PKM Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

PKM: Biological Significance

PKM is a glycolytic enzyme that catalyzes the final step of glycolysis, converting phosphoenolpyruvate (PEP) to pyruvate while generating ATP. Two splice variants exist:

  • PKM1: Expressed in tissues with high energy demands (e.g., muscle, brain).

  • PKM2: Predominantly found in proliferating cells, including cancer cells, where it supports aerobic glycolysis (the Warburg effect).

PKM2’s dimeric form promotes lactate production and nucleotide biosynthesis, making it a key driver of tumor growth . Elevated PKM2 levels in blood or stool are associated with gastrointestinal, lung, and breast cancers, positioning it as a biomarker for early diagnosis and prognosis .

Production Methodology

PKM mAbs are generated through hybridoma technology:

  1. Immunization: Mice are immunized with recombinant PKM protein .

  2. Hybridoma Formation: Splenic B cells are fused with myeloma cells to create antibody-producing hybridomas .

  3. Screening: Hybridomas are selected for high-affinity PKM binding .

  4. Purification: Antibodies are purified via Protein G chromatography .

Research Applications

PKM mAbs are widely used in:

  • Cancer Research: Detecting PKM2 overexpression in tumor tissues to assess metabolic reprogramming .

  • Diagnostics: Quantifying PKM2 in plasma or stool as a non-invasive biomarker for cancers .

  • Mechanistic Studies: Investigating PKM2’s interaction with oncogenic transcription factors (e.g., Oct-4) in stem cell regulation .

Validation Data

  • Western Blot: Anti-PKM (C-11) detects a ~60 kDa band in HeLa lysates .

  • Flow Cytometry: EPR10138(B) shows strong intracellular PKM staining in permeabilized HeLa cells .

  • Immunohistochemistry: Clone 6C3C7 localizes PKM2 in colorectal cancer biopsies .

Clinical and Therapeutic Implications

While PKM mAbs are primarily research tools, their diagnostic utility is well-established:

  • Biomarker Potential: PKM2 levels correlate with tumor stage and metastasis in gastric, lung, and liver cancers .

  • Therapeutic Targeting: PKM2’s role in glycolysis and tumor growth positions it as a candidate for small-molecule inhibitors or antibody-drug conjugates .

Challenges and Future Directions

  • Isoform Specificity: Many PKM mAbs cross-react with both PKM1 and PKM2, necessitating improved isoform-selective antibodies .

  • Standardization: Batch-to-batch variability in antibody concentration and affinity requires rigorous quality control .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

This monoclonal anti-PKM antibody (mouse IgG1 isotype) is produced from a hybridoma generated by fusing mouse myeloma cells with splenocytes from an immunized mouse. Splenocytes were isolated from a mouse immunized with recombinant human pyruvate kinase PKM protein (amino acids 2-531). The resulting antibody secreting hybridoma produces this unconjugated PKM antibody, which is purified using protein A chromatography to achieve >95% purity. This antibody exhibits cross-reactivity with human and mouse PKM and is suitable for ELISA, Western blotting (WB), immunohistochemistry (IHC), immunoprecipitation (IP), immunofluorescence (IF), and flow cytometry (FC) applications.

PKM is a glycolytic enzyme that catalyzes the conversion of phosphoenolpyruvate and ADP to pyruvate and ATP. It plays a crucial role in regulating cellular metabolism.

Form
Liquid
Lead Time
Orders typically ship within 1-3 business days of receipt. Delivery times may vary depending on shipping method and destination. Please contact your local distributor for specific delivery timelines.
Synonyms
CTHBP antibody; Cytosolic thyroid hormone-binding protein antibody; KPYM_HUMAN antibody; OIP-3 antibody; Opa-interacting protein 3 antibody; p58 antibody; pkm antibody; PKM1 antibody; PKM2 antibody; Pyruvate kinase 2/3 antibody; Pyruvate kinase muscle isozyme antibody; Pyruvate kinase PKM antibody; THBP1 antibody; Thyroid hormone-binding protein 1 antibody; Tumor M2-PK antibody
Target Names
PKM
Uniprot No.

