CTGF Antibody Pair

Shipped with Ice Packs
In Stock

Description

Functional Mechanisms in Research Applications

CTGF antibody pairs enable precise quantification of CTGF levels, which regulate:

  • Fibroblast growth factor (FGF)-mediated DNA synthesis

  • TGF-β1-induced collagen production

  • JNK2 phosphorylation in cardiac fibroblasts

A 2019 preclinical study demonstrated therapeutic potential in myocardial infarction models :

ParameterMI + IgG (n=5)MI + CTGF mAb (n=8)p-value
LVEDD (mm)5.84 ± 0.455.51 ± 0.75<0.05
LVESD (mm)5.41 ± 0.504.81 ± 0.74<0.01
LVED Vol (μl)170 ± 30.0151 ± 47.7<0.05

Treatment with CTGF monoclonal antibody (mAb) improved left ventricular dimensions and upregulated cardiac repair genes including Nkx2-5 (2.1-fold) and Cited4 (1.8-fold) .

Key Research Findings

RNA sequencing analysis of post-MI cardiac tissue revealed:

  • 72 significantly altered transcripts with CTGF mAb treatment

  • 42 upregulated genes associated with cardiac development

  • GATA-4 induction in treated groups (p<0.05)

In vitro studies using human cardiac fibroblasts showed:

  • 35% reduction in TGF-β1-induced collagen production

  • JNK-dependent inhibition of fibrotic pathways

  • No significant effect on fibroblast proliferation

Quality Assurance and Validation

Leading commercial providers implement stringent validation protocols:

  • Batch-to-batch consistency via recombinant production

  • Cross-reactivity testing across human/murine samples

  • Performance validation in SimpleStep ELISA® platforms

Research Applications

CTGF antibody pairs facilitate:

  • Quantification of CTGF in fibrosis models

  • Mechanistic studies of cardiac remodeling

  • High-throughput drug discovery screening

Product Specs

Buffer
**Capture Buffer:** 50% Glycerol, 0.01M PBS, pH 7.4
**Detection Buffer:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch orders within 1-3 business days of receipt. Delivery times may vary depending on the purchasing method and location. For specific delivery details, please consult your local distributor.
Notes
We recommend using the capture antibody at a concentration of 0.5 µg/mL and the detection antibody at a concentration of 0.25 µg/mL. Optimal dilutions should be determined experimentally by the researcher.
Synonyms
CCN family member 2,Hypertrophic chondrocyte-specific protein 24,Insulin-like growth factor-binding protein 8,CTGF,CCN2, HCS24, IGFBP8
Target Names

Q&A

What is CTGF and why is it an important target for antibody-based detection?

CTGF (Connective Tissue Growth Factor, also known as CCN2) is a matricellular protein that functions as a major connective tissue mitoattractant secreted primarily by vascular endothelial cells. It promotes proliferation and differentiation of chondrocytes while mediating heparin- and divalent cation-dependent cell adhesion in multiple cell types including fibroblasts, myofibroblasts, endothelial and epithelial cells. CTGF also enhances fibroblast growth factor-induced DNA synthesis .

The significance of CTGF as a detection target stems from its critical role in various pathological processes, particularly as an essential regulator in fibrosis, cancer progression, neurological disorders, and eye diseases . CTGF's elevated expression correlates with disease progression in multiple conditions, making it both a valuable biomarker and a therapeutic target worthy of precise detection and quantification in experimental settings.

How do I determine the optimal epitope region when selecting a CTGF antibody pair?

