APOBEC3C Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to APOBEC3C and Its Antibody

APOBEC3C (A3C) is a single-domain cytidine deaminase that restricts viral replication by introducing mutations in viral DNA. It also plays roles in cancer mutagenesis and immune modulation . The APOBEC3C antibody enables detection and functional analysis of this protein in experimental settings, with applications ranging from basic virology to oncology research.

Antiviral Mechanisms

  • APOBEC3C inhibits HIV-1 and murine leukemia virus (MLV) by deaminating viral DNA, causing G→A mutations. Its activity is partially counteracted by HIV-1 Vif protein .

  • Synthetic tandem-domain APOBEC3C constructs show enhanced antiviral activity and resistance to Vif, providing insights into evolutionary adaptations of restriction factors .

Cancer Biology

  • Genomic Mutagenesis: APOBEC3C overexpression in pancreatic ductal adenocarcinoma (PDAC) correlates with C>X mutations, kataegis clusters, and poor prognosis .

  • Tumor Microregulation: In gliomas, APOBEC3C modulates the tumor microenvironment by influencing CD4+/CD8+ T-cell infiltration and cancer stemness .

Cellular Localization and Stress Response

  • APOBEC3C localizes to nucleoli under basal conditions but redistributes upon genotoxic stress (e.g., etoposide exposure). This behavior is regulated by p53 and linked to DNA damage responses .

Clinical and Experimental Relevance

Study FocusKey InsightCitation
Antiviral ActivityAPOBEC3C targets TC/CC motifs, differing from A3G (CC) and A3F (TC)
Cancer Mutational BurdenHigh APOBEC3C expression in PDAC predicts shorter survival
Immune ModulationAPOBEC3C enhances CTL infiltration but may aid viral immune escape

Technical Considerations

  • Cross-Reactivity: APOBEC3C shares sequence homology with other APOBEC3 family members (e.g., A3A, A3B), necessitating rigorous validation to ensure antibody specificity .

  • Functional Assays: Combining APOBEC3C antibodies with CRISPR knockouts or deaminase-inactive mutants (e.g., C97S/C100S) helps distinguish catalytic vs. non-catalytic roles .

Future Directions

  • Therapeutic Targeting: APOBEC3C’s dual role in antiviral defense and cancer mutagenesis makes it a candidate for immune-oncology therapies .

  • Biomarker Development: Quantifying APOBEC3C expression in tumors could guide immunotherapy strategies, particularly in PDAC and gliomas .

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze-thaw cycles.
Lead Time
We typically dispatch products within 1-3 business days of receiving your order. Delivery time may vary based on your purchase method and location. For specific delivery times, please consult your local distributors.
Synonyms
A3C antibody; ABC3C_HUMAN antibody; APOBEC1 like antibody; APOBEC1-like antibody; APOBEC1L antibody; APOBEC3C antibody; Apolipoprotein B mRNA editing enzyme catalytic polypeptide like 3C antibody; ARDC2 antibody; ARDC4 antibody; ARP5 antibody; bK150C2.3 antibody; DNA dC >dU editing enzyme APOBEC 3C antibody; MGC19485 antibody; PBI antibody; Phorbolin I antibody; Phorbolin I protein antibody; Probable DNA dC dU editing enzyme APOBEC 3C antibody; Probable DNA dC->dU-editing enzyme APOBEC-3C antibody
Target Names
APOBEC3C
Uniprot No.

