SOV Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Terminology and Nomenclature

  • Antibodies are typically named based on their target antigen, structural features, or clinical application (e.g., rituximab targets CD20, adalimumab is anti-TNFα) .

  • Standardized naming conventions for antibodies include:

    • Prefix: Unique identifier (e.g., "mavu" for mavrilimumab).

    • Infix: Target or disease (e.g., "-li-" for immune system targets).

    • Suffix: "-mab" for monoclonal antibodies .

  • The term "SOV" does not align with established nomenclature rules or known antibody classifications .

Search Results Analysis

The provided sources cover diverse antibody-related topics, including:

  • Structural features: Variable and constant regions, CDRs (complementarity-determining regions), and Fc domains .

  • Therapeutic applications: Bispecific antibodies (e.g., targeting cancer or viral epitopes) , monoclonal antibodies (e.g., COVID-19 therapies) .

  • Diversity mechanisms: V(D)J recombination, somatic hypermutation .

  • Databases: Thera-SAbDab, which catalogs therapeutic antibodies and their structural data .

No reference to "SOV" appears in any context—structural, functional, or clinical—across these sources.

Potential Explanations for the Absence of Data

  1. Typographical Error: "SOV" may be a misspelling (e.g., "SARS-CoV-2 antibodies" are extensively studied ).

  2. Emerging Research: The term might refer to a novel, unpublished antibody not yet cataloged in public databases.

  3. Proprietary Compound: "SOV" could be an internal code name for a confidential therapeutic under development.

Recommendations for Further Inquiry

To resolve ambiguity, consider:

  • Verifying the term with the original source or context.

  • Expanding the search to proprietary clinical trial registries (e.g., ClinicalTrials.gov) or patent databases.

  • Consulting recent preprints (e.g., bioRxiv, medRxiv) for unpublished studies.

Comparative Table of Antibody Types Mentioned in Sources

Antibody TypeKey FeaturesExampleSource
Bispecific AntibodiesTargets two antigens/epitopes; used in cancer and viral therapiesBlinatumomab (anti-CD19/CD3)
Monoclonal AntibodiesSingle specificity; neutralizes pathogens or modulates immune responsesIxekizumab (anti-IL17A)
Broadly Neutralizing AbsTargets conserved viral epitopes; cross-reactive against variantsAntibody 2526 (anti-HIV/influenza)
Fc-Silenced AntibodiesEngineered to reduce Fc receptor binding; minimizes immune activationVariants with loop grafting

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
SOV antibody; DIS3L2DIS3-like exonuclease 2 antibody; EC 3.1.13.- antibody; Protein SUPPRESSOR OF VARICOSE antibody
Target Names
SOV
Uniprot No.

Target Background

Function
This antibody targets a 3'-5'-exoribonuclease that specifically recognizes RNAs polyuridylated at their 3' end and mediates their degradation. It is a component of an exosome-independent RNA degradation pathway that likely mediates the degradation of cytoplasmic mRNAs that have been deadenylated and subsequently uridylated at their 3' end.
Protein Families
RNR ribonuclease family, DIS3L2 subfamily
Subcellular Location
Cytoplasm. Cytoplasm, P-body.
Tissue Specificity
Widely expressed.

Q&A

How do IgG, IgM, and IgA responses differ temporally in SARS-CoV-2 infection?

IgM and IgA responses to SARS-CoV-2 antigens demonstrate relatively rapid decline following peak optical density (OD) measurements. This decline occurs between 20-30 days post-onset of symptoms for IgM and IgA respectively . For some individuals sampled at time points beyond 60 days POS, the IgM and IgA responses approach baseline levels . This pattern is consistent with the expected antibody kinetics following acute viral infection . IgG responses show different kinetics, with more persistent detection. Understanding these temporal differences is essential when selecting appropriate timepoints for sample collection and when interpreting antibody persistence data.

How should researchers validate antibodies for SARS-CoV-2 research applications?

Proper antibody validation is essential to address reproducibility concerns in research. Validation must occur for each specific application in which an antibody will be used . The validation process should focus on both specificity (ability to correctly detect the target epitope) and selectivity (ability to differentiate from similar epitopes) .

Three key principles for antibody validation include:

  • Demonstrating selectivity is an essential aspect of validation

  • Validation needs to be performed in each application where the antibody is used

  • Validation needs to be performed in samples containing varying, experimentally relevant concentrations and ratios of intended target and non-intended off-target proteins

For immunohistochemistry applications, researchers must consider that chemical fixation and subsequent antigen retrieval can affect selectivity depending on the epitope being detected . Therefore, antibody performance depends significantly on sample preparation methodology.

What considerations apply when using antibodies in dual-recognition assays?

When designing sandwich assays that employ dual-recognition (two antibodies per protein), researchers can enforce higher selectivity, enhancing reliable detection of target antigens . In such experimental designs, it may be acceptable to use a less specific (polyclonal) antibody for capture, combined with a highly specific (monoclonal) antibody for detection . This approach leverages the strengths of both antibody types while minimizing their respective limitations. Researchers should carefully assess the epitope targets to ensure the antibody pairs do not compete for binding sites.

What patterns of decline do neutralizing antibodies follow after SARS-CoV-2 infection?