Target Background

Function

Pyruvate kinase M (PKM) is a glycolytic enzyme that catalyzes the transfer of a phosphoryl group from phosphoenolpyruvate (PEP) to ADP, producing ATP. The equilibrium between the highly active tetrameric form and the less active dimeric form regulates the flux of glucose carbons into either biosynthetic pathways or glycolytic ATP production. This dynamic equilibrium contributes to glycolytic control and is critical for tumor cell proliferation and survival. Beyond its glycolytic role, PKM also modulates transcription, stimulating POU5F1-mediated transcriptional activation and promoting, in a STAT1-dependent manner, the expression of the immune checkpoint protein CD274 in ARNTL/BMAL1-deficient macrophages. Furthermore, PKM functions as a translational regulator for specific mRNAs, independent of its kinase activity. It associates with subsets of endoplasmic reticulum-associated ribosomes, directly binds to mRNAs translated at the endoplasmic reticulum, and promotes the translation of these ER-destined mRNAs. PKM also plays a general role in caspase-independent cell death of tumor cells.

Gene References Into Functions
  1. nBP1a/PKM2 interaction activates lipid metabolism genes in cancer cells; Thr-59 phosphorylation of SREBP-1a is crucial for cancer cell proliferation. PMID: 29514980
  2. PKM2 knockdown inhibits gastric cancer (GC) cell proliferation, G1-S phase transition, and cell migration, while promoting autophagy, potentially via the PI3K-Akt signaling pathway. PMID: 28588255
  3. PKM1 activates glucose catabolism and stimulates autophagy/mitophagy, promoting malignancy. PMID: 29533781
  4. miRNA-139-5p inhibits cell proliferation, migration, and glycolysis in gallbladder cancer (GBC) by repressing pyruvate kinase M2. PMID: 30105813
  5. The mammalian target of rapamycin (mTOR) pathway promotes aerobic glycolysis in esophageal squamous cell carcinoma by upregulating PKM2. PMID: 29916308
  6. PKM2 promotes tumor cell exosome release by phosphorylating SNAP23. PMID: 28067230
  7. PKM2 influences bladder cancer progression via the mitogen-activated protein kinase signaling pathway, affecting proteins such as MMP2, MMP9, and p38. PMID: 30249877
  8. Hypoxic stress in hepatocellular carcinoma (HCC) cells promotes YAP binding to HIF-1α, stabilizing HIF-1α to bind the PKM2 gene and activate its transcription, accelerating glycolysis. PMID: 30180863
  9. Overexpressed PKM2 increases CCND1 and decreases CDKN1A expression, while underexpressed PKM2 shows the opposite effect in ovarian cancer cells. PMID: 29752805
  10. PKM2 plays a regulatory role in HIV-1-host interactions, offering potential therapeutic targets. PMID: 29607934
  11. PKM2 is a novel target of RUNX1-ETO and is downregulated in RUNX1-ETO-positive acute myeloid leukemia (AML) patients, suggesting diagnostic potential. PMID: 28092997
  12. High PKM2 expression is associated with pancreatic and peri-ampullary cancer. PMID: 29540198
  13. PKM2 is essential for the development and metastasis of osteosarcoma (OS). PMID: 29155364
  14. Elevated PKM2 levels are necessary for myeloid dendritic cell (mDC) activation and improve the immune status of patients with systemic amyloidosis (SAA) by enhancing mDC and cytotoxic T lymphocyte (CTL) function. PMID: 29636835
  15. EGFR activation leads to c-Src-mediated Cdc25A phosphorylation at Y59, which interacts with nuclear PKM2. PMID: 27485204
  16. PKM2 plays an important role in the metabolic activity and malignancy of pancreatic ductal adenocarcinoma cells. PMID: 29393401
  17. PKM2 mediates the interaction between pancreatic cancer cells and pancreatic stellate cells. PMID: 29619774
  18. HSP90 regulates PKM2 expression in hepatocellular carcinoma (HCC) by enhancing its stability through Thr-328 phosphorylation. PMID: 29262861
  19. O-GlcNAcylation regulates PKM2 in cancer cells, linking it to metabolic reprogramming characteristic of the Warburg effect. PMID: 29229835
  20. PKM2 expression is positively correlated with Sp1 expression, and Sp1 directly regulates PKM2 expression in castration-resistant prostate cancer. PMID: 29094170
  21. High PKM2 expression is associated with lung adenocarcinoma. PMID: 28489603
  22. Epigenetic silencing of miR-338 promotes glioblastoma progression by preventing suppression of the PKM2/β-catenin axis. PMID: 28858851
