TCIRG1 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to TCIRG1 Antibody

TCIRG1 antibodies are immunological reagents designed to detect and quantify the TCIRG1 protein, encoded by the TCIRG1 gene. This protein is a subunit of the V-ATPase proton pump, essential for lysosomal acidification, osteoclast-mediated bone resorption, and immune cell activation . Antibodies targeting TCIRG1 are widely used in molecular biology and clinical research to explore its pathological roles in osteopetrosis, cancer metastasis, and immune dysregulation.

Antibody Characteristics

TCIRG1 antibodies vary in host species, clonality, and applications. Key examples include:

ProductHostClonalityApplicationsReactivityTarget Region
6H3 (sc-293491)MouseMonoclonalWB, IP, ELISAHumanFull-length TCIRG1
12649-1-APRabbitPolyclonalWB, IHC, IF/ICCHuman, Mouse, PigFusion protein Ag3378
PA5-90425RabbitPolyclonalWB, IHC, IFHuman, Mouse, RatN-terminal (AA 1-130)
CSB-PA615690LA01HURabbitPolyclonalIHC, IFHumanFull-length TCIRG1

These antibodies are validated for detecting TCIRG1 isoforms (OC116 and TIRC7) and their expression patterns across tissues .

Role in Bone Metabolism

  • Osteopetrosis: TCIRG1 mutations impair osteoclast acidification, leading to autosomal recessive osteopetrosis (OPTB1). Knockdown of Tcirg1 in mouse osteoclasts reduces NFATc1 nuclear translocation, inhibiting large osteoclast formation .

  • Mechanism: TCIRG1 deficiency disrupts V-ATPase function, lowering intracellular calcium signaling and osteoclast differentiation .

Cancer Progression

  • Renal Cell Carcinoma (ccRCC): TCIRG1 overexpression correlates with poor prognosis, increased tumor mutation burden (TMB), and immune infiltration (CD8+ T cells, Tregs). Knockdown inhibits ccRCC cell migration .

  • Hepatocellular Carcinoma (HCC): TCIRG1 promotes metastasis by modulating epithelial-mesenchymal transition (EMT) proteins (e.g., E-cadherin, Snail). Antibody-based inhibition reduces tumor growth and invasion .

  • Glioma: TCIRG1 is a biomarker for mesenchymal glioblastoma (GBM) and correlates with immune cell infiltration .

Immune Regulation

  • T-Cell Activation: TCIRG1 isoform TIRC7 is induced during immune activation, co-localizing with the T-cell receptor and CTLA4 in the immune synapse. Antibody targeting suppresses T-cell activation and IL-2 secretion .

  • Autoimmunity: TIRC7-knockout mice exhibit hyperactive T/B-cell responses and reduced CTLA4 expression, linking TCIRG1 to immune checkpoint regulation .

Therapeutic and Diagnostic Applications

  • Immunotherapy: TCIRG1 expression predicts response to immune checkpoint inhibitors (ICIs) in ccRCC .

