CD55 Mouse

CD55 Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Complement Regulation

CD55 protects host cells from autologous complement-mediated damage by:

  • Accelerating decay of C3/C5 convertases .

  • Cooperating with CD46 and CD59 to prevent C3 deposition and membrane attack complex (MAC) formation .

Granulocyte Homeostasis

CD55-deficient mice exhibit two-fold increases in circulating granulocytes due to enhanced granulopoiesis in the bone marrow . This phenotype is independent of complement activity and instead involves interactions with the adhesion GPCR CD97 .

Host Defense

  • Cd55−/− mice show improved survival during Streptococcus pneumoniae infections, with reduced bacteremia .

  • CD55 deficiency enhances neutrophil-mediated bacterial clearance by altering granulocyte margination and proliferation .

Wound Healing

CD55 inhibition via antibody blockade accelerates wound closure in burn and corneal injury models by potentiating C3a/C5a receptor (C3ar1/C5ar1) signaling .

Table 1: Phenotypic Effects of CD55 Deficiency in Mice

ParameterCd55−/− vs. Wild-TypeMechanism
Circulating granulocytes↑ 2-fold in blood, spleen, and marginated pool Enhanced granulopoiesis
Bone marrow activity↑ Gr-1+ cells in S/G2/M phase Increased granulopoietic proliferation
Bacterial clearance↓ Bacteremia in S. pneumoniae infection Enhanced neutrophil function
Wound healingAccelerated epithelialization and angiogenesis Potentiated C3ar1/C5ar1 signaling

Table 2: CD55 Overexpression in iPSCs

ParameterCD55-Overexpressing vs. ControlImplications
C3 deposition↓ 50–60% on iPSCs and kidney organoids Reduced complement activation
C5b-9 (MAC) formation↓ 30–40% Protection against MAC-mediated lysis

Research Tools and Antibodies

Key reagents for studying mouse CD55 include:

  • Clone 3D5: Used for Western blot (WB) and immunohistochemistry (IHC) .

  • Clone RIKO-3: Recognizes both GPI- and TM-CD55 isoforms; validated for flow cytometry (FC) .

  • Anti-CCP2–3 antibody: Blocks CD55 function, accelerating wound healing in vivo .

Therapeutic Implications

  • Infectious Disease: CD55 blockade may enhance neutrophil responses in bacterial infections .

  • Transplantation: Overexpression in iPSCs reduces complement-mediated damage, improving graft survival .

  • Autoimmunity: Modulating CD55-C3ar1/C5ar1 signaling could suppress inflammatory diseases .

Product Specs

Introduction
CD55, a member of the RCA (regulators of complement activation) family, is characterized by the presence of 4-30 SCRs (short consensus repeats) in their plasma-exposed regions. This protein is widely expressed in various malignant tumors and functions as an inhibitor of the complement system.
Description
CD55, produced in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain consisting of 336 amino acids (35-362 a.a.). It has a molecular mass of 36.8kDa and migrates at 40-57kDa on SDS-PAGE under reducing conditions. This protein is expressed with an 8 amino acid His tag at the C-Terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered colorless solution
Formulation
The CD55 protein solution (0.5mg/ml) is supplied in Phosphate Buffered Saline (pH 7.4) with 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For long-term storage, it is recommended to store the product frozen at -20°C. To ensure stability during long-term storage, the addition of a carrier protein (0.1% HSA or BSA) is recommended. Avoid multiple freeze-thaw cycles.
Purity
The purity of CD55 is greater than 95.0% as determined by SDS-PAGE.
Synonyms
Complement decay-accelerating factor, GPI-anchored, DAF-GPI, CD_antigen: CD55.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
DCGPPPDIPN ARPILGRHSK FAEQSKVAYS CNNGFKQVPD KSNIVVCLEN GQWSSHETFC EKSCVAPERL SFASLKKEYL NMNFFPVGTI VEYECRPGFR KQPPLPGKAT CLEDLVWSPV AQFCKKKSCP NPKDLDNGHI NIPTGILFGS EINFSCNPGY RLVGVSSTFC SVTGNTVDWD DEFPVCTEIH CPEPPKINNG IMRGESDSYT YSQVVTYSCD KGFILVGNAS IYCTVSKSDV GQWSSPPPRC IEKSKVPTKK PTINVPSTGT PSTPQKPTTE SVPNPGDQPT PQKPSTVKVS ATQHVPVTKT TVRHPIRTST DKGEPNTGLE HHHHHH.

