The CT domain (residues 182–250) is critical for:
Binding interactions: Facilitates binding to heparan sulfate proteoglycans (HSPGs), integrins, and growth factors like VEGF .
Cellular regulation: Modulates adhesion, migration, and differentiation via ERK and NF-κB signaling .
Pathological roles: Truncated CTGF isoforms (like this fragment) may promote proliferation and tumorigenesis, contrasting full-length CTGF’s antiproliferative effects .
Fibrosis and cancer: Used to study CTGF’s role in TGF-β-driven fibrosis and tumor angiogenesis .
Bone development: Investigates CTGF’s dual role in osteoblast differentiation and skeletal repair .
Antibody targeting: Serves as an antigen for generating CTGF-neutralizing antibodies (e.g., FG-3019 in clinical trials) .
Biomaterial engineering: Incorporated into scaffolds for bone regeneration due to angiogenic and osteogenic properties .
Skeletal repair: CTGF (182-250 a.a.) enhances chondrocyte proliferation and matrix mineralization during fracture healing .
Neuronal regulation: Modulates oligodendrocyte differentiation and Aβ deposition in Alzheimer’s disease models .
Pancreatic β-cells: Promotes proliferation under stress conditions, mediated by macrophage signaling .
The 182-250 amino acid region of human CTGF corresponds to part of the protein's conserved modular domains that are crucial for its diverse biological functions. CTGF contains four conserved domains, and this specific region participates in protein-protein interactions that mediate CTGF's regulatory effects. The cysteine-rich nature of CTGF (with 38 cysteines distributed throughout the protein) contributes to its tertiary structure through disulfide bonding, with 22 cysteines in the N-terminal and 16 in the C-terminal regions . This specific amino acid segment likely contributes to interactions with key signaling molecules like TGF-β or BMP, which are essential for CTGF's diverse regulatory functions in different biological systems .
This domain of CTGF contributes to various physiological processes through specific molecular interactions. In normal physiology, CTGF plays crucial roles in the development and function of multiple systems including the skeletal system, pancreas, central nervous system, eyes, and skin . The 182-250 a.a. region likely participates in the regulation of pancreatic β-cell proliferation, which is essential for glucose homeostasis . Additionally, this domain may be involved in CTGF's role in nervous system development, particularly in regulating oligodendrocyte differentiation and modulating olfactory behaviors . Understanding the specific contribution of this amino acid segment provides insights into how CTGF coordinates diverse physiological processes across multiple organ systems.
CTGF expression has been documented across multiple human tissues with varying expression levels. In the nervous system, CTGF has widespread localization, indicating important neurological functions . In the visual system, CTGF expression has been detected in cornea, iris, ciliary body, and choroid . In the vascular system, CTGF is expressed in endothelial cells and Sm22-positive vascular smooth muscle cells (VSMCs) . Notably, during fibrotic processes, CTGF expression increases in VSMCs during early stages (days 7-14) of fibrosis development, while endothelial cells show increased expression in middle to late stages (days 14-28) . The 182-250 a.a. region retains functionality across these diverse tissue types, suggesting conservation of critical interaction interfaces.
The 182-250 a.a. region of CTGF participates in critical protein-protein interactions that underlie its diverse functions. CTGF demonstrates varied functions by interacting with specific molecules such as VEGF, TGF-β, and BMP via its four conserved modular domains . Though the exact interactions specific to the 182-250 a.a. region aren't fully characterized, this segment likely participates in binding to growth factors or extracellular matrix components. Research examining truncated versions of CTGF has shown that different domains mediate specific interactions - the 182-250 a.a. region may be particularly important for interactions that regulate cell adhesion, migration, or matrix remodeling. Experimental approaches using targeted mutations or domain-specific blocking antibodies would help elucidate the precise molecular partners of this region.