  • Drug Development: TCIRG1 knockdown or antibody blockade reduces metastasis in HCC and renal cancer models .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery timelines may vary depending on your chosen shipping method and location. Please consult with your local distributors for specific delivery estimates.
Synonyms
a3 antibody; Atp 6i antibody; Atp6i antibody; ATP6N1C antibody; ATP6V0A3 antibody; ATPase H+ transporting 116kD antibody; OC 116 antibody; OC 116 kDa antibody; OC 116kDa antibody; OC-116 kDa antibody; OC116 antibody; OPTB 1 antibody; OPTB1 antibody; Osteoclastic proton pump 116 kDa subunit antibody; Specific 116 kDa vacuolar proton pump subunit antibody; Stv 1 antibody; Stv1 antibody; T cell immune regulator 1 antibody; T cell immune regulator 1 ATPase H+ transporting lysosomal V0 subunit A antibody; T cell immune regulator 1 ATPase H+ transporting lysosomal V0 subunit A3 antibody; T cell immune response cDNA 7 antibody; T cell immune response cDNA7 protein antibody; T cell, immune regulator 1, ATPase, H+ transporting, lysosomal V0 protein a antibody; T-cell immune regulator 1 antibody; T-cell immune response cDNA7 protein antibody; TCIRG 1 antibody; TCIRG1 antibody; TIRC 7 antibody; TIRC7 antibody; V ATPase 116 kDa antibody; V ATPase 116 kDa isoform a3 antibody; V type proton ATPase 116 kDa subunit a antibody; V-ATPase 116 kDa isoform a3 antibody; V-type proton ATPase 116 kDa subunit a isoform 3 antibody; Vacuolar proton translocating ATPase 116 kDa subunit A antibody; Vacuolar proton translocating ATPase 116 kDa subunit a isoform 3 antibody; Vph 1 antibody; Vph1 antibody; VPP3_HUMAN antibody
Target Names
Uniprot No.

Target Background

Function
TCIRG1, also known as ATP6I, is a subunit of the vacuolar H+-ATPase (V-ATPase) proton channel. This protein plays a crucial role in the activation of T-cells.
Gene References Into Functions

