The ACA1 Antibody is a mouse-derived monoclonal antibody designed to detect ACA1 (Calcium-Transporting ATPase), a magnesium-dependent enzyme in Arabidopsis thaliana. ACA1 catalyzes ATP hydrolysis coupled with calcium transport, playing a critical role in cellular calcium homeostasis .
| Attribute | Detail |
|---|---|
| Target Protein | ACA1 (Calcium-Transporting ATPase; EC 3.6.3.8; UniProt ID: F4HUS8) |
| Host Species | Mus musculus (Mouse) |
| Reactivity | Specific to A. thaliana (Arabidopsis thaliana) |
| Clone | MO1393FC |
| Format | Liquid or lyophilized |
| Purity | >90% (SDS-PAGE confirmed) |
| Storage | 4°C (short-term), -20°C (long-term) |
Western Blot: Detects ACA1 protein in lysates from A. thaliana.
ELISA: Quantitative or qualitative analysis of ACA1 expression.
| Application | Optimal Dilution | Notes |
|---|---|---|
| ELISA | 1:1000–1:3000 | Requires optimization by user |
| WB | N/A (varies by sample) | Requires denaturing conditions |
While ACA1 Antibody is primarily a research tool, its utility is constrained by limited published studies. Key insights include:
Target Specificity: Binds exclusively to ACA1 in A. thaliana .
Cross-Reactivity: No reported cross-reactivity with human or other non-plant proteins .
Use Case: Employed in plant physiology studies to investigate calcium signaling pathways.
Note: No clinical relevance exists for this antibody, as ACA1 is not implicated in human diseases.
To avoid ambiguity, anticentromere antibodies (ACA) are distinct autoantibodies linked to human autoimmune diseases (e.g., limited cutaneous systemic sclerosis, primary biliary cirrhosis) . These target centromere proteins (e.g., CENP-B) and are unrelated to ACA1 .
ACAP1, also known as CENTB1 (Centaurin beta1), is a protein that negatively regulates ARF6 function and interacts with Akt to form protein complexes that serve as adaptors for endocytic recycling of intracellular cargos including transferrin receptor, cellubrevin, integrin β1, and Glut4 . ACAP1 is primarily expressed in lymphocytes and has been shown to be necessary for their normal function. Research has demonstrated that ACAP1 expression is regulated by promoter DNA methylation and the transcription factor SPI1 .
The protein has gained significant attention in immunology research because ACAP1 levels positively correlate with tumor-infiltrating lymphocyte (TIL) levels across multiple solid cancer types. Additionally, ACAP1 deficiency has been associated with poor prognosis and reduced immunotherapeutic response in multiple cancer types, making it a potential biomarker for immunotherapy response prediction .
ACAP1 functions primarily through its role in endocytic recycling, which is critical for lymphocyte activation, proliferation, and effector functions. In lymphocytes, proper recycling of receptors and signaling molecules is essential for maintaining immunological synapse formation and sustained signaling. ACAP1 mediates these processes by functioning as an adaptor protein that interacts with multiple cellular components .
Beyond endocytic recycling, ACAP1 interacts with NOD1/NOD2 and inhibits NF-κB activation in response to bacterial component stimulation in intestinal epithelial cells. Its expression can be stimulated by pro-inflammatory cytokines and is significantly upregulated in ulcerative colitis, particularly in inflammatory infiltrates . This suggests that ACAP1 plays multiple roles in immune regulation beyond its canonical function in endocytic recycling.
Detection of ACAP1 in research samples typically involves antibody-based techniques. For protein-level detection, Western blotting using specific anti-ACAP1 antibodies is commonly employed, while qPCR is used for mRNA expression analysis . In tissue samples, immunohistochemistry (IHC) using validated ACAP1 antibodies can reveal expression patterns and localization.
For more sophisticated analyses, researchers utilize single-cell RNA sequencing to examine ACAP1 expression at the cellular level, particularly in heterogeneous tissue samples like tumors. ChIP-PCR has been successfully used to investigate transcriptional regulation of ACAP1, particularly the role of SPI1 as a transcription factor . When selecting antibodies for ACAP1 detection, researchers should prioritize recombinant monoclonal antibodies, which offer higher specificity and reproducibility compared to polyclonal alternatives.
