Target Function: ABCB7 exports iron-sulfur clusters from mitochondria to the cytosol, critical for cytosolic iron-sulfur protein assembly and iron homeostasis .
Disease Relevance: Mutations in ABCB7 cause X-linked sideroblastic anemia with ataxia (XLSA/A) .
Validation: Antibodies are validated in Western blot (WB), immunohistochemistry (IHC), and immunoprecipitation (IP) across human, mouse, and rat samples .
While ABC1K7 antibodies are not commercially available, research on Arabidopsis thaliana reveals:
Function: ABC1K7 regulates abscisic acid (ABA) signaling, oxidative stress responses, and chloroplast iron distribution .
Genetic Interactions: Knockout mutants (abc1k7) exhibit hypersensitivity to ABA, accelerated leaf senescence, and impaired stomatal closure under stress .
Expression: ABC1K7 is upregulated by ABA, cadmium, and osmotic stress, indicating roles in abiotic stress adaptation .
Nomenclature Confusion: ABC1K7 is a plant-specific kinase, whereas ABCB7 is a human mitochondrial transporter.
Commercial Availability: Antibodies targeting ABC1K7 may not exist due to limited demand or technical challenges in plant protein antibody development.
Research Focus: Current studies on ABC1K7 focus on genetic and functional analyses rather than antibody-based detection .
Verify Target Identity: Confirm whether the intended target is ABC1K7 (plant kinase) or ABCB7 (human transporter).
Alternative Strategies: For plant studies, consider generating custom polyclonal antibodies using peptide sequences from ABC1K7 (e.g., residues 691–740 in homologous regions) .
Explore Cross-Reactivity: Assess whether existing ABCB7 antibodies cross-react with ABC1K7 orthologs in model organisms (unverified in current literature).
ABCA7 is an ATP-binding cassette (ABC) transporter that has been identified as an important risk gene for Alzheimer's disease (AD). Since its identification, ABCA7 has been extensively researched for its role in AD pathophysiology. The gene encodes a protein involved in lipid metabolism, phagocytosis, and amyloid deposition processes, all of which are relevant to AD pathogenesis .
Notably, ABCA7 has been shown to have an even stronger effect size in AD than APOE in African Americans, highlighting its significant role in disease risk across different populations . The protein belongs to the same structural family as ABCA1 and ABCA4, sharing 52.8% and 48.2% sequence homology with these transporters, respectively . Functionally, ABCA7 is closely related to ABCA1, as both recognize and translocate cholesterol and phospholipids .
Multiple genetic variants of ABCA7 have been associated with increased risk for AD. These include both common variants identified through genome-wide association studies (GWAS) and rare variants with larger effect sizes .
Research has shown:
Premature termination codon (PTC) variants in ABCA7 are enriched in AD patients with odds ratios ranging from 1.4 to 5
The lowest odds ratio was observed in African American cohorts, influenced by a common 44 bp deletion (rs142076058, p.R578fs)
In large burden analyses and case-control meta-analyses in Caucasian populations, odds ratios for ABCA7 PTC variants were determined to be 1.7 and 2.6, respectively
Frequencies of these variants in AD patients range from 0.39% to 4.4%
Multiple GWAS sentinel SNPs in ABCA7 have been associated with AD endophenotypes, particularly increased amyloid pathology, which has been replicated in several studies. This suggests that ABCA7 dysfunction may play a role in the amyloid pathway of AD .
Studies have utilized both loss-of-function and gain-of-function approaches to understand ABCA7's role in AD. Specifically:
Abca7 knockout models have demonstrated increased amyloid-β (Aβ) load, supporting ABCA7's role in Aβ metabolism
Conversely, overexpression of ABCA7 in mouse models has led to reduction of Aβ, further confirming its involvement in amyloid processing
Certain polymorphisms have been associated with plaque formation in patients, which correlated with increased expression of ABCA7, suggesting a compensatory mechanism in response to elevated Aβ load
These experimental approaches have functionally linked ABCA7 to cholesterol metabolism and phagocytosis, processes that may influence Aβ distribution and degradation .
ABCA7 has been structurally characterized primarily through homology modeling based on related ABC transporters. The protein is large, consisting of 2,146 amino acids, comparable to the 2,261 and 2,273 amino acids of ABCA1 and ABCA4, respectively .
Recent advances include:
Development of homology models based on the recently reported cryogenic-electron microscopy (cryo-EM) structures of ABCA1 and ABCA4
Structural analysis revealing that ABCA7 is a "common" ABCA transporter from a structural perspective
Identification of important structural elements shared across ABCA family members, as marked in comparative sequence analyses
These structural insights are crucial for understanding the molecular mechanisms of ABCA7 function and for developing potential therapeutic interventions targeting this protein.
