PKM2 (Pyruvate Kinase M2) is a glycolytic enzyme isoform critical in cancer metabolism, particularly in the Warburg effect. PKM2 antibodies are immunological reagents designed to detect and study this protein in various biological contexts. These antibodies are polyclonal or monoclonal, targeting specific epitopes of PKM2, including phosphorylated residues (e.g., Ser37) or isoform-specific regions. Their applications span Western blotting (WB), immunohistochemistry (IHC), immunofluorescence (IF/ICC), and enzyme-linked immunosorbent assay (ELISA) .
PKM2 antibodies are pivotal in elucidating PKM2’s non-metabolic roles, such as regulating apoptosis, angiogenesis, and transcriptional activity. Below are key applications supported by experimental data:
PKM2 antibodies enable tracking of its subcellular distribution, which correlates with therapeutic outcomes:
Cytosolic PKM2: Dominant in aggressive NSCLC; depletion reduces tumor growth .
Nuclear PKM2: Associated with resistance to PKM2-targeted therapies (e.g., shRNA) .
Circulating PKM2 promotes tumor angiogenesis by increasing CD31+ endothelial cell density. Neutralizing antibodies (e.g., IgGPK) reduce vascularization in SW620 and PC-3 xenografts .
Phosphorylated PKM2 (e.g., p-Ser37) is critical for metabolic reprogramming. The SAB antibody detects this modification, enabling studies on HIF-1α stabilization in glioblastoma .
PKM2 antibodies have unveiled therapeutic targets:
PAT1B10AT.
PKM2 (Pyruvate Kinase M2) is a metabolic enzyme that catalyzes the final step of glycolysis, converting phosphoenolpyruvate (PEP) to pyruvate while generating ATP. Beyond its canonical glycolytic function, PKM2 exhibits unique regulatory properties that distinguish it from other pyruvate kinase isoforms. PKM2 is highly expressed during embryogenesis and remains expressed at varying levels throughout adult tissues, becoming the predominant isoform in proliferating cells and most cancer types .
The significance of PKM2 in research stems from its dual functionality:
As a metabolic enzyme regulating the balance between glycolysis and biosynthetic pathways
As a protein kinase capable of phosphorylating various targets including histone H3 and Erk1/2
This dual role positions PKM2 at the intersection of cellular metabolism and signaling, making it an important target for understanding how metabolic alterations contribute to disease states, particularly cancer.
For PKM2 detection and characterization, researchers have access to several antibody types:
Methodologically, researchers should select antibodies based on the intended application and required specificity. For applications requiring discrimination between PKM1 and PKM2 isoforms, antibodies targeting PKM2-specific regions (like the one mentioned in the search results targeting the peptide sequence "LRRLAPITSDPTEATAVGAV") are essential .
PKM2 antibodies are widely employed across multiple experimental techniques:
Western Blotting: For quantifying PKM2 protein expression levels and detecting post-translational modifications. Recommended dilutions typically range from 1:1000 to 1:5000, depending on antibody sensitivity and sample type.
Immunoprecipitation (IP): For isolating PKM2 complexes and identifying interacting partners. As demonstrated in the literature, PKM2 forms complexes with proteins including PDC-E2, p300, and AhR on chromatin .
Immunohistochemistry (IHC): For examining PKM2 expression patterns in tissue sections, particularly useful in cancer pathology studies.
Immunofluorescence (IF): For visualizing subcellular localization of PKM2, which can shuttle between cytoplasmic and nuclear compartments.
Chromatin Immunoprecipitation (ChIP): For detecting PKM2 binding to chromatin, particularly at gene enhancers as observed for AhR-target genes like CYP1A1 .
When designing experiments, researchers should include appropriate controls, such as PKM2-depleted cells (using shRNA or CRISPR) to validate antibody specificity .
Validating PKM2 antibody specificity is crucial given the high sequence similarity (>90%) between PKM1 and PKM2 isoforms, which differ primarily in the mutually exclusive inclusion of exon 9 (PKM1) or exon 10 (PKM2). A comprehensive validation approach includes:
Knockout/Knockdown Controls: Generate PKM2-specific knockouts using CRISPR-Cas9 targeting exon 10, or knockdowns using exon 10-targeting shRNAs as described in the literature . Examples from the search results include shPKM2#1 (5'-GCTGTGGCTCTAGACACTA-3') and shPKM2#2 (5'-GTTCGGAGGTTTGATGAAA-3') .