  23. Tumor M2 pyruvate kinase (TuM2PK) correlates with tumor size, C-reactive protein (CRP), and CA 15-3 in metastatic breast carcinomas. PMID: 28869444
  24. PKM2 modulates glycolysis and extracellular matrix generation, playing a vital role in osteoarthritis (OA) pathogenesis. PMID: 29356574
  25. Osteoarthritis increases the PKM1/PKM2 ratio, activating HNF-4α and promoting hepatoma differentiation. PMID: 28726775
  26. PKM2 and glutaminase 1 (GLS1) are associated with oxaliplatin resistance in colorectal cancer (CRC). PMID: 28498807
  27. Elevated PKM2 expression predicts poor clinical outcomes in gallbladder cancer (GBC), suggesting involvement in GBC progression. PMID: 27283076
  28. PKM2 knockdown increases p53 expression and prolongs its half-life by binding to both p53 and MDM2, promoting MDM2-mediated p53 ubiquitination; dimeric PKM2 significantly suppresses p53 expression. PMID: 27801666
  29. Inhibition of the mTOR pathway abolishes transforming growth factor-beta 1 (TGF-β1)-induced epithelial-mesenchymal transition (EMT) and reduces mTOR/p70s6k signaling, downregulating PKM2 expression. PMID: 28446743
  30. Nitric oxide induces PKM2 nuclear translocation and promotes glycolysis in ovarian cancer cells. PMID: 28380434
  31. AKT directly interacts with and phosphorylates PKM2 at Ser-202, crucial for nuclear translocation under insulin-like growth factor 1 (IGF-1) stimulation; nuclear PKM2 binds STAT5A and induces IGF-1-stimulated cyclin D1 expression. PMID: 27340866
  32. Targeting PKM2 with an oncolytic adenovirus produces a strong antitumor effect. PMID: 28569774
  33. miR-let-7a inhibits cell proliferation, migration, and invasion by downregulating PKM2 in cervical cancer. PMID: 28415668
  34. Cytomegalovirus US28 signaling activates the HIF-1α/PKM2 feedforward loop in fibroblasts and glioblastoma cells. PMID: 27602585
  35. High PKM2 expression is associated with urothelial tumorigenesis. PMID: 26992222
  36. PKM2 is succinylated at lysine 498; succinylation increases its activity; SIRT5 desuccinylates and inhibits PKM2; increased reactive oxygen species (ROS) decrease succinylation and activity by increasing SIRT5 binding. PMID: 28036303
  37. Overexpression of PKM2 is associated with poor prognosis in most solid cancers and may be a useful prognostic biomarker. PMID: 27911861
  38. GLO I and PKM2 are interdependent in the metabolic shift to escape apoptosis in GLO I-dependent cancer cells. PMID: 29225125
  39. Lapachol inhibits glycolysis in cancer cells by targeting PKM2, unlike shikonin, which also targets mitochondria. PMID: 29394289
  40. p53 (N340Q/L344R) promotes hepatocarcinogenesis by upregulating PKM2. PMID: 27167190
  41. PKM2 silencing promotes apoptosis and inhibits aerobic glycolysis, proliferation, migration, and invasion in colorectal cancer cells. PMID: 28543190
  42. There is insufficient evidence for the diagnostic and prognostic use of PKM2 in ovarian cancer. PMID: 29277786
  43. UCP2 stimulates hnRNPA2/B1, GLUT1, and PKM2 expression and sensitizes pancreatic cancer cells to glycolysis inhibition. PMID: 27989750
  44. Mitochondrial PKM2 phosphorylates Bcl2 and directly inhibits apoptosis. PMID: 28035139
  45. PKM2 and GLS play important roles in the proliferation of hypoxic gastric cancer cells. PMID: 29032577
  46. lincRNA-p21 inhibits prostate cancer cell proliferation and tumorigenesis by downregulating PKM2. PMID: 28994148
  47. SHP-1 dephosphorylates PKM2Y105 to inhibit the Warburg effect and nucleus-dependent cell proliferation; this dephosphorylation determines the efficacy of targeted drugs for hepatocellular carcinoma. PMID: 26959741
  48. PKM2 shRNA reduces PKM2 expression and increases p53 and p21 expression. PMID: 28746922
  49. The CARM1-PKM2 axis is a metabolic reprogramming mechanism in tumorigenesis. PMID: 29058718
  50. PKM2 activity is higher in non-small cell lung cancer (NSCLC) patients and is associated with advanced cancer stages. PMID: 27683215
Database Links

HGNC: 9021

OMIM: 179050

KEGG: hsa:5315

STRING: 9606.ENSP00000320171

UniGene: Hs.534770

Protein Families
Pyruvate kinase family
Subcellular Location
Cytoplasm. Nucleus.
Tissue Specificity
Specifically expressed in proliferating cells, such as embryonic stem cells, embryonic carcinoma cells, as well as cancer cells.