Q&A

What is CD55 and what is its function in mice?

CD55, also known as Decay Accelerating Factor (DAF), is a member of the Receptors of complement activation (RCA) protein family. It functions as a critical regulator of the complement system by recognizing C4b and C3b fragments that interact with cell-surface hydroxyl or amino groups during complement activation. In mice as in humans, CD55 is expressed on the plasma membranes of cells that come into direct contact with plasma complement proteins . Its primary function is to accelerate the decay of C3 and C5 convertases, thereby inhibiting complement activation and protecting host cells from complement-mediated damage .

How do mouse and human CD55 differ structurally and functionally?

While mouse and human CD55 share core functional properties as complement regulators, they exhibit several structural and functional differences:

  • Molecular Weight: Human CD55 has a reported length of 381 amino acid residues and a mass of approximately 41.4 kDa, whereas mouse CD55 has some variations in size and glycosylation patterns .

  • Isoform Diversity: Human CD55 has up to 7 different isoforms reported, while mouse CD55 may have fewer documented variants .

  • Complement Inhibition Efficacy: Human CD55 appears to have stronger inhibitory effects on complement activation in certain experimental contexts, which is why transgenic mice expressing human CD55 are often used to study enhanced complement regulation .

  • Post-translational Modifications: Both mouse and human CD55 undergo O-glycosylation, but the specific patterns and extent may differ between species .

What are the common methods to detect CD55 expression in mouse tissues?

CD55 expression in mouse tissues can be detected using several methodological approaches:

MethodApplicationsAntibody TypesTypical Tissue PreparationDetection Sensitivity
Western Blot (WB)Protein quantificationMonoclonal or polyclonalTissue lysatesModerate to high
Immunohistochemistry (IHC)Tissue localizationPrimarily monoclonalFixed, paraffin-embedded or frozen sectionsModerate
Flow Cytometry (FCM)Cellular expressionFluorophore-conjugated monoclonalSingle-cell suspensionsHigh
Immunofluorescence (IF)Subcellular localizationUnconjugated primary + fluorescent secondaryFixed cells or tissuesHigh
ELISASoluble protein detectionCapture and detection antibody pairsSerum or tissue homogenatesVery high

For optimal results, researchers should select antibodies specifically validated for mouse CD55 detection. Available antibodies include unconjugated forms and those conjugated to fluorophores like PE, APC, or FITC for direct detection .

How does overexpression of human CD55 in transgenic mice affect complement regulation in renal ischemia-reperfusion injury models?

Transgenic mice expressing human CD55 (hCD55) demonstrate enhanced protection against renal ischemia-reperfusion injury (IRI) through superior complement regulation. In mild IRI models (18 minutes of warm renal ischemia), hCD55 transgenic mice show significantly reduced serum creatinine and urea levels compared to wild-type littermates, indicating preserved renal function .

The protective mechanisms include:

  • Reduced C3b/c Deposition: hCD55 transgenic mice show decreased deposition of C3b/c in renal tissue following IRI, indicating effective inhibition of the C3 convertase .

  • Decreased C9 Deposition: These mice also demonstrate reduced C9 deposition, suggesting inhibition of the terminal complement pathway .

  • Attenuated Inflammatory Cell Infiltration: Neutrophil and macrophage infiltration into injured renal tissue is significantly reduced in hCD55 transgenic mice .

What are the synergistic effects of combined human CD55 and CD59 expression in complement-mediated pathologies?