Target Background

Function
APOBEC3C is a DNA deaminase (cytidine deaminase) that acts as an inhibitor of retrovirus replication and retrotransposon mobility. Its mechanism involves both deaminase-dependent and -independent pathways. After retroviral nucleocapsids enter target cells and reverse transcription begins, APOBEC3C can convert cytosine to uracil in the minus-sense single-strand viral DNA. This leads to G-to-A hypermutations in the subsequent plus-strand viral DNA. These detrimental mutations in the proviral genome, combined with a deamination-independent mechanism that operates before proviral integration, effectively inhibit retroviral replication within infected cells. APOBEC3C selectively targets single-stranded DNA and does not deaminate double-stranded DNA or single- or double-stranded RNA. Its antiviral activity extends to various viruses including simian immunodeficiency virus (SIV), hepatitis B virus (HBV), herpes simplex virus 1 (HHV-1), and Epstein-Barr virus (EBV). Additionally, it may inhibit the mobility of LTR and non-LTR retrotransposons. APOBEC3C might also play a role in the epigenetic regulation of gene expression through the process of active DNA demethylation.
Gene References Into Functions
  1. Studies indicate that heat shock proteins, particularly Hsp90, stimulate APOBEC3-mediated DNA deamination activity against hepatitis B viral DNA. This suggests a potential physiological role for APOBEC3 in mutagenesis/carcinogenesis and viral innate immunity. Hsp90 stimulates deamination activity of APOBEC3G, APOBEC3B, and APOBEC3C during co-expression in human liver HepG2 cells. PMID: 28637869
  2. Research suggests that APOBEC3C plays a significant role in protecting hosts from lentiviruses. PMID: 27732658
  3. APOBEC3C demonstrates antiviral functions against HIV-1 and exhibits binding capacity to the virus. PMID: 28315663
  4. Findings indicate that the functional potential of APOBEC3B and APOBEC3C in cancer mutagenesis is linked to estrogen receptor status. PMID: 26682542
  5. High APOBEC3C levels are associated with the pathogenesis of primary effusion lymphoma. PMID: 25650088
  6. Expression of APOBEC3A or 3C in 293FT cells reduced the infectivity of HPV16 pseudovirions. The decreased infectivity of virions assembled in the presence of APOBEC3A, but not 3C, was attributed to a lower copy number of the encapsidated reporter plasmid. PMID: 25576866
  7. APOBEC3 deaminases, upregulated by IFN-beta, induce E2 hypermutation of HPV16 in cervical keratinocytes. PMID: 24227842
  8. The mechanism of APOBEC3C (A3C)-mediated LINE-1 inhibition was found to be deaminase independent, requiring an intact dimerization site. The RNA-binding pocket mutation R122A abolished L1 restriction by A3C. PMID: 24101588
  9. This study confirmed the association of the APOBEC3 deletion with breast cancer risk among women of European ancestry. PMID: 23715497
  10. Decreases in APOBAC3A and APOBAC3C in the cortex of psychotic patients support the hypothesis that epigenetic misregulation of gene expression may contribute to the pathogenesis of psychotic disorders. PMID: 22948384
  11. A high-resolution crystal structure of APOBEC3C with the HIV-1 viral infectivity factor (Vif)-interaction interface has been determined. PMID: 23001005
  12. Findings suggest that APOBEC3-mediated editing of HIV-1 could be modulated by host and virus genetic characteristics in the context of pediatric infection. PMID: 22145963
  13. Results suggest that Core-A3C may be a potential candidate for a novel antiviral agent against human HBV infection. PMID: 21746770
  14. The authors identified a single cysteine at position 320 (C320) that disrupts A3DE activity. PMID: 21430060
  15. Somatic hypermutation of human mitochondrial and nuclear DNA by APOBEC3 cytidine deaminases represents a pathway for DNA catabolism. PMID: 21368204
  16. The basic biology of the interactions between human APOBEC3 proteins and HIV-1 Vif is reviewed. PMID: 20096141
  17. Different APOBEC3 family members exhibit distinct functions in neutralizing specific lentiviruses. PMID: 15466872
  18. APOBEC3 proteins may help prevent the zoonotic infection of humans by simple retroviruses and provide a mechanism for how simple retroviruses can avoid inhibition by APOBEC3 family members. PMID: 15956565
  19. Evidence suggests a role of host-encoded APOBEC3 proteins in regulating L1 retrotransposition. PMID: 16735504
  20. This review examines current knowledge on the mechanism of APOBEC3-dependent retrovirus restriction and discusses whether this innate host-defense system actively contributes to HIV genetic variation. PMID: 17078485
  21. Differences in promiscuity of monocytes, macrophages, and DCs can be defined, at least partly, by disparities in APOBEC expression. This has implications for enhancing cellular defenses against HIV-1. PMID: 17371941
  22. The catalytic domain of APOBEC3 proteins may be essential for proper folding and interactions with target factors such as RNA or proteins, in addition to cytidine deamination. PMID: 17582006
  23. These findings demonstrate that HBV is highly susceptible to the editing activity of an endogenous human deaminase, suggesting that A3C could contribute to innate anti-HBV host responses. PMID: 17625792
  24. APOBEC3 suppresses HBV replication in hepatocytes by inhibiting hnRNP K-mediated transcription and expression of HBV genes, as well as HBV core DNA synthesis. PMID: 17672864
  25. This study demonstrates that APOBEC3C is necessary and sufficient for G-to-A mutations in certain HIV-1 strains. PMID: 17967058
  26. Stress causes APOBEC3A, APOBEC3B, APOBEC3C, and APOBEC3F to colocalize efficiently with Vif(IIIB) and mRNA-PABP1 complexes in stress granules. PMID: 17977970
  27. This study compared the antiviral activities of human and murine Apobec3 (A3) and found that HIV is able to resist human A3G but is sensitive to murine A3, while murine leukemia virus is relatively resistant to murine A3 but sensitive to human A3G. PMID: 18032489
  28. Partially active Vif alleles leading to incomplete neutralization of cytoplasmic APOBEC3 molecules are directly responsible for the generation of a highly diverse, yet G-to-A biased, proviral reservoir. PMID: 18391217
  29. Cul5-E3 ubiquitin ligase appears to be a common pathway hijacked by HIV-1 and SIV Vif to overcome APOBEC3 proteins. PMID: 18419775
  30. These results indicate that APOBEC3G, APOBEC3C, and APOBEC3H have the ability to edit HBV DNA, and each protein is likely to contribute to various degrees to the generation of modified genomes in human liver cells. PMID: 18420796
  31. Human APOBEC3 proteins inhibit porcine endogenous retrovirus replication, which may reduce the risk of infection of human cells as a consequence of pig-to-human xenotransplantation. PMID: 18555089
  32. IAPE and HERV-K elements are restricted at the entry, amplification, and integration into their target genomes by the host APOBEC3 proteins. PMID: 18702815
  33. Distinct determinants in HIV-1 Vif and human APOBEC3 proteins are required for the suppression of diverse host antiviral proteins. PMID: 19088851
  34. Major cellular components of the human blood-retinal barrier (BRB) could be primarily cultured in vitro, and different expression of APOBEC3 in the human BRB was examined. PMID: 19369234
  35. APOBEC3 plays roles in limiting virus pathogenicity by partially restricting infection. PMID: 19390611
  36. The structure of APOBEC3C (A3C), a single-domain A3 with strong antilentiviral activity, was modeled. PMID: 19581596

