DCK Antibody

Shipped with Ice Packs
In Stock

Description

Structure and Function of DCK

DCK is a homodimer (31 kDa) with dual conformations ("open" and "closed") that regulate substrate binding based on phosphoryl donor interactions (e.g., ATP/UDP) . It catalyzes the first step in nucleoside analog activation, critical for antiviral/anticancer agents like gemcitabine . Overexpression of DCK correlates with drug sensitivity, while deficiency confers resistance .

DCK Antibody Characteristics

DCK antibodies are polyclonal or monoclonal, with varying specificities and applications:

Antibody TypeReactivityApplicationsDilutionCitations
Rabbit Polyclonal (ab186128)Human, MouseIP, WB, IHC-PWB: 0.1 µg/mL
Monoclonal (OTI16G6)Human, MouseWB, IPWB: 1:1000–1:3000
Rabbit Polyclonal (17758-1-AP)HumanWB, ELISA, IF, IP, IHC (p)WB: 1:1000–1:3000
Mouse Monoclonal (16G6)Human, MouseWB, IHC, IFWB: 1:1000–1:3000

Clinical Relevance of DCK

DCK expression levels significantly impact cancer prognosis and treatment outcomes:

  • Hepatocellular carcinoma (HCC): High DCK expression is associated with poor survival and immune infiltration (Tregs, CD8+ T cells) .

  • Gemcitabine resistance: DCK is a rate-limiting enzyme for gemcitabine activation. Low DCK activity correlates with chemoresistance .

  • Therapeutic targeting: Inhibiting DCK enhances antiproliferative effects in leukemia models when combined with deoxythymidine (dT) .

Research Applications of DCK Antibodies

DCK antibodies are used in:

  • Protein detection: Western blotting (WB) and immunoprecipitation (IP) to study DCK expression in cancer cells .

  • Tumor analysis: Immunohistochemistry (IHC) to assess DCK levels in HCC and bladder cancer tissues .

  • Drug development: Screening DCK inhibitors in combination therapies (e.g., dT + dCKi derivatives) .

Key Research Findings

  • Prognostic biomarker: DCK overexpression predicts unfavorable outcomes in early-stage HCC .

  • Immune modulation: DCK correlates with tumor-infiltrating immune cells (TAMs, CD8+ T cells) and exhaustion markers (PD-1, LAG3) .

  • Therapeutic synergy: Co-targeting DCK with ribonucleotide reductase (RNR) inhibitors enhances gemcitabine efficacy .

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship your orders within 1-3 business days after receiving them. Delivery times may vary depending on the order method and location. For specific delivery timeframes, please consult your local distributors.
Synonyms
dCK antibody; DCK protein antibody; DCK_HUMAN antibody; Deoxycytidine kinase antibody; Deoxynucleoside kinase antibody; EC 2.7.1 antibody; EC 2.7.1.74 antibody; MGC117410 antibody; MGC138632 antibody; OTTHUMP00000219118 antibody; OTTHUMP00000219119 antibody
Target Names
DCK
Uniprot No.