TCIRG1 Gene: Functions and Research

  1. Whole exome sequencing has identified mutations in TCIRG1, along with other genes, as a cause of malignant infantile osteopetrosis (MIOP). PMID: 27187610
  2. Case studies have documented TCIRG1-related osteopetrosis with a mild clinical course in Chinese patients. PMID: 28816234
  3. Research suggests that TCIRG1 is involved in inflammation associated with multiple sclerosis. Anti-TCIRG1 antibodies can potentially prevent immune activation by selectively inhibiting the expression of Th1- and Th17-associated cytokines. PMID: 24526664
  4. Mutations in the a3 subunit of the V-ATPase complex, which TCIRG1 is a part of, can disrupt bone resorption processes, leading to infantile osteopetrosis. PMID: 29237407
  5. TCIRG1 may play a role in endolysosomal transport, a process implicated in the development of early-onset Alzheimer's disease. PMID: 28738127
  6. Mutations in the TCIRG1 gene have been associated with infantile malignant osteopetrosis in a Chinese family. PMID: 28604959
  7. Rare variants in TCIRG1 have been linked to lower absolute neutrophil counts. PMID: 27229898
  8. Highly invasive human breast cancer cell lines exhibit higher levels of the a3 isoform of TCIRG1 compared to less invasive lines. Knockdown of a3 reduces V-ATPase expression at the plasma membrane and inhibits in vitro invasion of breast tumor cells. PMID: 26906430
  9. Specific intronic regions in TCIRG1 are prone to splicing mutations, which can result in a reduced amount of protein, potentially mitigating the severity of phenotypes typically associated with TCIRG1 defects. PMID: 25829125
  10. TCIRG1 may be involved in the pathogenesis of aplastic anemia. PMID: 26049920
  11. A specific transversion mutation in TCIRG1 has been identified as a cause of osteopetrosis in the Ashkenazi Jewish population. PMID: 24989235
  12. TCIRG1 may be associated with the pathogenesis of ITP, and its levels could serve as an indicator for evaluating patient response to HD-DXM treatment. PMID: 24617318
  13. Increased TCIRG1 expression in plasma has been linked to the severity of acute graft-versus-host disease. PMID: 25623380
  14. Research suggests that the effects of epiregulin (EREG) and TCIRG1 single nucleotide polymorphism (SNP) on pulmonary tuberculosis susceptibility may be dependent on gene-gene interactions in West African populations. PMID: 24898387
  15. TCIRG1 mutations have been associated with congenital neutropenia. PMID: 24753205
  16. Analysis indicates that mutations in CLCN7 and TCIRG1 differentially impact bone matrix mineralization, suggesting a need to revise the current classification of osteopetrosis. PMID: 24108692
  17. Research has investigated the role of V-ATPase a subunit isoforms in the invasiveness of human breast cancer cells. PMID: 24072707
  18. Large genomic deletions and mutations in the 5' UTR of TCIRG1 are significant for patient management and prenatal diagnosis. PMID: 22231430
  19. The N termini of a-subunit isoforms in V-ATPase are involved in signaling between V-ATPase and cytohesin-2. PMID: 23288846
  20. A novel mutation in TCIRG1, c.242delC, has been identified in infantile malignant osteopetrosis. PMID: 21042819
  21. V-ATPase localization and activity in kidney cells is regulated by PKA-dependent phosphorylation of the A subunit at Ser-175. PMID: 20525692
  22. Mutations in CLCN7 can lead to a phenotype as severe as that caused by TCIRG1 loss of function, indicating the importance of these residues for chloride channel or chloride/proton-exchanger function. PMID: 20424301
  23. TCIRG1 has been localized to chromosome 11q12-13 in autosomal dominant osteopetrosis type I. PMID: 12054167
  24. Linkage and association studies have examined the relationship between peak bone mineral density and the TCIRG1 gene locus on chromosome 11p12-13, which encodes the osteoclast-specific subunit of the vacuolar proton pump. PMID: 12161516
  25. Multiple novel single nucleotide mutations in TCIRG1 have been identified, affecting splice acceptor or donor sites and resulting in aberrant transcription products. PMID: 12552563
  26. A polymorphism affecting an API binding site in the TCIRG1 gene promoter has been associated with bone mass in Scottish women. PMID: 14523594
  27. Nine new TCIRG1 mutations were identified in patients with recessive osteopetrosis. A significant proportion of these patients exhibited specific mutations affecting splicing or protein sequence. PMID: 15300850
  28. Six new alternative splice events in TCIRG1 have been validated by RT-PCR in various human tissues. PMID: 15809087
  29. TCIRG1 acts as an upstream regulator of cytotoxic T-lymphocyte antigen 4 (CTLA-4) expression. PMID: 17082597
  30. The HLA-DR alpha 2 domain (sHLA-DRalpha2) induces negative signals by engaging TCIRG1 on lymphocytes, inhibiting proliferation and inducing apoptosis in CD4+ and CD8+ T-cells via the intrinsic pathway. PMID: 18270567
  31. Analysis of a novel Alu-Alu recombination-mediated genomic deletion in the TCIRG1 gene has been conducted in five osteopetrotic patients. PMID: 18715141
  32. Linkage disequilibrium (LD) mapping of the OPTB locus at the TCIRG1 region has identified a unique splice site mutation in all Chuvashian OPTB patients studied. PMID: 19172990
  33. Mutations in TCIRG1, OSTM1, ClCN7, and TNFRSF11A genes have been detected in patients with infantile malignant osteopetrosis. PMID: 19507210

Show More

Hide All

Database Links

HGNC: 11647

OMIM: 259700

KEGG: hsa:10312

STRING: 9606.ENSP00000265686

UniGene: Hs.495985

Involvement In Disease
Osteopetrosis, autosomal recessive 1 (OPTB1)
Protein Families
V-ATPase 116 kDa subunit family
Subcellular Location
Membrane; Multi-pass membrane protein.
Tissue Specificity
Isoform long is highly expressed in osteoclastomas. Isoform short is highly expressed in thymus.

Q&A

What is TCIRG1 and what are its main biological functions?

TCIRG1 functions as a critical component of vacuolar ATPase (V-ATPase), specifically as the V0 subunit A3. This protein is essential for cellular acidification processes that support metabolism, membrane transport, and intracellular signaling pathways. Research has revealed two main isoforms: TCIRG1-isoa (the longer variant) and TCIRG1-isob (which lacks the first 5 exons of isoa). The protein is expressed in multiple tissues including heart, liver, kidney, lung, and pancreas, with particularly notable roles in immune regulation and bone resorption. Recent studies have also identified TCIRG1's involvement in congenital neutropenia and its potential role as a prognostic biomarker in certain cancers, most notably clear cell renal cell carcinoma (ccRCC), where it appears to influence tumor progression .