Selecting the optimal epitope regions requires consideration of both the protein's functional domains and accessibility. CTGF contains distinct domains that mediate different biological activities. Based on available antibody data, common epitope regions include:

  • N-terminal region (AA 27-349): Contains binding sites for IGF and likely maintains full biological activity

  • Middle region (AA 80-200): Includes key functional domains for interaction with cell receptors

  • C-terminal region (AA 221-349): Contains domains involved in receptor binding and matrix interactions

For optimal antibody pair selection:

  • Choose antibodies targeting non-overlapping epitopes to prevent competitive binding

  • Consider using pairs where one antibody targets the N-terminal region and another targets the C-terminal region

  • Review cross-reactivity data—human CTGF shares approximately 95.5% and 95.9% amino acid sequence identity with mouse and rat CTGF respectively

  • Evaluate published validation data demonstrating successful epitope binding in your application of interest

The highest sensitivity is typically achieved when antibodies recognize conformational epitopes that remain accessible after protein immobilization in your detection platform.

What are the key differences between polyclonal and monoclonal CTGF antibody pairs for research applications?

Polyclonal CTGF Antibody Pairs:

  • Recognize multiple epitopes across the CTGF protein

  • Generally provide higher sensitivity for detection due to binding multiple sites

  • May show batch-to-batch variation requiring validation between lots

  • Often used in applications where maximum signal detection is the priority

  • Examples include rabbit polyclonal antibodies targeting AA 58-349 that exhibit strong reactivity with human and rat CTGF

Monoclonal CTGF Antibody Pairs:

  • Recognize a single epitope with high specificity

  • Provide more consistent results with minimal batch-to-batch variation

  • May offer lower sensitivity than polyclonal pairs but higher specificity

  • Preferable for quantitative applications requiring reproducibility over time

  • Examples include mouse monoclonal antibodies like clone AB01/1E5 that specifically target human CTGF

For optimal research outcomes, consider using a monoclonal capture antibody paired with a polyclonal detection antibody to balance specificity with signal amplification. This approach is particularly effective in sandwich ELISA applications, where sensitivity ranges of 0.19 ng/mL have been documented .

How should I optimize a sandwich ELISA protocol using CTGF antibody pairs?

Optimizing a sandwich ELISA for CTGF requires careful attention to several critical parameters:

  • Antibody Selection and Orientation:

    • Use a capture antibody targeting a different epitope than the detection antibody

    • Evaluate several antibody pairings, as sensitivity can vary significantly between combinations

    • Polyclonal capture with monoclonal detection or vice versa can improve performance

  • Protocol Optimization:

    • Coating concentration: Titrate capture antibody (typically 1-10 μg/mL) to determine optimal coating density

    • Sample dilution: Create a dilution series of samples to ensure measurements fall within the linear range

    • Detection antibody concentration: Titrate to maximize signal while minimizing background

    • Incubation conditions: Optimize temperature and duration for all steps

  • Standard Curve Preparation:

    • Use recombinant human CTGF protein for standard curve (range: 0.47-30 ng/mL is typical)

    • Prepare standards in the same matrix as samples to account for matrix effects

    • Include multiple replicates of each standard concentration

  • Validation Parameters:

    • Determine lower limit of detection (documented as low as 0.19 ng/mL for optimized pairs)

    • Assess intra- and inter-assay variability (<10% and <15% CV respectively is desirable)

    • Perform spike-recovery experiments to validate accuracy in your specific sample types

For human samples specifically, matched antibody pairs have been validated for detection in serum, plasma, and tissue homogenates with sensitivity in the sub-nanogram range .

What are the most effective sample preparation methods for detecting CTGF in different tissue types?

Sample preparation methods must be tailored to tissue type and experimental goals:

For Serum/Plasma Samples:

  • Collect blood in appropriate anticoagulant tubes (EDTA for plasma, no anticoagulant for serum)

  • Centrifuge at 1000-2000×g for 10 minutes at 4°C

  • Carefully remove supernatant without disturbing interface

  • Dilute samples in assay buffer (typically 1:2 to 1:10) to minimize matrix effects

  • For maximum sensitivity, avoid repeated freeze-thaw cycles

For Tissue Homogenates:

  • Collect tissues and immediately snap-freeze in liquid nitrogen

  • Weigh frozen tissue and add homogenization buffer (PBS with protease inhibitors) at 1:10 ratio (w/v)

