IgG Mouse, His

Immunoglobulin Heavy Chain Constant Region Gamma 2a Mouse Recombinant, His Tag
Shipped with Ice Packs
In Stock

Description

Definition and Structure of IgG Mouse, His

"IgG Mouse, His" refers to mouse-derived immunoglobulin G (IgG) antibodies engineered or purified to specifically bind His-tagged proteins. These antibodies are typically monoclonal IgG1 isotype antibodies produced via recombinant expression or isolated from mouse serum. The "His" designation indicates their primary function: recognizing histidine-rich peptide sequences (e.g., 4xHis, 5xHis, or RGSHHHH motifs) used for affinity purification or detection of recombinant proteins .

Production and Purification Methods

Mouse anti-His tag antibodies are produced via two primary approaches:

MethodDescriptionSource
Recombinant ExpressionEngineered mouse IgG1 antibodies expressed in HEK293 or other mammalian systems.
Native IsolationPurified from mouse serum via ion exchange chromatography. Contains trace contaminants.

Purity and Form:

  • Recombinant: >95% purity (SDS-PAGE/SEC-MALS), lyophilized in PBS with trehalose .

  • Native: 90–95% purity, liquid formulation in phosphate-buffered saline (PBS) with sodium azide .

Detection of His-Tagged Proteins

Mouse IgG1 anti-His tag antibodies are optimized for:

  • Western Blot: Detects 2–4 ng of His-tagged protein per lane .

  • ELISA: Sensitive to 0.1–16 ng/mL in competitive or sandwich assays .

  • Flow Cytometry: Stains transfected cells expressing His-tagged surface proteins .

  • Immunoprecipitation: Efficiently pulls down His-tagged targets from cell lysates .

Epitope Specificity

Antibody TypeTarget EpitopeDetection SensitivityCross-ReactivitySource
RGS·HisN-terminal RGSHHHH0.5 ng (dot blot)Negligible
Tetra·His4xHis (internal/N/C-terminal)0.5 ng (dot blot)Negligible
THE™ His Tag (GenScript)4xHis or 5xHis (any position)2 ng (Western blot)Low (E. coli, yeast)

Binding Affinity and Consistency

  • Biolayer Interferometry (BLI): THE™ His Tag Antibody (GenScript) shows consistent binding to His-tagged proteins across multiple lots, with no significant batch-to-batch variability .

  • Lot-to-Lot Consistency: Western blot experiments confirm reproducible detection of His-tagged fusion proteins (e.g., 52 kDa target) .

Cross-Species Reactivity

Mouse IgG1 anti-His tag antibodies exhibit negligible cross-reactivity with non-His-tagged proteins or endogenous IgGs from E. coli, yeast, or mammalian systems .

Subclass-Specific Properties

Mouse IgG1 antibodies differ from other subclasses (e.g., IgG2a, IgG2b) in hinge flexibility and Fcγ receptor binding. While IgG1 shows moderate ADCC activity, its primary role in anti-His tag antibodies is antigen recognition rather than effector functions .

Critical Considerations

  • Reconstitution: Lyophilized antibodies require precise rehydration (e.g., PBS with trehalose) to maintain activity .

  • Storage: Stable at ≤−20°C in lyophilized form; avoid repeated freeze-thaw cycles .

  • Strain-Specific IgG Subclass: Mouse strains like C57Bl/6 express IgG2c, while BALB/c express IgG2a; subclass-specific secondary antibodies may be needed for precise detection .