How do I select the appropriate TCIRG1 antibody for my specific research application?

Selecting the optimal TCIRG1 antibody requires careful consideration of several factors:

  • Target epitope: Different antibodies target distinct regions of TCIRG1. For example, some antibodies recognize the N-terminal cytoplasmic domain (AA 1-130), while others target regions AA 121-220 or other segments. Choose an antibody that targets the region most relevant to your research question and accessible in your experimental conditions .

  • Isoform specificity: Consider whether your research requires detection of specific TCIRG1 isoforms. Some antibodies detect both TCIRG1-isoa (~45kDa) and TCIRG1-isob, while others are isoform-specific .

  • Application compatibility: Verify that the antibody has been validated for your specific application (WB, IHC, IF, ELISA, FACS). For example, antibody ABIN7270707 is validated for WB, IF, and IHC applications .

  • Host species: Consider the host species (rabbit, mouse) in relation to your experimental design, especially for multi-color immunostaining where avoiding cross-reactivity is essential .

  • Clonality: Polyclonal antibodies offer broader epitope recognition but potential batch variation, while monoclonal antibodies provide higher specificity and reproducibility for quantitative analyses .

What are the validated cross-reactivity profiles of common TCIRG1 antibodies?

Most commercial TCIRG1 antibodies have been validated for human samples, with varying cross-reactivity to mouse and rat TCIRG1. For instance, the polyclonal antibody ABIN7270707 (targeting AA 1-130) demonstrates cross-reactivity with human, mouse, and rat samples, making it suitable for comparative studies across these species. When selecting antibodies for cross-species research, prioritize those with explicitly validated cross-reactivity rather than relying on sequence homology predictions. The antibody's validated applications may also differ between species - some antibodies work for Western blotting in human samples but only for immunofluorescence in mouse/rat samples. Always review the validation data specific to your species and application of interest .

What are the optimal protein extraction methods for Western blot detection of TCIRG1?

For effective TCIRG1 Western blot analysis, the protein extraction method must preserve the membrane-associated properties of this V-ATPase subunit while minimizing protein degradation:

  • Buffer selection: Use RIPA buffer supplemented with protease inhibitors for general applications. For enhanced membrane protein extraction, consider using buffers containing 1-2% NP-40 or Triton X-100.

  • Sample preparation: For peripheral blood mononuclear cells (PBMCs), which have been successfully used in TCIRG1 studies, use RBC lysis followed by gentle membrane disruption to maintain protein integrity .

  • Protein quantification: Due to TCIRG1's varying expression levels across tissues, standardize loading using housekeeping proteins that are stably expressed in your experimental context.

  • Denaturation conditions: Heat samples at 70°C (not 95°C) for 10 minutes in Laemmli buffer containing 2-5% β-mercaptoethanol to reduce aggregation of this membrane protein.

  • Gel selection: Use 10-12% polyacrylamide gels for optimal resolution of TCIRG1's ~45kDa fragment and other isoforms .

When analyzing Western blot results, note that different antibodies may detect distinct fragments of TCIRG1. For instance, polyclonal antibodies against the N-terminal domain detect the ~45kDa TCIRG1 fragment (likely TCIRG1-isob), while other antibodies may detect different isoforms or processed variants .

How can I optimize immunohistochemistry protocols for TCIRG1 detection in different tissue types?

Optimizing IHC protocols for TCIRG1 requires adjustments based on tissue type and fixation method:

  • Antigen retrieval: For formalin-fixed paraffin-embedded (FFPE) tissues, heat-induced epitope retrieval with citrate buffer (pH 6.0) typically yields optimal results for TCIRG1 antibodies. For tissues with high extracellular matrix content, consider extending retrieval time.

