The HK1/2/3 antibody is a mouse IgG1 antibody generated via hybridoma technology. Key characteristics include:
The antibody is validated for ELISA and Western blotting , enabling researchers to:
Quantify hexokinase expression in cell lysates or tissues.
Localize hexokinase isoforms in subcellular compartments.
Study glucose metabolism in diseases such as cancer, diabetes, and leukemia.
While not explicitly tested for flow cytometry, other HK1-specific antibodies are available for this purpose .
HK3 in Acute Myeloid Leukemia (AML): HK3 is upregulated during ATRA-induced neutrophil differentiation in AML cells. Knocking out HK3 increases reactive oxygen species (ROS) and DNA damage but does not affect glycolysis, suggesting a non-canonical role in cell survival .
HK1/2 in Glycolysis: HK2 is critical for glycolysis in AML cells, whereas HK3 is dispensable for this pathway. HK1 and HK2 interact with mitochondrial proteins to regulate apoptosis .
Autoantibodies in Diabetic Macular Edema (DME): Anti-HK1 autoantibodies have been proposed as biomarkers for DME, though this remains under investigation .
Pancreatic Islet Studies: HK1 expression is analyzed in pancreatic beta-cells to understand glucose sensing and insulin secretion regulation .
Hexokinases (HK1, HK2, HK3) phosphorylate glucose to glucose-6-phosphate, committing it to glycolysis. Key differences include:
Specificity: Cross-reactivity with HK1, HK2, and HK3 necessitates isoform-specific validation (e.g., siRNA knockdown or CRISPR editing) .
Clinical Utility: While HK1 autoantibodies are explored in DME , diagnostic applications remain experimental.
Supplier Variability: Other HK1 antibodies may differ in epitope recognition or conjugation, requiring optimization per assay .
Hexokinase-3, EC 2.7.1.1, Hexokinase type III, HK III, HXK3, HK3.
HK1/2/3 antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
PAT4C12AT.
Anti-human HK1/2/3mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human HK1/2/3amino acids 1-923 purified from E. coli.
Mouse IgG1 heavy chain and κ light chain.
H7.HK1, H7.HK2, and H7.HK3 are human monoclonal antibodies isolated from a 2013 H7N9 convalescent case in Hong Kong. These antibodies were obtained through single B cell RT-PCR after sorting IgG+ B cells that showed reactivity to H7-PE bait. They are part of a set of four H7-reactive monoclonal antibodies (including H7.HK4) recovered from peripheral blood mononuclear cells (PBMCs) of the convalescent donor . H7.HK1 and H7.HK2 are clonally related and target the hemagglutinin (HA) head domain (HA1), while H7.HK3 also targets HA1 but with different binding characteristics .
These antibodies primarily target the hemagglutinin protein of influenza viruses. Specifically, H7.HK1 and H7.HK2 bind to a β14-centered surface on the HA globular head domain (HA1) and disrupt the 220-loop that makes hydrophobic contacts with sialic acid on an adjacent protomer. This mechanism effectively blocks viral entry by interfering with receptor binding . H7.HK3 also binds to the HA1 domain but displays different cross-reactivity patterns compared to H7.HK1 and H7.HK2, suggesting it targets a different epitope within HA1 .
The HK antibodies utilize a different neutralization mechanism compared to traditional anti-influenza antibodies. While many HA1-directed antibodies typically neutralize by directly interfering with the receptor binding site (RBS), H7.HK1 and H7.HK2 bind to a conserved lateral patch on the HA head, disrupting the interaction between the virus and host cell receptors through an alternative mechanism .
In comparative studies, H7.HK2 showed superior or comparable neutralization potency against both 2013 and 2017 H7N9 strains when compared to previous RBS-directed antibodies like L4A-14 and H7.167. H7.HK2 matched the performance of the non-RBS antibody 07-5F01 in neutralization assays .
H7.HK1 and H7.HK2 utilize a unique neutralization mechanism by binding to a conserved lateral patch on the HA head rather than the typical receptor binding site. Cryo-EM structural analyses revealed that these antibodies bind to a β14-centered surface and disrupt the 220-loop that makes hydrophobic contacts with sialic acid on an adjacent protomer . This distinctive binding mechanism:
Targets a relatively conserved region of the HA head
Disrupts the viral attachment process through an allosteric mechanism
Maintains effectiveness against emerging strains that have undergone antigenic drift in the traditional antigenic sites A and B
This mechanism differs from traditional anti-influenza antibodies that directly compete with sialic acid binding at the receptor binding site, which is often subject to greater antigenic variation .
The differential neutralization potency between these antibodies can be attributed to their distinct epitope targeting and binding mechanisms:
A critical advantage of H7.HK1 and H7.HK2 antibodies is their maintained efficacy against antigenically drifted strains. While significant antigenic drift was documented in H7N9 strains from 2016-2017 compared to 2013 isolates, both H7.HK1 and H7.HK2 retained their binding and neutralization capacity against these later strains .
