DGKZ Antibody

Shipped with Ice Packs
In Stock

Description

What is DGKZ Antibody?

DGKZ antibodies are immunodetection tools targeting the DGKZ protein, which converts diacylglycerol (DAG) to phosphatidic acid (PA) to modulate DAG/PA-dependent signaling pathways . These antibodies enable researchers to:

  • Quantify DGKZ expression in tissues or cell lines

  • Study DGKZ’s role in immune regulation, cancer metastasis, and lipid metabolism

  • Validate DGKZ knockout or overexpression models

Immunological Functions

  • B Cells: DGKζ deficiency enhances B cell activation, proliferation, and antibody responses to antigens by prolonging DAG-mediated ERK signaling .

  • T Cells: DGKζ knockout (KO) CD8+ T cells exhibit increased ERK1/2 activation, proliferation, and antitumor activity compared to DGKα KO .

  • Immune Synapse: DGKζ promotes actin remodeling and antigen uptake in B cells by balancing DAG/PA signaling .

Cancer Research

  • Breast Cancer: DGKZ drives metastasis in triple-negative breast cancer (TNBC) by activating TGFβ/TGFβR2/Smad3 signaling and suppressing caveolin-dependent TGFβR2 degradation .

    • Clinical relevance: High DGKZ correlates with poor prognosis in TNBC patients (HR = 2.21, p = 0.021) .

  • Osteosarcoma: DGKZ overexpression accelerates proliferation and is linked to aggressive tumor behavior .

Experimental Applications

  • Western Blot: Detects endogenous DGKZ in Jurkat (T-cell leukemia) and HuT-78 (cutaneous T lymphoma) lysates .

  • Immunohistochemistry: Strong cytoplasmic/nuclear staining in brain neurons (rat/mouse) and immune cells (human tonsil) .

  • Functional Studies:

    • Knockout Models: CRISPR-Cas9-mediated DGKZ deletion reduces TNBC cell migration and lung metastasis in mice .

    • Overexpression: Enhances DAG-to-PA conversion, altering plasma membrane fluidity and receptor trafficking .

Technical Considerations

  • Antigen Retrieval: Required for IHC (Tris/EDTA buffer, pH 9.0) .

  • Cross-Reactivity: Some antibodies (e.g., ab239080) recognize rodent DGKZ but show lower affinity for human isoforms .

  • Controls: Include isotype-matched IgG and secondary antibody-only assays to rule out nonspecific binding .

Product Specs

Buffer
Liquid in PBS containing 50% glycerol, 0.5% BSA and 0.02% sodium azide.
Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your order. The delivery time may vary depending on the purchasing method or location. Please contact your local distributor for specific delivery time details.
Synonyms
DAG kinase zeta antibody; DAGK5 antibody; DAGK5 PEN antibody; DAGK6 antibody; DGK ZETA antibody; DGK-zeta antibody; Dgkz antibody; DGKZ_HUMAN antibody; Diacylglycerol kinase zeta antibody; Diglyceride kinase zeta antibody; hDGKzeta antibody
Target Names
Uniprot No.

Target Background

Function
Diacylglycerol kinase zeta (DGKζ) is an enzyme that converts diacylglycerol (DAG) into phosphatidic acid (PA). This conversion plays a pivotal role in regulating the levels of these two bioactive lipids, which are crucial second messengers in various cellular processes. DGKζ acts as a central switch between DAG and PA signaling pathways, influencing a wide range of biological functions.

DGKζ is involved in the biosynthesis of complex lipids and exhibits no acyl chain-dependent substrate specificity among diacylglycerol species. Although less efficiently, it can also phosphorylate 1-alkyl-2-acylglycerol in vitro, with a preference for alkylacylglycerols containing an arachidonoyl group.

DGKζ plays a critical role in T-cell activation by negatively regulating T-cell receptor signaling, a process partly mediated by diacylglycerol. By producing phosphatidic acid, DGKζ stimulates PIP5KIA activity, which regulates actin polymerization. Through this mechanism, DGKζ can also positively regulate insulin-induced translocation of SLC2A4 to the cell membrane. Additionally, DGKζ regulates RASGRP1 activity, but does not regulate RASGRP1 activity.
Gene References Into Functions
  1. Studies have shown that DGKζ is downregulated in bone marrow mononuclear cells and is associated with the severity of aplastic anemia (AA). Notably, DGKζ is a downstream target gene of miR34a. The dysregulation of these molecules enhances T-cell activation in AA cells. PMID: 28008152
  2. Research has established a positive correlation between DGKζ expression and glioma grade. PMID: 26452358
  3. Evidence suggests that DGKζ activation downstream of antigen recognition provides a mechanism to ensure the activation of PA-dependent signaling, directly influenced by the strength of TCR-dependent DAG mobilization. PMID: 27999176
  4. Diacylglycerol kinases alpha and zeta are upregulated in cancer and contribute to tumor immune evasion and T cells clonal anergy. (Review) PMID: 27697466
  5. Knockdown of DGKζ leads to an enhancement of the NF-kappaB pathway in response to TNF-alpha. [review] PMID: 26521214
  6. Diacylglycerol kinases alpha and zeta play redundant and specialized roles in controlling T cell functions. PMID: 25921290
  7. This study demonstrates that DGKζ knockdown facilitates the degradation of IkappaB, followed by the nuclear translocation of the NF-kappaB p65 subunit. PMID: 25450975
  8. Elevated DGKζ expression contributes to increased Rho GTPase activation and enhanced motility of metastatic cancer cells. PMID: 24646293
  9. Data indicate that after P2Y6 receptor stimulation, both phospholipase D (PLD) and DGKζ enzymes are responsible for producing phosphatidic acid (PA). PMID: 23723068
  10. DGK-zeta translocates rapidly to the plasma membrane at early stages of immunological synapse (IS) formation independent of enzyme activity. The study highlights a DGKζ-specific function for local diacylglycerol metabolism at the IS and provides new insights into its regulation. PMID: 21937721
  11. Antigen-specific CD8-positive T cells from DGKζ-deficient transgenic mice show enhanced expansion and increased cytokine production after lymphocytic choriomeningitis virus infection. However, DGK-deficient memory CD8+ T cells exhibit impaired expansion. PMID: 22271650
  12. Nucleosome assembly protein (NAP) 1-like 1 (NAP1L1) and NAP1-like 4 (NAP1L4) have been identified as novel DGKζ binding partners. PMID: 21996351
  13. Data demonstrate that 2-arachidonoyl glycerol is a very poor substrate for both the epsilon and zeta isoforms of diacylglycerol kinases. PMID: 21194521
  14. This research investigates the structural domain requirements for translocation and activity. PMID: 12015310
  15. PKC alpha phosphorylates diacylglycerol kinase zeta in cells, and this phosphorylation inhibits its kinase activity to remove cellular diacylglycerol, thereby affecting cell growth. PMID: 12890670
  16. DGKζ plays a role in controlling the induction of luteinizing hormone beta transcription by ERK1/2. PMID: 14707140
  17. DGKζ-derived phosphatidic acid acts as a mediator of mTOR signaling. PMID: 15632115
  18. DGKζ may function in vivo as a downstream effector of pRB to regulate nuclear levels of diacylglycerol and phosphatidic acid. PMID: 16286473
  19. 2,3-dioleoylglycerol binds to a site on the alpha and zeta isoforms of diacylglycerol kinase that is exposed as a consequence of substrate binding to the active site. PMID: 18004883
  20. PKD activation is induced by DGKζ, suggesting that DGK is an upstream regulator of oxidative stress-induced activation of the PKD signaling pathway in intestinal epithelial cells. PMID: 18694729
  21. In DGKζ-deficient fibroblasts, PAK1 phosphorylation and Rac1-RhoGDI dissociation were attenuated, leading to reduced Rac1 activation after platelet-derived growth factor stimulation. PMID: 19211846
  22. None of the SNPs of diacylglycerol kinase zeta tested showed association with bipolar disorder in the Sardinian sample. PMID: 19308020
  23. DGKζ negatively regulates T cell receptor-induced activation of the Ras-Erk1/2-AP1 pathway. PMID: 12070163