  • Antibody concentration: Titrate antibody concentrations beginning with the manufacturer's recommended dilution. TCIRG1 antibodies typically require optimization between 1:100-1:500 dilutions depending on tissue type and fixation conditions .

  • Incubation conditions: For stronger signals in tissues with low TCIRG1 expression, extend primary antibody incubation to overnight at 4°C rather than standard 1-hour incubations.

  • Detection system: For tissues with low expression levels, amplification systems like tyramide signal amplification (TSA) or polymer-based detection may enhance sensitivity while maintaining specificity.

  • Validation controls: Include positive controls (such as renal cell carcinoma tissues for TCIRG1 overexpression) and negative controls (antibody diluent only) to validate staining specificity .

For multi-color IHC detecting TCIRG1 alongside other markers, sequential staining protocols may be necessary to avoid antibody cross-reactivity, particularly when studying TCIRG1 in immune cell populations.

What are the recommended cell models for studying TCIRG1 function in vitro?

Based on the available research data, several cell models have been validated for TCIRG1 functional studies:

Cell TypeApplicationsTCIRG1 ExpressionNotable Characteristics
SKOV-3Western blot, functional studiesDetectableOvarian cancer cell line with validated TCIRG1 expression
HepG2Western blot, functional studiesDetectableLiver cancer cell line with moderate TCIRG1 expression
JurkatWestern blot, immune function studiesDetectableT-cell line suitable for immune regulatory studies of TCIRG1
DaudiWestern blot, immune function studiesDetectableB-cell line for studying TCIRG1 in B lymphocytes
Peripheral blood mononuclear cellsWestern blot, primary cell studiesVariablePrimary cells used in congenital neutropenia studies

When selecting cell models, consider the specific TCIRG1 isoform expression patterns in different cell types. For functional studies involving TCIRG1 knockdown, ccRCC cell lines have been successfully used to demonstrate the role of TCIRG1 in cancer cell migration . For studies focused on TCIRG1's role in immune regulation, primary immune cells or appropriate immune cell lines should be selected based on the specific immune cell population of interest.

How can I address non-specific binding issues when using TCIRG1 antibodies?

Non-specific binding is a common challenge with TCIRG1 antibodies, particularly in applications like Western blotting and immunohistochemistry. To address this issue:

  • Blocking optimization: Extended blocking (2 hours at room temperature) with 5% non-fat milk in TBST for Western blots, or 10% normal serum from the same species as the secondary antibody for IHC/IF, can significantly reduce non-specific binding.

  • Antibody dilution: Titrate your antibody carefully, as both insufficient and excessive concentrations can contribute to non-specific binding. Start with the manufacturer's recommended dilution and adjust based on signal-to-noise ratio .

  • Washing stringency: Increase the stringency of washing steps by adding 0.1-0.3% Tween-20 to washing buffers and extending washing durations. For particularly problematic samples, consider including a high-salt (500mM NaCl) wash step.

  • Alternative antibodies: If persistent non-specific binding occurs, compare results using antibodies targeting different epitopes of TCIRG1. For example, if an antibody against the N-terminal domain (AA 1-130) shows non-specific binding, try an antibody targeting AA 121-220 .

  • Validation controls: Always include appropriate negative controls, such as tissues or cells known to lack TCIRG1 expression, or ideally, TCIRG1 knockout/knockdown samples, to identify true non-specific binding patterns.

How should I interpret discrepancies in TCIRG1 detection between different antibodies or techniques?

Discrepancies in TCIRG1 detection are often attributable to several factors that require careful interpretation:

  • Isoform specificity: TCIRG1 has multiple isoforms, including TCIRG1-isoa and TCIRG1-isob, which lack the first 5 exons of the longer variant. Different antibodies may preferentially detect specific isoforms, leading to apparent discrepancies in expression levels .