Q&A

What is the mechanism of action for PKM monoclonal antibodies in research applications?

PKM monoclonal antibodies function through selective binding to specific epitopes on pyruvate kinase M isoforms. These laboratory-produced molecules are engineered to serve as substitute antibodies that can specifically recognize and bind to PKM antigens with high affinity and specificity. When an antibody binds to PKM, it serves as a flag to attract disease-fighting molecules or as a detection tool in various experimental procedures. Unlike polyclonal antibodies, monoclonal antibodies offer unparalleled specificity and uniformity, making them ideal for targeting specific domains or isoforms of the PKM enzyme. These antibodies can be designed to function through various mechanisms, including direct binding to active sites, recognition of post-translational modifications, or identification of specific conformational states of the PKM protein .

How should researchers validate the specificity of PKM monoclonal antibodies before experimental use?

Validation of PKM monoclonal antibody specificity requires a multi-faceted approach to ensure experimental rigor. Initially, researchers should conduct Western blot analysis using positive controls (cells known to express PKM) and negative controls (PKM knockout cells or tissues). Immunoprecipitation followed by mass spectrometry can confirm that the antibody captures the intended PKM isoform. Cross-reactivity testing against related proteins (like PKL or PKR) is essential to establish specificity boundaries. Additional validation should include immunohistochemistry on tissues with known PKM expression patterns and ELISA titration to determine optimal working concentrations. For knockdown validation, researchers should test the antibody against samples from PKM-silenced cells (using siRNA or CRISPR) to confirm signal reduction proportional to protein depletion. This comprehensive validation ensures that experimental outcomes truly reflect PKM-specific effects rather than non-specific antibody interactions .

What are the key considerations for incorporating PKM monoclonal antibodies in immunohistochemistry protocols?

When incorporating PKM monoclonal antibodies into immunohistochemistry protocols, researchers must carefully optimize several parameters. First, tissue fixation and antigen retrieval methods must be calibrated specifically for PKM epitopes, as over-fixation can mask antigenic sites while insufficient fixation leads to poor tissue morphology. The selection of blocking reagents should account for potential non-specific binding characteristics of the particular PKM antibody clone. Researchers should determine optimal antibody concentration through titration experiments, typically starting with 1-10 μg/ml depending on the antibody's affinity. Incubation conditions (temperature, duration, buffer composition) significantly impact staining quality and should be systematically optimized. Importantly, researchers must include appropriate controls: positive tissue controls with known PKM expression, negative controls lacking PKM, isotype controls to distinguish specific from non-specific binding, and antibody absorption controls using recombinant PKM protein. Signal amplification systems should be selected based on the expected abundance of PKM in the target tissue. Finally, quantification methods must be standardized, preferably using digital image analysis to ensure objective assessment of staining patterns and intensity .

How can researchers determine the optimal dose of PKM monoclonal antibodies for in vivo experiments?

Determining the optimal dose for PKM monoclonal antibodies in vivo requires a systematic approach that accounts for both pharmacokinetic and pharmacodynamic parameters. Researchers should begin with dose-ranging pilot studies that evaluate multiple concentrations, typically spanning at least three orders of magnitude. The minimal anticipated biological effect level (MABEL) approach is particularly valuable for novel PKM antibodies, where receptor occupancy should initially be targeted at no more than 10% for safety considerations. For antagonistic PKM antibodies, higher receptor occupancy may be acceptable. PK/PD modeling that incorporates both target-mediated and non-specific clearance mechanisms should be employed to predict optimal dosing intervals and exposure levels.

Importantly, researchers must consider species-specific differences in PKM binding affinity and distribution when translating doses between experimental models. Biomarker analysis (measuring downstream effects of PKM inhibition) should complement direct measurements of antibody concentration. When designing repeat-dose studies, researchers must account for potential immunogenicity that may alter clearance rates over time. Target saturation analysis using ex vivo samples can confirm whether the selected dose achieves the desired level of PKM binding in target tissues. This comprehensive approach ensures that in vivo experiments are conducted with doses that achieve consistent and physiologically relevant modulation of PKM activity .

What approaches can reliably distinguish between PKM1 and PKM2 isoforms when using monoclonal antibodies?

