processes, we seek to unravel their implications in cellular metabolism, oncogenesis, and metabolic disorders.

Methodological Framework

The study employs a multidisciplinary approach, integrating enzyme kinetics, proteomic profiling, and genome-editing technologies to dissect enzymatic regulation and its metabolic consequences.

1. Enzyme Kinetic Profiling

  • Principles: Kinetic analyses evaluate enzymatic activity by tracking substrate depletion or product formation. For glycolytic enzymes, factors such as substrate availability, pH, and allosteric modulators (inhibitors/activators) are assessed. Real-time spectrophotometric monitoring of cofactors (e.g., NADH/NADPH) provides insights into energy coupling and enzyme efficiency.
  • Protocols:
    • Hexokinase (HK2) Activity: Measure ATP consumption via NADH fluorescence as HK2 catalyzes glucose phosphorylation.
    • Pyruvate Kinase (PKM2) Activity: Quantify ATP synthesis and NADH oxidation during phosphoenolpyruvate (PEP) conversion to pyruvate.
  • Analytical Outcomes:
    • Michaelis-Menten Parameters: Determine maximal velocity (Vmax) and substrate affinity (Km) under varying conditions.
    • Mechanistic Insights: Assess how environmental variables (e.g., temperature, pH) modulate catalytic behavior.

2. Proteomic Profiling of Post-Translational Modifications

  • Principles: Proteomics identifies PTMs (e.g., phosphorylation, acetylation) that regulate enzyme function. Techniques include mass spectrometry (LC-MS/MS) and phospho-specific electrophoretic methods.
  • Applications:
    • Phosphorylation Analysis of PFK1:
      • Phos-tag Gel Electrophoresis: Resolves phospho-isoforms of fructose-1,6-bisphosphatase (PFK1) based on migration shifts.
      • Immunoblotting: Phospho-specific antibodies (e.g., anti-Ser32-PFK1) quantify inhibitory phosphorylation states.
  • Acetylation Profiling of PKM2:
    • LC-MS/MS Workflow:
      • Enrichment: Immunoprecipitation or chemical capture of acetylated peptides.
      • Identification: High-resolution mass spectrometry pinpoints acetylation sites (e.g., Lys305).
      • Quantification: Correlate acetylation levels with enzymatic activity changes.

3. CRISPR-Cas9-Mediated Genetic Manipulation

  • Principles: CRISPR-Cas9 enables precise gene editing to ablate or overexpress glycolytic enzymes, facilitating functional studies.
  • Applications:
    • Gene Knockout: Ablate HK2 or LDHA to assess impacts on glycolytic flux and cell proliferation.
    • Gene Overexpression: Engineer PKM2 or HK2 overexpression to study metabolic reprogramming in cancer models.
  • Integrated Metabolic Analysis:
    • Seahorse XF Technology:
      • Glycolytic Rate (ECAR): Measure extracellular acidification post-glucose challenge.
      • Oxidative Phosphorylation (OCR): Monitor oxygen consumption to evaluate mitochondrial activity.
      • Workflow: Analyze CRISPR-edited cells to link genetic perturbations to metabolic phenotypes.

Significance and Future Directions

By delineating PTM-driven regulatory networks and genetic dependencies in glycolysis, this study advances our understanding of metabolic dysregulation in diseases. Future work will leverage single-cell omics and dynamic flux models to capture spatiotemporal metabolic heterogeneity, offering novel therapeutic targets for cancer and metabolic syndromes.

Advanced Analytical Techniques

1. Spatial-Resolved Metabolomics

Spatial metabolomics investigates the distribution and dynamics of metabolites across tissue or cellular microenvironments using advanced imaging technologies such as mass spectrometry imaging (MSI) and Raman spectroscopy integrated with fluorescent probes. These approaches reveal how metabolite localization correlates with physiological or pathological states in complex biological systems.

Methodological Foundations

  • Mass Spectrometry Imaging (MSI): Enables direct, label-free metabolite mapping at cellular to organ-level resolution by analyzing molecular ions from sample surfaces.
  • Raman-Fluorescence Hybrid Systems: Combine vibrational spectroscopy for structural insights with fluorescent probes to enhance detection of low-abundance metabolites in biological specimens.

Applications

  • Tumor Microenvironment Studies: Identify hypoxia-driven metabolic adaptations, such as lactate accumulation in glycolytic tumor regions, elucidating cancer cell survival strategies.
  • Neuroscience Research: Map metabolic variations among neuronal populations, linking regional glycolysis rates to functional neural activity.

Challenges

  • Resolution Limits: Balancing spatial detail with sensitivity remains technically demanding.
  • Data Complexity: Integrating multidimensional metabolic datasets requires advanced computational frameworks.