  • Epitope accessibility: The conformation of TCIRG1 differs between applications - denatured in Western blots versus native in immunoprecipitation or flow cytometry. Antibodies targeting conformational epitopes may show different results across techniques.

  • Post-translational modifications: Modifications may mask epitopes in certain contexts. For example, studies have shown detection of reduced amounts of ~45kDa TCIRG1 product fragments in individuals with mutations, suggesting possible altered processing or stability .

  • Technical sensitivity: Western blotting typically offers higher sensitivity for low-abundance proteins compared to IHC. When discrepancies occur, quantitative PCR for TCIRG1 mRNA can provide complementary data to resolve conflicts.

When faced with discrepancies, employ multiple antibodies targeting different epitopes and use complementary techniques (e.g., RNA-seq or qPCR) to validate protein expression findings. Document the specific antibody used, detection method, and sample preparation for accurate interpretation of results across studies.

What considerations should I take into account when using TCIRG1 antibodies for co-localization studies?

Co-localization studies involving TCIRG1 require specific considerations due to its subcellular distribution and multiple isoforms:

  • Fixation effects: TCIRG1's membrane association makes it sensitive to fixation artifacts. Compare paraformaldehyde fixation (2-4%) with methanol fixation to determine optimal epitope preservation while maintaining cellular architecture.

  • Permeabilization optimization: As a membrane protein component of V-ATPase, TCIRG1 requires balanced permeabilization - sufficient for antibody access but not excessive to disrupt membrane structures. Try 0.1% Triton X-100 or 0.05% saponin, adjusting based on signal quality.

  • Sequential staining: For co-localization with other markers, especially when antibodies are raised in the same species, sequential staining protocols with appropriate blocking steps between primary antibodies are recommended.

  • Spectral considerations: Choose fluorophores with minimal spectral overlap when designing multi-color immunofluorescence experiments, particularly important when co-localizing TCIRG1 with other V-ATPase components.

  • Super-resolution techniques: Consider super-resolution microscopy techniques (STED, STORM, SIM) for precise subcellular localization of TCIRG1, especially when studying its distribution relative to endosomal/lysosomal markers.

When interpreting co-localization results, quantify overlap using appropriate coefficients (Pearson's, Mander's) rather than relying on visual assessment alone. This provides more objective assessment of TCIRG1's spatial relationship with other proteins of interest.

How can TCIRG1 antibodies be utilized to study its role in congenital neutropenia?

Investigating TCIRG1's role in congenital neutropenia requires specialized approaches using antibodies:

  • Genetic-protein correlation: Use Western blot analysis with TCIRG1 antibodies to assess protein expression in neutropenic patients with known TCIRG1 mutations. Previous studies have shown reduced levels of ~45kDa TCIRG1 product fragment in affected individuals carrying mutations like ARG736SER, compared to healthy controls .

  • Isoform-specific detection: Employ antibodies with known isoform specificity to distinguish between TCIRG1-isoa and TCIRG1-isob expression, as mutations may differentially affect these isoforms. A polyclonal antibody developed against the N-terminal cytoplasmic domain can detect the shorter TCIRG1-isob, while monoclonal antibodies against partial recombinant TCIRG1 isoa 121aa-220aa detect distinct patterns .

  • Functional assays: Combine antibody-based detection with functional assays measuring neutrophil development and survival in patient samples. Correlation between TCIRG1 protein levels and functional outcomes can provide mechanistic insights.

  • Tissue distribution analysis: Use immunohistochemistry with validated TCIRG1 antibodies to compare TCIRG1 distribution in bone marrow samples from patients versus controls, focusing on granulocyte precursor populations.

  • Protein interaction studies: Employ co-immunoprecipitation with TCIRG1 antibodies to identify altered protein interactions in neutropenic patients that might explain the link between TCIRG1 mutations and neutrophil development.