  • Homogenize using mechanical disruption (e.g., tissue homogenizer or bead-beater)

  • Centrifuge at 12,000×g for 15 minutes at 4°C to remove cellular debris

  • Collect supernatant and determine protein concentration

  • Normalize samples to equal protein concentration (typically 1-5 mg/mL)

For Cell Culture Supernatants:

  • Collect culture medium from cells grown in serum-free conditions for 24-48 hours

  • Centrifuge at 2000×g for 10 minutes to remove cellular debris

  • Filter through a 0.22 μm filter if necessary

  • Concentrate samples using centrifugal filter units if CTGF levels are expected to be low

For all sample types, inclusion of protease inhibitors is critical as CTGF can undergo proteolytic degradation. Additionally, samples should be processed consistently between experimental groups to ensure comparable results .

How can I validate CTGF antibody specificity for immunohistochemistry applications?

Validating antibody specificity for IHC applications requires a systematic approach:

  • Positive and Negative Controls:

    • Positive tissue controls: Use tissues known to express CTGF (vascular endothelial cells, fibrotic tissues)

    • Negative tissue controls: Use tissues with minimal CTGF expression

    • CTGF-knockout or siRNA-treated samples provide excellent negative controls when available

  • Antibody Validation Tests:

    • Peptide competition assay: Pre-incubate antibody with purified CTGF peptide before staining

    • Multiple antibody comparison: Use antibodies targeting different CTGF epitopes and compare staining patterns

    • Correlation with other detection methods: Compare IHC results with Western blot or in situ hybridization

  • Technical Controls:

    • Isotype control: Use matched isotype antibody at same concentration as CTGF antibody

    • Secondary-only control: Omit primary antibody to assess non-specific binding of secondary antibody

    • Endogenous peroxidase blocking: Verify complete blocking of endogenous activity

  • Optimization Parameters:

    • Antigen retrieval method: Compare heat-induced (citrate buffer, pH 6.0) versus enzymatic methods

    • Antibody dilution: Test dilution series (typically 1:50 to 1:500) to optimize signal-to-noise ratio

    • Incubation conditions: Optimize temperature (4°C, room temperature) and duration (1 hour, overnight)

Research has confirmed that reliable CTGF detection in paraffin-embedded sections requires careful antigen retrieval and validation against tissues with known expression patterns, such as fibrotic liver samples where CTGF is co-expressed with integrin αvβ6 in reactive ducts .

How should I normalize CTGF expression data across different experimental conditions?

Proper normalization is essential for meaningful comparison of CTGF expression data:

  • For Western Blot Analysis:

    • Use housekeeping proteins (GAPDH, β-actin, or α-tubulin) as loading controls

    • Calculate relative CTGF expression as ratio of CTGF band intensity to loading control

    • For secreted CTGF in culture media, normalize to cell number or total cellular protein

    • Consider using Ponceau S staining of membrane as alternative normalization method

  • For ELISA Quantification:

    • For tissue samples: Normalize to total protein concentration determined by BCA or Bradford assay

    • For serum/plasma: Express as absolute concentration (ng/mL)

    • For cultured cells: Normalize to cell number or cellular protein content

    • For in vivo studies: Consider additional normalization to tissue weight

  • For qRT-PCR Analysis:

    • Use validated reference genes (18S rRNA has been successfully used for CTGF studies)

    • Apply multiple reference gene normalization for higher accuracy

    • Verify reference gene stability across experimental conditions

    • Use the 2^(-ΔΔCt) method for relative quantification

  • For Immunohistochemistry:

    • Score staining intensity using consistent criteria (0-3+ scale)

    • Quantify percentage of positive cells in defined tissue areas

    • Use digital image analysis software for objective quantification

    • Express results as H-score (intensity × percentage) or modified scoring system

Research on CTGF often employs multiple normalization strategies. In studies investigating CTGF expression in liver injury models, 18S rRNA normalization for qRT-PCR and GAPDH for Western blot analysis have provided consistent results .