The binding curves of H7.HK1, H7.HK2, and H7.HK3 to both 2016 and 2017 HA1 proteins were fully retained, demonstrating remarkable conservation of their epitopes despite ongoing viral evolution. This stability is explained by structural data showing that the antigenic mutations during this timeframe occurred at the peripheries of their epitopes rather than in the core binding regions .
In contrast, many previously identified neutralizing antibodies against H7N9 lost activity against the 2016-2017 isolates, with only 3 out of 17 neutralizing monoclonal antibodies isolated from earlier cases maintaining activity against the newer strains .
Based on the successful isolation of H7.HK1, H7.HK2, and H7.HK3, the following methodological approach is recommended:
Antigen preparation: Prepare soluble recombinant HA protein (e.g., based on A/Shanghai/2/2013 H7N9) for biotinylation, followed by streptavidin-PE conjugation to create an antigen bait .
B cell isolation and sorting: Stain PBMCs from convalescent donors with the HA-PE bait and sort IgG+ B cells (defined as CD3-CD19+CD20+IgG+) that are HA-PE+ .
Antibody recovery: Perform single B cell RT-PCR on the sorted B cells to recover antibody sequences .
Expression and purification: Clone recovered antibody sequences into expression vectors and produce recombinant antibodies in appropriate expression systems.
Characterization:
ELISA binding assays against various HA antigens to determine specificity and cross-reactivity
Pseudovirus neutralization assays using luciferase reporters
Live virus neutralization assays to confirm activity in more physiologically relevant systems
Structural analysis using cryo-EM to determine binding sites and mechanisms of action
The choice of neutralization assay significantly impacts the measured potency of HK antibodies:
Sensitivity differences: Pseudovirus neutralization assays typically yield IC₅₀ values approximately 10-fold lower (indicating higher apparent potency) than those obtained using live replicating viruses. For instance, H7.HK1 and H7.HK2 showed IC₅₀ values of 20 ng/mL against pseudovirus but higher values against live virus .
Assay characteristics:
Pseudovirus assays:
Use single-round infection with luciferase readout
More sensitive for initial screening of neutralizing mAbs
Less biohazardous, requiring lower containment levels
May not fully recapitulate all aspects of viral entry
Live virus assays:
Application recommendations: Pseudovirus neutralization is recommended for initial screening of neutralizing antibodies due to its higher sensitivity, followed by confirmation with live replicating viruses for more definitive characterization .
While not directly described for the H7.HK antibodies in the provided search results, the general approach for using antibodies in IP-MS studies can be adapted from result :
Sample preparation: Coincubate sera containing antibodies of interest with antigen pools derived from relevant cell lysates (e.g., cells susceptible to viral infection) .
Immunoprecipitation: Enrich antigen-antibody complexes using protein A/G beads .
On-bead digestion: Perform in-solution on-bead trypsin digest of the complexes .
Peptide labeling and analysis: Label peptide fragments and perform quantitative proteomics analysis using platforms like DEEP SEQ .
Data analysis: Apply bioinformatics analysis to identify over-represented proteins in the experimental samples compared to controls .
This approach can be modified to use purified HK1/2/3 antibodies instead of sera, potentially providing insights into additional binding partners or conformational epitopes not identified through traditional structural studies.
The mechanism of action of H7.HK1 and H7.HK2 shares conceptual similarities with other virus-neutralizing antibodies:
Comparison with anti-EBV antibodies: Like the anti-EBV gB neutralizing antibodies 3A3 and 3A5 that effectively neutralize EBV infection of both B and epithelial cells , H7.HK1 and H7.HK2 demonstrate broad neutralization against H7N9 strains. Both sets of antibodies target conserved regions essential for viral entry .
Comparison with other influenza antibodies:
H7.HK1 and H7.HK2 target the HA head domain but bind to a conserved lateral patch rather than the typical receptor binding site (RBS)
Traditional RBS-directed antibodies (e.g., H7.167 and L4A-14) showed weaker neutralization against 2013 H7N9 than H7.HK2, and H7.167's activity was further reduced against the 2017 virus
The non-RBS antibody 07-5F01 showed comparable potency to H7.HK2 against both 2013 and 2017 H7N9 viruses
While the search results don't specifically address synergistic effects for H7.HK antibodies with other therapeutic agents, there is evidence for beneficial combination effects:
H7.HK4 augmentation: The HA2-directed mAb H7.HK4, despite lacking neutralizing activity on its own, moderately augmented mouse protection when used in combination with H7.HK2. This suggests that targeting different domains of the same viral protein can produce enhanced protective effects .
Potential for combination approaches: By analogy with other antibody studies, the unique binding sites of HK antibodies suggest they could be valuable components in antibody cocktails targeting multiple epitopes simultaneously to prevent viral escape .