Show More

Hide All

Database Links

HGNC: 17353

OMIM: 607750

KEGG: hsa:27350

STRING: 9606.ENSP00000355340

UniGene: Hs.441124

Protein Families
Cytidine and deoxycytidylate deaminase family
Subcellular Location
Nucleus. Cytoplasm.
Tissue Specificity
Expressed in spleen, testes, peripherical blood lymphocytes, heart, thymus, prostate and ovary.

Q&A

What is APOBEC3C and why is it important in antiviral research?

APOBEC3C is a single-domain cytidine deaminase that contributes to innate immunity against retroviruses. It functions by deaminating cytidines to uridines during the reverse transcription of viral genomes, particularly HIV-1. While A3C inhibits HIV-1 only weakly compared to other APOBEC3 family members, it has significant antilentiviral activity against other viruses like SIV. Understanding A3C is crucial for comprehending host defense mechanisms against viral infections and potentially developing novel antiviral therapies .

The APOBEC3 locus in humans encodes seven genes (A3A to A3H), with A3C being particularly interesting due to human polymorphisms that affect its antiviral activity. For instance, while the common form with serine at position 188 (A3C S188) has limited activity against HIV-1, approximately 10% of African individuals carry a polymorphism encoding isoleucine at position 188 (A3C I188), which correlates with increased antiviral activity due to enhanced dimer formation and cytidine deaminase activity .

What applications are validated for commercial APOBEC3C antibodies?

Commercial APOBEC3C antibodies have been validated for multiple applications in research settings, including:

  • Western Blot (WB): Typically used at dilutions of 1:500-1:2000

  • Immunohistochemistry (IHC): Typically used at dilutions of 1:600-1:2400

  • Immunofluorescence (IF)

  • ELISA

Specific cell lines where positive Western blot detection has been confirmed include:

  • Caco-2 cells

  • HEK-293 cells

  • K-562 cells

  • SW480 cells

For IHC applications, detection in human ovary cancer tissue and human ovary tumor tissue has been validated. For optimal results, antigen retrieval with TE buffer pH 9.0 is suggested, with citrate buffer pH 6.0 as an alternative .

What is the molecular weight and structure of APOBEC3C protein?

APOBEC3C has a calculated molecular weight of 23 kDa, which matches its observed molecular weight in experimental settings . Unlike some other APOBEC3 family members like A3F and A3G that contain two cytidine deaminase domains, A3C possesses only a single cytidine deaminase domain .

Three-dimensional models of A3C have been developed through comparative protein structure modeling using the crystal structures of APOBEC2 (A2) and the catalytic domain of A3G as templates. These models have revealed important structural features including:

  • A zinc-coordinating domain essential for enzymatic activity

  • A putative substrate binding pocket distal from the zinc-coordinating deaminase motif

  • Regions important for protein dimerization, which is crucial for antiviral activity

How should APOBEC3C antibodies be validated for specificity in different experimental systems?

Validating APOBEC3C antibodies for specificity requires a multi-faceted approach:

  • Positive and negative controls: Include cell lines known to express APOBEC3C (such as Caco-2, HEK-293, K-562, and SW480 cells) as positive controls. For negative controls, use APOBEC3C knockout cells or cells treated with APOBEC3C siRNA.