What are the expected CTGF expression patterns in different pathological conditions?

CTGF expression varies significantly across pathological conditions, providing valuable diagnostic and research insights:

In Fibrotic Diseases:

  • Liver Fibrosis: Sustained co-induction of CTGF and integrin β6 mRNAs observed from days 7 to 33 after DDC treatment, with 5-fold increase of CTGF mRNA in CCl₄-induced damage

  • Pulmonary Fibrosis: Significantly elevated CTGF in idiopathic pulmonary fibrosis (IPF), contributing to excessive extracellular matrix production

  • Cardiac Fibrosis: Increased CTGF expression in post-myocardial infarction remodeling, with CTGF inhibition reducing heart weight-to-body weight ratio

  • Skin Fibrosis: Transgenic mice overexpressing CTGF in fibroblasts show accelerated dermal fibrosis with marked expansion of myofibroblast populations

In Cancer:

  • Hepatocellular Carcinoma: Increased expression associated with worse prognosis; CTGF protein from tumor samples (~42 kDa) shows different migration pattern than non-tumor tissues

  • Pancreatic Cancer: Elevated CTGF expression contributes to desmoplastic reaction; targeted in clinical trials with pamrevlumab (FG-3019)

In Neurological Disorders:

  • Alzheimer's Disease: Elevated CTGF expression in astrocytes surrounding amyloid plaques; facilitates Aβ uptake and degradation via MMP-dependent mechanisms

  • Cerebrovascular Disease: Increased in vascular endothelial cells after ischemic injury

Expected Expression Ranges:
For quantitative ELISA measurements in human samples:

  • Normal serum/plasma: Typically <5 ng/mL

  • Fibrotic conditions: Often elevated 2-10 fold above normal

  • Cancer patients: Variable elevation depending on tumor type and stage

Understanding these expression patterns helps in experimental design and interpretation of results across different disease models .

How do I interpret contradictory CTGF expression data between protein and mRNA levels?

Discrepancies between CTGF protein and mRNA levels are common and can provide valuable biological insights:

  • Potential Mechanisms Explaining Discrepancies:

    • Post-transcriptional regulation: miRNAs may target CTGF mRNA without affecting transcription

    • Protein stability differences: CTGF protein undergoes proteolytic processing that affects detection

    • Secretion dynamics: As a secreted protein, cellular CTGF levels may not reflect mRNA production

    • Tissue-specific processing: CTGF from different tissues shows variable migration patterns on Western blot (~42 kDa in tumor vs. different in non-tumor)

  • Analysis Approaches:

    • Temporal analysis: Examine time course data to identify delays between mRNA induction and protein accumulation

    • Cellular fractionation: Analyze intracellular vs. secreted CTGF separately

    • Protein half-life studies: Use cycloheximide chase experiments to determine CTGF protein stability

    • Multiple detection methods: Complement Western blot with ELISA, IHC, and mass spectrometry

  • Interpretation Framework:

    • High mRNA/low protein suggests post-transcriptional inhibition or rapid protein turnover

    • Low mRNA/high protein suggests protein stabilization or accumulation of secreted CTGF in matrix

    • Different isoforms or processed forms may be detected preferentially by different methods

  • Validation Strategies:

    • Use conditional knockout models (e.g., Ctgf flox/flox with inducible Cre) to verify antibody specificity

    • Perform pulse-chase experiments to track protein synthesis and degradation

    • Investigate proteolytic processing by matrix metalloproteinases which can generate CTGF fragments

In research on liver injury, studies found ubiquitous induction of both CTGF mRNA and protein in non-tumor and tumor areas, but Western blot revealed different migration patterns, suggesting tissue-specific post-translational modifications that may explain functional differences .

How can I address poor sensitivity issues in CTGF detection assays?

