CRT2 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to CRT2 Antibody

The CRT2 antibody is a monoclonal antibody that recognizes a 150 kDa S-layer protein found in Campylobacter rectus, a Gram-negative bacterium associated with periodontitis . In the context of complement and adaptive immunity, complement receptor type 2 (CR2) acts as an interface between the complement system and adaptive immunity . CR2 is also involved in innate immunity, particularly in the development of natural antibodies targeting neo-epitopes in ischemic tissue during reperfusion .

CRT2 Antibody in Campylobacter rectus Research

CRT2 antibody specifically targets Campylobacter rectus, a bacterium implicated in periodontitis .

Key findings:

  • CRT2 recognizes a 150 kDa S-layer protein on C. rectus .

  • CRT2 reacts with all C. rectus strains, except for the S-layer negative strain C. rectus ATCC 33238 .

  • CRT2 antibody can detect as few as 10³ organisms in subgingival dental plaque samples using the dot-blot method .

  • Detection of C. rectus using CRT2 significantly correlates with clinical findings such as probing depth (P < 0.001), bleeding on probing (P < 0.001), and gingival index (P < 0.001) .

These findings suggest that C. rectus infection, detected by CRT2, is an indicator of periodontal disease status .

Complement Receptor Type 2 (CR2) and Immunity

CR2 serves as a crucial link between the complement system and adaptive immunity .

Functions of CR2:

  • Interface between complement and adaptive immunity .

  • Involvement in innate immunity .

  • Development of natural antibodies targeting neo-epitopes in ischemic tissue undergoing reperfusion .

  • Binds C3 complement fragments and is involved in the uptake and retention of immunocomplexes .

  • Role in the survival of memory B cells and in the development and maintenance of the humoral response to T-dependent antigens .

  • Serves as a receptor for Epstein-Barr virus (EBV) .

  • Binds to CD23 and associates with CD19, CD81, and Leu13 to form a signal-transduction complex involved in B cell activation .

  • Acts as a receptor for C3d, C3dg, iC3b complement components, EBV, and IFNalpha .

Applications of Anti-Complement Receptor Type 2 CR2 Antibody

The anti-complement receptor type 2 CR2 antibody has various applications in biological research .

ApplicationRecommended Dilution
Western blot (WB)1:500-1:2000
Immunohistochemistry (IHC)1:50-1:100
Immunocytochemistry/Immunofluorescence (ICC/IF)1:50-1:100

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks (made-to-order)
Synonyms
CRT2 antibody; CRTL antibody; At1g09210 antibody; T12M4.8Calreticulin-2 antibody
Target Names
CRT2
Uniprot No.

Target Background

Function
CRT2 is a molecular calcium-binding chaperone that plays a crucial role in endoplasmic reticulum (ER) quality control. It promotes the proper folding and oligomeric assembly of glycoproteins through the calreticulin/calnexin cycle. This lectin transiently interacts with nearly all monoglucosylated glycoproteins synthesized within the ER.
Gene References Into Functions
  • In Arabidopsis, a triple knockout lacking calreticulins CRT1, CRT2, and CRT3 exhibited increased sensitivity to water stress. (PMID: 24022063) [Link]
  • Arabidopsis CRT2 is a pathogen-responsive gene potentially involved in disease resistance. CRT2 overexpression leads to pleiotropic phenotypes and elevated salicylic acid levels. (PMID: 21974727) [Link]
Database Links

KEGG: ath:AT1G09210

STRING: 3702.AT1G09210.1

UniGene: At.1529

Protein Families
Calreticulin family
Subcellular Location
Endoplasmic reticulum lumen.

Q&A

What is CRT2 and why is it significant in cancer research?

CRT2 is a cancer-testis antigen that demonstrates restricted expression in normal tissues (primarily in elongated spermatids of testis) while showing aberrant expression in various cancer cell lines and tissues, particularly melanoma. This distinctive expression pattern makes it a promising target for cancer immunotherapy and diagnosis. The significance of CRT2 lies in its recognition by serum immunoglobulin G (IgG) from cancer patients and its ability to induce tumor-reactive cytotoxic T lymphocytes (CTLs), suggesting potential applications in both cancer diagnosis and immunotherapeutic approaches .

What is the expression pattern of CRT2 in normal and malignant tissues?

In normal tissues, CRT2 expression is primarily restricted to the elongated spermatids of testis, exhibiting the characteristic tissue-restricted expression pattern of cancer-testis antigens. In pathological contexts, CRT2 demonstrates expression in various cancer cell lines and tissue samples, with particularly high expression observed in melanoma. This expression pattern was verified using a specifically generated anti-CRT2 antibody, providing further evidence of its potential as a cancer biomarker .