Show More

Hide All

Database Links

HGNC: 2857

OMIM: 601441

KEGG: hsa:8525

STRING: 9606.ENSP00000412178

UniGene: Hs.502461

Protein Families
Eukaryotic diacylglycerol kinase family
Subcellular Location
Nucleus. Cytoplasm, cytosol. Cell membrane. Cell projection, lamellipodium.
Tissue Specificity
Highest levels in brain, and substantial levels in skeletal muscle, heart, and pancreas.; [Isoform 2]: Predominantly expressed in muscle.

Q&A

What is DGKZ and why is it important in immunological research?

Diacylglycerol kinase zeta (DGKZ) is a critical enzyme that catalyzes the conversion of diacylglycerol (DAG) to phosphatidic acid (PA), thereby regulating the levels of these two important bioactive lipids in cell signaling pathways . DGKZ serves as a central molecular switch between DAG-mediated and PA-mediated signaling mechanisms, which have distinct cellular targets and often opposing effects in numerous biological processes .

In immunological research, DGKZ is particularly significant because it negatively regulates T cell receptor (TCR) signaling by terminating DAG-mediated activation . The importance of DGKZ extends to:

  • Modulation of T cell activation thresholds

  • Regulation of anti-viral and anti-tumor immune responses

  • Control of B cell development and antibody production

  • Influence on T helper cell differentiation and function

DGKZ represents a 124.1 kilodalton protein with 1117 amino acids and is predominantly expressed in lymphoid tissues, where it functions as the dominant DGK isoform in T cells compared to DGKα .

What are the key applications for DGKZ antibodies in research?

DGKZ antibodies serve multiple essential applications in fundamental and translational research:

ApplicationCommon UsageTechnical Considerations
Western Blot (WB)Protein expression quantification, molecular weight verification (~124kDa)Working dilutions typically 1:500-1:2000
Immunohistochemistry (IHC)Tissue localization and expression patternsBoth paraffin-embedded (IHC-p) and frozen section protocols available
Immunocytochemistry (ICC)Subcellular localization (cytoplasmic, nuclear, membrane-associated)Often requires optimization of fixation conditions
Flow Cytometry (FCM)Quantitative analysis in immune cell populationsMay require permeabilization for intracellular detection
Immunoprecipitation (IP)Protein-protein interaction studiesSelect antibodies validated for IP applications
ELISAQuantitative measurement in biological samplesValidated for human and rodent samples

When selecting antibodies for these applications, researchers should consider the specific epitope recognition, species reactivity, and validation data available for each antibody .

How do I select the appropriate DGKZ antibody for my experimental system?

Selection of an appropriate DGKZ antibody should be guided by several critical factors:

  • Experimental application: Different applications require antibodies with specific characteristics. For example, antibodies for Western blotting may not perform well in immunohistochemistry applications.

  • Species reactivity: Verify cross-reactivity with your experimental model organism. Available DGKZ antibodies show reactivity to human (Hu), mouse (Ms), rat (Rt), and some to monkey (Mk) DGKZ .

  • Epitope recognition: Consider which region of DGKZ the antibody recognizes:

    • N-terminal (aa 1-200) antibodies may detect all isoforms

    • C-terminal (aa 1050 to C-terminus) antibodies might distinguish specific isoforms

  • Validation data: Review published validation studies and supplier validation data:

    • Western blot showing expected ~124 kDa band

    • Appropriate subcellular localization in immunostaining

    • Citations in peer-reviewed literature (e.g., ab105195 has been cited in 8 publications)

  • Polyclonal vs. monoclonal: Polyclonal antibodies offer broader epitope recognition but potentially more background, while monoclonal antibodies provide greater specificity but might be sensitive to epitope modifications .

Select antibodies with comprehensive validation data in applications and cell/tissue types similar to your experimental system for optimal results.

How should DGKZ antibodies be stored and handled to maintain optimal performance?

Proper storage and handling of DGKZ antibodies is critical for maintaining their performance and extending their usable lifespan:

Storage recommendations:

  • Store antibodies at -20°C for long-term storage (one year or more)

  • For frequent use and short-term storage (up to one month), store at 4°C

  • Many commercial DGKZ antibodies are supplied in buffer containing 50% glycerol, PBS with 0.02% sodium azide, pH 7.2 to enhance stability

Handling guidelines:

  • Avoid repeated freeze-thaw cycles which can denature antibodies and reduce activity

  • Aliquot antibodies upon first thaw if multiple uses are anticipated

  • Centrifuge briefly before opening vials to collect liquid at the bottom

  • Use sterile technique when handling antibody solutions

  • Reconstitute lyophilized antibodies with deionized water or specified buffer to the recommended volume

Working solution preparation:

  • For Western blot applications, typical dilutions range from 1:500 to 1:2000

  • Prepare working solutions fresh on the day of experiment when possible

  • Always include appropriate controls (positive control lysates, negative controls)

Following these storage and handling practices will help ensure consistent antibody performance across experiments and maximize the value of your research reagents.

How do DGKZ knockout models inform our understanding of antibody specificity and functional studies?

