Syrian Hamster IgG Isotype Control — Purified in vivo GOLD™ Functional Grade

Syrian Hamster IgG Isotype Control — Purified in vivo GOLD™ Functional Grade

Product No.: I-444

- -
- -
Clone
Polyclonal
Formats AvailableView All
Product Type
Polyclonal Isotype Control
Isotype
IgG
Applications
FA
,
FC
,
in vivo

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Host Species
Syrian Hamster
Recommended Dilution Buffer
Product Concentration
≥ 5.0 mg/ml
Endotoxin Level
< 1.0 EU/mg as determined by the LAL method
Purity
≥95% monomer by analytical SEC
>95% by SDS Page
Formulation
This monoclonal antibody is aseptically packaged and formulated in 0.01 M phosphate buffered saline (150 mM NaCl) PBS pH 7.2 - 7.4 with no carrier protein, potassium, calcium or preservatives added. Due to inherent biochemical properties of antibodies, certain products may be prone to precipitation over time. Precipitation may be removed by aseptic centrifugation and/or filtration.
Product Preparation
Functional grade preclinical antibodies are manufactured in an animal free facility using in vitro cell culture techniques and are purified by a multi-step process including the use of protein A or G to assure extremely low levels of endotoxins, leachable protein A or aggregates.
Storage and Handling
Functional grade preclinical antibodies may be stored sterile as received at 2-8°C for up to one month. For longer term storage, aseptically aliquot in working volumes without diluting and store at ≤ -70°C. Avoid Repeated Freeze Thaw Cycles.
Country of Origin
USA
Shipping
Next Day 2-8°C
Applications and Recommended Usage?
Quality Tested by Leinco
FC This isotype control antibody should be used at the same concentration as the primary antibody.
Each investigator should determine their own optimal working dilution for specific applications. See directions on lot specific datasheets, as information may periodically change.

Description

Specificity
This isotype control antibody is for use as a negative control in the measurement of the non-specific binding of Syrian hamster monoclonal antibodies of isotype IgG to human, mouse or rat tissues.

Leinco Antibody Advisor

Powered by AI: AI is experimental and still learning how to provide the best assistance. It may occasionally generate incorrect or incomplete responses. Please do not rely solely on its recommendations when making purchasing decisions or designing experiments.

Common in vivo applications of polyclonal antibodies in mice include use as experimental controls, immune depletion or neutralization, and testing antibody efficacy and safety in models of infection, cancer, and autoimmune disease. While "clone Polyclonal" is not a standard designation (polyclonals originate from multiple B-cell clones), polyclonal antibodies as a reagent class are widely used for various in vivo investigations.

Key in vivo applications in mice:

  • Isotype or Non-reactive Controls: Polyclonal mouse IgG is frequently used as a non-reactive control to account for non-specific effects in antibody dosing studies, including immunotherapy, immunomodulation, and toxicity studies. This helps distinguish between specific and non-specific effects of test antibodies.

  • Immune Modulation: Polyclonal antibodies can be administered to deplete specific cell types (e.g., using anti-lymphocyte serum), neutralize circulating toxins or cytokines, or block particular immune pathways to study immune function or disease progression.

  • Therapeutic Evaluation: In transgenic models (such as humanized mice), polyclonal antibodies produced in response to immunization with specific pathogens or proteins are evaluated for their ability to neutralize targets, clear infection, or alter disease outcomes.

  • Pathogen Neutralization and Vaccine Studies: Mice are immunized to produce polyclonal antisera, which can then be harvested and re-injected into naïve or infected mice to test efficacy in neutralizing pathogens, providing passive immunity, or assessing protective mechanisms.

  • Validation and Dosage Optimization: Researchers use polyclonal antibodies in vivo to validate candidate therapeutic targets (e.g., by neutralizing growth factors, receptors, or cytokines) and optimize dosing regimens for preclinical studies.

Additional details:

  • Polyclonal antibodies are generated in mice through immunization with target antigens, leveraging the mouse’s immune response to create a diverse pool of antigen-specific antibodies that recognize multiple epitopes, which enhances their robustness in detecting or neutralizing targets in complex biological settings.
  • While rabbits and larger mammals are often used for large-scale antibody production, mice remain a standard model for generating, characterizing, and applying polyclonal antibodies, especially when testing humanized antibodies or conducting mechanistic studies.