Target Background

Function
Deoxycytidine Kinase (DCK) catalyzes the phosphorylation of deoxyribonucleosides such as deoxycytidine, deoxyguanosine, and deoxyadenosine. It exhibits broad substrate specificity and lacks selectivity based on the substrate's chirality. Notably, DCK is an essential enzyme for the phosphorylation of various nucleoside analogs commonly used as antiviral and chemotherapeutic agents.
Gene References Into Functions
  • Studies indicate that DCK knockdown promotes apoptosis while inhibiting proliferation and tumorigenicity in vivo of cervical cancer HeLa cells. PMID: 28820179
  • Activated DCK, through phosphorylation, can suppress radiation-induced cell death, including apoptosis and mitotic catastrophe. It also promotes radiation-induced autophagy via the PI3K/Akt/mTOR pathway. PMID: 27879648
  • Low deoxycytidine kinase expression has been associated with periampullary adenocarcinoma. PMID: 26362587
  • The expression and A9846G AA genotype of DCK are significantly associated with reduced mortality in pancreatic ductal adenocarcinoma. PMID: 26962792
  • Researchers have identified a DCK mutant (G12) that sensitizes cancer cell lines to gemcitabine treatment. A subsequent mutant derived from G12 exhibits even greater sensitization to gemcitabine. PMID: 26485161
  • RNA expression of deoxycytidine kinase (DCK), human equilibrative nucleoside transporter-1 (ENT1), and ribonucleotide reductase M1 (RRM1) were significantly higher, while cytidine deaminase (CDA) expression was significantly lower in ex vivo Ara-C sensitive samples. PMID: 26083014
  • Multivariate Cox regression analysis revealed that age at diagnosis, wild-type genotype of the CDA A79C polymorphism, and wild-type genotype of the dCK C360G polymorphism were the most significant prognostic factors for predicting the risk of death. PMID: 26090398
  • Results strongly suggest that the E197K alteration in DCK causes inactivation of the enzyme, and that loss of the normal E197 allele is the crucial mechanism in acquiring gemcitabine resistance in the RMKN28 tumor cell line. PMID: 26196746
  • DCK rs12648166 and DCK rs4694362 SNPs were associated with hematologic toxicity (OR = 2.63, CI 95% = 1.37-5.04, P = 0.0036 and OR = 2.53, CI 95% = 1.34-4.80, P = 0.0044, respectively). PMID: 25557962
  • These findings indicate that the decitabine metabolic pathway influences its therapeutic effects. Lower hENT1 expression may contribute to primary resistance, and down-regulated DCK expression may be associated with secondary resistance. PMID: 25533931
  • Induction of DCK expression after hypoxia plays a role in the progression of pulmonary fibrosis by contributing to alveolar epithelial cell proliferation. PMID: 25358054
  • DCK expression levels have prognostic value and predict sensitivity to 5-FU in pancreatic cancer. PMID: 24618665
  • DCK can regulate the migration and invasion of fibroblast-like synoviocytes through the AKT pathway in rheumatoid arthritis patients. PMID: 24294360
  • PP2A constitutively dephosphorylates dCK in cells and negatively regulates its activity. PMID: 24462681
  • This study reports genetic variation in deoxycytidine kinase and its subsequent impact on gemcitabine metabolism. PMID: 23230131
  • The DCK and CDA polymorphisms may serve as important markers for the therapy outcomes of AML patients in a Chinese population. PMID: 22884143
  • Hematologic toxicity, such as neutropenia, thrombocytopenia, and anemia, were not associated with three tagged single-nucleotide polymorphisms of deoxycytidine kinase or haplotypes. Genetic variations did not influence the survival of patients. PMID: 23035362
  • High levels of dCK in pancreatic ductal adenocarcinoma predict longer survival times in patients treated with adjuvant gemcitabine. PMID: 22705007
  • These results suggest that the inactivation of DCK is a crucial mechanism in acquiring gemcitabine resistance. PMID: 22490663
  • While dFdU increased the net intracellular radioactivity of [5-(3)H]dFdC at 24 h in control cells, this increase was abolished in the absence of dCK activity. PMID: 21832002
  • Data indicate that the CDA/DCK ratio was 3-fold higher in non-responders compared to responders (P<.05), suggesting that this could be a mechanism of primary resistance. PMID: 21858090