  • Multiple detection methods: Validate antibody specificity using complementary techniques:

    • Western blot: Look for a single band at approximately 23 kDa

    • Immunoprecipitation followed by mass spectrometry

    • Immunofluorescence with co-localization studies

  • Cross-reactivity assessment: Test against other APOBEC3 family members, especially those with high sequence similarity.

  • Epitope mapping: Determine which region of APOBEC3C the antibody recognizes to ensure it doesn't cross-react with conserved domains present in other APOBEC proteins.

  • Titration experiments: Perform antibody dilution series to determine optimal concentration for specific detection while minimizing background .

What are the optimal conditions for detecting APOBEC3C in different tissue types using immunohistochemistry?

Optimizing IHC for APOBEC3C detection requires careful consideration of several parameters:

  • Fixation: Formalin-fixed, paraffin-embedded tissues typically require antigen retrieval. Fresh frozen tissues may provide better epitope preservation but poorer morphology.

  • Antigen retrieval:

    • Primary recommendation: TE buffer at pH 9.0

    • Alternative method: Citrate buffer at pH 6.0

    • Heating methods: Water bath (95-98°C for 20-30 min) or pressure cooker (high pressure for 3-5 min)

  • Antibody dilution: Start with 1:600 and optimize up to 1:2400 based on signal-to-noise ratio

  • Detection system:

    • For weakly expressed samples: Use polymer-based detection systems or tyramide signal amplification

    • For routine detection: Standard avidin-biotin or polymer systems

  • Counterstaining: Light hematoxylin counterstaining to visualize tissue architecture without obscuring specific staining

  • Tissue-specific considerations:

    • Ovarian tissue: Validated with consistent results

    • Lymphoid tissues: May require additional blocking steps due to high background

    • Other tissues: May require optimization of antibody concentration and incubation time

What experimental controls are essential when studying APOBEC3C-mediated cytidine deamination activity?

When investigating APOBEC3C deaminase activity, several critical controls should be included:

  • Enzyme activity controls:

    • Positive control: Active APOBEC3C enzyme (wild-type)

    • Negative control: Catalytically inactive APOBEC3C mutant (e.g., mutation at the zinc-coordinating residue C98S)

    • Related enzyme control: Another APOBEC3 family member with known activity profile

  • Substrate controls:

    • Target sequence containing preferred APOBEC3C motifs

    • Non-targetable control sequence lacking motifs

    • Single-stranded versus double-stranded templates

  • Experimental validation controls:

    • Sequencing-based assay: Include a plasmid-only control to identify sequencing errors and PCR artifacts

    • "No-A3" control: To distinguish mutations made by reverse transcriptase from those introduced by APOBEC3 variants

    • Sequence-tagged templates: Use uniquely barcoded primers to distinguish true mutations from technical artifacts

  • Quantification controls:

    • Standard curves with known amounts of deaminated product

    • Input controls to normalize for template quantity variation

A robust hypermutation assay might use Illumina sequencing to assess G-to-A mutations in viral DNA products, with appropriate bioinformatic filtering to distinguish real editing events from background errors .

How do synthetic tandem domain APOBEC3C proteins differ from natural single-domain APOBEC3C in antiviral activity?

Synthetic tandem domain APOBEC3C proteins represent a fascinating research direction with significant implications for understanding APOBEC biology and developing potential antiviral strategies. These engineered constructs show several important differences from natural single-domain APOBEC3C:

  • Enhanced antiviral activity: All A3C tandem domain variants demonstrated greater antiviral activity against HIV-1 than their single-domain counterparts. These "super restriction factors" significantly inhibit viral replication .

  • Increased virion packaging: Tandem domain A3C proteins are packaged into virions at higher levels than single-domain versions, which appears to be the primary mechanism for their enhanced antiviral activity .

  • Active site requirements: Surprisingly, disabling one of the active sites in the C-terminal domain of A3C S188-A3C S188 resulted in even greater antiviral activity than the same protein with two active sites. This recapitulates similar evolutionary patterns seen in natural double-domain A3 proteins like A3F and A3G, which use only a single catalytically active deaminase domain .

  • Mechanism of action: While natural A3C primarily acts through cytidine deamination, the tandem domain proteins show inhibition of reverse transcription products that is largely independent of increased mutational load. Instead, they form larger higher-order complexes in cells that appear to physically impede reverse transcription .

  • Vif resistance: The synthetic A3C-A3C super restriction factors largely escape antagonism by the HIV-1 viral protein Vif, which normally counteracts APOBEC3 proteins .

This research demonstrates that protein engineering approaches can create enhanced restriction factors with potentially therapeutic applications.

How does the mutation signature of APOBEC3C differ from other APOBEC3 family members?