The combined expression of human CD55 and CD59 (hCD55/hCD59) in transgenic mice provides superior protection against complement-mediated pathologies compared to hCD55 expression alone. This synergistic effect was demonstrated in a moderate renal IRI model, where:

  • Enhanced Protection Against Moderate IRI: While hCD55 alone failed to preserve renal function in moderate IRI (22 minutes ischemia), mice expressing both hCD55 and hCD59 maintained significantly better renal function with reductions in all measured damage parameters .

  • Complementary Regulatory Mechanisms: This synergy arises from the complementary modes of action - hCD55 inhibits the C3 and C5 convertases (early and mid-complement pathway), whereas hCD59 regulates the terminal complement pathway by preventing formation of the membrane attack complex .

  • Additive Anti-inflammatory Effects: The combined expression resulted in more profound reductions in neutrophil and macrophage infiltration compared to hCD55 alone, suggesting enhanced anti-inflammatory properties .

This suggests that comprehensive regulation of the complement cascade at multiple levels provides optimal protection against complement-mediated tissue damage, particularly in severe pathological conditions .

What are the technical considerations for using recombinant human CD55 (rhCD55) as a therapeutic agent in mouse models?

The therapeutic application of recombinant human CD55 (rhCD55) in mouse models requires several technical considerations:

  • Timing of Administration: Research suggests that rhCD55 can be protective when administered immediately after reperfusion in renal IRI models, indicating that early intervention during the acute phase of complement activation is crucial .

  • Dosage Optimization: Effective dosing must be determined based on the severity of the model and complement activation extent. In published studies, rhCD55 demonstrated protection in moderate IRI models, suggesting adequate cross-species activity .

  • Delivery Method: The mode of delivery (intravenous, intraperitoneal, or site-specific) affects bioavailability and efficacy. Systemic delivery may be suitable for acute conditions like IRI, while local delivery might be preferable for chronic or localized pathologies.

  • Cross-species Functionality: Although human CD55 is being administered to mice, research confirms its ability to regulate mouse complement, though possibly with different efficiency compared to native mouse CD55 .

  • Pharmacokinetics and Half-life: The soluble rhCD55 likely has a shorter half-life compared to membrane-bound CD55, potentially necessitating repeated dosing for sustained complement inhibition.

What are the optimal protocols for assessing complement activation in CD55 mouse models?

Assessment of complement activation in CD55 mouse models requires rigorous methodological approaches:

  • In Vitro Complement Activation Assay:

    • Modified mouse complement ELISA assays can be used to measure complement activation in mouse serum

    • Mouse IgM-coated microplates incubated with 5% normal mouse serum (NMS) at 37°C for 60 minutes

    • Activation can be assessed with or without mouse PBMCs (50,000 cells per well) to evaluate cellular involvement

  • Tissue Complement Deposition Analysis:

    • Immunohistochemistry or immunofluorescence using anti-C3b/c and anti-C9 antibodies

    • Quantification through digital imaging analysis for objective measurement

    • Comparison between transgenic (hCD55, hCD55/hCD59) and wild-type littermates

  • Functional Complement Assays:

    • Hemolytic assays using sensitized erythrocytes to assess complement-mediated lysis

    • CH50 assays to measure total complement activity

    • Analysis of activated complement components in serum (C3a, C5a) by ELISA

  • Regulatory Protein Expression Analysis:

    • Flow cytometry for cell surface expression using specific anti-CD55 antibodies

    • Western blot for total protein levels in tissue homogenates

    • qRT-PCR for mRNA expression levels

How should researchers design experiments to distinguish CD55-specific effects from other complement regulatory mechanisms?