DGKZ knockout (KO) models have become instrumental in validating antibody specificity and understanding DGKZ function in complex biological systems:

Antibody validation using knockout controls:

  • DGKZ KO tissues/cells provide the gold standard negative control for antibody specificity

  • True specific antibodies should show no signal in DGKZ KO samples while detecting the expected ~124 kDa band in wild-type samples

  • Studies utilizing DGKζ^-/- mice have demonstrated enhanced specificity validation compared to traditional blocking peptide approaches

Functional insights from knockout phenotypes:

  • DGKζ^-/- T cells display hyperresponsiveness to TCR stimulation with enhanced ERK activation and cytokine production

  • DGKζ^-/- mice show improved viral clearance in LCMV Armstrong infection models

  • Enhanced anti-tumor responses are observed in DGKζ^-/- mice, particularly against MC38 tumors, where tumor rejection is more pronounced than in DGKα^-/- mice

  • Interestingly, DGKζ^-/- mice do not develop spontaneous autoimmunity despite enhanced T cell activation, possibly due to concurrent enhancement of regulatory T cell development

Technical considerations for knockout experiments:

  • Some studies use conditional knockout approaches (ERCre system with DGKζ^f/f^ mice) to avoid developmental compensation

  • Double knockout models (DGKα^-/-DGKζ^-/-) demonstrate more profound phenotypes, suggesting partial functional redundancy

  • Reconstitution experiments with wildtype DGKZ in knockout cells can confirm specificity of observed phenotypes

Researchers using DGKZ antibodies should consider performing parallel experiments in knockout systems when possible, as this approach provides the most definitive validation of antibody specificity and functional relevance.

What are the critical methodological considerations when using DGKZ antibodies to study T cell signaling?

When investigating T cell signaling with DGKZ antibodies, several methodological considerations are essential:

T cell activation conditions:

  • DGKZ's regulatory role is most evident during suboptimal TCR stimulation, as differences between wildtype and DGKZ-deficient cells may be masked under strong stimulation conditions

  • Significant differences in activation markers (CD69), proliferation, and cytokine production between wildtype and DGKZ-deficient T cells are observed at lower concentrations of anti-CD3/CD28 antibodies

  • DAG analog phorbol-12-myristate-13-acetate (PMA) treatment bypasses TCR activation and abolishes differences between DGKZ KO and WT T cells, confirming that hyperactivation phenotypes are due to enhanced DAG signaling

Timing considerations:

  • DGKZ activity dynamically regulates DAG levels during T cell activation

  • Early time points (minutes to hours) are critical for studying immediate signaling events

  • Later time points (hours to days) reveal effects on proliferation and differentiation

Subcellular localization analysis:

  • DGKZ shuttles between cytoplasmic and nuclear compartments

  • Proper cell fixation and permeabilization protocols are essential

  • Confocal microscopy with appropriate controls should be used to determine subcellular distribution changes during T cell activation

Downstream signaling analysis:

  • Monitor ERK phosphorylation as a primary readout of DAG-mediated signaling

  • Assess PKCθ membrane translocation through fractionation or imaging approaches

  • Examine RasGRP1 activation, which is directly regulated by DGKZ isoform 1 but not isoform 2

Functional outcome measurements:

  • Proliferation assays (CFSE dilution or thymidine incorporation)

  • Cytokine production (ELISA or intracellular staining)

  • Cytotoxicity assays for CD8+ T cells

  • In vivo models of infection or tumor challenge

These methodological considerations help ensure that experiments using DGKZ antibodies accurately reveal the biological significance of DGKZ in T cell signaling cascades.

How does DGKZ interact with DGKα in immune function, and what are the implications for antibody-based studies?

DGKZ and DGKα exhibit complex interactions in immune function that have important implications for antibody-based studies:

Hierarchical and cooperative roles:

  • DGKZ appears to be the dominant isoform in T cells based on direct comparisons of TCR signal strength between DGKα^-/- and DGKζ^-/- T cells

  • DGKζ exerts greater control than DGKα over CD8+ T cell activity and tumor control in vivo, particularly evident in the MC38 tumor model

  • Single knockout of either DGKα or DGKζ selectively impairs TH1 cell differentiation, while double knockout enhances both TH1 and TH17 differentiation, indicating complex, non-redundant roles

Methodological approaches for distinguishing isoform-specific functions:

  • Isoform-specific antibodies that recognize unique domains (e.g., MARCKS domain in DGKζ)

  • Side-by-side comparison of single knockout models (DGKα^-/- vs. DGKζ^-/-)

  • Double knockout models followed by reconstitution with individual isoforms

  • isoform-specific inhibitors (e.g., the more specific DGKα inhibitor recently developed)

Experimental design considerations:

  • Include both isoforms in expression analysis studies

  • Compare phenotypes between single and double knockout models

  • Design antibody-based experiments that can distinguish between isoforms

  • Consider potential compensatory mechanisms when one isoform is targeted

Signaling pathway differential regulation:

  • DGKζ more strongly regulates the Ras-ERK pathway

  • DGKα may preferentially affect PKC-dependent pathways

  • Both isoforms impact NF-κB signaling but through potentially different mechanisms

Understanding these distinctions is crucial when designing experiments with DGKZ antibodies to avoid misinterpreting results due to compensatory mechanisms or overlapping functions between DGK isoforms.

What are the challenges in detecting DGKZ in different subcellular compartments, and how can antibody-based approaches address them?

Detecting DGKZ in various subcellular compartments presents significant technical challenges that require specialized antibody-based approaches:

Subcellular distribution complexity:

  • DGKZ exhibits dynamic distribution between cytoplasmic, nuclear, and membrane-associated compartments

  • This distribution may change during cell activation, differentiation, or in response to stimuli

  • Multiple isoforms may show distinct localization patterns

Technical challenges in immunostaining:

  • Fixation-dependent epitope accessibility:

    • Over-fixation can mask epitopes

    • Under-fixation can cause protein redistribution during processing

    • Different fixatives (paraformaldehyde, methanol, acetone) may preserve different epitopes

  • Membrane-associated protein extraction:

    • Standard lysis buffers may incompletely solubilize membrane-associated DGKZ

    • Specialized detergent combinations may be required for complete extraction

  • Nuclear localization detection:

    • Nuclear envelope can create barriers to antibody penetration

    • Nuclear extraction protocols may disrupt native protein interactions

Methodological solutions:

  • Subcellular fractionation combined with Western blotting:

    • Separate cytoplasmic, membrane, nuclear, and cytoskeletal fractions

    • Use fraction-specific markers (e.g., GAPDH, Na+/K+-ATPase, Histone H3) to verify fractionation quality

    • Quantify DGKZ distribution across fractions using validated antibodies

  • Confocal microscopy optimization:

    • Test multiple fixation and permeabilization protocols

    • Use z-stack imaging to assess three-dimensional distribution

    • Perform co-localization studies with compartment-specific markers

    • Consider super-resolution microscopy for detailed localization studies

  • Live-cell imaging approaches:

    • Use fluorescently-tagged DGKZ constructs to complement antibody-based detection

    • Validate localization with antibody staining of fixed cells

  • Proximity ligation assays:

    • Detect DGKZ interaction with compartment-specific proteins

    • Provide spatial resolution beyond standard co-localization

By addressing these challenges with appropriate methodological approaches, researchers can gain more accurate insights into the dynamic subcellular distribution of DGKZ and its functional implications.