APOBEC3C exhibits a distinct mutation signature compared to other APOBEC3 family members, particularly APOBEC3A and APOBEC3B:

  • Sequence context preference:

    • APOBEC3C shows an over-representation of thymine (T) at the -2 position that is not observed for APOBEC3B and is stronger than in APOBEC3A

    • APOBEC3C exhibits an under-representation of cytosine (C) at -2 that is not apparent for APOBEC3A or APOBEC3B

    • Cytosine is neutral at -1 in the APOBEC3C motif, whereas it is under-represented in APOBEC3A and APOBEC3B motifs

  • Triplet motif representation:

    • APOBEC3A and APOBEC3B: High fractions of TCA and TCT variants

    • APOBEC3C: These variants are substantially diminished, while CCA, CCT, and CCC variants each approach 10% (9.4%, 7.7%, and 9.8%, respectively), compared to fewer than 3% for APOBEC3A and fewer than 4% for APOBEC3B

  • Mutation clustering behavior:

    • APOBEC3C caused strand-coordinated clustered mutations at a high frequency (4.8% of SNV pairs were clustered)

    • APOBEC3A and APOBEC3B caused clustered mutations at lower frequencies (1.1% and 1.3%, respectively)

    • APOBEC3C and APOBEC3B clustered mutations were more likely to be strand-coordinated, whereas APOBEC3A clustered mutations were not

  • Replication strand bias:

    • APOBEC3A and APOBEC3B display a clear preference to deaminate the lagging strand template during DNA replication

    • Surprisingly, APOBEC3C does not show a substantial bias and deaminates the leading and lagging strand templates approximately equally

These distinct mutation signatures have important implications for identifying the specific APOBEC3 enzyme responsible for mutations in cancer genomes and understanding their roles in various biological processes.

What is the role of APOBEC3C dimerization in its antiviral function?

Dimerization of APOBEC3C plays a critical role in its antiviral activity:

  • Structure-function relationship: Three-dimensional protein modeling of A3C has revealed that its antiviral activity requires protein dimerization. Mutations that disrupt dimer formation significantly reduce antiviral potency .

  • Polymorphism impact: The common human A3C variant with serine at position 188 (A3C S188) shows weak antiviral activity and limited dimerization. The less common variant with isoleucine at position 188 (A3C I188), found in approximately 10% of African individuals, demonstrates increased antiviral activity against HIV-1, correlating with enhanced dimer formation .

  • Mechanistic basis: Dimerization appears to influence:

    • Cytidine deaminase activity in vitro

    • RNA binding capacity

    • Virion incorporation efficiency

    • Stability of the protein complex

  • Experimental evidence: Studies using the Δvif simian immunodeficiency virus (SIV) reporter virus assay have confirmed that mutations affecting dimerization directly impact antiviral activity .

  • Evolutionary context: The importance of dimerization for A3C activity provides insight into why double-domain APOBEC3 proteins like A3F and A3G show greater antiviral potency, as they essentially contain permanently "dimerized" deaminase domains in a single polypeptide .

Understanding the molecular determinants of A3C dimerization could potentially inform the design of compounds that enhance this interaction and increase antiviral activity.

How can researchers address discrepancies in APOBEC3C antibody detection between different experimental systems?

When encountering discrepancies in APOBEC3C antibody detection across different experimental systems, consider the following troubleshooting strategies:

  • Antibody validation:

    • Verify that the antibody recognizes the correct epitope within APOBEC3C

    • Test multiple antibodies targeting different regions of APOBEC3C

    • Ensure the antibody is compatible with your specific application (WB, IHC, IF)

  • Sample preparation variations:

    • Fixation methods may affect epitope accessibility (formalin vs. alcohol fixation)

    • Antigen retrieval conditions (pH, temperature, duration)

    • Protein extraction methods may influence protein conformation

    • Post-translational modifications might mask epitopes

  • Expression levels and localization:

    • APOBEC3C expression varies across cell types and tissues

    • Subcellular localization may differ depending on cellular activation state

    • Viral infection or immune stimulation may alter expression patterns

  • Technical considerations:

    • Buffer compatibility (reducing vs. non-reducing conditions for WB)

    • Blocking reagents (milk vs. BSA may affect background)

    • Secondary antibody cross-reactivity

    • Detection system sensitivity

  • Experimental controls:

    • Include recombinant APOBEC3C protein as a positive control

    • Use APOBEC3C-overexpressing and APOBEC3C-knockout samples

    • Compare results with orthogonal detection methods (RT-PCR, mass spectrometry)

Discrepancies between different experimental platforms might reflect real biological differences rather than technical issues. For example, findings from the HPV16 system showed conflicting results that could be attributed to differences in the HPV16 clone used and/or differences in the methods used to measure infectivity .