Poor sensitivity in CTGF detection can stem from multiple factors requiring systematic troubleshooting:

  • Antibody-Related Factors:

    • Epitope accessibility: CTGF undergoes proteolytic processing; antibodies targeting processed regions may show reduced binding

    • Antibody quality: Use antibodies specifically validated for your application with documented sensitivity

    • Antibody pairs: Test multiple capture-detection antibody combinations; some pairs (documented sensitivity as low as 0.19 ng/mL) significantly outperform others

  • Sample-Related Solutions:

    • Proteolytic degradation: Add protease inhibitor cocktails to all buffers during sample preparation

    • Sample concentration: Use immunoprecipitation or centrifugal concentration filters for dilute samples

    • Matrix effects: Optimize sample dilution to minimize interference from complex matrices

    • Binding interference: Pre-clear samples with protein A/G if high background is observed

  • Protocol Optimization:

    • Signal amplification: Implement tyramide signal amplification or poly-HRP detection systems

    • Incubation conditions: Extend primary antibody incubation time (overnight at 4°C)

    • Blocking optimization: Test different blocking agents (BSA, casein, commercial blockers)

    • Buffer optimization: Adjust salt concentration and pH to enhance antibody-antigen binding

  • Instrument and Detection Optimization:

    • Substrate selection: Switch to more sensitive chemiluminescent or fluorescent substrates

    • Extended exposure: Optimize imaging parameters for detection near sensitivity limits

    • Equipment calibration: Ensure detection instruments are properly calibrated

For particularly challenging samples, consider targeted enrichment strategies using heparin-sepharose chromatography, as CTGF binds heparin with high affinity, enabling purification and concentration prior to analysis .

What approaches should I use to distinguish between different CTGF isoforms or fragments?

Distinguishing between CTGF isoforms and proteolytic fragments requires specialized techniques:

  • Molecular Weight-Based Differentiation:

    • Western blot with gradient gels: Use 4-20% gradient gels to maximize resolution of different CTGF forms

    • Size comparison: Full-length CTGF appears at ~38-42 kDa; proteolytic fragments range from 10-30 kDa

    • Multiple antibodies: Use antibodies targeting different epitopes to identify specific fragments

    • Standard comparison: Include recombinant full-length and fragment controls

  • Domain-Specific Detection:

    • Domain-specific antibodies: Use antibodies that recognize specific domains (N-terminal vs. C-terminal)

    • Selective immunoprecipitation: Capture with domain-specific antibody followed by detection with another

    • Competing peptides: Use domain-specific blocking peptides to confirm fragment identity

  • Advanced Analytical Techniques:

    • Mass spectrometry: LC-MS/MS can precisely identify fragments and post-translational modifications

    • 2D gel electrophoresis: Separate isoforms based on both molecular weight and isoelectric point

    • Size exclusion chromatography: Fractionate samples before immunodetection

  • Functional Validation:

    • Domain-specific bioassays: Different domains mediate distinct biological activities

    • Binding partner analysis: Various CTGF fragments interact selectively with different binding partners

    • Cell-based assays: Test fragments for specific activities (e.g., fibroblast proliferation)

The CC samples from hepatocellular carcinoma patients revealed CTGF protein bands migrating differently (~42 kDa) compared to non-tumor tissues, highlighting the importance of distinguishing between tissue-specific forms for accurate interpretation .

How can I minimize cross-reactivity when detecting CTGF in multi-species studies?

Managing cross-reactivity in multi-species CTGF studies requires careful antibody selection and validation:

  • Species Homology Considerations:

    • Human CTGF shares 95.5% and 95.9% amino acid sequence identity with mouse and rat CTGF, respectively

    • Regions of highest conservation are typically in functional domains

    • Target antibodies to species-unique epitopes when species-specific detection is required

  • Antibody Selection Strategies:

    • Validated cross-reactivity: Choose antibodies explicitly validated for multiple species of interest

    • Epitope mapping: Select antibodies recognizing epitopes with known sequence conservation or divergence

    • Monoclonal specificity: For distinguishing closely related species, monoclonal antibodies can offer higher specificity