How can DGKZ antibodies be utilized in cancer immunotherapy research?

DGKZ antibodies serve as valuable tools in cancer immunotherapy research, particularly for understanding T cell dysfunction and developing strategies to enhance anti-tumor immunity:

Mapping T cell dysfunction mechanisms:

  • DGKZ expression in tumor-infiltrating lymphocytes (TILs) correlates with reduced anti-tumor activity

  • DGKZ antibodies enable assessment of expression levels in TILs compared to peripheral blood T cells

  • Immunohistochemistry with DGKZ antibodies can map spatial distribution within the tumor microenvironment

Therapeutic targeting validation:

  • Studies show that DGKζ^-/- mice demonstrate enhanced control of MC38 tumors compared to wildtype or DGKα^-/- mice

  • DGKZ antibodies can confirm successful genetic or pharmacological targeting in various experimental models

  • Combining DGKZ deficiency with checkpoint inhibition (anti-PD1) shows additive effects in tumor control

CAR-T cell engineering applications:

  • DGKζ deficiency promotes chimeric antigen receptor (CAR) T cell-mediated anti-tumor responses

  • DGKZ antibodies can validate knockdown efficiency in engineered T cells

  • Expression analysis before and after tumor exposure can track potential adaptive upregulation

Potential combinatorial strategies:

  • The MARCKS domain of DGKZ represents a potential target for therapeutic intervention

  • Antibodies recognizing this domain can help validate targeting approaches

  • Simultaneous targeting of both DGKα and DGKζ may provide enhanced anti-tumor effects

Considerations for clinical translation:

  • DGKZ knockout enhances T cell proliferation without inducing spontaneous autoimmunity

  • Antibody-based monitoring of DGKZ expression could help predict responsiveness to immunotherapy

  • Combinatorial approaches targeting both DGKZ and immune checkpoint molecules may enhance efficacy

These applications highlight how DGKZ antibodies contribute to both mechanistic understanding and therapeutic development in cancer immunotherapy research.

What are the methodological approaches for studying DGKZ in B cell development and antibody responses?

Investigating DGKZ in B cell development and antibody responses requires specialized methodological approaches utilizing DGKZ antibodies:

B cell developmental analysis:

  • Flow cytometry with DGKZ antibodies can quantify expression across developmental stages

  • DGKZ mRNA transcripts increase as B cells progress from early transitional to mature follicular stages

  • Immunohistochemistry of lymphoid tissues can map spatial expression patterns in follicular versus marginal zone compartments

Signaling threshold regulation:

  • DGKZ controls B cell receptor (BCR) activation threshold particularly in mature follicular B cells

  • Western blot analysis of ERK phosphorylation and IκBα degradation under varying BCR stimulation conditions reveals DGKZ-dependent signaling differences

  • Flow cytometry with phospho-specific antibodies alongside DGKZ detection enables single-cell correlation of expression with signaling output

Experimental approaches for functional studies:

  • In vitro activation models:

    • Purified B cells stimulated with anti-IgM under varying conditions

    • Measurement of activation markers (CD69), proliferation, and differentiation markers

    • DGKZ antibodies for protein expression correlation with functional outcomes

  • In vivo immunization protocols:

    • T-dependent antigens (e.g., NP-KLH, SRBC)

    • T-independent antigens (e.g., TNP-Ficoll, NP-LPS)

    • Germinal center formation assessment

    • Plasma cell differentiation analysis

  • Antibody response evaluation:

    • ELISA for antigen-specific antibody titers

    • ELISPOT for enumeration of antibody-secreting cells

    • Flow cytometry to quantify antigen-specific B cells

Key findings from DGKZ-deficient B cell studies:

  • DGKζ KO mice show enhanced antibody responses to both T-dependent and T-independent antigens

  • Enhanced antigen-specific expansion of germinal center B cells and plasma cells is observed in DGKζ-deficient mice

  • Effects are most pronounced under suboptimal BCR stimulation conditions

These methodological approaches enable comprehensive analysis of how DGKZ regulates B cell development, activation thresholds, and antibody production in both physiological and pathological contexts.

What are the discrepancies in DGKZ research findings, and how might antibody selection contribute to these inconsistencies?

Several discrepancies exist in the DGKZ research literature, and antibody selection may contribute significantly to these inconsistencies:

Observed research discrepancies:

  • Subcellular localization variations:

    • Some studies report predominantly cytoplasmic localization

    • Others describe significant nuclear presence

    • Membrane association has varying descriptions across studies

  • Functional impact disparities:

    • While most studies show enhanced T cell activation in DGKZ-deficient models, the magnitude varies considerably

    • Some studies suggest DGKZ deficiency promotes autoimmunity, while others note no spontaneous autoimmunity despite enhanced T cell activation

    • Effects on TH differentiation show complex patterns: single knockout impairs TH1 differentiation, but double knockout enhances both TH1 and TH17

  • Molecular weight inconsistencies:

    • Reported as 124.1 kDa based on amino acid sequence

    • Western blots sometimes show bands at different molecular weights

Antibody-related factors contributing to discrepancies:

  • Epitope specificity:

    • Antibodies targeting different domains may detect distinct subsets of DGKZ molecules

    • Post-translational modifications may mask certain epitopes

    • C-terminal (aa 1050 to C-terminus) versus N-terminal (aa 1-200) targeting antibodies may yield different results

  • Isoform recognition:

    • Multiple DGKZ isoforms exist with different functional properties

    • Isoform 1 regulates RASGRP1 activity, while isoform 2 does not

    • Antibodies may differentially detect these isoforms

  • Technical variables:

    • Application-specific performance differences (antibodies optimized for WB may perform poorly in IHC)

    • Batch-to-batch variation in polyclonal antibody preparations

    • Differences in immunogen design and antibody production methods

Recommendations for addressing discrepancies:

  • Detailed antibody reporting:

    • Specify catalog number, lot number, and dilution

    • Describe validation methods used

    • Include knockout/knockdown controls when possible

  • Multi-antibody approach:

    • Use multiple antibodies targeting different epitopes

    • Compare monoclonal and polyclonal antibodies

    • Validate with complementary techniques (e.g., mass spectrometry)

  • Standardized protocols:

    • Develop community standards for DGKZ detection methods

    • Share detailed protocols including critical parameters

    • Conduct interlaboratory validation studies

Understanding these discrepancies and their potential sources is essential for accurate interpretation of DGKZ research findings and for designing experiments that can resolve existing contradictions in the literature.