Distinguishing between PKM1 and PKM2 isoforms requires careful antibody selection and experimental design due to their high sequence homology (differing only in ~45 amino acids from mutually exclusive exons). Researchers should employ monoclonal antibodies specifically raised against the unique exon regions (exon 9 for PKM1, exon 10 for PKM2). Validation must include competitive binding assays with recombinant PKM1 and PKM2 proteins to confirm isoform specificity. Western blot analysis should demonstrate distinct molecular weight bands corresponding to each isoform (both approximately 58 kDa but often showing slight mobility differences).

For more complex samples, researchers can implement a sequential immunoprecipitation strategy where one isoform is depleted using a specific antibody, followed by analysis of the remaining protein. Immunohistochemistry requires careful optimization of antigen retrieval conditions that preserve the conformational differences between isoforms. Mass spectrometry analysis of immunoprecipitated samples can provide definitive confirmation by identifying isoform-specific peptides. Researchers should also consider employing orthogonal methods, such as RNA analysis (RT-PCR or RNA-seq targeting the alternative exons) to corroborate protein-level findings. Finally, functional assays that exploit the different kinetic properties of PKM1 (constitutively active) versus PKM2 (allosterically regulated) can provide additional confirmation of antibody specificity in complex biological systems .

How should researchers address potential cross-reactivity issues with PKM monoclonal antibodies?

Addressing cross-reactivity issues with PKM monoclonal antibodies requires comprehensive screening and validation protocols. Researchers should first conduct in silico analysis to identify proteins with sequence or structural homology to PKM, particularly other pyruvate kinase isoforms (PKL, PKR) and related metabolic enzymes. Cross-reactivity testing must include Western blot analysis against tissue panels expressing variable levels of PKM and potentially cross-reactive proteins. Competitive binding assays using recombinant proteins can quantitatively determine relative affinities for intended versus unintended targets.

Epitope mapping techniques are essential to identify the precise binding region of the antibody, allowing researchers to predict potential cross-reactive proteins based on shared epitope sequences. When cross-reactivity is detected, researchers should implement dual-labeling approaches to distinguish between specific and non-specific signals, using a second validated PKM antibody targeting a different epitope. Knockout/knockdown validation experiments provide the most definitive assessment of specificity, as signals that persist in PKM-depleted samples indicate cross-reactivity. For critical applications, researchers should consider using multiple PKM monoclonal antibodies targeting different epitopes and confirming consistent results across antibodies. Finally, pre-absorption controls, where the antibody is pre-incubated with excess recombinant PKM protein before application, can help distinguish specific from non-specific binding patterns in complex samples .

How do the pharmacokinetic properties of PKM monoclonal antibodies differ across experimental models?

The pharmacokinetic properties of PKM monoclonal antibodies exhibit significant variations across experimental models due to species-specific differences in distribution, clearance mechanisms, and target expression. In rodent models, PKM antibodies typically demonstrate faster clearance rates compared to non-human primates or humans, with half-lives often 3-5 times shorter. This difference stems from species-specific variations in neonatal Fc receptor (FcRn) binding, which is critical for antibody recycling and extended circulation.

The distribution of PKM antibodies into tissues is governed by vascular reflection coefficients, which vary by tissue type and exhibit some cross-species consistency. Tissues with tight endothelium (σ₁ averaging 0.908 across species) show limited antibody extravasation, while tissues with leaky endothelium (σ₂ averaging 0.579) allow greater antibody penetration. Target-mediated drug disposition (TMDD) significantly influences PKM antibody pharmacokinetics, particularly at lower doses where target binding contributes substantially to clearance. This effect is more pronounced in models with higher PKM expression levels.

Importantly, allometric scaling of clearance follows the relationship CL = a·BW^b, where the exponent b for PKM monoclonal antibodies typically ranges from 0.695 to 1.27 (averaging 0.91). This scaling relationship enables researchers to translate dosing regimens across species, though additional adjustments may be necessary to account for differences in binding affinity and target expression. When transitioning between experimental models, researchers should conduct species-specific binding studies and employ physiologically-based pharmacokinetic modeling to accurately predict antibody behavior in the new system .

What methodological approaches can accurately assess tissue distribution of PKM monoclonal antibodies?

Accurate assessment of PKM monoclonal antibody tissue distribution requires a multi-modal approach that combines both direct and indirect measurement techniques. The gold standard for direct measurement involves radiolabeling the antibody (typically with 125I or 111In) followed by quantitative whole-body autoradiography (QWBA) or gamma counting of isolated tissues. Researchers must ensure that labeling does not alter binding properties by comparing the affinity of labeled versus unlabeled antibody. Near-infrared fluorescence (NIRF) imaging of fluorophore-conjugated antibodies offers non-radioactive visualization of distribution, though quantification is less precise than radiolabeling approaches.