    • Negative controls: Include samples from CTGF knockout animals or tissues known to lack CTGF expression

  • Experimental Validation Approaches:

    • Peptide competition: Conduct blocking experiments with species-specific peptides

    • Recombinant protein controls: Include species-specific recombinant CTGF proteins as standards

    • Cross-absorption: Pre-absorb antibodies with recombinant proteins from non-target species

    • Sequential immunodepletion: Deplete cross-reactive antibodies using immobilized proteins

  • Analysis Methods to Address Cross-Reactivity:

    • Comparative titration: Determine relative affinities for target species

    • Species-specific PCR primers: Complement protein detection with species-specific mRNA quantification

    • Mass spectrometry: Identify species-specific peptides for targeted proteomics

    • Domain-specific detection: Target domains with greater sequence divergence between species

For studies using human CTGF antibody pairs in animal models, researchers have successfully implemented these approaches to distinguish endogenous from transgenic CTGF, particularly in fibrosis and cancer models where both human and murine forms may be present .

How can CTGF antibody pairs be utilized for in vivo imaging of fibrotic diseases?

Advanced in vivo imaging of CTGF represents a frontier in fibrosis research:

  • Antibody Modification for In Vivo Applications:

    • Fluorescent conjugation: Direct labeling with near-infrared fluorophores for optical imaging

    • Radioisotope labeling: Conjugation with PET tracers (e.g., ⁸⁹Zr, ¹²⁴I) for deep tissue detection

    • Nanoparticle conjugation: Attachment to gold nanoparticles or quantum dots for multimodal imaging

    • Fragment development: Use Fab or scFv fragments for improved tissue penetration and faster clearance

  • Novel CTGF-Targeting Approaches:

    • Peptide-coated nanocluster probes: Recently developed gold nanoclusters specifically target CTGF with high affinity (KD ~21.9 nM), penetrating the blood-brain barrier for NIR-II imaging

    • Bispecific antibodies: Engineer antibodies targeting both CTGF and tissue-specific markers

    • Activatable probes: Design probes that fluoresce only upon binding to CTGF, reducing background

    • MMP-responsive systems: Create detection systems responsive to CTGF-associated MMP activity

  • Multimodal Imaging Applications:

    • Combined PET-CT: Correlate CTGF distribution with anatomical features

    • Optical-MRI fusion: Use dual-labeled antibodies for complementary resolution and sensitivity

    • Intravital microscopy: Apply fluorescently labeled antibodies for real-time cellular imaging

    • Chromogenic imaging: Leverage peroxidase-like activity for postmortem tissue analysis

  • Validation and Quantification Methods:

    • Dynamic imaging protocols: Track CTGF changes longitudinally during disease progression

    • Co-registration with fibrosis markers: Correlate CTGF signal with established fibrosis measurements

    • Pharmacokinetic modeling: Optimize imaging timepoints based on antibody biodistribution

    • ICP-MS quantitation: Provide precise measurement of probe accumulation in tissues

These advanced approaches enable early detection of fibrotic changes before conventional clinical manifestations appear, as demonstrated with peptide-coated gold nanoclusters that detect elevated CTGF in Alzheimer's disease models before amyloid plaque formation .

What are the current approaches for developing therapeutic antibodies targeting CTGF?

Development of therapeutic CTGF antibodies has progressed significantly, offering insights for researchers:

  • Current Therapeutic Antibody Development:

    • Pamrevlumab (FG-3019): Human monoclonal antibody against CTGF that has shown efficacy in clinical trials for idiopathic pulmonary fibrosis, pancreatic cancer, and Duchenne muscular dystrophy

    • FG-3149: Chimeric antibody with the binding motif of FG-3019 but a mouse IgG2a constant region, designed to reduce immunogenicity in rodent models

    • Mechanism-specific antibodies: Development of antibodies targeting specific CTGF domains to inhibit distinct functions

  • Experimental Design for Anti-CTGF Therapy Assessment:

    • Disease-specific models: Cardiac remodeling post-MI shows reduced heart weight-to-body weight ratio with CTGF mAb treatment

    • Timing optimization: Early vs. late intervention studies assess preventive vs. therapeutic efficacy

    • Combination approaches: Test CTGF antibodies with existing standards of care

    • Biomarker identification: Develop companion diagnostics to identify responders

  • Molecular and Cellular Mechanisms:

    • Signaling pathway analysis: CTGF mAb treatment induces ERK and JNK2 phosphorylation in infarct scars

    • Cell-specific effects: Differential impact on various cell types (fibroblasts vs. immune cells)

    • Extracellular matrix remodeling: Effects on collagen deposition and matrix organization

    • Immune cell recruitment: Potential impact on CD11b+ cell accumulation in pulmonary hypertension models

  • Emerging Therapeutic Strategies:

    • Antibody-drug conjugates: Delivery of anti-fibrotic or anti-inflammatory agents directly to CTGF-expressing cells

    • Bispecific formats: Dual targeting of CTGF and complementary pathways (e.g., TGF-β)

    • Domain-specific targeting: Selective inhibition of CTGF domains mediating specific pathological effects

    • Cell-type selective delivery: Development of antibodies preferentially targeting CTGF in specific cellular contexts

Experimental studies with anti-CTGF antibodies have revealed promising therapeutic potential, demonstrating reduced infarct expansion index and improved cardiac function in MI models, suggesting broad applications across fibrotic diseases .

How can CTGF antibody pairs be applied to study the relationship between CTGF and matrix metalloproteinases in tissue remodeling?

The complex relationship between CTGF and matrix metalloproteinases (MMPs) represents a crucial area for advanced research:

  • Detection of CTGF-MMP Interactions:

    • Co-immunoprecipitation: Capture CTGF complexes with anti-CTGF antibodies and probe for associated MMPs

    • Proximity ligation assays: Visualize and quantify CTGF-MMP interactions in situ with subcellular resolution

    • FRET-based reporters: Develop fluorescent biosensors to detect CTGF-MMP binding in real-time

    • Surface plasmon resonance: Determine binding kinetics and affinities between purified CTGF and MMPs

  • Functional Relationship Studies:

    • Cell-specific expression: CTGF enhances extracellular Aβ degradation via membrane-bound MMP14 in glia and extracellular MMP13 in neurons

    • Conditional knockouts: Use Ctgf flox/flox with inducible Cre systems to study temporal relationships with MMPs

    • Domain-specific mutations: Engineer CTGF variants with altered MMP binding sites to dissect functional interactions

    • Substrate specificity: Investigate how CTGF modulates MMP substrate preferences

  • Tissue Remodeling Assessment:

    • Multiplex immunostaining: Simultaneously visualize CTGF, MMPs, and matrix components

    • In situ zymography: Correlate CTGF expression with localized MMP activity

    • Second harmonic generation imaging: Assess collagen reorganization in relation to CTGF-MMP activity

    • Gene expression profiling: Analyze coordinated regulation of CTGF and MMP family members

  • Translational Applications:

    • Biomarker development: Measure CTGF-MMP ratios as indicators of active remodeling

    • Therapeutic targeting: Design interventions targeting CTGF-MMP interactions rather than individual proteins

    • Diagnostic imaging: Develop probes detecting active CTGF-MMP complexes

    • Personalized medicine: Stratify patients based on CTGF-MMP profiles for targeted therapies

Research on Alzheimer's disease has demonstrated that CTGF facilitates Aβ uptake and degradation through specific MMP pathways, with glial cells using membrane-bound MMP14 and neurons utilizing extracellular MMP13, providing a functional link in neuron-glia communication that protects against Aβ-induced CNS degeneration .

What are the emerging technologies for single-cell analysis of CTGF expression in heterogeneous tissues?