Product Specs

Introduction
Immunoglobulin G (IgG) antibodies are molecules composed of four peptide chains: two heavy chains and two light chains. Each IgG molecule possesses two antigen-binding sites and plays a crucial role in the secondary immune response. The presence of specific IgG typically indicates a mature antibody response. IgG is also essential for Antibody-dependent cell-mediated cytotoxicity (ADCC) and Intracellular antibody-mediated proteolysis. In these processes, IgG binds to TRIM21 (the receptor with the highest affinity for IgG in humans) to target marked virions for degradation by the proteasome within the cytosol.
Description
Recombinant Mouse IgG, produced in Hi-5 cells, is a single polypeptide chain comprising 242 amino acids (98-330 a.a) with a molecular weight of 27.5 kDa. The IgG protein is tagged with a 6-amino acid His-tag at the C-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The IgG protein is supplied as a 1 mg/ml solution in 20 mM Tris-HCl buffer (pH 8.0) containing 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity is determined to be greater than 90% by SDS-PAGE analysis.
Synonyms
Ig gamma-2A chain C region, A allele, Immunoglobulin heavy chain gamma polypeptide, Ighg, Igh-1, Igh-1a, 1810060O09Rik.
Source
Hi-5 cells.
Amino Acid Sequence
ADPEPRGPTI KPCPPCKCPA PNLLGGPSVF IFPPKIKDVL MISLSPIVTC VVVDVSEDDP DVQISWFVNN VEVHTAQTQT HREDYNSTLR VVSALPIQHQ DWMSGKEFKC KVNNKDLPAP IERTISKPKG SVRAPQVYVL PPPEEEMTKK QVTLTCMVTD FMPEDIYVEW TNNGKTELNY
KNTEPVLDSD GSYFMYSKLR VEKKNWVERN SYSCSVVHEG LHNHHTTKSF SRTPGKHHHH HH.

Q&A

What are the different mouse IgG subclasses and how do they compare functionally?

Mouse IgG exists in four primary subclasses: IgG1, IgG2a, IgG2b, and IgG3. Each subclass demonstrates unique binding characteristics to various Fc gamma receptors (FcγRs), resulting in distinct functional properties. Mouse FcγRI is a high-affinity receptor for IgG2a and a low-affinity receptor for IgG2b and IgG3. Mouse FcγRIII binds IgG1, IgG2a, and IgG2b with low affinity. Mouse FcγRIV demonstrates high affinity for IgG2a and IgG2b but does not bind IgG1 or IgG3. The inhibitory receptor FcγRIIB shows low-affinity binding to IgG1, IgG2a, and IgG2b .

These differential binding profiles make specific subclasses more suitable for certain research applications. For instance, mouse IgG2a strongly engages activating receptors FcγRI and FcγRIV, making it effective for studies requiring potent effector functions. IgG1 interacts with both inhibitory FcγRIIB and activating FcγRIII, providing a more balanced immune activation profile. Understanding these binding characteristics is essential when selecting appropriate antibody subclasses for immunological studies.

How should researchers select the appropriate mouse IgG subclass for specific immunological studies?

Selection of mouse IgG subclasses significantly impacts experimental outcomes due to their differential FcγR binding profiles. For studies requiring potent effector functions like antibody-dependent cellular cytotoxicity (ADCC), mouse IgG2a is optimal as it strongly binds activating receptors FcγRI and FcγRIV. This makes IgG2a particularly effective for cancer immunotherapy research or studies targeting pathogen clearance .

Mouse IgG1 engages both inhibitory FcγRIIB and activating FcγRIII, making it suitable for studies requiring more balanced immune activation. This subclass is often preferred for immunomodulatory studies or when excessive immune activation could be detrimental. For studies focused on complement-dependent cytotoxicity (CDC), mouse IgG3 may be preferable since it has limited FcγR engagement but effectively activates complement .

When designing experiments, researchers should consider the FcγR expression profile on target cell populations. For instance, studies focusing on neutrophil-mediated functions should account for the expression of FcγRIII and FcγRIV on mouse neutrophils. Additionally, researchers must recognize that "mouse FcγRs and FcRn efficiently bind human IgG subclasses, whereas human FcγRs and FcRn do not or poorly bind mouse IgG subclasses," which has important implications for translational studies .

What are the critical considerations when adding a His-tag to mouse IgG antibodies?

When incorporating histidine tags into mouse IgG antibodies, researchers must carefully consider several factors to maintain antibody functionality. The position of the His-tag is particularly critical, as inappropriate placement can interfere with antigen binding or Fc-mediated functions. C-terminal heavy chain His-tags are generally preferred as they position the tag away from both the antigen-binding region and most FcγR interaction sites, although they may potentially affect binding to certain FcγRs that interact with the CH2-CH3 domain interface .

Tag length and composition also require optimization. While 6×His tags are most common, longer tags may provide better purification efficiency but increase the risk of functional interference. Including flexible linker sequences between the antibody and His-tag can minimize structural constraints that might affect antibody function.

When studying FcγR interactions specifically, researchers should validate that His-tagged mouse IgG variants maintain similar binding affinity profiles to native antibodies. This can be accomplished through surface plasmon resonance (SPR) or bio-layer interferometry (BLI) binding assays comparing tagged and untagged versions . For in vivo applications, potential immunogenicity of the His-tag should be evaluated, particularly in repeat-dose studies where anti-tag antibodies might develop and compromise experimental results.