2. Single-Cell Metabolomics

Single-cell metabolomics deciphers metabolic heterogeneity at the individual cell level, offering insights into disease mechanisms and personalized therapeutic strategies.

Core Technologies

  • Microfluidic Single-Cell Trapping: Enables precise isolation and metabolic profiling of individual cells.
  • Nano-Electrospray Ionization MS (nanoESI-MS): Achieves zeptomole-level sensitivity for detecting rare metabolites in minute cellular volumes.
  • Multi-Omics Integration: Couples single-cell RNA sequencing (scRNA-seq) with metabolite profiling to correlate gene expression with metabolic activity.

Applications

  • Oncology: Uncover metabolic rewiring in cancer cells, informing targeted therapies based on tumor-specific pathways.
  • Immunology: Characterize metabolic states of immune cell subsets to optimize checkpoint inhibitor or adoptive cell therapies.

Challenges

  • Low-Abundance Metabolites: Technical noise and quantification inaccuracies hinder detection of trace analytes.
  • Cellular Heterogeneity: Distinguishing biological variation from technical artifacts demands robust statistical models.
  • Data Synthesis: Managing high-dimensional omics datasets necessitates AI-driven analytical pipelines.

Future Directions

  • Multi-Omics Convergence: Integrate metabolomic data with genomic, proteomic, and epigenomic layers for holistic cellular portraits.
  • Technological Innovations: Enhance MSI resolution and nanoESI-MS sensitivity through novel ion sources and microfluidic designs.
  • Clinical Translation: Leverage spatial and single-cell insights for early disease diagnostics and precision medicine.

Application Scenarios and Case Analysis

  • Reveal the regulatory effect of metabolomics: In this study, the regulatory effects of methionine restriction (MetR) and glucose restriction (LowCarb) on the metabolomics of mouse L929 cells were compared by dynamic closed perfusion culture system (lasting for 7 days). Combined with LC-MS metabolomics analysis, it was found that although the restricted nutrients were different (methionine vs. glucose), both interventions significantly reduced glycolytic flux (pyruvate and lactic acid levels decreased) and exhausted ATP, resulting in high similarity of metabolomics. Further experiments show that glycolysis inhibitor 2- deoxy -D- glucose (2-DG) can rapidly induce energy depletion by blocking glycolysis flow within 72 hours, which verifies the key role of glycolysis inhibition (Volland JM et al., 2024).
  • Evaluation of cancer prognosis: In this study, the glycolytic metabolic activity (marked by FDG uptake) of patients with locally advanced esophageal squamous cell carcinoma (ESCC) was evaluated by sequential 18F-FDG PET/CT technique (before treatment and 3 months after treatment), and it was found that the metabolic tumor volume (MTV2) after treatment was significantly related to the prognosis. MTV 2 > 5.7 ml suggested that the residual glycolysis activity of tumor was high, which was related to worse overall survival (OS), progression-free survival (PFS) and local control rate (LRC) (p<0.05). By quantifying the metabolic load (such as MTV and TLG) driven by glycolysis, this technology provides a noninvasive imaging biomarker for dynamically monitoring tumor metabolic remodeling and identifying patients with high recurrence risk after radiotherapy and chemotherapy, and supports personalized treatment decision (Ha LN et al., 2023).
  • Cell stress metabolism: In this study, the high-sensitivity 13C metabolic flux analysis (combined with mathematical modeling) and single-cell ATP real-time detection technology were integrated to reveal the mechanism that mouse hematopoietic stem cells (HSC) dynamically regulate glycolysis to quickly respond to energy demand when proliferation or oxidative phosphorylation (OXPHOS) was inhibited. Single cell metabolic tracking accurately quantifies the glycolytic flux of rare HSC based on glucose tracing, avoiding the limitations of traditional batch pyrolysis methods; It is found that OXPHOS inhibition or proliferation stress activates PFKFB3 in a few seconds through AMPK phosphorylation or PRMT1 methylation, which drives the explosive production of glycolytic ATP (Yabushita T et al., 2024).
  • Developing biomarkers: In this study, hyperpolarized C-pyruvate magnetic resonance (MR) spectroscopy (real-time monitoring of pyruvate metabolic flow) combined with dynamic contrast-enhanced MR imaging was used to reveal the mechanism of long-term high-fat diet (HFD) driving brain metabolic disorder by enhancing glycolysis. Hyperpolarized MR technology can capture the changes of glycolysis flux in the brain non-invasively and dynamically, which proves that long-term HFD induces metabolic stress through the loop of "glycolysis dependence-oxidative metabolism inhibition" and provides early image markers for dementia risk (Choi YS et al., 2018).