Overexpression Validation: Perform rescue experiments with shRNA-resistant PKM2 constructs to confirm antibody specificity, as demonstrated in published protocols .
Isoform-Specific Peptide Competition: Pre-incubate antibodies with PKM1 and PKM2-specific peptides to determine epitope specificity.
Mass Spectrometry Correlation: Validate western blot results with mass spectrometry-based quantification of PKM1 and PKM2 peptides.
Multiple Antibody Concordance: Use multiple antibodies targeting different PKM2 epitopes to confirm consistent results.
The specific validation method should be selected based on the research question and experimental context, with results systematically documented to establish antibody reliability.
Distinguishing between PKM2's pyruvate kinase and protein kinase activities requires specialized experimental designs:
For Pyruvate Kinase Activity:
Spectrophotometric assays measuring the conversion of PEP to pyruvate coupled to NADH oxidation
Isotopic tracing with U-13C glucose to track carbon flux through glycolysis
For Protein Kinase Activity:
In vitro kinase assays with recombinant PKM2, target proteins, and ATP
Detection of specific phosphorylation events using phospho-specific antibodies
SAICAR-mediated activation of PKM2's protein kinase activity in controlled reactions
Differential Analysis Approaches:
Site-Directed Mutagenesis: Use of the K367M mutant, which disrupts protein kinase activity while preserving pyruvate kinase function
Metabolite Modulation: Application of SAICAR to enhance protein kinase activity
Subcellular Fractionation: Separate analysis of cytosolic versus nuclear PKM2 functions
ChIP-Seq Combined with Metabolic Analysis: Correlate PKM2 chromatin binding with metabolic shifts in the same experimental system
When reporting results, researchers should clearly distinguish which PKM2 function they are measuring and include appropriate controls for each activity.
PKM2's non-canonical roles include nuclear translocation and chromatin association, which require specific detection methodologies:
Subcellular Fractionation Protocol:
Chromatin Immunoprecipitation (ChIP):
Sequential ChIP (Re-ChIP):
For detecting PKM2 co-occupancy with transcription factors like AhR
Perform first IP with anti-AhR antibody, followed by second IP with anti-PKM2 antibody
Immunofluorescence Microscopy:
Visualize PKM2 subcellular localization in fixed cells
Co-stain with DAPI for nuclear visualization
Quantify nuclear/cytoplasmic ratios under different conditions
When analyzing results, researchers should consider that nuclear translocation often occurs in response to specific stimuli (e.g., growth factor signaling, metabolic stress) and may involve post-translational modifications of PKM2.
Based on recent findings, PKM2 plays a critical role in regulating oxidative stress, particularly in cardiomyocytes . Effective experimental designs include:
Genetic Manipulation Models:
Oxidative Stress Assessment Panel:
Measure total reactive oxygen species (ROS) using 2',7'-dichlorofluorescin diacetate (DCFDA)
Quantify mitochondrial superoxide using MitoSOX Red
Assess lipid peroxidation through malondialdehyde (MDA) or 4-hydroxynonenal (4-HNE) levels
Measure antioxidant enzyme activities (SOD, catalase, GPx)
Metabolic Flux Analysis:
Integrated Functional Assessment:
Research has demonstrated that PKM2−/− hearts show increased ROS levels, reduced ATP content, and altered mitochondrial function, suggesting PKM2 acts as an important regulator of metabolic flux and oxidative stress in cardiomyocytes .
Researchers frequently encounter variability in PKM2 antibody results due to several factors:
Post-Translational Modifications:
PKM2 undergoes numerous modifications (phosphorylation, acetylation, oxidation) that can affect epitope recognition
Solution: Use multiple antibodies targeting different regions, or use modification-insensitive antibodies for total PKM2 detection
Conformation-Dependent Epitope Accessibility:
PKM2 exists in different oligomeric states (monomer, dimer, tetramer) with varying epitope exposure
Solution: Optimize sample preparation to maintain native states or deliberately denature all forms
Cross-Reactivity with PKM1:
High sequence homology can lead to non-specific detection
Solution: Validate with recombinant PKM1 and PKM2 controls; use PKM2-specific antibodies targeting unique regions
Batch-to-Batch Variability:
Commercial antibody lots can vary in performance
Solution: Test and validate each new lot against previous standards; maintain reference samples
Sample Preparation Effects:
Extraction methods can affect PKM2 detection, particularly when assessing subcellular localization
Solution: Standardize lysis buffers and fractionation protocols; include protease and phosphatase inhibitors
Implementing rigorous validation protocols and maintaining detailed records of antibody performance across experiments will help minimize inconsistencies and improve reproducibility.