Docking of novel, diverse, and potent pan-ABC transporter inhibitors to ABCA7 homology models
Exploration of a proposed "multitarget binding site" that might allow for targeting of multiple ABC transporters including ABCA7
Screening of compounds with activity across multiple ABC transporters
Below is a table of compounds that have been identified as pan-ABC transporter inhibitors that might have relevance for ABCA7 research:
Compound No. | Original Name | Molecular Weight | Calc Log P | Targeted ABC Transporters |
---|---|---|---|---|
6 | benzbromarone | 424.09 | 5.55 | B1, B11, C1–6, G2 |
7 | imatinib | 493.62 | 4.38 | A3, B1, B11, C1, C10, G2 |
8 | quercetin | 302.24 | 2.16 | B1, C1–2, C4–5, C11, G2, G6 |
9 | verapamil | 454.61 | 5.04 | A8, B1, B4–5, B11, C1, C4, C11, G2 |
10 | verlukast | 515.08 | 5.67 | A8, B4, B11, C1–C5, C10–C11, G2 |
Additional focused pan-ABC transporter inhibitors with potential relevance to ABCA7 research include compounds based on various chemical scaffolds such as quinoline/1,2,4-oxadiazole, quinazoline, and pyrrolopyrimidine derivatives .
ABCA7 variants show different distributions and effects across ethnic populations. Research has found:
ABCA7 SNPs contribute to AD pathophysiology in Caucasian, African American, and East Asian cohorts
Most associations have been found in Caucasian cohorts, followed by East Asian populations
This distribution likely reflects research disparities rather than biological differences, as European ancestry is overrepresented in large GWAS studies
In African American populations, ABCA7 has shown a stronger effect size in AD than APOE, the most well-established genetic risk factor for the disease
The 44 bp deletion (rs142076058, p.R578fs) is common in African Americans (found in up to 21.7% of AD patients) but has a weaker risk-increasing effect compared to other variants
These findings underscore the importance of expanding research beyond European populations to fully understand ABCA7's role in AD risk across different ethnicities .
Recent advances in antibody design combine experimental selection with computational modeling to achieve customized specificity profiles. A key approach involves:
Phage display experiments for antibody library selection
High-throughput sequencing of selected antibodies
Computational modeling to identify different binding modes
Design of novel antibodies with customized specificity profiles
This integrated approach allows researchers to:
Generate antibodies that specifically bind to particular target ligands
Create antibodies with cross-specificity for multiple target ligands
Mitigate experimental artifacts and biases in selection experiments
One particularly effective methodology uses a minimal antibody library based on a single naïve human V domain with systematic variation of four consecutive positions in the third complementarity determining region (CDR3). This approach allows for high-coverage of the library composition by high-throughput sequencing while still containing antibodies that bind specifically to diverse ligands .
Computational models play a crucial role in predicting and designing antibody specificity, particularly when very similar epitopes need to be discriminated. These models:
Identify different binding modes associated with particular ligands
Disentangle these modes even when they involve chemically similar ligands
Enable the design of antibodies with customized specificity profiles
The computational approach involves optimizing energy functions associated with each binding mode. For cross-specific sequences, the functions associated with the desired ligands are jointly minimized. For specific sequences, the function associated with the desired ligand is minimized while those associated with undesired ligands are maximized .
This biophysics-informed modeling, combined with extensive selection experiments, offers broad applicability beyond antibodies, providing a powerful toolset for designing proteins with desired physical properties .
Research on SARS-CoV-2 variants, particularly B.1.1.7 and B.1.351, has revealed important challenges in understanding antibody resistance:
B.1.1.7 is refractory to neutralization by most monoclonal antibodies targeting the N-terminal domain of the spike protein and is relatively resistant to some monoclonal antibodies against the receptor-binding domain
B.1.351 is not only resistant to neutralization by N-terminal domain antibodies but also by multiple individual monoclonal antibodies against the receptor-binding motif, primarily due to the E484K substitution
Compared to wild-type SARS-CoV-2, B.1.351 shows marked resistance to neutralization by convalescent plasma (9.4-fold) and sera from vaccinated individuals (10.3-12.4-fold)
These findings highlight the challenges for monoclonal antibody therapies and raise concerns about the protective efficacy of current vaccines against emerging variants .
While the search results don't directly connect ABCA7 research with antibody development, several methodological parallels can be drawn:
Structural insights: Just as homology modeling of ABCA7 enables understanding of its function and potential drug binding sites , structural biology approaches can inform antibody design by revealing epitope structures and binding interfaces.
Population variations: Understanding how ABCA7 variants affect disease risk across different populations parallels the need to account for viral variant emergence when developing antibody therapeutics against pathogens like SARS-CoV-2 .
Computational approaches: The computational methods used to predict ABCA7-drug interactions share similarities with those used to design antibodies with custom specificity profiles , suggesting potential for cross-disciplinary methodological exchange.
Both research areas rely on rigorous experimental validation strategies:
Genetic association studies: For ABCA7, case-control studies and meta-analyses establish links between variants and disease risk , while antibody research requires validation of binding profiles through selection experiments .
Functional assays: ABCA7 research uses knockout and overexpression models to understand protein function , similar to how antibody research requires functional assays to confirm binding specificity and neutralization capacity .
Structural validation: Homology models of ABCA7 require validation , just as computationally designed antibodies need experimental confirmation of their predicted binding properties .
Several promising directions for integrated research emerge from the search results:
Development of specific antibodies against ABCA7: Creating highly specific antibodies against ABCA7 using the computational design approaches described for custom antibody specificity could provide valuable research tools for studying ABCA7's role in AD.
Therapeutic targeting: The identification of pan-ABC transporter inhibitors could be complemented by development of specific antibodies or antibody-drug conjugates targeting ABCA7 in AD.
Diagnostic applications: Antibodies with custom specificity profiles could be developed to detect specific variants or conformational states of ABCA7, potentially enabling early detection of AD risk.