What are the common pitfalls in interpreting APOBEC3C mutation signature data, and how can they be avoided?

Interpreting APOBEC3C mutation signature data requires careful consideration of several potential pitfalls:

The development of more sophisticated computational models that integrate sequence context, clustering patterns, and strand specificity can improve attribution of mutations to specific APOBEC3 enzymes. For instance, tools like RNAsee have been developed for predicting APOBEC3-mediated editing sites with various performance metrics (recall, precision, F1 score, and Matthew's correlation coefficient) .

How can researchers distinguish between hypermutation activity and non-catalytic antiviral mechanisms of APOBEC3C?

Distinguishing between the cytidine deaminase-dependent (hypermutation) and independent (non-catalytic) antiviral mechanisms of APOBEC3C requires carefully designed experiments:

  • Catalytic site mutants:

    • Create catalytically inactive APOBEC3C mutants (e.g., C98S mutation in the zinc-coordinating motif)

    • Compare antiviral activity of wild-type and mutant APOBEC3C

    • If mutants retain significant antiviral activity, this suggests non-catalytic mechanisms are important

  • Hypermutation assessment:

    • Develop assays to directly measure G-to-A mutations in viral genomes

    • Use deep sequencing with unique molecular identifiers to distinguish true mutations from PCR or sequencing artifacts

    • Compare mutation frequencies between wild-type and catalytic mutants of APOBEC3C

  • Reverse transcription product analysis:

    • Quantify early and late reverse transcription products in the presence of APOBEC3C variants

    • Reduced RT products without corresponding increase in mutations suggests non-catalytic inhibition

    • The A3C synthetic tandem domain studies demonstrated that inhibition of reverse transcriptase products can occur largely independent of increased mutational load

  • Virion incorporation studies:

    • Assess packaging efficiency of APOBEC3C variants into viral particles

    • Determine correlation between packaging and antiviral activity

    • Identify domains required for packaging versus catalytic activity

  • Protein-protein interaction analysis:

    • Investigate interactions between APOBEC3C and viral components (e.g., nucleocapsid, reverse transcriptase)

    • Determine if these interactions are required for antiviral activity

    • Map interaction domains and compare with catalytic domains

A comprehensive experimental approach that combines these methods can reveal the relative contributions of deaminase-dependent and independent mechanisms to APOBEC3C's antiviral activity.

How might APOBEC3C contribute to cancer development, and what methods can be used to study this relationship?

APOBEC3C, like other APOBEC3 family members, has potential roles in cancer development through its cytidine deaminase activity, which can create mutations in host DNA. This relationship can be studied through several approaches:

  • Cancer genomic analysis:

    • Identify APOBEC3C-specific mutation signatures in cancer genomes

    • Compare frequency of these signatures across different cancer types

    • Correlate with APOBEC3C expression levels in corresponding tumor samples

    • Distinguish APOBEC3C-specific mutations from those caused by other APOBEC3 enzymes using extended sequence context analysis

  • Mechanistic studies:

    • Investigate how chronic viral infections may dysregulate A3C expression

    • Study how BER (Base Excision Repair) processes A3C-induced lesions

    • Examine whether A3C-induced mutations can overwhelm DNA repair mechanisms

    • Determine if clustered mutations (kataegis) produced by A3C can lead to double-strand breaks and chromosomal rearrangements

  • Experimental models:

    • Develop cell lines with controlled APOBEC3C expression

    • Create mouse models with human APOBEC3C variants

    • Use CRISPR/Cas9 to introduce or knock out APOBEC3C

    • Analyze transformation potential and genomic instability

  • Viral co-factor analysis:

    • Investigate how specific viral infections alter APOBEC3C expression and function

    • Study virus-host interactions that may redirect APOBEC3C activity to host DNA

    • Examine long-term effects of persistent viral infections on APOBEC3C activity

  • Quantitative assessments:

    • Measure relative contributions of different APOBEC3 enzymes to mutation burden

    • Determine threshold levels of APOBEC3C activity associated with increased cancer risk

    • Develop biomarkers for APOBEC3C-mediated genomic instability

Recent research indicates that viral infections can lead to persistently high APOBEC3 levels, which combined with other tumor-initiating events, can intensify non-specific targeting of host DNA by these enzymes, thereby driving oncogenesis. The BER pathway often ineffectively resolves clustered mutations (kataegis) produced by APOBEC enzymes, potentially contributing to genomic instability .

How can structural knowledge of APOBEC3C's binding pocket be leveraged for antiviral drug development?