What approaches are used to generate and validate anti-CRT2 antibodies?

Generating and validating anti-CRT2 antibodies typically involves:

  • Antigen preparation: Recombinant CRT2 protein is expressed and purified, often using bacterial or mammalian expression systems.

  • Immunization: Laboratory animals (typically rabbits or mice) are immunized with the recombinant CRT2 protein to generate polyclonal antibodies. For monoclonal antibodies, hybridoma technology following mouse immunization is employed.

  • Antibody purification: Serum from immunized animals is collected and antibodies are purified using affinity chromatography.

  • Validation: The specificity and sensitivity of the anti-CRT2 antibody are evaluated through:

    • Western blot analysis using recombinant CRT2 protein and lysates from tissues/cell lines with known CRT2 expression

    • Immunohistochemistry on tissue sections from testis and cancer samples

    • Flow cytometry on cell lines expressing CRT2

    • ELISA using purified recombinant CRT2 protein

This systematic approach ensures the antibody specifically recognizes CRT2 and can be reliably used in various experimental applications .

What techniques are employed to assess CRT2 expression in clinical samples?

The assessment of CRT2 expression in clinical samples employs multiple complementary techniques:

  • Reverse transcription-PCR (RT-PCR): Used to detect CRT2 mRNA expression in tissue samples, providing information about gene transcription.

  • Immunohistochemistry (IHC): Utilizes anti-CRT2 antibodies to visualize protein expression and localization within tissue sections, enabling the assessment of expression patterns in different cell types.

  • Western blot analysis: Provides quantitative information about CRT2 protein levels in tissue lysates and allows comparison between different samples.

  • Flow cytometry: Enables the detection of CRT2 on the surface or intracellularly in dissociated cells from clinical samples.

Each technique offers distinct advantages, and researchers often use multiple approaches to comprehensively characterize CRT2 expression in clinical specimens .

How is the immunogenicity of CRT2 evaluated in research settings?

Assessing CRT2 immunogenicity involves multiple experimental approaches:

  • Serological analysis: Western blot and ELISA analyses are used to detect anti-CRT2 IgG antibodies in sera from cancer patients. This approach helps evaluate the natural immunogenicity of CRT2 in the context of cancer.

  • T-cell epitope identification: Potential T-cell epitopes within the CRT2 sequence are identified using prediction algorithms and validated through in vitro peptide binding assays with HLA molecules (particularly HLA-A24).

  • CTL induction assays: CRT2-derived peptides are tested for their ability to induce tumor-reactive cytotoxic T lymphocytes (CTLs) using:

    • HLA-A24 transgenic mice immunization

    • In vitro stimulation of human peripheral blood lymphocytes with peptide-pulsed antigen-presenting cells

  • Cytotoxicity assays: The functional activity of CRT2-specific CTLs is evaluated through their ability to recognize and kill tumor cells expressing CRT2 in an HLA-restricted manner.

These methods collectively provide comprehensive evidence of CRT2's potential as an immunotherapeutic target .

What are the challenges in developing CRT2-specific monoclonal antibodies for research applications?

Developing CRT2-specific monoclonal antibodies presents several challenges:

  • Protein structure complexity: CRT2's tertiary structure may contain conformational epitopes that are difficult to replicate with synthetic peptides or recombinant proteins expressed in prokaryotic systems.

  • Cross-reactivity concerns: Ensuring specificity against potential homologous proteins requires extensive validation to prevent misleading experimental results.

  • Epitope accessibility: Some epitopes may be inaccessible in the native protein conformation, limiting antibody utility in certain applications (e.g., flow cytometry of live cells vs. fixed samples).

  • Application-specific optimization: Different experimental techniques (IHC, Western blot, flow cytometry) may require antibodies recognizing different epitopes of CRT2, necessitating development of application-specific antibodies.

  • Reproducibility challenges: Variability between antibody batches can affect experimental reproducibility, requiring stringent quality control measures.

Addressing these challenges requires comprehensive characterization and validation strategies to ensure antibody reliability in research applications .

How can researchers distinguish between false positive and true positive results when detecting CRT2 expression?

Distinguishing between false positive and true positive CRT2 detection requires a multi-faceted approach:

  • Multiple detection methods: Employing orthogonal techniques (RT-PCR, IHC, Western blot) provides corroborating evidence of CRT2 expression.

  • Appropriate controls:

    • Positive controls: Testis tissue or CRT2-transfected cell lines

    • Negative controls: Normal tissues known not to express CRT2

    • Isotype controls: For antibody-based detection methods

    • Blocking peptide controls: To confirm antibody specificity

  • Antibody validation: Using multiple antibodies targeting different CRT2 epitopes helps confirm detection specificity.