Post-translational modifications of the 182-250 a.a. region can significantly alter CTGF's binding affinity, stability, and signaling capabilities. While specific modifications of this exact region aren't detailed in the available research, CTGF is known to undergo various modifications including glycosylation, phosphorylation, and proteolytic processing that affect its bioactivity. The cysteine-rich nature of CTGF suggests that disulfide bond formation is crucial for maintaining proper protein structure and function . Researchers investigating this domain should consider employing mass spectrometry approaches to identify specific modifications, coupled with site-directed mutagenesis studies to determine their functional consequences. Modifications within this region may be particularly relevant in pathological conditions where CTGF signaling is dysregulated.
Matrix stiffness has been shown to significantly regulate CTGF expression, which has important implications for understanding how mechanical forces might influence the functionality of the 182-250 a.a. region. Experimental evidence demonstrates that fibroblasts cultured on matrices of different stiffness show varied CTGF expression patterns. On physiologically soft matrices (1 kPa, similar to normal lung tissue), CTGF expression remains low, while on pathologically stiff matrices (50 kPa, similar to fibrotic tissue) or extremely stiff tissue culture plastic (TCP, ~10^6 kPa), CTGF expression increases significantly . After 72 hours of culture, CTGF mRNA was 5.5 times higher on 50 kPa matrices and 10.5 times higher on TCP compared to 1 kPa matrices . These mechanical cues may alter the conformation or accessibility of the 182-250 a.a. region, potentially affecting its interaction with binding partners and downstream signaling effects.
The 182-250 a.a. region of CTGF likely plays a critical role in fibrotic disorders through its participation in TGF-β signaling and extracellular matrix regulation. In pulmonary fibrosis models, CTGF expression is significantly upregulated following TGF-β1 exposure, as demonstrated in rat models where intratracheal administration of AdTGF-β1 increased CTGF expression in lung tissues . Fibroblasts cultured in fibrotic lung scaffolds show elevated CTGF expression compared to those in normal scaffolds, indicating the importance of the matrix microenvironment in regulating CTGF activity . Specifically, fibrotic fibroblasts placed in normal scaffolds exhibited approximately 45% reduced CTGF mRNA expression compared to those in fibrotic scaffolds, highlighting the plasticity of these cells in response to matrix cues . The 182-250 a.a. region may contain binding sites crucial for interactions with fibrosis-promoting factors or matrix components.
The 182-250 a.a. domain of CTGF may have significant implications for neurodegenerative diseases through its effects on neuroinflammation and tissue repair. In amyotrophic lateral sclerosis (ALS), CTGF upregulation has been observed in reactive astrocytes, contributing to astrogliosis . In Alzheimer's disease (AD), CTGF expression is elevated in plaque-associated regions of the brain, though its precise role remains debated. Some studies suggest that CTGF contributes to amyloid plaque formation and disease progression, while others propose it serves a protective function by facilitating amyloid-beta uptake and degradation . The 182-250 a.a. region may contain structural elements that determine whether CTGF exerts protective or detrimental effects in neurodegenerative contexts. Understanding the specific interactions mediated by this domain could provide insights into novel therapeutic approaches for these challenging conditions.
Cell-based assays for studying the CTGF (182-250 a.a.) region should focus on measuring specific biological responses related to known CTGF functions. Fibroblast proliferation assays using BrdU incorporation or Ki-67 immunostaining provide direct measures of CTGF's mitogenic activity. Migration assays (wound healing or Boyden chamber) assess CTGF's chemotactic properties, which were among the first identified functions of this protein . For mechanistic studies, reporter assays measuring activation of TGF-β, Hippo, or NF-κB pathways can reveal signaling mechanisms . Notably, matrix stiffness significantly affects CTGF responsiveness, so culturing cells on matrices of defined stiffness (1kPa for normal conditions, 50kPa for fibrotic conditions) is critical for physiologically relevant results . For studying domain-specific functions, comparing responses to full-length CTGF versus the isolated 182-250 a.a. fragment helps delineate this region's specific contributions. Co-immunoprecipitation followed by mass spectrometry can identify binding partners specific to this domain.