How can researchers generate and purify bispecific mouse IgG antibodies with His-tags?

Generating bispecific mouse IgG antibodies with His-tags presents unique challenges due to the limited technologies available for producing native IgG-like mouse bispecific antibodies . Based on current methodologies, several approaches can be employed:

The controlled Fab-arm exchange (cFAE) protocol has been successfully extended to mouse antibodies. This approach requires parental antibodies with bispecific-enabling mutations to be expressed separately, mixed in vitro under mild reducing conditions to facilitate exchange, and subsequently purified. This technique has been validated for creating surrogate CD3/GP75 bispecific antibodies for targeting mouse melanoma in syngeneic models .

Alternatively, researchers have developed "a simple co-expression system for one-step purification of intact mouse IgG1 and IgG2a bispecific antibodies from any antibody pair" . This system employs complementary charged mutations in CH3 domains to promote heterodimerization, strategic cysteine residues to form inter-chain disulfide bonds, and a His-tag on one chain for selective purification. When both chains are co-expressed in a single cell, proper assembly occurs, allowing for efficient one-step purification.

For effective purification, immobilized metal affinity chromatography (IMAC) using Ni²⁺, Co²⁺, or Cu²⁺ columns can be employed. This is typically followed by size-exclusion chromatography to separate properly assembled bispecific antibodies from other species. Proof-of-concept has been demonstrated with CD3/CD20 bispecific antibodies, which "highlighted both the quality and efficacy of materials generated by this technology" .

What methodological approaches help distinguish between specific and non-specific binding when using His-tagged mouse IgG in binding assays?

Distinguishing between specific and non-specific binding is critical when using His-tagged mouse IgG in binding assays, particularly in FcγR interaction studies. Robust experimental design incorporating multiple controls is essential. Researchers should include non-specific isotype control antibodies with matching His-tag configurations to establish baseline non-specific binding levels. Additionally, comparing binding profiles between tagged and untagged versions of the same antibody can reveal tag-induced artifacts .

Binding kinetics analysis provides valuable information for distinguishing binding types. Specific interactions typically display saturable binding curves with defined KD values, while non-specific binding often presents as linear, non-saturable binding. Comparative kinetic analysis of His-tagged and native antibodies using surface plasmon resonance can identify tag-related alterations in binding characteristics .

Multiple detection methods should be employed to confirm binding through orthogonal approaches. Researchers can use both tag-dependent detection (anti-His antibodies) and tag-independent detection (anti-Fc antibodies) to verify consistent results across different detection systems. Testing binding under varied buffer conditions (different salt concentrations, pH, detergents) can further differentiate between specific and non-specific interactions, as the latter are typically more sensitive to changing buffer compositions .

Competition assays provide additional confirmation of specificity. If excess untagged antibody can compete with His-tagged antibody binding, this strongly suggests that the observed interaction is specific rather than driven by the His-tag itself.

How do mouse strain differences impact the interpretation of results using mouse IgG in FcγR-dependent assays?

Mouse strain differences significantly impact the interpretation of FcγR-dependent assays through variations in receptor expression, polymorphisms, and functional responses. Different mouse strains express varying levels of FcγRs on immune cell populations, which can substantially alter antibody-mediated effector functions. For example, C57BL/6 mice demonstrate different FcγR expression patterns compared to BALB/c mice, potentially leading to divergent responses in antibody-dependent assays .

Genetic polymorphisms further complicate interpretation. The search results indicate that "allelic polymorphism introducing a stop codon" can affect receptor expression patterns . These polymorphisms can result in functional differences in FcγR-mediated responses across strains, even when using identical antibody preparations. Researchers should document the specific strain background and relevant polymorphisms in all experimental reports to facilitate proper interpretation and reproducibility.

When evaluating critical findings, including multiple mouse strains is recommended to establish the generalizability of results. Additionally, normalizing data to strain-specific baseline responses helps account for inherent differences in FcγR expression and function. The use of FcγR knockout mice on different backgrounds can validate receptor dependency and isolate strain-independent effects .

How do cross-species interactions between mouse and human IgG and FcγRs impact experimental design?