When confronted with conflicting results from different PKM2 antibodies, a systematic troubleshooting approach is essential:
Epitope Mapping Assessment:
Compare the specific epitopes recognized by each antibody
Different antibodies may detect different PKM2 populations based on post-translational modifications or conformational states
Validation with Multiple Techniques:
Controlled Rescue Experiments:
Functional Correlation Analysis:
Correlate antibody signals with functional readouts of PKM2 activity
For instance, correlate detected protein levels with pyruvate kinase activity measurements
Statistical Meta-Analysis:
When possible, perform quantitative analysis across multiple experiments
Report confidence intervals rather than single values
When reporting discrepant results, researchers should transparently document the specific antibodies used, their sources, catalog numbers, and the exact experimental conditions to facilitate interpretation and reproducibility.
The interaction between PKM2 and SAICAR (succinylamino-imidazolecarboxamide ribose-5′-phosphate) represents a key regulatory mechanism activating PKM2's protein kinase function . To investigate this complex:
Co-Immunoprecipitation Strategy:
Use PKM2 antibodies for pull-down experiments from cells with manipulated SAICAR levels
Identify complex formation through western blotting or mass spectrometry
Compare results under conditions that alter SAICAR abundance (e.g., proliferation states)
In Vitro Reconstitution Assays:
Combine purified recombinant PKM2 with synthetic SAICAR
Measure protein kinase activity using:
Radioactive ATP incorporation assays
Phospho-specific antibodies to detect substrate phosphorylation
Kinase activity sensing systems
Substrate Profiling:
Structural Studies:
Use antibody fragments to stabilize PKM2-SAICAR complexes for structural analysis
Apply X-ray crystallography or cryo-EM to determine complex structure
Cellular Signaling Investigation:
This methodological approach allows researchers to dissect the unique non-metabolic functions of PKM2 when complexed with SAICAR and understand how this interaction links metabolic status to cellular signaling.
PKM2's nuclear functions include participation in transcriptional complexes and chromatin modification . To investigate these processes:
Chromatin Immunoprecipitation (ChIP) Protocol:
Sequential ChIP (Re-ChIP):
First immunoprecipitate with antibodies against known PKM2-interacting transcription factors (e.g., AhR)
Follow with PKM2 antibodies to confirm co-occupancy at specific genomic sites
Histone Modification Analysis:
Transcriptional Complex Characterization:
Functional Reporter Assays:
Employ luciferase reporters driven by PKM2-responsive promoters/enhancers
Compare activity in PKM2 wild-type, depleted, and rescue conditions
This integrated approach enables researchers to decode PKM2's contribution to transcriptional regulation beyond its metabolic functions, particularly in contexts like xenobiotic metabolism and cancer cell proliferation.
Recent evidence suggests PKM2 plays a crucial role in cardiac metabolism and protection against injury . To explore this cross-talk:
Cardiac-Specific Genetic Models:
Generate cardiomyocyte-specific PKM2 knockout or overexpression models
Use Cre-loxP systems for temporal control of PKM2 expression
Ischemia-Reperfusion Studies:
Metabolic Characterization:
Mitochondrial Function Assessment:
Measure oxygen consumption rates in isolated mitochondria
Quantify reactive oxygen species production
Assess mitochondrial membrane potential and calcium handling
Signaling Pathway Integration:
Research indicates that PKM2 may act as a metabolic rheostat in the heart, maintaining ATP levels while limiting oxidative stress . Understanding these mechanisms could lead to novel therapeutic approaches for cardiac protection.