  • dCK expression level in fludarabine-sensitive patients was significantly higher than in fludarabine-resistant patients. PMID: 20137114
  • High deoxycytidine kinase expression is associated with response to pemetrexed-gemcitabine combination therapy in patients with advanced non-small cell lung cancer. PMID: 21336182
  • Data indicate that dCK undergoes a transition between open and closed states during the catalytic cycle. PMID: 21351740
  • Overexpressed dCK and knocked down p8 are associated with enhanced gemcitabine sensitivity in pancreatic cancer. PMID: 21225236
  • The variant C28624T showed a lower risk of lymphopenia (P=0.04) but a higher risk of neutropenia. PMID: 21030078
  • ABCG2 influence on clofarabine cytotoxicity is significantly impacted by dCK activity. PMID: 21245102
  • The dCK-360G allele was found to increase the risk of mucositis after exposure to low-dose cytarabine in childhood ALL therapy. PMID: 20890066
  • Site-directed mutagenesis demonstrated that only Ser-74 phosphorylation is involved in dCK activation by casein kinase 1 delta, reinforcing the crucial role of this residue in regulating dCK activity. PMID: 20637175
  • Phosphorylation of the three other sites located in the N-terminal extremity of the protein does not significantly alter dCK activity, but phosphorylation of Thr-3 may promote dCK stability. PMID: 20544527
  • Methylation was detected in one of the SP1 binding sites of the dCK promoter in most tested cancer cell lines and in patient samples from brain tumors and leukemia. Methylation might therefore regulate the transcription of dCK. PMID: 20544528
  • Residue Asp133 of dCK appears to be primarily responsible for discriminating against the thymine base. PMID: 20614893
  • Gemcitabine sensitivity of two pancreatic cancer cell lines transduced with deoxycytidine kinase was significantly elevated compared to control cells. PMID: 20043109
  • Alternatively spliced dCK forms found in acute myeloid leukemia cells play a significant role in cytarabine resistance. PMID: 11830489
  • This study explores the molecular basis of 2',3'-dideoxycytidine-induced drug resistance in human cells. PMID: 11952160
  • Inorganic tripolyphosphate (PPP(i)) serves as a good donor for human deoxycytidine kinase and deoxyguanosine kinase. PMID: 12535661
  • The activity of the human deoxycytidine kinase promoter is regulated by USF and Sp1. PMID: 14514691
  • DCK can function as a phosphorylase, similar to the nucleoside phosphorylase family of enzymes. PMID: 15561147
  • This study examines the binding of antitumor drugs to deoxycytidine kinase. PMID: 15571255
  • DCK expression varies among individual samples and between different types of malignancies, and may play a role in resistance to ara-C in specific tumor types. PMID: 15571257
  • Deoxycytidine kinase activity is stimulated by 2-chlorodeoxyadenosine and aphidicolin in a cellular context. PMID: 15571258
  • Deoxycytidine kinase activity is regulated by reversible phosphorylation. PMID: 15571259
  • Increased expression of mRNA, specific for thymidine kinase 1, dCK, and thymidine phosphorylase, may be involved in carcinogenic processes in the human thyroid. PMID: 15978330
  • DCK activity can be controlled by phosphorylation in intact cells, and Ser-74 is essential for its activity. PMID: 16361699
  • Crystal structures of a deoxycytidine kinase variant lacking a flexible insert (residues 65-79) reveal significant changes in the donor base binding loop (residues 240-247) between UDP-bound and ADP-bound forms, involving substantial main-chain rearrangement. PMID: 16401075
  • An increase in the activity of dCK, TK1, and TK2 might be involved in an adaptive response of cultured human squamous lung carcinoma cells to radiation by facilitating DNA repair. PMID: 16969512
  • Deoxycytidine kinase plays a role in lymphoma cell sensitivity to cladribine. PMID: 17065053
  • This study analyzes phosphorylation sites on human deoxycytidine kinase. PMID: 17065079
  • This study analyzes the reversible phosphorylation of deoxycytidine kinase in normal human lymphocytes. PMID: 17065080
  • Deoxycytidine kinase activity is enhanced after pulsed low dose rate and single dose gamma irradiation. PMID: 17065085