Cross-species interactions between mouse and human IgG and FcγRs create complex experimental considerations that must be carefully managed for accurate data interpretation. A fundamental asymmetry exists in these interactions: "mouse FcγRs and FcRn efficiently bind human IgG subclasses, whereas human FcγRs and FcRn do not or poorly bind mouse IgG subclasses" . This has significant implications for translational research and the development of therapeutic antibodies.

Specific cross-species binding patterns require particular attention. Human FcγRI binds mouse IgG2a and IgG2b but not mouse IgG1, while human FcγRIIA binds mouse IgG1, IgG2a, and IgG2b but not mouse IgG3. Human FcγRIIIB does not bind mouse IgG at all . These interaction patterns can create misleading results when testing mouse antibodies in human systems or vice versa.

In FcγR-humanized mouse models, these cross-species interactions present additional challenges. The search results note that "this cross-binding can potentially lead to a competition in vivo for IgG binding between the transgenic human FcγR and the endogenous mouse FcγR. This phenomenon may also induce the aggregation of receptors originating from different species, which result in artifactual situations" . To address this, researchers should ideally express human FcγRs in mice deficient for endogenous FcγRs to avoid competition and artifacts.

Surrogate antibody development requires careful engineering to recapitulate the functionality of human therapeutic antibodies in mouse models. Researchers must consider whether the mouse surrogate engages the appropriate mouse FcγRs to model the intended human therapeutic mechanism . This is particularly important for bispecific antibodies used in syngeneic mouse models.

What are the key differences between human and mouse FcγR expression patterns across immune cell populations?

Human and mouse FcγR expression patterns differ substantially across immune cell populations, which significantly impacts the translation of findings between species. The following key differences warrant careful consideration:

In mice, FcγRI expression is restricted to monocyte-derived dendritic cells, whereas in humans, FcγRI (CD64) is expressed on monocytes/macrophages, dendritic cells, and can be induced on neutrophils and mast cells . This differential expression affects the interpretation of myeloid cell-mediated effector functions between species.

The inhibitory receptor FcγRIIB shows markedly different expression patterns between species. In humans, FcγRIIB (CD32B) expression "is mainly restricted to B cells and basophils," with poor expression on only a small percentage of monocytes and neutrophils . By contrast, mouse FcγRIIB is more broadly expressed across multiple immune cell types, including mast cells. This has profound implications for inhibitory regulation of immune responses.

Human mast cells express only activating FcγRIIA (CD32A) and inducible FcγRI, but lack inhibitory FcγRIIB. In contrast, mouse mast cells express both activating (FcγRIII) and inhibitory (FcγRIIB) receptors . This fundamental difference affects the interpretation of IgG-mediated allergic reactions between species.

NK cells in humans express FcγRIIIA (CD16A) and sometimes FcγRIIC (CD32C), while mouse NK cells express FcγRIII . These differences impact antibody-dependent cellular cytotoxicity (ADCC) mechanisms in cancer and infection models.

Human neutrophils uniquely express FcγRIIIB (CD16B), a GPI-anchored receptor without a direct mouse equivalent . This receptor plays important roles in neutrophil activation and function that cannot be directly modeled in mice.

These differences highlight why "human activating IgG receptors may be negatively regulated by hFcγRIIB only on B cells and basophils, but not on mast cells, NK cells, and on most neutrophils and monocytes that do not express this inhibitory receptor" , creating fundamentally different regulatory landscapes between species.

How do human and mouse IgG subclasses differ in their binding to FcγRs and what are the implications for translational research?

Human and mouse IgG subclasses exhibit fundamental differences in their FcγR binding profiles that have profound implications for translational research. Three critical differences emerge from the search results: First, "all human activating FcγRs bind the major human IgG subclass IgG1, whereas only mouse activating FcγR FcγRIII binds mouse IgG1" . This differential engagement of activating receptors means that human IgG1 antibodies typically drive stronger effector functions than mouse IgG1 antibodies.

Second, "human inhibitory hFcγRIIB has a lower affinity for IgG1, IgG2, and IgG3 than all other hFcγRs, which is not the case in mice for IgG1 and IgG2b" . This creates a fundamentally different balance between activating and inhibitory signaling across species, potentially leading to different functional outcomes even when targeting the same antigen with species-matched antibodies.

Third, while "no human FcγR binds human IgE, 3 of the 4 mouse FcγRs (FcγRIIB, FcγRIII, FcγRIV) bind mouse IgE" . This cross-isotype binding in mice creates unique regulatory mechanisms without human parallels, complicating the interpretation of studies involving IgE-mediated allergic responses.