ACAP1 deficiency has been linked to poor immunotherapy response across multiple cancer types, making ACAP1 antibodies valuable tools for studying resistance mechanisms. Researchers can use ACAP1 antibodies to stratify patient samples based on ACAP1 expression levels and correlate these with response to immune checkpoint inhibitors . This approach can reveal whether ACAP1 deficiency is a driver or merely a marker of resistance.
To thoroughly investigate resistance mechanisms, researchers can combine ACAP1 antibody staining with multiplex immunofluorescence techniques to simultaneously assess ACAP1 expression and the presence of various immune cell populations. This allows for spatial analysis of the tumor microenvironment, revealing how ACAP1 expression in lymphocytes correlates with their infiltration patterns and functional states . Additionally, co-immunoprecipitation experiments using ACAP1 antibodies can identify interacting partners that might contribute to resistance mechanisms.
ACAP1 expression positively correlates with TIL levels across multiple solid cancer types, suggesting a mechanistic relationship between ACAP1 and lymphocyte infiltration into tumors . This correlation has been validated across eight different algorithms used to estimate immune cell infiltration, including TIMER, CIBERSORT, xCELL, and MCPCOUNTER.
The mechanistic basis for this relationship likely stems from ACAP1's role in lymphocyte function. ACAP1 regulates endocytic recycling of key receptors and adhesion molecules that lymphocytes need for proper migration and tumor infiltration. When ACAP1 is deficient, lymphocytes may have impaired ability to respond to chemotactic signals, adhere to endothelium, or migrate through tissue, resulting in reduced tumor infiltration . This creates a "cold" tumor immune microenvironment, which is associated with poor response to immunotherapy.
When designing antibodies to target specific ACAP1 epitopes, researchers should consider rational design approaches that optimize binding specificity and affinity. This process begins with epitope selection, ideally targeting functionally significant regions of ACAP1 that are accessible in the protein's native conformation .
For disordered regions within ACAP1, researchers can adopt complementary peptide approaches, where peptides complementary to the target epitope are designed and grafted onto an antibody scaffold, particularly within the CDR3 loop . This method has been successfully used to create single-domain antibodies targeting disordered proteins involved in neurodegenerative diseases and could be applied to ACAP1.
Stability testing is essential during antibody design, as modifications to recognize specific epitopes might compromise structural integrity. Researchers should verify that designed antibodies maintain native-like structure through circular dichroism spectroscopy and assess binding affinity through techniques like ELISA . Advanced techniques such as yeast or phage display can be employed to further optimize binding characteristics before proceeding to functional studies.
Validating ACAP1 antibodies requires a multi-faceted approach to ensure specificity, sensitivity, and reproducibility. First, researchers should verify antibody specificity using positive and negative controls, including ACAP1 knockout or knockdown cells alongside wild-type controls . Western blotting with recombinant ACAP1 protein can confirm that the antibody recognizes the correct target.
For immunohistochemistry applications, validation should include testing on tissues known to express ACAP1 (lymphoid tissues) and those with minimal expression. Cross-reactivity with related proteins, particularly other ACAP family members, should be assessed to ensure specificity. Researchers should also validate lot-to-lot consistency when using commercial antibodies, as variation can significantly impact experimental results.
For quantitative applications, establishing a standard curve using recombinant ACAP1 protein is essential to determine the linear range of detection. Additionally, validation across multiple experimental platforms (Western blot, IHC, flow cytometry) ensures the antibody performs consistently across different applications. Finally, confirming that the antibody recognizes the native protein conformation is crucial for applications like immunoprecipitation or flow cytometry.
To effectively use ACAP1 antibodies in immunotherapy response studies, researchers should implement a comprehensive experimental design that addresses both mechanistic questions and clinical correlations. A typical approach would begin with stratifying patient samples based on ACAP1 expression levels using validated antibodies in IHC or multiplex immunofluorescence .