For high-resolution analysis, immunohistochemistry using anti-idiotypic antibodies (that recognize the PKM antibody itself) allows visualization of antibody localization at the cellular level, revealing important information about tissue penetration and cellular binding patterns. Mass spectrometry imaging (MSI) combined with liquid chromatography-mass spectrometry (LC-MS) enables precise quantification of antibody concentration in tissue sections with spatial context. For longitudinal studies, researchers can implement microdialysis techniques in accessible tissues to monitor antibody concentrations over time.

Mathematical modeling approaches, particularly minimal physiologically-based pharmacokinetic (mPBPK) models, can integrate sparse tissue data with plasma concentration curves to provide comprehensive distribution predictions. These models account for vascular reflection coefficients (σ₁ and σ₂) that govern antibody extravasation into different tissue types. When designing distribution studies, researchers should collect samples at multiple timepoints to capture both distribution and elimination phases, typically extending observations to at least 3-5 half-lives of the antibody. This comprehensive approach provides a holistic understanding of PKM antibody biodistribution, which is essential for correlating tissue exposure with biological effects .

How can researchers account for non-linear clearance mechanisms when studying PKM monoclonal antibodies?

Non-linear clearance presents a significant challenge in PKM monoclonal antibody research, primarily arising from target-mediated drug disposition (TMDD) where binding to PKM contributes to elimination. To accurately account for this phenomenon, researchers should implement a structured analytical approach beginning with dose-ranging studies covering at least three dosing levels to characterize the transition from non-linear to linear kinetics as PKM becomes saturated. Sampling schedules should be designed to capture both distribution and elimination phases, with more frequent sampling during the early distribution phase and extended sampling during terminal elimination.

Mathematical modeling using two-compartment models with parallel linear and non-linear elimination pathways can effectively describe the observed PK profiles. More mechanistic approaches employ quasi-steady-state or full TMDD models that incorporate parameters for PKM binding, internalization, and degradation. To distinguish between specific and non-specific clearance mechanisms, researchers should conduct experiments with a non-binding control antibody of the same isotype to establish the baseline linear clearance component.

Receptor occupancy assays using ex vivo samples can directly measure the relationship between antibody concentration and target engagement, providing crucial data for model parameterization. For translational research, researchers should determine species-specific binding affinities and target expression levels to accurately scale between experimental models. When designing multi-dose studies, time-dependent changes in clearance (due to target downregulation or anti-drug antibody formation) must be monitored and incorporated into PK analyses. This comprehensive approach enables researchers to develop robust predictive models that account for the complex non-linear clearance of PKM monoclonal antibodies across different experimental conditions .

How should researchers interpret contradictory results from different PKM monoclonal antibody clones?

When faced with contradictory results from different PKM monoclonal antibody clones, researchers must implement a systematic troubleshooting approach to identify the source of discrepancies. First, epitope mapping should be conducted to determine if the antibodies recognize different regions of the PKM protein, which may explain divergent results if conformational changes or protein interactions differentially affect epitope accessibility. Researchers should examine the validation history of each antibody clone, particularly regarding specificity for PKM1 versus PKM2 isoforms, as many reported discrepancies stem from unrecognized isoform preferences.

Experimental conditions must be scrutinized, as buffer components, fixation methods, or denaturation procedures can disproportionately affect certain antibody clones. Side-by-side testing under multiple conditions can reveal condition-dependent concordance patterns. For each antibody, researchers should implement orthogonal validation using techniques like mass spectrometry to confirm target identity, or functional assays to correlate antibody binding with PKM enzymatic activity.

When discrepancies persist, researchers should consider biological explanations such as post-translational modifications or protein-protein interactions that might mask or create epitopes recognized by different antibodies. In such cases, phospho-specific or conformation-specific antibodies may be required to fully characterize the system. Publication bias toward positive results means that negative findings about antibody limitations are often unreported; therefore, researchers should conduct their own validation even for commercially available antibodies with published precedents. The most robust approach is to report results from multiple antibody clones alongside appropriate controls, acknowledging discrepancies transparently while using orthogonal techniques to establish which findings most accurately reflect true PKM biology .

What strategies can overcome the challenges of studying PKM in complex tissue microenvironments?