Show More

Hide All

Database Links

HGNC: 2704

OMIM: 125450

KEGG: hsa:1633

STRING: 9606.ENSP00000286648

UniGene: Hs.709

Protein Families
DCK/DGK family
Subcellular Location
Nucleus.

Q&A

What is Deoxycytidine Kinase (DCK) and why is it important in research?

Deoxycytidine kinase (DCK) is an essential enzyme involved in the phosphorylation of several deoxyribonucleosides: deoxycytidine (dC), deoxyguanosine (dG), and deoxyadenosine (dA). It demonstrates broad substrate specificity without selectivity based on substrate chirality.

DCK is critically important in research for several reasons:

  • It serves as a key enzyme in the nucleoside salvage pathway

  • It phosphorylates numerous nucleoside analogs used as antiviral and chemotherapeutic agents

  • DCK deficiency is associated with resistance to certain chemotherapeutic agents, while increased activity correlates with enhanced cytotoxicity of these compounds

  • It has emerging roles in immune cell development and homeostasis, particularly in T cell populations

What are the common applications of DCK antibodies in laboratory research?

DCK antibodies are versatile tools employed in multiple experimental techniques:

ApplicationDescriptionCommon Dilutions
Western Blotting (WB)Detection of DCK protein in cell/tissue lysates1:1000-1:5000
Immunohistochemistry (IHC-P)Visualization of DCK in paraffin-embedded tissues1:50-1:1000
Immunofluorescence (IF)Cellular localization studies1:50-1:200
Immunoprecipitation (IP)Isolation of DCK protein complexes1:50-1:500
Flow CytometryQuantification of DCK in cell populationsVariable by antibody
ELISAQuantitative detection of DCK≈1 μg/ml

Most commercially available DCK antibodies are validated against human samples, though many cross-react with mouse and rat DCK due to sequence homology .

How should I optimize DCK antibody concentration for Western blotting experiments?

When optimizing DCK antibody concentration for Western blotting, follow this methodological approach:

  • Initial titration: Begin with the manufacturer's recommended dilution (typically 1:1000-1:5000)

  • Sample preparation:

    • Use 30-50 μg of total protein per lane

    • Standard conditions: 12% SDS-PAGE separation is sufficient for DCK detection (≈31 kDa)

  • Validation controls:

    • Include positive controls such as HEK-293T, HeLa, or Jurkat cell lysates, which express detectable levels of endogenous DCK

    • For specificity confirmation, consider DCK knockout/knockdown samples if available

  • Optimization steps:

    • If signal is weak: increase antibody concentration or extend exposure time

    • If background is high: increase blocking time or washing steps, or dilute antibody further

  • Expected results:

    • DCK typically appears as a single band at approximately 29-31 kDa

    • Multiple bands may indicate degradation or post-translational modifications

Most DCK antibodies perform well with standard PVDF or nitrocellulose membranes and conventional ECL detection systems .

What are the critical considerations for successfully immunoprecipitating DCK?

Successful immunoprecipitation of DCK requires attention to several key factors:

  • Antibody selection: Choose antibodies specifically validated for IP applications

  • Lysis buffer composition:

    • Use non-denaturing buffers containing 1% NP-40 or Triton X-100

    • Include protease inhibitors to prevent degradation

    • Consider phosphatase inhibitors if studying DCK phosphorylation

  • Protocol optimization:

    • Recommended antibody amount: 1-5 μg per 500 μg of total protein

    • Pre-clear lysates with protein A/G beads to reduce non-specific binding

    • Optimal incubation: overnight at 4°C with gentle rotation

  • Controls:

    • IgG isotype control to detect non-specific binding

    • Input control (5-10% of lysate used for IP)

    • If available, DCK-deficient samples as negative controls

  • Verification:

    • Confirm successful IP by Western blotting using a separate DCK antibody targeting a different epitope

    • Expected molecular weight: approximately 30-31 kDa

DCK has been reported to exist as a homodimer in some contexts, which may affect immunoprecipitation efficiency depending on epitope accessibility .

How can DCK antibodies be used to investigate correlations between DCK expression and tumor-infiltrating immune cells?