This comprehensive approach can help elucidate how TCIRG1 mutations contribute to the pathogenesis of congenital neutropenia, potentially revealing novel therapeutic targets.

What methodologies are recommended for studying TCIRG1's role in tumor immunity and cancer progression?

Research into TCIRG1's oncogenic functions can be approached through several antibody-dependent methodologies:

  • Expression profiling: Use validated TCIRG1 antibodies for IHC analysis of tissue microarrays to correlate TCIRG1 expression with clinical outcomes across cancer types. TCIRG1 overexpression has been documented in several cancers, particularly ccRCC, where it predicts unfavorable clinical outcomes .

  • Immune infiltrate characterization: Combine TCIRG1 IHC with immune cell markers to analyze relationships between TCIRG1 expression and tumor-infiltrating lymphocytes. TCIRG1 has shown strong associations with CD8+ T-cell, Treg, and CD4+ T-cell infiltration patterns in tumors .

  • Cell line models: Implement TCIRG1 knockdown/overexpression in cancer cell lines followed by Western blot confirmation of altered expression, enabling functional studies of migration and invasion. Experimental evidence indicates that TCIRG1 knockdown inhibits migration of ccRCC cells .

  • Therapeutic response prediction: Correlate TCIRG1 expression levels (detected by IHC or Western blot) with response to immunotherapy treatments. TCIRG1 levels have been associated with Tumor Immune Dysfunction and Exclusion (TIDE) scores, suggesting potential value in predicting immunotherapy response .

  • Mechanistic pathway analysis: Use phospho-specific antibodies alongside TCIRG1 antibodies to investigate downstream signaling pathways activated in TCIRG1-overexpressing tumors, potentially revealing therapeutic targets.

This multi-faceted approach allows for comprehensive investigation of TCIRG1's roles in both tumor progression and immune evasion mechanisms.

How can I design effective validation strategies for TCIRG1 antibody specificity?

Rigorous validation of TCIRG1 antibody specificity is essential for confident data interpretation. A comprehensive validation strategy should include:

  • Genetic knockout/knockdown controls: The gold standard for antibody validation involves testing the antibody in TCIRG1 knockout or knockdown models. Compare Western blot or IHC signals between wild-type and TCIRG1-depleted samples to confirm specificity.

  • Peptide competition assays: Pre-incubate the TCIRG1 antibody with excess immunizing peptide before application to samples. Specific binding should be competitively inhibited, while non-specific binding will persist.

  • Multiple antibody concordance: Test at least two antibodies targeting different epitopes of TCIRG1 (e.g., N-terminal domain versus middle region) and compare detection patterns. Consistent results across different antibodies increase confidence in specificity .

  • Cross-species reactivity assessment: If the antibody claims cross-reactivity with multiple species, test samples from each species to confirm consistent detection of the appropriately sized protein band or expected localization pattern .

  • Correlation with orthogonal methods: Compare protein detection results with mRNA expression data from qPCR or RNA-seq to confirm that protein and transcript levels correlate in the experimental system.

  • Mass spectrometry validation: For ultimate confirmation, perform immunoprecipitation with the TCIRG1 antibody followed by mass spectrometry analysis to verify the identity of the captured proteins.

Document these validation steps thoroughly when publishing research using TCIRG1 antibodies, as this improves reproducibility and confidence in the findings.

What methodological approaches are recommended for studying TCIRG1 isoform-specific functions?

Investigating isoform-specific functions of TCIRG1 requires specialized methodological approaches:

  • Isoform-specific detection: Select antibodies with validated specificity for either TCIRG1-isoa or TCIRG1-isob. For instance, antibodies targeting the first 5 exons will detect only the longer TCIRG1-isoa, while those targeting shared regions will detect both isoforms but require careful band size interpretation in Western blots .

  • Isoform-selective knockdown: Design siRNA or shRNA constructs targeting unique regions of each isoform, followed by validation of isoform-specific reduction using appropriate antibodies. This allows for functional studies distinguishing the roles of different TCIRG1 variants.