In summary, the most common in vivo uses of polyclonal antibodies in mice are as controls in immunotherapy/immunology research, experimental depletion/neutralization tools, and preclinical efficacy and safety testing for infectious or immune-mediated diseases.

In the literature, polyclonal antibodies are often used alongside other antibodies or proteins, particularly in research and therapeutic contexts. Here are some commonly used antibodies or proteins in conjunction with polyclonal antibodies:

1. Monoclonal Antibodies

  • Use: Monoclonal antibodies are frequently used in combination with polyclonal antibodies for enhanced specificity and broader antigen recognition. They are particularly useful in assays requiring high specificity, such as ELISA, Western Blotting, and immunohistochemistry.
  • Examples in Use: Monoclonal antibodies like anti-PD-L1 are combined with polyclonal antibodies to enhance antitumor effects.

2. Immune Checkpoint Inhibitors (ICIs)

  • Use: ICIs, such as anti-PD-L1 antibodies, are used in combination with polyclonal antibodies to enhance their therapeutic efficacy, particularly in cancer treatment.
  • Examples in Use: The combination of polyclonal antibodies with ICIs like anti-PD-L1 has shown improved survival in certain tumor models.

3. Proteins and Peptides

  • Use: These are often used as targets for polyclonal antibodies in research and diagnostics. For example, polyclonal antibodies are used to detect specific proteins in Western Blotting and ELISA.
  • Examples in Use: Polyclonal antibodies against proteins such as those involved in oncology or infectious diseases are used for detection and therapeutic purposes.

4. Other Antibody Therapies

  • Use: Combinations of polyclonal and monoclonal antiviral antibodies are used for optimal protection against certain infections.
  • Examples in Use: The use of polyclonal antiviral antibodies alongside monoclonal antibodies can provide broader protection against viral infections.

5. Single Particle Cryo-Electron Microscopy (cryoEM) and Mass Spectrometry (MS)

  • Use: These techniques are used to sequence and map epitopes for polyclonal antibodies, helping to understand their specificity and function.

These combinations and techniques highlight the versatility and utility of polyclonal antibodies in various research and therapeutic applications.

Based on the search results, the key findings related to polyclonal research in scientific literature span two distinct areas: tumor biology and antibody development.

Polyclonality in Cancer Biology

Recent research has revealed that polyclonal tumors possess distinct structural and functional characteristics that influence cancer progression. In Apc-driven tumorigenesis, polyclonal tumors maintain a complex architecture comprising subclones with different Apc mutations and transcriptional states. These tumors typically contain both major and minor clones, each harboring exclusive Apc-inactivating mutations.

A critical discovery is that early truncating mutations occurring N-terminal to the Armadillo repeat region are under-represented in monoclonal tumors compared to the minor clones of polyclonal tumors. This suggests that polyclonality helps overcome certain fitness barriers during tumor development. The clonal architecture directly influences transcriptional heterogeneity within tumors, with different numbers of differentially expressed genes observed between major and minor clones.

Interestingly, mutations in other cancer-related genes including Trp53, Ctnnb1, and Kras were present but not differentially enriched within tumor clones. The research proposes that poorly-transforming APC mutant intestinal stem cells can overcome competitive disadvantages through polyclonal cooperation, particularly when paired with another APC mutant exhibiting supercompetitor behavior.

Polyclonal Antibody Sequencing and Characterization

In immunology research, significant advances have been made in understanding and utilizing polyclonal antibodies. Modern de novo sequencing methods enable the identification of individual antibodies in serum or plasma without requiring a B cell source. These techniques are particularly valuable for converting existing polyclonal antibodies to monoclonal antibodies, analyzing biorepository samples with low cell viability, and cataloging dominant clones in patient samples.

The key advantage of de novo polyclonal antibody sequencing is its ability to capture the most abundant antibodies in samples, which typically have the highest functional impact. However, these methods require more extensive mass spectrometry data generation and higher sample requirements compared to alternative approaches. Phage display technology has also been employed to characterize polyclonal antibody repertoires, using multiple panning cycles to identify and sequence specific antibody-binding peptides.