DCK expression shows significant correlations with tumor-infiltrating immune cells (TIICs), making DCK antibodies valuable tools for cancer immunology research. Methodological approach:

  • Tissue preparation and analysis:

    • Perform multiplexed immunohistochemistry or immunofluorescence using DCK antibodies alongside markers for specific immune cell populations

    • Analyze serial sections with DCK staining and immune cell markers

    • For quantitative assessment, use digital image analysis platforms

  • Key correlations to investigate:
    Research has demonstrated significant positive correlations between DCK expression and various immune cell populations in hepatocellular carcinoma, including:

    • CD4+ T cells (correlation coefficient = 0.419, p = 4.61×10⁻¹⁶)

    • CD8+ T cells (correlation coefficient = 0.310, p = 4.57×10⁻⁹)

    • B cells (correlation coefficient = 0.351, p = 1.97×10⁻¹¹)

    • Tumor-associated macrophages (positive correlation)

    • Dendritic cells (correlation coefficient = 0.485, p = 2.00×10⁻²¹)

    • Neutrophils (correlation coefficient = 0.556, p = 2.50×10⁻²⁹)

  • Functional implications:

    • Investigate correlations between DCK expression and regulatory T cell markers (CCR8, STAT5b, TGFB1)

    • Examine relationships with exhaustion-related markers (PD-1, CTLA-4, LAG3, TIM-3, GZMB)

    • These correlations suggest DCK's potential involvement in immunoregulation and tumor immune escape mechanisms

  • Validation approaches:

    • Compare antibody-based findings with transcriptomic data (e.g., using TIMER or GEPIA databases)

    • Consider functional validation through DCK inhibition or knockdown studies

What methodological approaches can resolve contradictory DCK antibody staining results in clinical specimens?

Resolving contradictory DCK antibody staining in clinical specimens requires systematic troubleshooting:

  • Antibody validation hierarchy:

    • Test multiple antibodies targeting different DCK epitopes

    • Prioritize antibodies validated by knockout/knockdown controls

    • Compare monoclonal (higher specificity) vs. polyclonal (potentially higher sensitivity) antibodies

  • Technical optimization:

    • Antigen retrieval methods: Compare heat-induced epitope retrieval (citrate vs. EDTA buffers)

    • Fixation impact: Test freshly fixed vs. archived specimens

    • Signal amplification systems: Standard ABC vs. polymer-based detection

    • Blocking protocols: Extend blocking steps to reduce non-specific binding

  • Controls framework:

    • Positive tissue controls: Include tissues with known high DCK expression (lymphoid tissues)

    • Negative controls: Include both antibody omission and isotype controls

    • Cellular-level controls: Use immunofluorescence to confirm appropriate subcellular localization (primarily nuclear)

  • Cross-validation approaches:

    • Correlate IHC findings with Western blot results from the same specimens

    • Compare protein expression with mRNA levels (RT-PCR or RNA-seq)

    • Digital quantification of staining to establish objective intensity thresholds

  • Clinical interpretation standardization:

    • Establish scoring criteria specific to DCK (e.g., H-score, Allred score)

    • Consider tumor heterogeneity through multiple sampling

    • Document the relationship between staining patterns and clinical outcomes

How do DCK expression patterns correlate with clinical outcomes in cancer research?

DCK expression patterns have significant prognostic implications in cancer research, particularly in hepatocellular carcinoma (HCC):

  • Expression analysis findings:

    • Higher DCK expression is observed in HCC tissues compared to adjacent normal tissues

    • Elevated DCK expression correlates with poorer prognosis in HCC patients

    • This correlation is particularly pronounced in early-stage and early-grade HCC

  • Methodological approach for expression analysis:

    • IHC staining using validated DCK antibodies on tumor microarrays

    • Western blotting of paired tumor/normal tissues

    • Correlation with clinical databases and survival data

  • Mechanistic insights:

    • DCK expression positively correlates with markers of regulatory T cells, suggesting potential involvement in immunosuppression

    • Association with exhaustion-related inhibitory receptors indicates possible roles in immune escape mechanisms

  • Potential applications:

    • DCK expression could serve as a prognostic biomarker in HCC

    • May help identify patients who might benefit from specific therapeutic approaches

    • Could represent a novel therapeutic target in certain cancer types

What methodological considerations are important when using DCK antibodies to study the efficacy of DCK inhibitors?