How can researchers optimize DGKZ antibody-based approaches for studying T helper cell differentiation?

Optimizing DGKZ antibody-based approaches for studying T helper (TH) cell differentiation requires careful consideration of several technical and experimental design factors:

T helper differentiation complexity:

  • DGKZ deficiency shows complex effects on TH differentiation: single knockout of either DGKα or DGKζ selectively impairs TH1 differentiation, but double knockout enhances both TH1 and TH17 differentiation

  • These paradoxical findings suggest context-dependent roles requiring sophisticated experimental approaches

Optimized differentiation protocols:

  • In vitro differentiation systems:

    • Isolate naïve CD4+ T cells (CD4+CD44lowCD62Lhigh) using FACS or magnetic separation

    • Culture in IMDM supplemented with 10% FBS, antibiotics, and 2-mercaptoethanol

    • Apply polarizing conditions:

      • TH1: IL-12 + anti-IL-4

      • TH2: IL-4 + anti-IFN-γ

      • TH17: TGF-β + IL-6 + anti-IFN-γ + anti-IL-4

      • Treg: TGF-β + IL-2

  • Multiparameter flow cytometry panels:

    • Surface markers: CD4, CD44, CD62L, activation markers

    • Transcription factors: T-bet (TH1), GATA3 (TH2), RORγt (TH17), Foxp3 (Treg)

    • Cytokines: IFN-γ (TH1), IL-4 (TH2), IL-17A/F (TH17)

    • Include DGKZ staining to correlate expression with differentiation state

Analytical approaches:

  • Time-course analysis:

    • Examine DGKZ expression dynamics during differentiation process (days 0, 1, 3, 5, 7)

    • Correlate with acquisition of lineage-specific markers

  • Signaling pathway interrogation:

    • Phospho-flow cytometry for key pathways (mTOR, STAT, MAPK)

    • Western blot analysis at critical time points

    • Correlate DGKZ expression with signaling intensity

  • Transcriptional analysis:

    • qPCR for lineage-defining transcripts

    • Single-cell approaches to capture heterogeneity

    • ChIP-seq to identify DGKZ-dependent epigenetic changes

In vivo validation approaches:

  • Adoptive transfer models:

    • Transfer DGKZ-deficient versus wildtype naïve CD4+ T cells into congenic recipients

    • Challenge with appropriate stimuli (infection, immunization)

    • Analyze fate using flow cytometry with antibodies against DGKZ and lineage markers

  • Disease models:

    • Experimental autoimmune encephalomyelitis (EAE) for TH17 responses

    • Allergic inflammation models for TH2 responses

    • Intracellular pathogen challenges for TH1 responses

  • Tissue-specific analysis:

    • Immunohistochemistry to localize DGKZ expression in lymphoid organs

    • Multi-color fluorescence microscopy to identify co-expression with lineage markers

These optimized approaches enable researchers to dissect the complex and context-dependent roles of DGKZ in T helper cell differentiation, potentially resolving current discrepancies in the literature.

What are the common technical challenges in DGKZ Western blotting, and how can they be addressed?

Western blotting for DGKZ presents several technical challenges that can be addressed through optimized protocols:

Challenge 1: Detecting the full-sized 124 kDa protein

  • Problem: High molecular weight proteins transfer inefficiently.

  • Solutions:

    • Use low percentage gels (6-8%) for better resolution of high MW proteins

    • Extend transfer time (overnight at low voltage) or use specialized high MW transfer systems

    • Add SDS (0.1%) to transfer buffer to aid in large protein migration

    • Consider semi-dry transfer systems optimized for high MW proteins

Challenge 2: Multiple bands or unexpected molecular weights

  • Problem: DGKZ antibodies may detect multiple isoforms, degradation products, or non-specific bands.

  • Solutions:

    • Include both positive controls (cells known to express DGKZ) and negative controls (DGKZ knockout cells or tissues if available)

    • Use freshly prepared lysates with complete protease inhibitor cocktails

    • Compare results with multiple antibodies targeting different epitopes

    • Verify bands using DGKZ-overexpressing cells as positive controls

    • For recombinant DGKZ antibody (A06678-1), the recommended dilution is 1:500-1:2000 for Western blot applications

Challenge 3: Inconsistent results across experiments

  • Problem: Variable band intensity or pattern between experiments.

  • Solutions:

    • Standardize lysate preparation (consistent lysis buffer, protein concentration, and handling)

    • Optimize antibody concentration through titration experiments

    • Prepare larger batches of working solutions to reduce preparation variability

    • Consider using automated Western blot systems for greater consistency

    • Normalize loading with appropriate housekeeping proteins

Challenge 4: High background

  • Problem: Non-specific binding creates high background that obscures specific signals.

  • Solutions:

    • Increase blocking time or concentration (5% BSA often performs better than milk for phospho-proteins)

    • Include 0.1-0.3% Tween-20 in wash buffers

    • Try alternative secondary antibodies

    • For polyclonal antibodies, pre-adsorb against tissues/cells lacking the target protein

Optimized DGKZ Western blot protocol:

  • Prepare lysates in RIPA buffer with protease inhibitors

  • Separate 30-50 μg protein on 8% SDS-PAGE

  • Transfer to PVDF membrane overnight at 30V, 4°C

  • Block in 5% BSA in TBST for 2 hours at room temperature

  • Incubate with primary antibody at 1:1000 dilution overnight at 4°C

  • Wash 4 × 10 minutes with TBST

  • Incubate with appropriate HRP-conjugated secondary antibody (1:5000) for 1 hour

  • Wash 4 × 10 minutes with TBST

  • Develop using enhanced chemiluminescence (ECL) detection system

  • Expected result: DGKZ band at approximately 124 kDa

These optimizations help ensure consistent and reliable detection of DGKZ in Western blotting applications.

How can researchers validate DGKZ antibody specificity in their experimental systems?