The identification and characterization of APOBEC3C's substrate binding pocket provides promising opportunities for antiviral drug development:

  • Structure-based drug design:

    • The 3D protein models of A3C derived through comparative modeling reveal a putative substrate binding pocket distal from the zinc-coordinating deaminase motif

    • Automated pocket extraction algorithms have identified specific regions that could be targeted for drug development

    • These pockets could serve as binding sites for small molecules that enhance APOBEC3C's antiviral activity

  • Critical residue targeting:

    • R122 at the pocket entrance has been identified as crucial for A3C incorporation into viral particles

    • Mutations in this region diminish antiviral activity

    • Compounds that stabilize this region or enhance its interaction with viral components could increase antiviral efficacy

  • RNA-interaction modulation:

    • Evidence suggests 5.8S RNA specifically binds to the substrate binding pocket and mediates incorporation of A3C into virus particles

    • Small molecules that mimic or enhance this RNA interaction could improve viral packaging of A3C

    • Conversely, viral inhibitors that block this interaction could be developed as antivirals

  • Dimerization enhancement:

    • A3C activity requires protein dimerization for optimal antiviral function

    • Drugs that stabilize A3C dimers could enhance its restriction activity

    • Targeting the dimerization interface with small molecules could convert the weakly active common variant (A3C S188) into a more potent restriction factor

  • Vif antagonism inhibition:

    • The synthetic A3C tandem domain proteins largely escape HIV-1 Vif-mediated antagonism

    • Understanding the structural basis for this resistance could inform the design of inhibitors that block Vif-mediated degradation of natural A3C

    • Such inhibitors would allow endogenous A3C to exert its antiviral activity more effectively

The structural modeling approaches described in the research, including comparative protein structure modeling with templates like the crystal structures of A2 and the catalytic domain of A3G, provide valuable frameworks for these drug development efforts. The models have demonstrated medium accuracy (approximately 85% of residues within 3.5 Å of the actual conformation), making them suitable to support structure-based drug design initiatives .

What are the best practices for designing experiments to study APOBEC3C-mediated RNA editing using antibody-based approaches?

Designing robust experiments to study APOBEC3C-mediated RNA editing requires careful consideration of several methodological aspects:

  • Sample preparation and RNA isolation:

    • Use RNase inhibitors throughout to prevent degradation

    • Consider subcellular fractionation to enrich for specific compartments where editing occurs

    • Employ specialized RNA extraction methods that preserve edited sites

    • Include controls to account for potential deamination during sample preparation

  • Antibody selection and validation:

    • Use antibodies that specifically recognize APOBEC3C (not cross-reactive with other APOBEC3 family members)

    • Validate antibody specificity using APOBEC3C knockout or knockdown controls

    • Consider epitope tags (FLAG, HA) for recombinant APOBEC3C to enable consistent immunoprecipitation

  • RNA immunoprecipitation (RIP) protocols:

    • Crosslink RNA-protein complexes before immunoprecipitation (UV or chemical crosslinking)

    • Use stringent washing conditions to reduce non-specific binding

    • Include appropriate negative controls (IgG, irrelevant antibody)

    • Verify enrichment by qRT-PCR before proceeding to more comprehensive analyses

  • Identifying RNA editing sites:

    • Employ deep sequencing with unique molecular identifiers to distinguish true editing events from sequencing errors

    • Use computational tools like RNAsee with appropriate thresholds:

      • Score threshold ≥10 for rules-based models

      • Probability threshold >0.5 for random forest models

    • Apply filtering criteria that account for APOBEC3C's unique sequence preferences

  • Validation strategies:

    • Confirm editing with independent methods (Sanger sequencing, site-specific PCR)

    • Perform parallel experiments with catalytically inactive APOBEC3C mutants

    • Compare results with predictions from computational models

    • Use targeted approaches like SITE-Seq or DART-seq to validate specific sites

  • Quantification approaches:

    • Accurately determine editing efficiency at individual sites

    • Consider the rarity of genuine editing sites (estimated at 0.2% of cytosines)

    • Use appropriate statistical methods that account for the imbalanced nature of the data

For experimental validation of predicted editing sites, a combination of antibody-based enrichment followed by high-throughput sequencing offers the most comprehensive approach, though researchers should be prepared for relatively low precision (26.5% for intersection models in realistic proportions) but still achieving over 100x enrichment relative to baseline .

How can researchers optimize Western blot protocols for detecting different APOBEC3C variants?