This concept is similar to approaches used with other viruses, such as the HK2 antisense oligonucleotide (HK2-ASO1) combined with oxidative phosphorylation inhibitor (DPI) and fatty acid oxidation inhibitor (perhexiline) to achieve synthetic lethality in certain cancer cells .
The conserved lateral patch on HA targeted by H7.HK1 and H7.HK2 represents a promising target for developing broader influenza vaccines:
Epitope conservation: Sequence analysis indicates that the lateral patch targeted by H7.HK1 and H7.HK2 is conserved among influenza subtypes, suggesting it could elicit broader protection than traditional vaccine approaches focused on highly variable regions .
Resistance to antigenic drift: These antibodies retain binding and neutralization capacity to later H7N9 isolates from 2016–2017, demonstrating that their epitopes remain conserved despite ongoing viral evolution .
Vaccine design strategies:
Structure-based immunogen design focusing on the β14-centered surface
Prime-boost strategies to direct immune responses toward this conserved epitope
Multivalent approaches combining this conserved epitope with other conserved targets
While cryo-EM has already provided valuable insights into the binding mechanisms of H7.HK1 and H7.HK2 , additional structural biology approaches could further enhance our understanding:
Single-particle cryo-EM with improved resolution: Higher resolution structures could reveal additional molecular details of the antibody-antigen interface.
X-ray crystallography: Complementary to cryo-EM, crystallography might reveal atomic-level details of specific interactions.
Hydrogen-deuterium exchange mass spectrometry (HDX-MS): This technique could provide information about conformational changes in HA induced by antibody binding, potentially revealing allosteric mechanisms.
Molecular dynamics simulations: Computational approaches could model the dynamic interactions between antibodies and their epitopes, providing insights into binding kinetics and energetics.
Structure-guided mutagenesis: Systematic mutation of residues at the antibody-antigen interface could precisely map the contribution of specific interactions to binding and neutralization.
Hexokinases are a family of enzymes that play a crucial role in glucose metabolism by catalyzing the phosphorylation of glucose to produce glucose-6-phosphate (G6P). This is the first step in most glucose metabolism pathways. There are four main isoforms of hexokinase in mammalian cells: Hexokinase-1 (HK1), Hexokinase-2 (HK2), Hexokinase-3 (HK3), and Hexokinase-4 (HK4, also known as glucokinase).
Hexokinase-1 (HK1) is encoded by the HK1 gene located on chromosome 10 in humans. It is a ubiquitous form of hexokinase that localizes to the outer membrane of mitochondria. HK1 is involved in the regulation of glucose metabolism and is essential for maintaining cellular energy homeostasis. Mutations in the HK1 gene have been associated with hemolytic anemia due to hexokinase deficiency . HK1 is also implicated in various diseases, including Alzheimer’s disease, where a reduction in HK1 levels in astrocytes has been observed .
Hexokinase-2 (HK2) is encoded by the HK2 gene on chromosome 2 in humans. It is the predominant form of hexokinase found in skeletal muscle and is also localized to the outer membrane of mitochondria. HK2 is insulin-responsive and plays a significant role in the increased rate of glycolysis observed in rapidly growing cancer cells . HK2 is highly expressed in many malignant tumors and is associated with poor prognosis in glioma patients . It is also involved in various biological processes, including cellular glucose homeostasis, regulation of mitochondrial membrane permeability, and response to hypoxia .
Hexokinase-3 (HK3) is encoded by the HK3 gene on chromosome 5 in humans. Unlike HK1 and HK2, HK3 does not bind to mitochondria. It is involved in glucose phosphorylation and plays a role in cellular protection against oxidative stress by increasing ATP levels, reducing reactive oxygen species (ROS) production, and preserving mitochondrial membrane potential . HK3 is also associated with poor prognosis in glioblastoma multiforme (GBM) and is correlated with immune response and drug resistance .
Mouse anti-human hexokinase antibodies are monoclonal antibodies derived from hybridoma cells produced by the fusion of mouse myeloma cells with spleen cells from immunized mice. These antibodies are highly specific and are used in various research applications, including Western blotting, immunohistochemistry, and flow cytometry.
Hexokinase-1 Antibodies: Mouse anti-human HK1 antibodies are used to detect HK1 in human tissues and cells. They are valuable tools for studying the role of HK1 in glucose metabolism and its involvement in diseases such as Alzheimer’s disease .
Hexokinase-2 Antibodies: Mouse anti-human HK2 antibodies are used to detect HK2 in human tissues and cells. These antibodies are particularly useful in cancer research, as HK2 is highly expressed in many tumors and is associated with poor prognosis .
Hexokinase-3 Antibodies: Mouse anti-human HK3 antibodies are used to detect HK3 in human tissues and cells. They are important for studying the role of HK3 in cellular protection against oxidative stress and its involvement in diseases such as glioblastoma multiforme .