Comprehensive validation of DGKZ antibody specificity is essential for generating reliable research data. Researchers should implement multiple complementary approaches:

Genetic approach validation strategies:

  • Knockout/knockdown controls:

    • Test antibodies on tissues/cells from DGKZ knockout mice or CRISPR-edited cell lines

    • Use siRNA or shRNA knockdown samples with verified reduction in DGKZ mRNA

    • Include heterozygous samples to confirm dose-dependent signal reduction

  • Overexpression systems:

    • Compare wildtype cells to those transfected with DGKZ expression constructs

    • Use tagged DGKZ constructs (e.g., GFP-fusion) to confirm co-localization with antibody signal

    • Test multiple DGKZ isoforms to determine isoform specificity

Immunological validation approaches:

  • Epitope blocking:

    • Pre-incubate antibody with immunizing peptide before application

    • Verify signal elimination or reduction in blocked samples

    • Use unrelated peptides as negative controls for blocking

  • Multiple antibody concordance:

    • Compare results using antibodies against different DGKZ epitopes

    • Confirm similar patterns in compatible applications

    • Reconcile differences by determining epitope accessibility in different contexts

Application-specific validation methods:

  • Western blot validation:

    • Verify single band at expected molecular weight (~124 kDa)

    • Compare migration pattern with recombinant DGKZ protein

    • Assess band disappearance in knockout/knockdown samples

  • Immunohistochemistry/Immunofluorescence validation:

    • Compare staining patterns with published DGKZ localization data

    • Perform dual-labeling with antibodies to known DGKZ-interacting proteins

    • Include absorption controls and isotype controls

    • Verify staining pattern differences between wildtype and knockout tissues

  • Flow cytometry validation:

    • Compare signal in positive and negative cell populations

    • Include fluorescence-minus-one (FMO) controls

    • Verify detection of overexpressed DGKZ in transfected cells

Advanced validation approaches:

  • Mass spectrometry confirmation:

    • Perform immunoprecipitation with the DGKZ antibody

    • Analyze pulled-down proteins by mass spectrometry

    • Confirm presence of DGKZ peptides in immunoprecipitated samples

  • Functional validation:

    • Correlate antibody-detected expression levels with known DGKZ-dependent functions

    • For example, verify that cells with higher antibody-detected DGKZ show reduced DAG-dependent signaling

Implementing these validation strategies provides confidence in antibody specificity and ensures that experimental results accurately reflect true DGKZ biology.

What are the best practices for quantifying DGKZ expression in tissue samples for comparative studies?

Accurate quantification of DGKZ expression in tissue samples requires standardized approaches to ensure reliable comparative analyses:

Sample preparation standardization:

  • Tissue collection and processing:

    • Minimize cold ischemia time (<30 minutes when possible)

    • Use consistent fixation protocols (duration, fixative composition)

    • Process all experimental groups simultaneously to minimize batch effects

    • For frozen sections, snap-freeze tissues in liquid nitrogen and store at -80°C

  • Extraction methods for protein analysis:

    • Use specialized extraction buffers that efficiently solubilize membrane-associated proteins

    • Include phosphatase and protease inhibitors to prevent degradation

    • Homogenize tissues using consistent mechanical disruption methods

    • Determine protein concentration using methods tolerant of detergents (e.g., BCA assay)

Quantification approaches:

  • Western blot quantification:

    • Include calibration standards on each gel (recombinant DGKZ or consistently expressing cell line)

    • Use internal loading controls appropriate for the experimental context

    • Employ digital image analysis with linear detection range verification

    • Present data as normalized DGKZ/loading control ratios

  • Immunohistochemistry quantification:

    • Use automated staining platforms when possible for consistency

    • Include positive and negative control tissues on each slide

    • Employ digital pathology approaches:

      • Whole slide scanning at standardized resolution

      • Automated region of interest (ROI) selection

      • Consistent thresholding algorithms

      • Report both staining intensity and percentage of positive cells

  • Flow cytometry quantification:

    • Use antibody-capture beads to establish standard curves

    • Report data as molecules of equivalent soluble fluorochrome (MESF)

    • Include fluorescence-minus-one (FMO) controls

    • Gate consistently across samples

Statistical approaches for comparative studies:

  • Normalization strategies:

    • Consider using multiple reference genes/proteins for normalization

    • Employ global normalization methods for large-scale studies

    • Verify that normalization controls are not affected by experimental conditions

  • Accounting for technical variability:

    • Include technical replicates to assess method precision

    • Use mixed-effects statistical models that account for batch effects

    • Consider randomization of sample processing order

  • Presentation of quantitative data:

    • Report both absolute and relative quantification when possible

    • Include scatter plots to show distribution rather than only bar graphs

    • Report effect sizes with confidence intervals rather than only p-values

Special considerations for DGKZ:

  • Account for potential differences in antibody affinity between species when comparing across organisms

  • Consider quantifying multiple DGKZ isoforms separately if using isoform-specific antibodies

  • For phosphorylation studies, report both total DGKZ and phosphorylated DGKZ

Implementing these best practices ensures that comparative studies of DGKZ expression generate reliable, reproducible, and biologically meaningful quantitative data.

How can researchers integrate multiple detection methods to gain comprehensive insights into DGKZ biology?

Integrating multiple detection methods provides a more complete understanding of DGKZ biology by overcoming the limitations of individual techniques:

Complementary method integration strategies:

  • Multi-omics approach:

    • Genomics: Analyze DGKZ gene variants, copy number, and mutations

    • Transcriptomics: Measure DGKZ mRNA expression and splicing variants

    • Proteomics: Quantify DGKZ protein levels and post-translational modifications

    • Metabolomics: Assess DAG and PA levels as functional readouts of DGKZ activity

    • Integration: Use computational approaches to correlate findings across platforms

  • Multi-scale biological analysis:

    • Molecular level: In vitro enzyme activity assays with recombinant DGKZ

    • Cellular level: Immunofluorescence for subcellular localization

    • Tissue level: Immunohistochemistry for expression patterns

    • Organism level: Phenotypic analysis of DGKZ knockout models

Technical integration approaches:

  • Correlative microscopy:

    • Perform immunofluorescence with super-resolution techniques

    • Use the same specimens for electron microscopy (immuno-EM)

    • Implement correlative light and electron microscopy (CLEM)

    • Example application: Precise localization of DGKZ at membrane microdomains during T cell activation

  • Flow cytometry with functional assays:

    • Combine DGKZ antibody staining with:

      • Phospho-flow detection of ERK activation

      • Calcium flux measurement

      • Cell proliferation tracking (CFSE dilution)