For translational research, these differences necessitate careful antibody engineering. When developing therapeutic antibodies in mouse models, researchers must consider that human IgG1 antibodies will engage multiple mouse FcγRs, potentially leading to distinct effector functions compared to human systems . Conversely, mouse therapeutic antibodies may exhibit unexpected behavior in humanized models due to different receptor binding profiles.

How can researchers optimize mouse IgG antibodies for studying therapeutic mechanisms in syngeneic tumor models?

Optimizing mouse IgG antibodies for syngeneic tumor models requires strategic selection of IgG subclasses based on the desired mechanism of action. For therapies intended to activate immune effector functions, mouse IgG2a represents the optimal subclass as it strongly engages activating FcγRs, particularly FcγRI and FcγRIV. This engagement enables potent antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) by NK cells and macrophages within the tumor microenvironment .

For checkpoint blockade antibodies where pure blocking activity without immune cell recruitment is desired, researchers might prefer mouse IgG1 or engineered variants with reduced FcγR binding. Understanding the FcγR expression profile on tumor-infiltrating immune cells in the specific model is critical for predicting antibody efficacy. As noted in the search results, studies using model systems have "led to the identification of specific cell populations responsible for the induction of various inflammatory diseases" , highlighting the importance of characterizing the local immune environment.

For bispecific antibody approaches, the search results indicate that "surrogate mouse bispecific antibodies are needed to accommodate repeated dosing schedules in immuno-oncology studies using syngeneic mouse models, which require intact immune systems" . The development of mouse bispecific platforms that enable "one-step purification of intact mouse IgG1 and IgG2a bispecific antibodies from any antibody pair" provides valuable tools for such studies .

When designing studies with repeated antibody administration, researchers should monitor for potential anti-drug antibody responses, particularly when using engineered formats. Additionally, incorporating imaging techniques to confirm tumor localization helps validate that antibody distribution correlates with observed therapeutic effects. Biodistribution studies comparing different IgG subclasses or formats can provide critical insights into tumor penetration and retention properties that influence efficacy.

What considerations should researchers address when using mouse IgG to study FcγR-mediated inflammatory diseases?

When studying FcγR-mediated inflammatory diseases using mouse IgG, researchers must carefully select appropriate disease models and antibody subclasses. The search results indicate that "the in vivo roles of IgG receptors have been addressed using specific FcR knockout mice or in mice expressing a single FcR, and have demonstrated a predominant contribution of mouse activating IgG receptors FcγRIII and FcγRIV to models of autoimmunity (eg, arthritis) and allergy (eg, anaphylaxis)" . This highlights the importance of understanding which FcγRs drive pathology in specific disease models.

Cell type-specific considerations are essential, as different immune cells express distinct FcγR profiles. The search results reveal that studies "led to the identification of specific cell populations responsible for the induction of various inflammatory diseases and have revealed, in particular, the unexpected contribution of neutrophils and monocytes to the induction of anaphylactic shock" . Researchers should characterize FcγR expression on disease-relevant cell populations to accurately interpret antibody-mediated effects.

The presence or absence of inhibitory FcγR signaling significantly impacts disease manifestation. While human FcγRIIB expression "is mainly restricted to B cells and basophils," mouse FcγRIIB is more broadly expressed . This means that inhibitory regulation of immune responses fundamentally differs between species, potentially leading to different disease outcomes or therapeutic responses. When developing therapeutic antibodies targeting inflammatory diseases, researchers should consider engineering Fc regions that selectively engage either activating or inhibitory FcγRs as appropriate for the disease mechanism.

Dosing and pharmacokinetic considerations are crucial, as antibody half-life and tissue distribution affect local concentrations at inflammatory sites. Researchers should determine antibody pharmacokinetic properties in the specific disease model and adjust dosing regimens accordingly. Monitoring for anti-drug antibody responses with repeated dosing is particularly important in chronic inflammatory disease models.

How do mouse bispecific IgG antibodies compare to other formats in T-cell engagement studies?