Studying PKM in complex tissue microenvironments presents unique challenges that require specialized methodological approaches. Researchers should implement multiparametric immunofluorescence techniques that simultaneously visualize PKM, cell type-specific markers, and relevant microenvironmental factors. This approach enables spatial correlation of PKM expression with specific cell populations and tissue structures. Laser capture microdissection combined with downstream protein analysis offers precise isolation of specific cellular populations from heterogeneous tissues for detailed PKM characterization.

For dynamic assessment, ex vivo tissue slice cultures maintain the native microenvironment while allowing controlled experimental manipulation and longitudinal imaging of PKM expression and activity. When spatial resolution is critical, techniques like imaging mass cytometry or multiplexed ion beam imaging can visualize dozens of proteins simultaneously at subcellular resolution, placing PKM expression in its full tissue context. Single-cell proteomics approaches can reveal cell-specific PKM expression patterns that would be masked in bulk tissue analysis.

To account for matrix effects on antibody penetration, researchers should optimize tissue clearing techniques compatible with PKM immunostaining. Computational deconvolution algorithms can help separate cell type-specific signals when analyzing bulk tissue data. When studying PKM enzyme activity within the tissue microenvironment, metabolic imaging techniques such as hyperpolarized 13C-MRI can provide spatial information about pyruvate metabolism in intact tissues. For the most comprehensive analysis, researchers should integrate multimodal data using spatial statistics and machine learning approaches to identify microenvironmental factors that correlate with PKM expression or activity patterns. This integrated approach overcomes the limitations of any single technique and provides a more complete understanding of PKM biology within complex tissues .

How can researchers effectively use PKM monoclonal antibodies in multiplex immunoassay systems?

Effective implementation of PKM monoclonal antibodies in multiplex immunoassay systems requires careful optimization to maintain specificity while enabling simultaneous detection of multiple targets. Researchers should begin by testing for potential cross-reactivity between the PKM antibody and other detection antibodies in the multiplex panel through preliminary single-plex assays followed by progressive addition of components. Antibody pairs (capture and detection) must be tested in various combinations to identify optimal pairings that minimize interference while maximizing signal-to-noise ratios.

Conjugation chemistry for reporter molecules (fluorophores, enzymes, or beads) should be optimized specifically for the PKM antibody, as standard protocols may not preserve activity. Researchers should determine the dynamic range of the PKM antibody in the multiplex context, which often differs from single-plex applications due to buffer compromises and intermolecular interactions. Calibration curves must be generated both in isolation and within the full multiplex system to quantify and compensate for any matrix effects.

To minimize non-specific binding in complex samples, specialized blocking reagents (often containing heterophilic blocking agents) should be employed. Signal spillover between detection channels requires careful compensation, particularly for spectrally similar fluorophores. For bead-based multiplexing, validation should include tests for potential antibody exchange between different bead populations during prolonged incubations. When extending to clinical samples, researchers must validate with a diverse sample set that represents the expected range of PKM concentrations and potential interfering factors. Finally, statistical approaches like principal component analysis can help identify and correct for systematic bias in multiplex data. This comprehensive optimization strategy ensures reliable PKM detection within multiplex systems, enabling researchers to efficiently analyze PKM in the context of broader signaling or metabolic networks .

What experimental approaches can effectively combine PKM monoclonal antibodies with metabolic profiling techniques?

Integrating PKM monoclonal antibodies with metabolic profiling requires thoughtful experimental design that links protein detection directly to metabolic function. Researchers should implement immunocapture-coupled metabolomics, where PKM is immunoprecipitated from cell lysates along with its interacting metabolites for subsequent mass spectrometry analysis. This approach reveals metabolites directly associated with PKM complexes. For spatial correlation, sequential staining protocols can be developed where tissue sections are first subjected to matrix-assisted laser desorption/ionization (MALDI) imaging for metabolite mapping, followed by PKM immunohistochemistry on the same section.

Stable isotope tracing combined with immunofluorescence enables researchers to track carbon flux through glycolysis while simultaneously visualizing PKM expression at the single-cell level. Researchers can develop activity-based PKM probes by conjugating metabolic sensors to anti-PKM antibodies, allowing simultaneous detection of PKM localization and local enzymatic activity. In complex tissue systems, laser capture microdissection guided by PKM immunostaining can isolate specific cellular populations for targeted metabolomic analysis.