      • Cytokine production (intracellular cytokine staining)

    • Example application: Correlate DGKZ expression levels with functional outcomes in single cells

  • Biochemical approaches with imaging validation:

    • Perform co-immunoprecipitation to identify DGKZ interaction partners

    • Validate interactions with proximity ligation assays in intact cells

    • Confirm co-localization with confocal microscopy

    • Example application: Characterizing the DGKZ interactome in different cell activation states

Case study: Comprehensive analysis of DGKZ in T cell activation

Integrated approach combining:

  • Expression analysis:

    • qPCR for mRNA quantification

    • Western blot for protein levels

    • Flow cytometry for single-cell quantification

  • Localization studies:

    • Subcellular fractionation followed by Western blot

    • Confocal microscopy with organelle markers

    • Live-cell imaging of fluorescently tagged DGKZ

  • Functional assessments:

    • Enzymatic activity assays measuring DAG conversion to PA

    • Phospho-flow analysis of downstream signaling

    • Proliferation and cytokine production assays

  • In vivo relevance:

    • Adoptive transfer experiments with DGKZ-deficient T cells

    • Challenge models (infection, tumor, autoimmunity)

    • Therapeutic targeting experiments

This integrated approach provides a comprehensive picture of DGKZ biology that cannot be achieved through any single methodology, enabling researchers to understand how DGKZ expression, localization, and activity collectively regulate T cell function in health and disease.

How might developing DGKZ phospho-specific antibodies advance our understanding of its regulation?

The development of phospho-specific antibodies recognizing specific DGKZ phosphorylation sites would significantly advance our understanding of its dynamic regulation:

Current knowledge gaps:

  • DGKZ contains multiple potential phosphorylation sites that may regulate its activity, localization, and protein interactions

  • Phosphorylation may mediate the shuttling of DGKZ between cytoplasmic and nuclear compartments

  • The kinetics and responsible kinases for DGKZ phosphorylation during T cell activation remain incompletely understood

Target phosphorylation sites for antibody development:

  • MARCKS domain phosphorylation - This domain is unique to DGKζ compared to DGKα and represents a potential specificity determinant

  • Nuclear localization sequence (NLS) phosphorylation - May regulate nuclear-cytoplasmic shuttling

  • Catalytic domain phosphorylation - Could directly modulate enzymatic activity

  • Protein interaction domain phosphorylation - May regulate binding to partners like RasGRP1

Methodological approaches for phospho-antibody development:

  • Identification of relevant phosphorylation sites:

    • Mass spectrometry analysis of DGKZ under various activation conditions

    • Bioinformatic prediction of kinase target sequences

    • Comparison with evolutionary conserved sites across species

  • Antibody generation strategy:

    • Synthetic phosphopeptides representing specific phosphorylation sites

    • Multiple rabbit immunization with phosphopeptide conjugates

    • Extensive screening with both phosphorylated and non-phosphorylated peptides

    • Affinity purification against phosphopeptide columns

  • Validation approaches:

    • Western blot comparison of wildtype versus phospho-mutant DGKZ

    • Treatment with phosphatases to confirm phospho-specificity

    • Kinase inhibitor studies to identify regulatory pathways

    • Stimulation time-course experiments to detect dynamic changes

Potential research applications:

  • Signaling dynamics analysis:

    • Mapping the kinetics of DGKZ phosphorylation during T cell activation

    • Determining the sequence of phosphorylation events on multiple sites

    • Correlating phosphorylation with changes in subcellular localization and activity

  • Identification of regulatory kinases:

    • Systematic kinase inhibitor screens to identify regulators

    • In vitro kinase assays with candidate kinases

    • Genetic approaches using kinase knockout models

  • Functional correlation studies:

    • Analysis of how phosphorylation status correlates with T cell functional outcomes

    • Development of phosphomimetic and phosphodeficient mutants for mechanistic studies

    • Therapeutic targeting of specific phosphorylation events

The development of phospho-specific DGKZ antibodies would provide valuable new tools for understanding the complex regulation of this important signaling enzyme and potentially reveal new therapeutic intervention points for modulating immune responses.

What novel antibody-based technologies might advance DGKZ research beyond traditional applications?

Emerging antibody-based technologies offer exciting opportunities to advance DGKZ research beyond traditional applications:

Single-cell proteomics approaches:

  • Mass cytometry (CyTOF):

    • Integrate DGKZ antibodies into multi-parameter panels (30+ markers)

    • Simultaneously detect DGKZ with activation markers, transcription factors, and signaling molecules

    • Identify rare cell populations with distinct DGKZ expression patterns

    • Application: Mapping DGKZ expression across immune cell subsets in normal and disease states

  • Single-cell Western blotting:

    • Analyze DGKZ expression in thousands of individual cells

    • Correlate with other signaling molecules at single-cell level

    • Reveal population heterogeneity masked in conventional Western blots

    • Application: Identifying distinct signaling states in activated T cell populations

Spatially resolved proteomics:

  • Imaging mass cytometry:

    • Combine DGKZ detection with tissue architecture preservation

    • Analyze dozens of proteins simultaneously in tissue sections

    • Provide spatial context for DGKZ expression and activity

    • Application: Mapping DGKZ expression in lymphoid tissue microenvironments

  • Multiplexed ion beam imaging (MIBI):

    • Achieve sub-cellular resolution with multiple antibodies

    • Study DGKZ co-localization with interaction partners

    • Examine tissue-level expression patterns with unprecedented detail

    • Application: Analyzing DGKZ distribution in tumor-infiltrating lymphocytes with spatial context

Temporal dynamics technologies:

  • Live-cell antibody fragment imaging:

    • Engineer Fab fragments or nanobodies against DGKZ

    • Label with cell-permeable fluorophores

    • Track DGKZ dynamics in living cells during activation

    • Application: Real-time visualization of DGKZ translocation during T cell activation

  • Intracellular protein-protein interaction detection:

    • Split fluorescent protein complementation with DGKZ fusion proteins

    • FRET/FLIM approaches to detect DGKZ interactions

    • Proximity ligation assays for endogenous protein interactions

    • Application: Mapping the dynamic DGKZ interactome during immune cell activation

Functional manipulation approaches:

  • Intrabodies:

    • Engineer antibody fragments that function inside living cells

    • Target specific domains of DGKZ to inhibit function

    • Provide greater specificity than small molecule inhibitors

    • Application: Domain-specific inhibition of DGKZ to dissect function

  • Antibody-directed protein degradation:

    • Develop DGKZ-targeting PROTACs (Proteolysis Targeting Chimeras)

    • Achieve rapid and specific DGKZ degradation

    • Provide temporal control over DGKZ depletion

    • Application: Acute DGKZ depletion during defined stages of immune responses

  • Optogenetic antibody-based tools:

    • Create light-sensitive antibody-based inhibitors of DGKZ

    • Enable precise spatiotemporal control of DGKZ activity

    • Study subcellular roles of DGKZ with unprecedented precision

    • Application: Investigating localized DGKZ function during immunological synapse formation

These innovative technologies will enable researchers to study DGKZ with greater precision, resolution, and physiological relevance, potentially revealing new aspects of its biology that have remained hidden using conventional approaches.