Mouse bispecific IgG antibodies offer distinct advantages for T-cell engagement studies compared to other formats, particularly in the context of syngeneic mouse models. The search results describe "CD3/CD20 bispecific antibodies" as proof of concept, which "highlighted both the quality and efficacy of materials generated by this technology" . Several aspects warrant consideration when comparing formats:

IgG-like bispecific antibodies maintain the advantages of natural antibody architecture including extended half-life through FcRn-mediated recycling and potential Fc-mediated functions. This contrasts with smaller formats like BiTEs (Bispecific T-cell Engagers) which typically demonstrate shorter half-lives but potentially better tumor penetration. The physiological half-life of IgG-based formats enables less frequent dosing in extended studies, which is particularly valuable for chronic models requiring repeated administration .

Production methods impact antibody quality and consistency. The search results describe "a simple co-expression system for one-step purification of intact mouse IgG1 and IgG2a bispecific antibodies from any antibody pair" . This single-batch production approach potentially reduces heterogeneity compared to post-production assembly methods like controlled Fab-arm exchange, which requires in vitro mixing under reducing conditions.

The spatial arrangement between T cells and target cells differs between formats, potentially affecting functional outcomes. IgG-like bispecific antibodies create different synaptic geometries compared to smaller formats, which may influence T-cell activation kinetics and cytolytic efficiency. When designing T-cell engaging bispecific antibodies, researchers should consider the distance between epitopes on target cells and T cells to optimize engagement.

For in vivo studies, researchers must evaluate pharmacokinetics, biodistribution, and potential immunogenicity of different formats. The intact IgG format typically demonstrates longer persistence and more restricted tissue distribution compared to fragment-based approaches. In syngeneic models where repeated dosing is required, the potential for anti-drug antibody responses should be assessed, particularly for highly engineered formats that might present novel epitopes to the immune system .

What is the binding affinity profile of different mouse IgG subclasses to mouse FcγRs compared to human IgG-FcγR interactions?

The binding affinity profiles of mouse IgG subclasses to mouse FcγRs reveal distinct patterns that differ significantly from human IgG-FcγR interactions. Mouse FcγRI demonstrates high affinity (KD < 10⁻⁸ M) for mouse IgG2a only, with low affinity for IgG2b and IgG3, and no binding to IgG1. In contrast, human FcγRI binds human IgG1, IgG3, and IgG4 with high affinity .

Mouse FcγRIII exhibits low-affinity binding (KD ~10⁻⁶ M) to mouse IgG1, IgG2a, and IgG2b, but not to IgG3. Critically, it is "the only mouse activating FcγR that binds mouse IgG1" . This differs markedly from human activating receptors, where "all human activating FcγRs bind the major human IgG subclass IgG1" .

Mouse FcγRIV, which has no direct human equivalent, shows high affinity for both IgG2a and IgG2b (KD < 10⁻⁸ M) but does not bind IgG1 or IgG3. This receptor has been described as "functionally similar to human FcγRIIIA or FcγRI" in certain contexts .

The inhibitory receptor mouse FcγRIIB binds mouse IgG1, IgG2a, and IgG2b with low affinity. Importantly, in mice, this inhibitory receptor has comparable affinity to activating receptors for some IgG subclasses, which is not the case in humans where "human inhibitory hFcγRIIB has a lower affinity for IgG1, IgG2, and IgG3 than all other hFcγRs" .

A unique feature of the mouse system is that "3 of the 4 mouse FcγRs (FcγRIIB, FcγRIII, FcγRIV) bind mouse IgE," whereas "no human FcγR binds human IgE" . This cross-isotype binding in mice creates regulatory mechanisms without human parallels.

These differences highlight why direct translation of findings between mouse and human systems requires careful interpretation, and why engineered antibodies with modified Fc regions are often necessary to recapitulate human antibody functions in mouse models.

What are the expression patterns of mouse FcγRs across immune cell populations compared to human FcγRs?

NK cells in mice express primarily FcγRIII, while human NK cells express FcγRIIIA (CD16A) and sometimes FcγRIIC (CD32C). Additionally, a recently identified polymorphism can lead to FcγRIIB expression on human NK cells, which "negatively regulate IgG-induced NK cell activation" . This fundamentally alters how antibody-dependent cellular cytotoxicity is regulated between species.

Neutrophils show particularly distinct receptor profiles. Mouse neutrophils express FcγRIII and FcγRIV with inducible FcγRI. Human neutrophils express FcγRIIA, FcγRIIIB (a GPI-anchored receptor unique to humans), inducible FcγRI, and low levels of FcγRIIB on a small percentage of cells . These differences affect how neutrophils respond to immune complexes and opsonized pathogens.