Optimizing Western blot protocols for APOBEC3C variants requires attention to specific technical considerations:

  • Sample preparation:

    • Use RIPA buffer with protease inhibitors for effective protein extraction

    • For clinical samples or tissues, consider specialized extraction methods to preserve native protein structure

    • When comparing APOBEC3C variants (e.g., S188 vs. I188), ensure equal loading by normalizing to total protein or housekeeping genes

    • For tandem domain constructs, adjust loading to account for their larger molecular weight (approximately 46 kDa)

  • Gel electrophoresis conditions:

    • Use 12-15% SDS-PAGE gels for optimal resolution of APOBEC3C's 23 kDa band

    • For tandem domain proteins, consider 10-12% gels

    • Run at lower voltage (80-100V) for better resolution of closely migrating variants

    • Include molecular weight standards that bracket the expected sizes

  • Antibody selection and dilution:

    • Primary antibody: Start with 1:500-1:2000 dilution and optimize based on signal-to-noise ratio

    • For variant-specific detection, consider using antibodies raised against specific peptides containing the variant residue

    • Secondary antibody: HRP-conjugated anti-rabbit IgG at 1:5000-1:10000

    • Consider using fluorescent secondary antibodies for multiplexing or quantitative analysis

  • Detection optimization:

    • For low expression levels: Use high-sensitivity ECL substrates or signal amplification systems

    • For quantitative comparison: Use digital imaging systems with linear dynamic range

    • For multiple variants: Consider multiplexing with differently labeled secondary antibodies

  • Controls and validation:

    • Positive controls: Include lysates from cells expressing recombinant APOBEC3C variants

    • Negative controls: Use APOBEC3C knockout cells or cells treated with APOBEC3C siRNA

    • Loading controls: Probe for housekeeping proteins (β-actin, GAPDH) or use total protein staining

    • Specificity controls: Pre-adsorb antibody with recombinant APOBEC3C to confirm specificity

  • Troubleshooting common issues:

    • Multiple bands: May indicate post-translational modifications or degradation products

    • No signal: Increase antibody concentration or protein amount, verify expression in sample

    • High background: Increase blocking time/concentration, reduce primary antibody concentration

    • Variable results: Standardize lysate preparation and storage conditions

For detecting dimerization of APOBEC3C, consider using non-reducing conditions and native PAGE, as reducing agents may disrupt protein-protein interactions that are essential for dimer formation .

What protocol modifications are needed when using APOBEC3C antibodies in co-immunoprecipitation studies with viral proteins?

Co-immunoprecipitation (Co-IP) studies examining interactions between APOBEC3C and viral proteins require specific protocol modifications:

  • Cell system selection:

    • Use cells that support viral replication (e.g., 293T, HeLa for HIV-1)

    • Consider physiologically relevant cell types (primary CD4+ T cells, macrophages)

    • For transfection-based studies, optimize to achieve near-physiological expression levels

  • Expression constructs:

    • For APOBEC3C: Consider epitope-tagged constructs (FLAG, HA, Myc) to facilitate detection

    • For viral proteins: Use codon-optimized constructs for improved expression

    • Include both wild-type and mutant constructs (e.g., A3C S188 vs. I188; wild-type vs. catalytically inactive C98S)

  • Lysis conditions:

    • Use mild lysis buffers to preserve protein-protein interactions:

      • NP-40 buffer (0.5% NP-40, 150 mM NaCl, 50 mM Tris-HCl pH 7.4)

      • Add protease inhibitors freshly before use

    • Include RNase A treatment in control samples to determine RNA-dependent interactions

    • Consider crosslinking (formaldehyde or DSP) for transient interactions

  • Immunoprecipitation strategy:

    • Antibody approach:

      • Anti-APOBEC3C antibody (10591-1-AP or equivalent) at 2-5 μg per sample

      • Anti-tag antibody for epitope-tagged constructs

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

    • Incubation conditions:

      • 4°C overnight with gentle rotation

      • Include BSA (0.1-0.5%) to reduce non-specific binding

  • Washing and elution:

    • Use progressively stringent washes (increasing salt concentration)

    • For RNA-mediated interactions, include control samples with RNase treatment

    • Elute with either low pH, high salt, or SDS-containing buffer

    • For epitope-tagged proteins, consider peptide elution for gentler conditions

  • Detection and analysis:

    • Western blot for co-precipitated proteins using specific antibodies

    • Include input controls (typically 5-10% of IP material)

    • For weak interactions, use more sensitive detection methods (ECL Advance, Femto)

    • Consider mass spectrometry for unbiased detection of interaction partners

  • Specific considerations for viral protein interactions:

    • For HIV-1 Vif studies:

      • Include proteasome inhibitors (MG132) to prevent APOBEC3C degradation

      • Compare wild-type Vif with non-functional mutants

    • For nucleocapsid/Gag interactions:

      • Test both immature and mature forms of viral proteins

      • Consider the role of RNA in mediating interactions

When studying interactions involving the R122 residue of APOBEC3C, which is critical for RNA-dependent virion incorporation, include controls that distinguish direct protein-protein interactions from those mediated by RNA bridging .

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.