For high-throughput applications, researchers should consider microfluidic platforms that combine on-chip immunoassays for PKM with real-time measurements of extracellular acidification rates or oxygen consumption. When studying metabolic regulation, proximity ligation assays can be employed to detect interactions between PKM and its regulatory partners while simultaneously measuring metabolic outcomes. Computational integration of these multimodal datasets requires specialized bioinformatic pipelines that align protein expression data with metabolic flux measurements, accounting for differences in temporal dynamics between protein expression and metabolic adaptation. This integrated approach provides unprecedented insights into the relationship between PKM expression, localization, post-translational modification states, and metabolic phenotypes in both normal and disease states .

How can researchers utilize PKM monoclonal antibodies in live-cell imaging applications?

Utilizing PKM monoclonal antibodies for live-cell imaging requires specialized approaches that preserve antibody specificity while maintaining cellular viability and function. Researchers should prioritize antibody fragments (Fab or scFv) over full IgG molecules, as their smaller size improves intracellular delivery efficiency and reduces steric hindrance. For membrane-impermeable antibodies, several delivery strategies can be employed: microinjection for precise single-cell delivery, cell-penetrating peptide conjugation for broader uptake, or electroporation for transient membrane permeabilization. The optimization of delivery protocols must balance transfection efficiency against cellular stress, which can alter PKM regulation and confound experimental interpretations.

Fluorophore selection is critical, with far-red and near-infrared dyes preferred for their minimal phototoxicity and reduced cellular autofluorescence interference. To minimize potential functional interference, researchers should target non-functional epitopes of PKM that do not affect enzymatic activity or protein-protein interactions. Validation must include side-by-side comparisons with fixed-cell staining patterns and confirmation that antibody binding does not alter PKM enzymatic activity or regulatory dynamics.

For extended imaging, researchers should implement photobleaching-resistant fluorophore conjugates and minimize acquisition frequency and intensity to reduce phototoxicity. To distinguish between different PKM isoforms in live cells, researchers can develop ratiometric imaging approaches using differentially labeled isoform-specific antibodies. When studying PKM translocation or complex formation, fluorescence resonance energy transfer (FRET) sensors utilizing labeled antibodies against PKM and its interaction partners provide real-time readouts of protein dynamics. Integration with genetically encoded metabolic sensors allows simultaneous visualization of PKM localization and metabolic activity. These advanced live-cell imaging approaches enable researchers to directly correlate PKM dynamics with cellular functions in real-time, providing insights into the spatial and temporal regulation of glycolysis that are not possible with fixed-cell techniques .

What are the optimal approaches for using PKM monoclonal antibodies in flow cytometry experiments?

Optimizing PKM monoclonal antibodies for flow cytometry requires careful consideration of fixation, permeabilization, and staining protocols to accurately detect intracellular PKM while maintaining compatibility with multiparameter panels. Researchers should begin by comparing multiple fixation methods (paraformaldehyde, methanol, or commercial fixatives) to identify conditions that best preserve PKM epitopes without compromising cellular integrity. Permeabilization protocols must be systematically optimized, with saponin typically preferred for cytoplasmic proteins like PKM, while harsher detergents like Triton X-100 may be necessary if PKM is associated with membrane fractions or nuclear compartments.

Antibody titration is essential to determine the optimal concentration that maximizes signal-to-noise ratio, with titration curves performed directly in flow cytometry rather than extrapolating from other applications. For multiparameter panels, spillover between fluorochromes must be meticulously addressed through proper compensation controls and selection of spectrally distinct fluorophores for PKM detection. When studying PKM isoforms, researchers should implement differential staining approaches using antibodies with validated isoform specificity, potentially employing a ratiometric analysis to quantify relative isoform expression.

To correlate PKM expression with functional parameters, researchers can develop protocols that combine PKM staining with real-time metabolic probes such as 2-NBDG for glucose uptake. Cell cycle analysis can be integrated by incorporating DNA stains like DAPI or propidium iodide, revealing cell cycle-dependent regulation of PKM expression. For rare cell populations, pre-enrichment strategies should be considered to improve statistical power. When studying clinical samples, standardization is critical; researchers should include calibration beads and develop standard operating procedures that ensure consistent staining intensity across batches. Finally, advanced analysis techniques such as t-SNE or UMAP can reveal subtle PKM expression patterns across heterogeneous cell populations that might be missed in conventional biaxial plots. This comprehensive approach enables researchers to precisely quantify PKM expression at the single-cell level while simultaneously evaluating multiple functional parameters .

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.