People Also Ask

What techniques are used to monitor glycolysis?

Commonly used methods include liquid chromatography-mass spectrometry (LC-MS/MS) to detect lactic acid level, hyperpolarized C-pyruvate magnetic resonance spectroscopy (real-time tracking pyruvate-lactic acid transformation), Seahorse cell energy analyzer (measuring extracellular acidification rate ECAR) and fluorescent probe (such as 2-NBDG to monitor glucose uptake) to comprehensively evaluate metabolic flux, enzyme activity and energy dynamics.

What is the major regulatory method for glycolysis?

Dynamically respond to cell energy state and metabolic demand through allosteric regulation of key rate-limiting enzymes (such as phosphofructokinase -1) (such as ATP/AMP dynamic balance) and hormone-mediated covalent modification (such as insulin activation and glucagon inhibition).

How to choose the most suitable detection technology?

According to the experimental requirements: Seahorse(ECAR) or HP-MRS (in vivo/in vitro tissue) is selected for real-time dynamic monitoring. Metabolic flux analytical selection isotope tracing+mass spectrometry (providing path information of the whole metabolic network). Microplate fluorescence method (such as lactic acid detection kit) is selected for Qualcomm screening. Spatial resolution requires the selection of micro-fluorescence imaging (glycolytic activity of single cells).

References

  1. Cordani M, Michetti F, Zarrabi A, Zarepour A, Rumio C, Strippoli R, Marcucci F. "The role of glycolysis in tumorigenesis: From biological aspects to therapeutic opportunities." Neoplasia. 2024 Dec;58:101076. doi: 10.1016/j.neo.2024.101076
  2. Volland JM, Kaupp J, Schmitz W, Wünsch AC, Balint J, Möllmann M, El-Mesery M, Frackmann K, Peter L, Hartmann S, Kübler AC, Seher A. "Mass Spectrometric Metabolic Fingerprinting of 2-Deoxy-D-Glucose (2-DG)-Induced Inhibition of Glycolysis and Comparative Analysis of Methionine Restriction versus Glucose Restriction under Perfusion Culture in the Murine L929 Model System." Int J Mol Sci. 2022 Aug 16;23(16):9220. doi: 10.3390/ijms23169220
  3. Ha LN, Chau ND, Bieu BQ, Son MH. "The Prognostic Value of Sequential 18 F-FDG PET/CT Metabolic Parameters in Outcomes of Upper-Third Esophageal Squamous Cell Carcinoma Patients Treated with Definitive Chemoradiotherapy." World J Nucl Med. 2023 Sep 13;22(3):226-233. doi: 10.1055/s-0043-1774417
  4. Watanuki S, Kobayashi H, Sugiura Y, Yamamoto M, Karigane D, Shiroshita K, Sorimachi Y, Fujita S, Morikawa T, Koide S, Oshima M, Nishiyama A, Murakami K, Haraguchi M, Tamaki S, Yamamoto T, Yabushita T, Tanaka Y, Nagamatsu G, Honda H, Okamoto S, Goda N, Tamura T, Nakamura-Ishizu A, Suematsu M, Iwama A, Suda T, Takubo K. "Context-dependent modification of PFKFB3 in hematopoietic stem cells promotes anaerobic glycolysis and ensures stress hematopoiesis. Elife. 2024 Apr 4;12:RP87674. doi: 10.7554/eLife.87674." Erratum in: Elife. 2024 Oct 22;13:e104576. doi: 10.7554/eLife.87674
  5. Choi YS, Kang S, Ko SY, Lee S, Kim JY, Lee H, Song JE, Kim DH, Kim E, Kim CH, Saksida L, Song HT, Lee JE. "Hyperpolarized [1-13C] pyruvate MR spectroscopy detect altered glycolysis in the brain of a cognitively impaired mouse model fed high-fat diet." Mol Brain. 2018 Dec 18;11(1):74. doi: 10.1186/s13041-018-0415-2
  6. Lee MH, Malloy CR, Corbin IR, Li J, Jin ES. "Assessing the pentose phosphate pathway using [2, 3-13 C2 ]glucose." NMR Biomed. 2019 Jun;32(6):e4096. doi: 10.1002/nbm.4096
* For Research Use Only. Not for use in diagnostic procedures.
Our customer service representatives are available 24 hours a day, 7 days a week. Inquiry

From Our Clients

Online Inquiry

Please submit a detailed description of your project. We will provide you with a customized project plan to meet your research requests. You can also send emails directly to for inquiries.

* Email
Phone
* Service & Products of Interest
Services Required and Project Description
* Verification Code
Verification Code

Great Minds Choose Creative Proteomics