  • Expression systems: For overexpression studies, construct expression vectors containing the specific TCIRG1 isoform of interest, and confirm expression using isoform-specific antibodies before proceeding to functional analyses.

  • Tissue distribution mapping: Use isoform-specific antibodies to map the differential distribution of TCIRG1 isoforms across tissues, particularly focusing on hematopoietic and immune cell populations relevant to neutropenia and immune regulation .

  • Co-immunoprecipitation analyses: Employ isoform-specific antibodies for co-immunoprecipitation studies to identify potential isoform-specific protein interaction partners that might explain their distinct biological functions.

These approaches allow for precise delineation of the distinctive roles of TCIRG1 isoforms in cellular processes, disease pathogenesis, and potential therapeutic targeting.

How can I integrate TCIRG1 antibody-based techniques with genomic and transcriptomic analyses for comprehensive studies?

Multi-omics integration with TCIRG1 antibody-based techniques enables deeper mechanistic insights:

  • Genotype-phenotype correlations: Pair TCIRG1 antibody detection (by Western blot or IHC) with genomic sequencing data to correlate specific TCIRG1 mutations or variants with protein expression levels and localization patterns. This approach has successfully identified association between TCIRG1 mutations and congenital neutropenia .

  • Transcriptome-proteome integration: Correlate TCIRG1 mRNA expression from RNA-seq or microarray data with protein levels detected by TCIRG1 antibodies to identify potential post-transcriptional regulatory mechanisms or protein stability factors.

  • Epigenetic regulation: Combine chromatin immunoprecipitation (ChIP) data on TCIRG1 promoter regions with antibody-based TCIRG1 protein detection to investigate epigenetic mechanisms controlling TCIRG1 expression. Studies have shown relationships between TCIRG1 expression and DNA methylation status .

  • Pathway analysis validation: Use antibody-based techniques to validate key protein interactions or modifications predicted from computational analyses of transcriptomic data, strengthening the biological relevance of in silico findings.

  • Single-cell integration: Pair single-cell RNA-seq data with single-cell protein detection methods (such as mass cytometry or imaging mass cytometry using TCIRG1 antibodies) to resolve cell-type specific expression patterns at both RNA and protein levels.

This integrated approach has been productive in cancer research, where TCIRG1 antibody-based detection combined with genomic analyses has revealed associations with cancer stemness indicators and somatic mutation patterns (particularly PBRM1 and BAP1 mutations in renal cancer) .

What considerations should be taken into account when developing TCIRG1-targeted therapeutic approaches?

Developing therapeutic strategies targeting TCIRG1 requires careful antibody-based validation steps:

  • Expression profiling: Use validated TCIRG1 antibodies to comprehensively map expression across healthy tissues to predict potential on-target toxicities of TCIRG1-directed therapies. Note TCIRG1's expression in heart, liver, kidney, lung, and pancreas .

  • Function validation: Before therapeutic targeting, confirm the causal role of TCIRG1 in disease pathogenesis through antibody-based detection following genetic manipulation in relevant model systems. Experimental evidence shows that TCIRG1 knockdown inhibits migration of cancer cells, supporting its potential as a therapeutic target .

  • Biomarker development: Develop reliable IHC protocols with TCIRG1 antibodies for potential use as companion diagnostics to identify patients likely to benefit from TCIRG1-targeted therapies, particularly in cancers where TCIRG1 overexpression correlates with poor outcomes .

  • Isoform selectivity: Determine whether therapeutic approaches should target all TCIRG1 isoforms or specific variants based on their differential expression in disease states. This requires isoform-specific antibodies for validation studies .

  • Immune impact assessment: Given TCIRG1's association with immune cell infiltration in tumors, use multiparameter immunofluorescence with TCIRG1 and immune cell marker antibodies to predict how TCIRG1-targeted therapies might alter tumor immunology .

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.