When using DCK antibodies to study DCK inhibitor efficacy, researchers should consider:

  • Antibody selection for inhibitor studies:

    • Choose antibodies whose epitopes do not overlap with inhibitor binding sites

    • Consider antibodies that can detect both free and inhibitor-bound DCK

    • Validate antibody performance in the presence of the inhibitor

  • Experimental design for inhibitor efficacy assessment:

    • Establish dose-response curves using multiple inhibitor concentrations

    • Include washout experiments to assess inhibitor reversibility

    • Compare total DCK protein levels vs. enzymatic activity

    • For studies with (R)-DI-87 (a clinical-stage DCK inhibitor), consider its protective effects against Staphylococcus aureus infection

  • Readout methodologies:

    • Direct enzyme activity assays to correlate with antibody-detected protein levels

    • Phosphorylation status of DCK substrates

    • Cell viability/functional assays (e.g., protection from death-effector deoxyribonucleosides in infection models)

  • Controls and validation:

    • Include genetic knockdown/knockout as positive controls for inhibition

    • Use structurally related but inactive compounds as negative controls

    • Consider time-course experiments to assess temporal dynamics of inhibition

  • Therapeutic context considerations:

    • For cancer applications: monitor resistance development

    • For infectious disease applications (e.g., S. aureus): assess protection of host immune cells

    • Consider combinatorial approaches with standard therapies

What are the most common causes of non-specific binding when using DCK antibodies and how can they be addressed?

Non-specific binding with DCK antibodies can originate from several sources:

  • Antibody-related factors:

    • Cross-reactivity with related kinases

    • Non-specific IgG binding

    • Lot-to-lot variability

    Solutions:

    • Use antibodies validated against DCK knockout/knockdown samples

    • Compare multiple antibodies targeting different DCK epitopes

    • Test pre-adsorption with immunizing peptide when available

  • Sample preparation issues:

    • Incomplete blocking

    • Inadequate washing

    • Excessive fixation (for IHC/ICC)

    Solutions:

    • Extend blocking time (1-2 hours)

    • Use alternative blocking agents (5% BSA, 5% milk, or commercial blockers)

    • Increase washing duration and number of washes

    • Optimize fixation protocols

  • Detection system problems:

    • Excessive signal amplification

    • High background from secondary antibody

    Solutions:

    • Titrate detection reagents

    • Use directly conjugated primary antibodies

    • Include secondary-only controls

    • Consider alternative detection systems

  • Protocol optimization:

    • For Western blotting: reduce primary antibody concentration (1:5000-1:20000)

    • For IHC: implement antigen retrieval optimization

    • For IF/ICC: add 0.1-0.3% Triton X-100 for membrane permeabilization

    • For all applications: consider using protein-free blocking buffers

How should researchers address epitope masking or unavailability when DCK antibody staining yields unexpectedly negative results?

When DCK antibody staining yields unexpectedly negative results despite presumed DCK expression, systematic troubleshooting for epitope masking is essential:

  • Antigen retrieval optimization:

    • Compare heat-induced epitope retrieval methods:

      • Citrate buffer (pH 6.0)

      • EDTA buffer (pH 9.0)

      • Tris-EDTA buffer (pH 8.0)

    • Test enzymatic retrieval (proteinase K, trypsin)

    • Vary retrieval duration (10-30 minutes)

  • Fixation considerations:

    • Overfixation may cause extensive protein crosslinking

    • Test shorter fixation times for prospective samples

    • Compare antibody performance on frozen vs. FFPE tissues

    • For cell lines, compare different fixatives (paraformaldehyde, methanol, acetone)

  • Protein-protein interaction interference:

    • DCK forms homodimers that might mask certain epitopes

    • Use denaturing conditions for Western blotting

    • For IP applications, consider tandem IP strategies

    • Test antibodies targeting different regions of DCK (N-terminal, center, C-terminal)

  • Post-translational modifications:

    • Phosphorylation may affect epitope recognition

    • Include phosphatase treatment in parallel samples

    • Consider antibodies specifically designed to detect modified forms

  • Technical approaches:

    • Signal amplification systems (TSA, polymer-based detection)

    • Extend antibody incubation times (overnight at 4°C)

    • Increase antibody concentration incrementally

    • Use fresh antibody aliquots to rule out degradation

  • Validation strategies:

    • Confirm DCK expression at the mRNA level

    • Use positive control tissues with known high DCK expression

    • Consider alternative detection methods (mass spectrometry)

How can DCK antibodies be utilized in studying the relationship between DCK and immune regulation in infectious diseases?