  • Quantitative thresholds: Establishing expression thresholds based on control samples helps differentiate background signal from true positive expression.

  • mRNA and protein correlation: Concordance between mRNA and protein detection provides stronger evidence of true expression.

  • Functional validation: In cases of uncertainty, functional assays (such as immunogenicity testing) can provide additional evidence of authentic CRT2 expression .

What considerations should be made when designing experiments to evaluate CRT2 as an immunotherapeutic target?

Designing robust experiments to evaluate CRT2 as an immunotherapeutic target requires consideration of multiple factors:

  • Expression heterogeneity: Account for potential variability in CRT2 expression between patients, tumor types, and even within different regions of the same tumor.

  • HLA restriction: Since T-cell responses are HLA-restricted, experiments should consider the HLA types relevant to the target population. The identified HLA-A24-restricted epitope may not be applicable to all patient populations.

  • Immune escape mechanisms: Investigate potential mechanisms tumors might employ to evade CRT2-directed immune responses, such as antigen loss or downregulation.

  • Combination approaches: Design experiments that evaluate CRT2-targeted immunotherapy in combination with other treatment modalities, such as checkpoint inhibitors or conventional therapies.

  • Pre-existing immunity: Assess whether patients have pre-existing immunity to CRT2 (antibodies or T-cells) and how this might influence treatment efficacy.

  • Off-target effects: Despite restricted expression in normal tissues, thorough evaluation of potential off-target effects is essential to predict safety profiles.

  • Delivery systems: Consider different approaches for targeting CRT2, including peptide vaccines, DNA vaccines, adoptive T-cell therapy, or antibody-drug conjugates .

How should researchers interpret discrepancies between CRT2 mRNA and protein expression data?

Discrepancies between CRT2 mRNA and protein expression are not uncommon and can arise from various biological and technical factors. When faced with such inconsistencies, researchers should consider:

  • Post-transcriptional regulation: mRNA may be present but not translated due to microRNA regulation, RNA-binding proteins, or other post-transcriptional mechanisms.

  • Protein stability: CRT2 protein might have different turnover rates in different tissues or under different conditions, affecting steady-state levels.

  • Detection sensitivity: Protein detection methods may have different sensitivity thresholds compared to mRNA detection methods.

  • Sampling variation: Biological heterogeneity within samples or between adjacent sections used for different analyses can contribute to apparent discrepancies.

  • Antibody specificity: Validate whether the antibody used truly recognizes CRT2 and not related proteins.

When discrepancies occur, additional validation using alternative methods or antibodies targeting different epitopes is recommended to clarify the actual expression status .

What statistical approaches are recommended for analyzing CRT2 expression in relation to clinical outcomes?

When analyzing the relationship between CRT2 expression and clinical outcomes, the following statistical approaches are recommended:

  • Categorical analysis:

    • Kaplan-Meier survival analysis with log-rank tests to compare outcomes between CRT2-positive and CRT2-negative groups

    • Chi-square or Fisher's exact tests to examine associations between CRT2 expression and categorical clinicopathological variables

  • Continuous variable analysis:

    • Cox proportional hazards regression to assess the relationship between CRT2 expression levels and survival outcomes

    • Multivariate models including relevant clinical covariates to determine independent prognostic value

  • Expression thresholds:

    • Receiver operating characteristic (ROC) curve analysis to determine optimal cutoff values for categorizing CRT2 expression

    • Sensitivity analyses using different thresholds to ensure robustness of findings

  • Multiple hypothesis testing correction:

    • Apply methods such as Bonferroni correction or false discovery rate (FDR) when conducting multiple comparisons

  • Power considerations:

    • Perform power calculations to ensure sufficient sample sizes for detecting clinically meaningful associations

These approaches help establish whether CRT2 expression has prognostic or predictive value in cancer patients .

What are the methodological considerations when developing CRT2-based cancer vaccines?

Developing CRT2-based cancer vaccines requires careful consideration of several methodological aspects:

  • Epitope selection:

    • Identify epitopes with strong HLA binding affinity

    • Confirm immunogenicity through in vitro T-cell stimulation assays

    • Consider population HLA distribution to maximize coverage

  • Vaccine formulation:

    • Peptide vaccines: Determine optimal length (short 8-11 amino acids vs. long 15-30 amino acids)

    • DNA vaccines: Optimize codon usage and promoter selection

    • Viral vector vaccines: Select appropriate vector with optimal immunogenicity profile

  • Adjuvant selection:

    • Choose adjuvants that promote appropriate T-cell responses (Th1-biased for CTL induction)

    • Consider combination adjuvants to enhance both innate and adaptive immunity

  • Delivery route and schedule:

    • Determine optimal administration route (subcutaneous, intradermal, etc.)