Mast cells show a critical regulatory difference: "human activating IgG receptors may be negatively regulated by hFcγRIIB only on B cells and basophils, but not on mast cells, NK cells, and on most neutrophils and monocytes that do not express this inhibitory receptor" . Mouse mast cells express inhibitory FcγRIIB alongside activating receptors, creating a different regulatory balance than in humans.

Dendritic cells exhibit different receptor distributions, with mouse FcγRI expression restricted to monocyte-derived DCs, whereas human FcγRI is more broadly expressed . This impacts antigen presentation and DC activation in response to immune complexes.

These expression differences underscore why mouse models may not always predict human responses to therapeutic antibodies, particularly for mechanisms relying on specific FcγR-expressing cell populations. Researchers must consider these species differences when designing experiments and interpreting results for translation to human applications.

How do transgenic mouse models expressing human FcγRs contribute to translational antibody research?

Transgenic mouse models expressing human FcγRs provide valuable systems for studying human antibody functions in vivo, though with important limitations. These models have been developed to express individual human FcγRs or combinations of receptors. The search results mention mice transgenic for human FcγRIIA, FcγRIIB, FcγRIIIA, and FcγRI, among others . These models enable the study of human receptor-mediated functions in the context of a living organism, bridging the gap between in vitro human cell studies and conventional mouse models.

Expression patterns of transgenic receptors may not perfectly recapitulate human patterns. Models with human FcγRs may show "abnormal expression on several cell populations" , potentially leading to non-physiological responses. Additionally, studies using mice transgenic for hFcγRIIA on a wild-type background reported "spontaneous autoimmune diseases (ie, pneumonitis, glomerulonephritis, and rheumatoid arthritis)" , highlighting potential adverse effects of introducing human receptors into the mouse immune system.

To address these limitations, researchers have developed more sophisticated models. The search results indicate that "the analysis of the properties of human FcRs in vivo in inflammatory models of disease requires that the transgenes encoding human FcRs are expressed in mice deficient for endogenous FcRs to avoid competition for ligands or artifacts" . These models provide cleaner systems for studying human FcγR functions.

Despite these challenges, FcγR-humanized mice have provided valuable insights. For example, "hFcγRI tg mice have revealed that hFcγRI enables antigen targeting to DCs for enhanced protective immunity and IgG-dependent protection against malaria infection" . These models continue to evolve, providing increasingly relevant systems for translational antibody research.

Product Science Overview

Introduction

The Immunoglobulin Heavy Chain Constant Region Gamma 2a (Mouse Recombinant, His Tag) is a recombinant protein derived from the mouse immunoglobulin heavy chain. This protein is significant in various research and clinical applications due to its role in the immune response.

Structure and Composition

The Immunoglobulin Heavy Chain Constant Region Gamma 2a is a part of the immunoglobulin G (IgG) class of antibodies. It is composed of a single polypeptide chain containing 242 amino acids, with a molecular mass of approximately 27.5 kDa . The protein is fused to a 6-amino acid His-tag at the C-terminus, which facilitates its purification through affinity chromatography techniques .

Expression System

This recombinant protein is produced in Hi-5 cells, a type of insect cell line commonly used for the expression of recombinant proteins . The use of Hi-5 cells ensures high yield and proper folding of the protein, which is crucial for its functionality in research applications.

Function and Applications

The Immunoglobulin Heavy Chain Constant Region Gamma 2a plays a critical role in the immune system. It is involved in the effector functions of antibodies, such as complement activation and binding to Fc receptors on immune cells . These functions are essential for the clearance of pathogens and the regulation of immune responses.

In research, this recombinant protein is used in various assays and experiments to study antibody functions, interactions with other immune components, and the mechanisms of immune responses. It is also utilized in the development of therapeutic antibodies and diagnostic tools.

Stability and Storage

The recombinant protein is provided as a sterile filtered colorless solution, typically at a concentration of 1 mg/ml in a buffer containing 20 mM Tris-HCl (pH 8.0) and 10% glycerol . For short-term storage, it can be kept at 4°C if used within 2-4 weeks. For long-term storage, it is recommended to store the protein at -20°C, with the addition of a carrier protein such as 0.1% HSA or BSA to prevent degradation . It is important to avoid multiple freeze-thaw cycles to maintain the protein’s stability and functionality.

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 2024 Thebiotek. All Rights Reserved.