As single-cell technologies advance, new approaches for PKM2 detection offer exciting research potential:
Single-Cell Antibody-Based Technologies:
Development of PKM2 antibodies compatible with CyTOF (mass cytometry)
Optimization for single-cell western blotting platforms
Integration with microfluidic antibody capture techniques
Proximity Ligation Assays (PLA):
Creation of PKM2-specific PLA probes to detect protein-protein interactions at single-molecule resolution
Application to visualize PKM2 dimers versus tetramers in situ
Combination with other markers to map PKM2 interactome in different cellular compartments
Antibody-Based Biosensors:
Development of FRET-based sensors using PKM2 antibody fragments
Creation of nanobodies against conformation-specific PKM2 epitopes
Design of split-GFP complementation systems for detecting PKM2 oligomerization states
Single-Cell Spatial Transcriptomics Integration:
Pairing PKM2 protein detection with transcriptomic analysis in tissue sections
Correlation of PKM2 localization with metabolic gene expression programs
Mapping of PKM2-dependent transcriptional changes at single-cell resolution
In Situ Conformational Analysis:
Development of antibodies that specifically recognize different PKM2 quaternary structures
Application to map the distribution of catalytically active versus inactive PKM2 within tissues
These approaches would enable researchers to understand PKM2's diverse functions with unprecedented spatial and temporal resolution in heterogeneous cell populations.
Understanding the dynamic regulation of PKM2 requires sophisticated conditional systems:
Optogenetic PKM2 Control Systems:
Development of light-inducible PKM2 expression or degradation systems
Creation of photoswitchable PKM2 variants with tunable kinase activities
Integration with PKM2 antibody-based detection for real-time monitoring
Chemical-Genetic Approaches:
Engineering PKM2 variants sensitive to small-molecule control
Development of rapid protein degradation systems (e.g., AID or dTAG) for PKM2
Creation of compartment-specific PKM2 regulation through localized enzyme activators
Temporal-Spatial Antibody-Based Detection:
Integration of destabilized fluorescent reporters with PKM2 antibodies
Development of antibodies sensitive to PKM2 post-translational modifications
Creation of FRET-based reporters of PKM2 enzymatic activities
Live-Cell Imaging Strategies:
Application of antibody fragments (Fabs) for live-cell tracking of PKM2
Development of split fluorescent protein complementation systems for monitoring PKM2 interactions
Integration with metabolic sensors to correlate PKM2 dynamics with metabolic state
Multi-Modal Integration:
Correlation of PKM2 antibody-based imaging with metabolic flux measurements
Simultaneous detection of PKM2 activity and downstream signaling events
Development of computational models to predict PKM2 behavior based on integrated datasets
These approaches would enable researchers to dissect the complex regulation of PKM2 in both physiological and pathological contexts, potentially revealing new therapeutic opportunities.
Tumor Type M2 Pyruvate Kinase (PKM2) is a key enzyme in the glycolytic pathway, specifically expressed at high levels in tumor cells. It plays a crucial role in the metabolic reprogramming of cancer cells, facilitating their rapid growth and proliferation. The enzyme is encoded by the PKM gene and is one of the four isozymes of pyruvate kinase found in mammals, the others being L, R, and M1 .
PKM2 catalyzes the transfer of a phosphate group from phosphoenolpyruvate (PEP) to adenosine diphosphate (ADP), generating adenosine triphosphate (ATP) and pyruvate in the final step of glycolysis. This reaction is essential for the production of energy in cells. Unlike other pyruvate kinase isozymes, PKM2 can exist in different oligomeric states, including a highly active tetrameric form and a less active dimeric form. The dimeric form is often found in cancer cells and is associated with the Warburg effect, where cancer cells preferentially produce energy through aerobic glycolysis even in the presence of oxygen .
PKM2 is not only involved in glycolysis but also plays non-metabolic roles that contribute to tumorigenesis. It can translocate to the nucleus and act as a protein kinase, phosphorylating various substrates involved in cell proliferation and survival. This dual functionality makes PKM2 a critical player in cancer metabolism and a potential target for cancer therapy .
Mouse anti-human PKM2 antibodies are monoclonal antibodies specifically designed to target the human form of PKM2. These antibodies are used in research and diagnostic applications to detect and quantify PKM2 levels in various biological samples. They are particularly useful in studying the role of PKM2 in cancer and in developing therapeutic strategies targeting this enzyme .
Given its central role in cancer metabolism, PKM2 has emerged as a promising target for cancer therapy. Inhibitors of PKM2 activity, as well as agents that promote its tetramerization, are being explored as potential therapeutic strategies. For instance, TEPP-46 is a small molecule that induces the tetramerization of PKM2, thereby enhancing its glycolytic activity and reducing its non-metabolic functions that promote tumor growth .