The neutralizing antibody response after SARS-CoV-2 infection follows a pattern typical of acute viral infections, with declining neutralizing antibody (nAb) titers observed following an initial peak . The decline trajectory varies significantly between individuals. In subjects who develop modest nAb titers (100-300 range) following infection, titers may become undetectable (ID50 <50) or approach baseline after approximately 50 days, highlighting the transient nature of the antibody response in some individuals . In contrast, individuals with high peak ID50 maintain nAb titers in the 1,000-3,500 range beyond 60 days POS . These findings have critical implications for studying potential protection against reinfection and for assessing durability of vaccine-induced immunity.

How do binding antibody responses to different SARS-CoV-2 antigens compare temporally?

Detailed analysis of antibody binding responses to SARS-CoV-2 Spike protein (S), receptor binding domain (RBD), and nucleocapsid protein (N) demonstrates varying patterns across individuals. Approximately 58.1% of individuals show synchronous seroconversion to S, RBD and N, whereas singular seroconversion to N or S occurs in about 16.1% of individuals for each antigen . IgG responses against S, RBD, and N antigens can be observed in 92.3%, 89.2%, and 93.8% of individuals respectively . Cumulative frequency analysis of positive IgG, IgA, and IgM responses against these antigens does not indicate more rapid elicitation of IgM and IgA responses against a particular antigen . These varied seroconversion patterns must be considered when designing diagnostic assays targeting specific antigens or when interpreting serological survey data.

How can large antibody datasets be leveraged for machine learning applications?

Large datasets containing quantitative binding scores of antibodies against SARS-CoV-2 target peptides provide valuable resources for developing and benchmarking machine learning models . For example, datasets containing binding scores for over 100,000 scFv-format antibodies with predicted affinity measurements ranging from 37 pM to 22 mM offer unprecedented opportunities to train models that can predict antibody-antigen interactions . Researchers can utilize these datasets to develop antibody-specific representation models that may accelerate the discovery and optimization of therapeutic antibodies. When working with such datasets, researchers should account for variables such as antibody format (e.g., scFv vs. full IgG) and chain orientation (heavy-light vs. light-heavy), as these factors can influence binding properties .

What methodologies exist for in silico design of SARS-CoV-2 antibody libraries?

In silico antibody library design typically begins with seed sequences identified from methods such as phage display campaigns using human naïve libraries . From these seeds, researchers can systematically generate variants through directed mutagenesis strategies, such as creating all k=1 mutations and random k=2 and k=3 mutations throughout the complementary-determining regions (CDRs) . This approach allows for the exploration of sequence space around promising lead antibodies. When designing such libraries, researchers should consider allocating a portion of the sequence budget for controls in binding experiments . Subsequent high-throughput screening methods like AlphaSeq can then evaluate the binding properties of these designed variants, enabling the identification of improved candidates with enhanced affinity or specificity against SARS-CoV-2 targets.

How should researchers address contradictory antibody response data in SARS-CoV-2 studies?

When encountering contradictory antibody response data across SARS-CoV-2 studies, researchers should systematically evaluate several factors that may contribute to discrepancies:

  • Timing of sample collection relative to symptom onset is critical, as antibody kinetics follow specific temporal patterns with IgM and IgA responses declining after 20-30 days post-symptom onset

  • Disease severity differences between study populations, as the magnitude (but not kinetics) of neutralizing antibody responses correlates with severity

  • Assay methodology variations, as different detection methods (ELISA, neutralization assays, etc.) may yield different results

  • Antibody type being measured (binding vs. neutralizing antibodies)

  • Target antigen differences (S, RBD, N proteins) which show varied patterns of seroconversion

When designing studies to resolve such contradictions, researchers should employ standardized protocols with consistent sampling timepoints, clearly defined clinical categories, and multiple complementary assay methods to comprehensively characterize antibody responses.

What are the key methodological considerations for evaluating antibody therapeutics against SARS-CoV-2 variants?

When evaluating antibody therapeutics against SARS-CoV-2 variants, researchers must address several methodological challenges. The virus's propensity for mutation has rendered most antibody treatments developed during the pandemic ineffective against newer variants . A promising approach involves engineering antibody pairs where one antibody serves as an anchor by binding to conserved viral regions while another blocks the virus's ability to infect cells . This strategy has demonstrated effectiveness against multiple variants through Omicron in laboratory testing .

Key methodological considerations include:

  • Testing against a panel of relevant circulating variants

  • Assessing both binding affinity and neutralization potency

  • Evaluating potential for viral escape through in vitro selection experiments

  • Characterizing the precise epitopes targeted using structural biology approaches

  • Determining whether neutralizing activity correlates with protection in animal models

When designing such studies, researchers should remember that demonstrating neutralizing activity requires assays using live or pseudotyped virus, which cannot be performed in a high-throughput fashion .

How should researchers interpret neutralizing vs. binding antibody data for immunity assessment?

When assessing immunity to SARS-CoV-2, researchers must carefully distinguish between neutralizing antibodies (nAbs) and binding antibodies. Total antibody measurements do not necessarily reflect protection after infection, nor do they indicate the efficacy of vaccination to ensure immunity . While binding antibodies (measured by ELISA against S, RBD, or N proteins) indicate exposure, neutralizing antibodies more directly correlate with potential protection.

  • Certain target epitopes of antibodies might enhance virus entry rather than neutralize it

  • Simple immunoassays that best reflect virus neutralization need to be developed based on identified protective antibody targets

  • The protective threshold of neutralizing antibody titers remains incompletely defined

  • Neutralizing antibody titers decline over time, with some individuals showing undetectable levels after ~50 days despite initial robust responses

These factors underscore the complexity of interpreting antibody data for immunity assessment and highlight the need for complementary approaches including cellular immunity evaluation.

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.