Designing experiments to isolate CD55-specific effects requires careful controls and methodological precision:

  • Genetic Approaches:

    • Use CD55 knockout mice as negative controls

    • Compare with other complement regulator knockouts (CD59, Crry) to distinguish specific pathways

    • Employ conditional or inducible CD55 expression systems to control timing of CD55 activity

  • Pharmacological Inhibition Strategy:

    • Use anti-CD55 blocking antibodies alongside appropriate isotype controls

    • Apply selective inhibitors targeting specific steps in complement activation

    • Compare with broad-spectrum complement inhibitors to identify CD55-specific contributions

  • Cross-breeding Experimental Design:

    • Generate double transgenic/knockout mice (e.g., CD55-/-/CD59-/-)

    • Cross CD55 transgenic mice with models of specific diseases to assess contextual importance

    • Compare hCD55 and hCD55/hCD59 transgenic mice to evaluate regulatory synergy

  • In Vitro Depletion/Reconstitution:

    • Deplete CD55 from cell surfaces using enzymatic methods

    • Reconstitute with recombinant CD55 to confirm specificity of observations

    • Use siRNA knockdown of CD55 in primary cells from transgenic models

  • Control for Species-Specific Differences:

    • Include both human and mouse CD55 in parallel experiments when using transgenic models

    • Account for potential differences in interaction with other complement components

What are the key considerations for immunohistochemical detection of CD55 in mouse tissue samples?

Successful immunohistochemical detection of CD55 in mouse tissues requires attention to several critical factors:

  • Antibody Selection:

    • Choose antibodies validated specifically for mouse CD55 (for native expression) or human CD55 (for transgenic models)

    • Consider using monoclonal antibodies for higher specificity, particularly in fresh frozen sections

    • If studying transgenic models, ensure antibodies can distinguish between human and mouse CD55

  • Tissue Fixation and Processing:

    • Fresh frozen sections often yield better results than formalin-fixed paraffin-embedded (FFPE) tissues

    • If using FFPE tissues, optimization of antigen retrieval is crucial (test both heat-induced epitope retrieval and enzymatic methods)

    • Fixation time should be minimized to prevent excessive cross-linking that may mask CD55 epitopes

  • Detection Systems:

    • Amplification systems (e.g., tyramide signal amplification) may be necessary for low expression levels

    • Fluorescent detection allows for co-localization studies with other markers

    • Chromogenic detection with DAB offers long-term stability and conventional microscopy compatibility

  • Controls:

    • Positive controls: tissues known to express high levels of CD55 (kidney, spleen)

    • Negative controls: CD55 knockout tissues or isotype antibody controls

    • Blocking peptide controls to confirm antibody specificity

  • Quantification Methods:

    • Digital image analysis for objective quantification

    • Establish clear scoring criteria for manual assessment (intensity, percentage of positive cells)

    • Use standardized reference samples across experiments for consistency

How can researchers accurately interpret CD55 expression data across different mouse strains and experimental conditions?

Accurate interpretation of CD55 expression data requires consideration of several variables that can influence results:

  • Strain-Dependent Variations:

    • Baseline CD55 expression varies significantly across different mouse strains (C57BL/6, BALB/c, CBA)

    • Some strains may have natural polymorphisms affecting CD55 expression or function

    • Always use appropriate strain-matched controls and report the precise strain background

  • Age and Sex Considerations:

    • CD55 expression can vary with age and sexual maturity

    • Sex-based differences in complement regulation may affect CD55 expression and function

    • Match experimental and control groups for age and sex, and consider analyzing these variables separately

  • Normalization Strategies:

    • For quantitative analyses, normalize CD55 expression to stable housekeeping genes or proteins

    • Consider using multiple reference genes/proteins to minimize bias

    • Report raw and normalized data when possible to allow for reinterpretation

  • Statistical Analysis:

    • Apply appropriate statistical tests based on data distribution

    • Account for multiple comparisons when analyzing CD55 across different tissues or conditions

    • Report effect sizes alongside p-values to convey biological significance

  • Experimental Condition Standardization:

    • Control environmental factors that may affect immune status and complement activation

    • Document housing conditions, microbiome status, and health screening results

    • Consider circadian variations in complement activity when planning experiments

What are the critical controls needed when studying transgenic mice expressing human CD55?