    • Establish dosing schedule (prime-boost intervals)

  • Monitoring immune responses:

    • Develop assays to track CRT2-specific T-cell responses (ELISPOT, tetramer staining)

    • Monitor antibody responses as potential biomarkers

  • Combination strategies:

    • Design protocols combining vaccination with immune checkpoint inhibitors

    • Consider combination with conventional therapies that may enhance antigen presentation

The identified HLA-A24-restricted epitope from CRT2 provides a starting point for such vaccine development efforts .

How can researchers optimize the use of anti-CRT2 antibodies for diagnostic applications?

Optimizing anti-CRT2 antibodies for diagnostic applications involves:

  • Antibody characterization:

    • Determine specificity, sensitivity, and linearity of detection

    • Establish optimal antibody concentrations for different applications

    • Characterize performance across fixed and frozen tissues

  • Protocol standardization:

    • Develop standardized protocols for sample preparation, staining, and interpretation

    • Establish quality control procedures to ensure consistent results

    • Define scoring systems for quantitative assessment

  • Clinical validation:

    • Perform studies comparing CRT2 detection with gold standard diagnostic methods

    • Establish reference ranges and positivity thresholds

    • Determine sensitivity and specificity for detecting specific cancer types

  • Technical considerations:

    • Optimize antigen retrieval methods for immunohistochemistry

    • Determine appropriate blocking conditions to minimize background

    • Validate detection systems (chromogenic vs. fluorescent)

  • Automation potential:

    • Assess compatibility with automated staining platforms

    • Explore digital pathology approaches for quantitative analysis

  • Companion diagnostics development:

    • Develop standardized assays for patient selection in CRT2-targeted therapy trials

    • Ensure reproducibility across different testing sites

These optimizations enhance the reliability and clinical utility of anti-CRT2 antibodies as diagnostic tools .

What emerging technologies could enhance CRT2 antibody development and application?

Several emerging technologies hold promise for advancing CRT2 antibody development and applications:

  • Single B-cell antibody cloning: Isolating CRT2-specific B cells from cancer patients and cloning their antibody genes could yield naturally occurring high-affinity antibodies.

  • Phage display and yeast display technologies: These platforms enable rapid screening of large antibody libraries to identify high-affinity anti-CRT2 antibodies with desired properties.

  • CRISPR/Cas9 gene editing: Creating CRT2 knockout cell lines facilitates antibody validation and provides valuable negative controls.

  • Spatial transcriptomics and proteomics: These technologies enable simultaneous analysis of CRT2 expression and the tumor microenvironment, providing insights into contextual factors affecting expression.

  • Antibody engineering approaches:

    • Bispecific antibodies targeting CRT2 and immune effector cells

    • Antibody-drug conjugates for targeted delivery of cytotoxic agents

    • Nanobodies or single-domain antibodies for improved tissue penetration

  • Mass cytometry (CyTOF): Allows simultaneous detection of CRT2 and numerous other markers at the single-cell level, enabling comprehensive phenotyping.

  • Artificial intelligence-assisted image analysis: Enhances detection and quantification of CRT2 expression in histopathological samples.

These technologies could significantly advance the utility of CRT2 antibodies in both research and clinical applications .

What are the key research questions that remain unanswered about CRT2 as a cancer-testis antigen?

Despite progress in understanding CRT2, several critical research questions remain:

  • Biological function:

    • What is the physiological function of CRT2 in testicular cells?

    • Does CRT2 expression contribute to tumor development or progression?

    • Are there specific signaling pathways activated by CRT2 in cancer cells?

  • Expression regulation:

    • What epigenetic mechanisms control CRT2 expression?

    • Which transcription factors regulate CRT2 in normal and malignant contexts?

    • How is CRT2 expression affected by tumor microenvironment factors?

  • Clinical implications:

    • Is CRT2 expression associated with specific cancer subtypes or stages?

    • Does CRT2 expression correlate with response to immunotherapies?

    • Can CRT2 serve as a prognostic or predictive biomarker?

  • Immunological aspects:

    • Beyond HLA-A24, what other HLA restrictions exist for CRT2 epitopes?

    • What is the prevalence of natural T-cell responses against CRT2 in cancer patients?

    • Can CRT2-specific T-cell receptors be identified for adoptive cell therapy?

  • Therapeutic potential:

    • What is the optimal vaccination strategy targeting CRT2?

    • Can anti-CRT2 antibodies be developed for therapeutic applications?

    • How might CRT2-targeted approaches be combined with other immunotherapies?

Addressing these questions will advance understanding of CRT2 and its potential applications in cancer management .

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.