For mechanistic studies, researchers can isolate TILs from patient samples and analyze ACAP1 expression in different lymphocyte subsets using flow cytometry with ACAP1 antibodies. This can be correlated with functional assays measuring cytokine production, proliferation, and cytotoxicity to determine how ACAP1 levels affect lymphocyte function .
Longitudinal sampling before and during immunotherapy treatment allows researchers to track changes in ACAP1 expression over time and correlate these with treatment response. To establish causality rather than mere correlation, researchers should complement human studies with animal models where ACAP1 is genetically manipulated, followed by treatment with immune checkpoint inhibitors to directly assess the impact of ACAP1 deficiency on response .
For multiplex imaging applications, researchers have several conjugation strategies available for ACAP1 antibodies. Direct conjugation with fluorophores is the most straightforward approach, but careful selection of fluorophores is essential to avoid spectral overlap when performing multiplexed analyses .
Metal isotope labeling for mass cytometry (CyTOF) offers superior multiplexing capabilities with minimal signal overlap. This approach allows simultaneous detection of ACAP1 along with dozens of other markers, providing comprehensive characterization of the immune landscape in relation to ACAP1 expression . When using this method, researchers should ensure that antibody function is not compromised by the conjugation process by validating with pre- and post-conjugation binding assays.
Oligonucleotide conjugation for techniques like CODEX or Immuno-SABER provides an alternative multiplexing strategy. The conjugation-ready format of many commercial antibodies makes them ideal for these applications . Regardless of the conjugation method chosen, researchers should optimize antibody concentration to achieve sufficient signal while minimizing background, and include appropriate controls to account for potential artifacts introduced by the conjugation process.
When confronted with contradictory findings regarding ACAP1 expression across cancer types, researchers should first consider methodological differences that might explain the discrepancies. Different antibodies, detection methods, or scoring systems can lead to apparently contradictory results even when the underlying biology is consistent .
Tissue heterogeneity is a common source of contradictory findings, as ACAP1 expression may vary significantly within a tumor. Single-cell RNA sequencing data should be leveraged to resolve such contradictions by examining expression patterns at cellular resolution . Additionally, researchers should distinguish between ACAP1 expression in tumor cells versus infiltrating lymphocytes, as these distinct populations might show different patterns.
Cox proportional hazards regression allows for multivariate analysis, which is essential to determine whether ACAP1 expression is an independent predictor of response or is confounded by other clinical or molecular variables. Researchers should include relevant covariates such as PD-L1 expression, tumor mutational burden, and baseline clinical characteristics .
For continuous measures of response, such as tumor shrinkage, linear regression models or generalized linear models may be more appropriate. To determine optimal cutoff values for ACAP1 expression, researchers can employ receiver operating characteristic (ROC) curve analysis, which balances sensitivity and specificity. Finally, machine learning approaches can be valuable for integrating ACAP1 expression with other biomarkers to create composite predictors of immunotherapy response.
Differentiating between correlation and causation in ACAP1 studies requires thoughtful experimental design and careful interpretation of results. While observational studies may identify correlations between ACAP1 expression and immune parameters, establishing causation requires intervention studies .
Genetic manipulation approaches, such as CRISPR-Cas9-mediated knockout or knockdown of ACAP1 in lymphocytes, followed by functional assays, can establish whether ACAP1 is causally related to observed phenotypes. Similarly, overexpression studies can determine whether restoring ACAP1 in deficient cells rescues functional defects .
Temporal relationships are important for establishing causation; therefore, time-course experiments tracking ACAP1 expression before and after immune activation can help determine whether changes in ACAP1 precede functional changes. Dose-response relationships, where varying levels of ACAP1 expression correspond to proportional changes in immune function, provide additional evidence for causation.
Animal models offer valuable systems for testing causal relationships in vivo. Conditional knockout models, where ACAP1 can be deleted specifically in lymphocytes or other cell types, allow researchers to determine the cell type-specific contributions of ACAP1 to immune function and immunotherapy response .