Dosing Regimens of Polyclonal Antibodies Across Mouse Models

The dosing regimens of polyclonal antibodies (such as those targeting mouse IFNAR-1 or polyclonal mouse IgG used as a control) can vary significantly across different mouse models and studies, depending on the experimental context, therapeutic goal, antigen, and mouse strain.

Key Factors Influencing Dosing

  • Purpose of Study: Dosing may be tailored for immune cell depletion, cytokine blockade, treatment of infection, or as a control antibody. For example, anti-mouse IFNAR-1 (MAR1-5A3) is often given as a single intraperitoneal injection in flavivirus or dengue studies in wild-type mice.
  • Mouse Strain and Model: Genetic background (e.g., wild-type vs. transgenic), age, sex, immune status, and disease model (e.g., tumor, infection, autoimmunity) all influence the effective dose and schedule. For instance, protocols for inducing anti-drug antibodies (ADAs) in transgenic mice may use multiple, low-dose injections over weeks, but these do not always translate directly to other models or antibodies.
  • Antibody Characteristics: The specificity, affinity, source (endogenous vs. exogenous), and Fc region of the polyclonal antibody affect pharmacokinetics and pharmacodynamics, hence dosing.
  • Outcome Measure: Efficacy (e.g., tumor shrinkage, viral clearance), pharmacodynamic (PD) markers, or immune modulation (e.g., cytokine neutralization) each require different dose optimization.

Examples of Variability

  • COVID-HIGIV (Polyclonal Hyperimmune Globulin): In a lethal SARS-CoV-2 mouse model, the dose, timing, and route of administration were optimized to achieve viral clearance and survival, with dosing schedules determined by pharmacokinetic (PK) studies.
  • Anti-Mouse IFNAR-1: The regimen can be a single dose (e.g., 250–500 μg/mouse, IP) in wild-type mice for short-term cytokine blockade, or repeated dosing in chronic disease models.
  • Control IgG: Polyclonal mouse IgG, used as a negative control, is typically dosed at similar amounts and frequencies to the experimental antibody to control for non-specific effects.

General Guidelines

  • Typical Doses: Polyclonal antibodies are often administered at 100–500 μg per mouse, intraperitoneally, but the actual dose and schedule must be empirically determined for each model and antibody.
  • Frequency: Single-dose regimens are common for acute studies, while repeated dosing (e.g., every 3–7 days) is used in chronic or therapeutic models.
  • Route: Intraperitoneal (IP) injection is standard, but intravenous (IV) or subcutaneous (SC) routes may be used depending on the study design.

Challenges and Considerations

  • Lack of Universal Dosing: There is no universal “correct” dose for polyclonal antibodies across all mouse models; dosing must be optimized experimentally for each context.
  • PK/PD Variability: The half-life, tissue distribution, and clearance of polyclonal antibodies can differ by model, strain, and antigen, necessitating pilot PK studies.
  • Outcome Variability: The relationship between antigen copy number and antibody dose can critically impact experimental outcomes, such as antibody-mediated immune suppression.

Summary Table: Example Dosing Regimens

Model/Study TypeExample AntibodyTypical DoseFrequencyRouteNotes
Flavivirus infectionAnti-IFNAR-1 (MAR1-5A3)250–500 μg/mouseSingle doseIPWild-type mice
SARS-CoV-2 lethal modelCOVID-HIGIVOptimized per PKVariableIP/IVDosing based on PK studies
Control (all models)Polyclonal mouse IgGMatch test AbMatch test AbIPNegative control

Conclusion

Dosing regimens for polyclonal antibodies in mouse models are highly variable and must be tailored to the specific experimental question, mouse strain, disease model, and antibody characteristics. While some general principles apply (e.g., IP administration, dose ranges around 100–500 μg/mouse), there is no one-size-fits-all protocol; optimization through pilot studies and PK/PD analysis is essential for reliable results.

References & Citations

FA
Flow Cytometry
in vivo Protocol

Certificate of Analysis

Formats Available

- -
- -
Disclaimer AlertProducts are for research use only. Not for use in diagnostic or therapeutic procedures.