Single-cell analysis of CTGF represents a frontier for understanding its role in complex tissue environments:

  • Single-Cell Protein Detection Methods:

    • Mass cytometry (CyTOF): Use metal-labeled anti-CTGF antibodies for high-dimensional analysis

    • Single-cell Western blotting: Separate and detect CTGF from individual cells in microfluidic platforms

    • Imaging mass cytometry: Visualize CTGF expression with subcellular resolution in tissue sections

    • Proximity extension assays: Detect CTGF with high sensitivity using oligonucleotide-conjugated antibody pairs

  • Integrated Multi-Omics Approaches:

    • CITE-seq: Combine CTGF antibody detection with single-cell transcriptomics

    • Single-cell spatial transcriptomics: Map CTGF mRNA expression with spatial context

    • Cellular indexing of transcriptomes and epitopes (CITE): Correlate CTGF protein with global transcriptional profiles

    • Single-cell secretome analysis: Measure CTGF secretion from individual cells

  • In Situ Analysis Techniques:

    • Multiplexed immunofluorescence: Co-localize CTGF with cell-type markers and signaling molecules

    • In situ hybridization with immunodetection: Combine CTGF mRNA and protein visualization

    • Live-cell imaging: Monitor CTGF dynamics in real-time using fluorescent protein fusions or antibody fragments

    • Reporter mouse models: Use CTGF-GFP reporter mice to track expression in specific cell populations

  • Computational Analysis Methods:

    • Trajectory inference: Reconstruct CTGF expression dynamics during cellular differentiation and activation

    • Cell-cell communication modeling: Infer CTGF-mediated intercellular signaling networks

    • Spatial statistics: Analyze neighborhood relationships between CTGF-expressing and responding cells

    • Multi-modal data integration: Combine proteomic, transcriptomic, and spatial information

Research utilizing CTGF-GFP mice has already provided insights into cell-specific expression patterns during liver injury, revealing differential expression in oval cells versus myofibroblasts, which would not be apparent in bulk tissue analysis .

How can we design antibody-based approaches to selectively target specific CTGF domains and functions?

Selective targeting of CTGF domains represents an advanced frontier for precise modulation of its multiple functions:

  • Domain-Specific Antibody Development:

    • Structure-guided design: Use crystallographic data to design antibodies against specific functional epitopes

    • Phage display selection: Screen antibody libraries against isolated CTGF domains

    • Yeast surface display: Evolve antibodies with enhanced specificity for particular domains

    • Site-directed mutagenesis: Generate antibodies targeting regions outside of conserved functional sites

  • Functional Domain Characterization:

    • CTGF contains four distinct domains with specific functions:

      • IGFBP domain: Insulin-like growth factor binding

      • VWC domain: TGF-β binding and receptor interaction

      • TSP1 domain: Cell attachment and extracellular matrix binding

      • CT domain: Heparin binding and dimerization

  • Application-Specific Targeting Strategies:

    • Fibrosis inhibition: Target domains mediating TGF-β signaling enhancement

    • Angiogenesis modulation: Focus on domains involved in endothelial cell function

    • Matrix interaction disruption: Target domains mediating binding to extracellular matrix proteins

    • Cell migration inhibition: Develop antibodies against domains involved in integrin binding

  • Advanced Antibody Formats:

    • Biparatopic antibodies: Target two different CTGF epitopes simultaneously

    • Intrabodies: Develop antibodies for intracellular targeting of newly synthesized CTGF

    • pH-dependent binding: Create antibodies that release in specific cellular compartments

    • Conditionally active antibodies: Design antibodies activated only in disease microenvironments

Recent research demonstrated differential protective effects of CTGF against Aβ neurotoxicity on neurons and glia, where CTGF enhanced Aβ degradation via distinct MMP pathways in different cell types, suggesting that domain-specific targeting could selectively modulate beneficial versus pathological CTGF functions .

By strategically targeting specific CTGF domains with domain-selective antibodies, researchers can dissect the multifunctional nature of this protein and potentially develop therapeutic approaches with enhanced specificity and reduced off-target effects.

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.