How might the development of conformation-specific DGKZ antibodies advance mechanistic understanding?

The development of conformation-specific antibodies that recognize distinct structural states of DGKZ would provide unprecedented insights into its activation mechanisms and regulation:

Conformational states of potential interest:

  • Active site conformation:

    • Open (catalytically active) versus closed (inactive) states

    • Substrate-bound versus unbound conformations

    • ATP-binding pocket occupancy states

  • Regulatory domain conformations:

    • Extended versus compact MARCKS domain configurations

    • Exposed versus masked nuclear localization signals

    • Protein interaction surface accessibility states

  • Oligomerization states:

    • Monomeric versus dimeric/multimeric forms

    • Interaction-specific conformational changes

Development strategies for conformation-specific antibodies:

  • Structure-guided design:

    • Use structural biology data (X-ray crystallography, cryo-EM) to identify conformation-specific epitopes

    • Design immunogens that stabilize specific conformations

    • Employ computational modeling to predict exposed regions in different states

  • Screening approaches:

    • Generate large antibody libraries (phage display, yeast display)

    • Screen against native DGKZ in different biochemical conditions

    • Select antibodies that discriminate between conformational states

    • Validate using mutants locked in specific conformations

  • Nanobody/single-domain antibody development:

    • Develop camelid nanobodies against DGKZ conformational states

    • Their small size allows access to cryptic epitopes

    • Can be used as crystallization chaperones to capture transient states

Research applications:

  • Enzymatic mechanism studies:

    • Track the transition between inactive and active conformations during signaling

    • Correlate conformational changes with catalytic activity

    • Identify allosteric regulators of DGKZ conformational states

  • Signaling dynamics visualization:

    • Visualize DGKZ activation state changes during T cell receptor engagement

    • Map the spatiotemporal dynamics of active DGKZ at the immunological synapse

    • Correlate with downstream signaling events

  • Protein interaction regulation:

    • Determine how conformational changes expose or mask interaction surfaces

    • Identify conformation-specific binding partners

    • Understand the structural basis for isoform-specific functions

Therapeutic implications:

  • Conformation-specific inhibitors:

    • Use conformation-specific antibodies as templates for designing small molecule inhibitors

    • Develop biologics that stabilize inactive conformations

    • Create activators that promote active conformations in specific contexts

  • Diagnostic applications:

    • Develop assays to measure the activation state of DGKZ in patient samples

    • Correlate DGKZ conformational states with disease progression or treatment response

    • Use as biomarkers for immune system activation state

The development of conformation-specific DGKZ antibodies would move beyond simple detection of protein presence to provide dynamic information about the functional state of DGKZ in various biological contexts, representing a significant advancement in our mechanistic understanding of this important signaling enzyme.

What are the emerging applications of DGKZ antibodies in precision medicine and biomarker development?

DGKZ antibodies are poised to play important roles in precision medicine approaches and biomarker development across several disease contexts:

Cancer immunotherapy applications:

  • Predictive biomarkers for immunotherapy response:

    • DGKZ expression levels in tumor-infiltrating T cells may predict responsiveness to checkpoint inhibitors

    • Rationale: High DGKZ levels correlate with T cell hyporesponsiveness

    • Approach: Multiplex immunohistochemistry panels including DGKZ and T cell markers

    • Clinical application: Patient stratification for checkpoint inhibitor therapy

  • Monitoring CAR-T cell functionality:

    • DGKZ levels may serve as a marker of CAR-T cell exhaustion or dysfunction

    • DGKζ deficiency promotes CAR T cell-mediated anti-tumor responses

    • Approach: Flow cytometric assessment of DGKZ in CAR-T products pre-infusion

    • Clinical application: Quality control metric for cell therapy products

Autoimmune disease applications:

  • Disease activity biomarkers:

    • DGKZ phosphorylation state as an indicator of aberrant T cell activation

    • Rationale: DGKZ regulates activation thresholds in both T and B cells

    • Approach: Phospho-specific antibodies to measure active vs. inactive DGKZ in patient samples

    • Clinical application: Monitoring disease activity in rheumatoid arthritis, lupus, or multiple sclerosis

  • Therapeutic response prediction:

    • DGKZ expression patterns may predict response to specific immunomodulatory therapies

    • Approach: Pre-treatment peripheral blood mononuclear cell (PBMC) profiling

    • Clinical application: Personalized selection of immunosuppressive regimens

Infectious disease applications:

  • Immune exhaustion monitoring:

    • DGKZ upregulation as a marker of T cell exhaustion in chronic infections

    • Rationale: Enhanced viral clearance in DGKZ-deficient models

    • Approach: Serial monitoring of DGKZ in virus-specific T cells during infection

    • Clinical application: Predicting spontaneous viral clearance vs. chronicity

  • Vaccination response prediction:

    • DGKZ levels in B cells may predict antibody response quality after vaccination

    • Rationale: Enhanced antibody responses in DGKZ-deficient mice

    • Approach: Pre-vaccination DGKZ assessment in memory B cells

    • Clinical application: Identifying individuals needing enhanced vaccination strategies

Technological approaches:

  • Single-cell analysis platforms:

    • Mass cytometry for high-dimensional analysis of DGKZ with other markers

    • Single-cell sequencing with protein detection (CITE-seq) including DGKZ antibodies

    • Application: Identifying specific immune cell subsets with altered DGKZ expression

  • Minimal invasive testing:

    • Development of DGKZ assays compatible with liquid biopsies

    • Application: Longitudinal monitoring without repeated tissue sampling

  • Point-of-care testing:

    • Simplified DGKZ detection platforms for clinical use

    • Application: Rapid assessment of immune activation state

Implementation challenges:

  • Standardization needs:

    • Reference standards for DGKZ quantification

    • Validated clinical cutoff values

    • Quality control systems

  • Combined biomarker strategies:

    • Integration of DGKZ with other immune activation markers

    • Algorithmic approaches to interpret multi-parameter data

    • Machine learning to identify complex patterns associating DGKZ with clinical outcomes

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.