When working with transgenic mice expressing human CD55, several critical controls should be implemented:

  • Genetic Background Controls:

    • Use wild-type littermates as primary controls to account for background genetics

    • If backcrossed to a specific strain (e.g., C57BL/6), document the number of generations to ensure genetic homogeneity

    • Consider using heterozygous mice as intermediate controls to assess gene dosage effects

  • Expression Level Verification:

    • Quantify human CD55 expression levels in each experimental cohort

    • Verify tissue-specific expression patterns if using tissue-specific promoters

    • Monitor expression stability across generations and with aging

  • Functional Validation:

    • Confirm that human CD55 is functionally active in the mouse environment

    • Assess complement regulation capacity using functional assays

    • Verify that the human protein localizes correctly to cell membranes

  • Phenotypic Characterization:

    • Document any baseline phenotypic differences in transgenic mice without experimental intervention

    • Assess for compensatory changes in other complement regulators

    • Evaluate potential developmental effects of constitutive human CD55 expression

  • Species-Specific Analysis:

    • Use antibodies that distinguish between human and mouse CD55

    • Include species-specificity controls when measuring functional outcomes

    • Consider potential interactions between human CD55 and mouse complement components

How should researchers design experiments to evaluate the therapeutic potential of CD55-targeted interventions?

Designing experiments to evaluate CD55-targeted therapeutics requires a comprehensive approach:

  • Model Selection:

    • Choose disease models where complement dysregulation is a known pathogenic factor

    • Consider both acute (e.g., ischemia-reperfusion injury) and chronic (e.g., autoimmune) models

    • Include models with varying severity to determine therapeutic windows

  • Intervention Timing:

    • Test both preventive (pre-insult) and therapeutic (post-insult) administration

    • Include time-course studies to determine optimal intervention points

    • Assess both immediate and long-term outcomes to capture full therapeutic impact

  • Dosing Strategy:

    • Conduct dose-response experiments to establish minimum effective doses

    • Compare single versus repeated dosing regimens

    • Evaluate different routes of administration for optimal bioavailability

  • Combination Approaches:

    • Test CD55-based interventions alone and in combination with other complement regulators

    • Compare with standard-of-care treatments to assess relative efficacy

    • Explore synergistic potential with non-complement targeted therapies

  • Comprehensive Outcome Assessment:

    • Measure multiple parameters: functional, histological, biochemical, and molecular

    • Include biomarkers of complement activation to confirm mechanism of action

    • Assess potential immunogenicity or adverse effects, especially with recombinant human proteins in mice

What are the common pitfalls in CD55 detection and quantification in mouse models?

Researchers frequently encounter several technical challenges when detecting and quantifying CD55:

  • Antibody Cross-Reactivity Issues:

    • Some antibodies may cross-react between mouse and human CD55, confounding results in transgenic models

    • Certain antibodies may recognize specific isoforms but not others, leading to incomplete detection

    • Thorough validation with positive and negative controls is essential before experimental use

  • Post-translational Modification Interference:

    • Heavy glycosylation of CD55 can mask epitopes and interfere with antibody binding

    • Sample preparation methods may alter glycosylation patterns

    • Consider using deglycosylation treatments in parallel experiments to confirm protein identity

  • Expression Level Variability:

    • CD55 expression can be modulated by inflammatory stimuli, potentially introducing variability

    • Expression may vary significantly between tissues and cell types within the same animal

    • Standardize sample collection timing and conditions to minimize variability

  • Soluble Versus Membrane-Bound Forms:

    • Detection methods may preferentially capture either membrane-bound or soluble forms

    • Some assays may not distinguish between these forms, affecting data interpretation

    • Specify which form is being measured and consider measuring both when relevant

  • Tissue Preparation Artifacts:

    • Fixation can alter antigenicity, especially for heavily glycosylated proteins like CD55

    • Freeze-thaw cycles may degrade the protein or affect membrane localization

    • Optimize and standardize tissue preparation protocols specifically for CD55 detection

How can researchers address data inconsistencies between in vitro and in vivo CD55 functional studies?