DCK antibodies are valuable tools for investigating the emerging roles of DCK in immune regulation during infectious diseases:

  • Infection models and DCK function:

    • Recent research has identified a critical role for DCK in Staphylococcus aureus infections

    • DCK mediates the phosphorylation of bacterial death-effector deoxyribonucleosides that can trigger host immune cell death

    • The pharmacological inhibition of DCK (using (R)-DI-87) protects host immune cells and mitigates S. aureus abscess formation

  • Methodological applications of DCK antibodies:

    • Infection progression monitoring:

      • Track DCK expression levels in different immune cell populations during infection

      • Correlate with markers of cell death and immune function

    • Mechanistic studies:

      • Co-localization studies with bacterial virulence factors

      • Phosphorylation status assessment of DCK during infection

      • Identification of DCK-interacting proteins in infection contexts

    • Therapeutic evaluation:

      • Monitor DCK inhibition efficacy in infection models

      • Assess effects on immune cell survival and function

      • Evaluate combinatorial approaches with antibiotics

  • Advanced applications:

    • Single-cell analysis of DCK expression in heterogeneous immune populations

    • In vivo imaging using fluorescently-labeled DCK antibodies

    • CyTOF/mass cytometry for comprehensive immune phenotyping

    • Spatial transcriptomics combined with DCK protein detection

  • Translational relevance:

    • DCK inhibitors may represent a novel host-directed therapeutic approach for bacterial infections

    • Particularly relevant for multidrug-resistant pathogens where bacterial resistance development is challenging

    • Potential applications beyond S. aureus to other pathogens that manipulate the purine salvage pathway

What emerging multiplexed immunoassay approaches can enhance DCK antibody applications in complex tissue microenvironments?

Advanced multiplexed immunoassay techniques are revolutionizing DCK antibody applications in complex tissue microenvironments:

  • Multiplexed immunofluorescence techniques:

    • Sequential immunostaining approaches:

      • Cyclic immunofluorescence (CyCIF) allowing 30+ markers on the same tissue section

      • Tyramide signal amplification (TSA) multiplexing for enhanced sensitivity

      • Integration with DCK antibodies validated for immunofluorescence applications

    • Spectral unmixing methods:

      • Multispectral imaging platforms enabling separation of overlapping fluorophores

      • Simultaneous visualization of DCK with multiple immune cell markers

      • Quantitative assessment of co-localization patterns

  • Mass cytometry-based tissue imaging:

    • Imaging Mass Cytometry (IMC):

      • Metal-conjugated DCK antibodies for high-dimensional analysis

      • Simultaneous detection of 40+ markers with subcellular resolution

      • Ideal for mapping DCK in relation to complex immune infiltrates

    • Multiplexed ion beam imaging (MIBI):

      • Secondary ion mass spectrometry for antibody detection

      • Compatible with FFPE tissues from clinical archives

      • High-resolution spatial mapping of DCK and interaction partners

  • Digital spatial profiling approaches:

    • Combines immunofluorescence with spatially resolved molecular profiling

    • Enables correlation of DCK protein expression with local transcriptome

    • Facilitates integration of proteomic and genomic data in spatial context

  • Computational analysis frameworks:

    • Cell segmentation algorithms for single-cell quantification

    • Spatial statistics to analyze DCK expression patterns

    • Machine learning approaches for biomarker identification

    • Network analysis to infer functional relationships

  • Applications in DCK research:

    • Mapping DCK expression in relation to tumor-immune interfaces

    • Spatial correlation with markers of immunosuppression

    • Therapeutic response monitoring with spatial resolution

    • Identification of novel DCK-expressing cellular niches

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.