Reconciling discrepancies between in vitro and in vivo CD55 functional studies requires systematic investigation:

  • Microenvironment Differences:

    • In vitro systems lack the complex microenvironment that may modulate CD55 function in vivo

    • Consider using more complex in vitro systems (e.g., 3D cultures, co-cultures) to better approximate in vivo conditions

    • Document differences in complement component concentrations between in vitro media and in vivo fluids

  • Temporal Dynamics:

    • In vitro experiments often represent static snapshots, while in vivo systems have dynamic regulation

    • Design time-course experiments that capture the kinetics of CD55 function

    • Consider using intravital imaging to observe CD55 function in real-time in vivo

  • Compensatory Mechanisms:

    • In vivo systems may engage compensatory complement regulators absent in simplified in vitro models

    • Assess the expression and activity of other complement regulators in parallel

    • Use combined inhibition approaches to account for redundant regulatory systems

  • Experimental Readout Alignment:

    • Ensure that in vitro and in vivo experiments measure comparable endpoints

    • Develop bridging assays that can be performed in both settings

    • When possible, collect samples from in vivo experiments for parallel in vitro analysis

  • Verification Through Multiple Approaches:

    • Use both genetic (knockout/transgenic) and pharmacological (antibodies/inhibitors) approaches

    • Employ both gain-of-function and loss-of-function methodologies

    • Validate key findings using primary cells isolated from experimental animals

Product Science Overview

Structure and Function

CD55 is a glycoprotein that is broadly distributed among hematopoietic and non-hematopoietic cells. It is a 70-75 kDa membrane protein that attaches to the cell membrane via a glycophosphatidylinositol (GPI) anchor . The protein contains four complement control protein (CCP) repeats, which are involved in its inhibition of the complement pathways. Specifically, CCP2 and CCP3 are involved in the inhibition of the classical pathway, while CCP2, CCP3, and CCP4, along with three consecutive lysine residues in a positively charged pocket between CCP2 and CCP3, are involved in the inhibition of the alternate complement pathway .

Role in Immune Regulation

CD55 regulates the complement system by recognizing C4b and C3b fragments that are created during the activation of C4 (classical or lectin pathway) or C3 (alternative pathway). Interaction of CD55 with cell-associated C4b interferes with the conversion of C2 to C2b, thereby preventing the formation of the C4b2a C3-convertase. Similarly, interaction of CD55 with C3b interferes with the conversion of factor B to Bb by factor D, thereby preventing the formation of the C3bBb C3 convertase of the alternative pathway . By limiting the amplification convertases of the complement cascade, CD55 indirectly blocks the formation of the membrane attack complex.

Recombinant Mouse CD55

Recombinant mouse CD55 is produced using a mouse myeloma cell line, NS0-derived mouse CD55/DAF protein. The recombinant protein is typically purified to a high degree of purity, often greater than 90%, as determined by SDS-PAGE under reducing conditions and visualized by silver stain . The recombinant protein is often used in various research applications, including functional assays and binding studies.

Applications and Research

Recombinant mouse CD55 has been used in various research areas, including studies on immune evasion by pathogens, granulocyte homeostasis, and anti-inflammatory responses . For example, CD55 has been shown to facilitate immune evasion by certain pathogens, such as Borrelia crocidurae, the causative agent of relapsing fever . Additionally, CD55 is a ligand of the adhesion class G protein-coupled receptor CD97, and this interaction has been studied in the context of granulocyte homeostasis .

Pathological Implications

CD55 is also implicated in certain pathological conditions. For instance, in paroxysmal nocturnal hemoglobinuria (PNH), a disorder characterized by complement-mediated hemolysis, the expression of CD55 is reduced due to mutations that affect GPI anchor synthesis . This reduction in CD55 expression leads to increased susceptibility of red blood cells to complement-mediated lysis, resulting in symptoms such as anemia, fatigue, and episodes of dark-colored urine.

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.