Quantifying interactions between CTGF (182-250 a.a.) and extracellular matrix components requires combining biophysical and cell-based approaches. Surface plasmon resonance (SPR) provides direct binding kinetics and affinity measurements, typically revealing KD values in the nanomolar range for specific ECM interactions. Isothermal titration calorimetry (ITC) offers complementary thermodynamic parameters of binding. For cell-context measurements, solid-phase binding assays using purified matrix proteins (fibronectin, various collagens, fibrinogen) coated on plates can determine relative binding strengths. Three-dimensional cell culture models using decellularized normal or fibrotic lung scaffolds recellularized with fibroblasts provide physiologically relevant systems to study how matrix composition affects CTGF function . In such systems, fibrotic fibroblasts placed in normal scaffolds showed approximately 45% reduction in CTGF expression compared to those in fibrotic scaffolds . For high-throughput screening of matrix interactions, protein microarrays containing various ECM proteins can identify novel binding partners of the 182-250 a.a. region.
Several approaches can effectively target the CTGF (182-250 a.a.) domain for potential therapeutic interventions. Monoclonal antibodies specifically recognizing this region represent the most clinically advanced approach, with existing anti-CTGF antibodies like FG3149 and FG3019 showing promising outcomes in clinical and preclinical trials . For more precise targeting, domain-specific aptamers offer advantages in terms of stability, immunogenicity, and tissue penetration compared to antibodies . Small-molecule inhibitors identified through structure-based drug design or high-throughput screening can disrupt specific interactions mediated by this domain. For genetic approaches, antisense oligonucleotides or siRNAs targeting CTGF mRNA can reduce expression, while CRISPR-based strategies could disrupt specific functions by introducing precise modifications to the 182-250 a.a. region. In cell-based therapies, engineering cells with modified CTGF domains could harness beneficial functions while eliminating detrimental effects. Efficacy evaluation should include both in vitro assays measuring specific interactions and in vivo models of relevant diseases like fibrosis or cancer.
Single-cell analysis techniques offer unprecedented opportunities to understand the cell-specific roles of CTGF (182-250 a.a.) across different biological contexts. Single-cell RNA sequencing (scRNA-seq) has already revealed cell-type specific expression patterns of CTGF, with data showing that fibroblasts and vascular cells are primary expressers of CTGF . Future research should employ spatial transcriptomics to map CTGF expression in relation to specific tissue structures and pathological features. Single-cell proteomics and phosphoproteomics would reveal how the 182-250 a.a. domain's post-translational modifications vary between cell types and under different conditions. Advanced techniques like CITE-seq (cellular indexing of transcriptomes and epitopes by sequencing) could correlate CTGF expression with cell surface marker profiles to identify novel CTGF-responsive cell populations. For mechanistic insights, single-cell ATAC-seq would reveal how CTGF signaling affects chromatin accessibility and transcriptional regulation in individual cells, potentially identifying cell-specific transcriptional signatures downstream of CTGF activation.
Targeting the CTGF (182-250 a.a.) region in combination with other anti-fibrotic approaches represents a promising therapeutic strategy for fibrotic diseases. CTGF acts as a downstream effector in multiple fibrogenic pathways, including TGF-β signaling, which makes it an attractive target for combination therapies . Potential synergistic approaches include combining anti-CTGF therapies with TGF-β pathway inhibitors, antioxidants, or anti-inflammatory agents. In pulmonary fibrosis models, CTGF expression increases in response to TGF-β stimulation in a dose-dependent manner, suggesting that dual inhibition could provide enhanced efficacy . The extracellular matrix environment significantly influences CTGF expression and function, with fibroblasts in fibrotic scaffolds showing higher CTGF expression than those in normal scaffolds . Therefore, combining CTGF inhibition with therapies that target matrix stiffness or composition could address multiple aspects of the fibrotic process. Future research should systematically evaluate such combination approaches in relevant animal models and eventually in clinical settings to determine optimal therapeutic strategies.
Computational approaches offer powerful tools for elucidating the structural dynamics of the CTGF (182-250 a.a.) region and its interactions. Molecular dynamics simulations can reveal conformational flexibility and identify potential binding sites within this domain. The cysteine-rich nature of CTGF suggests complex disulfide bonding patterns that could be predicted using specialized algorithms . Protein-protein docking simulations can model interactions between the 182-250 a.a. region and known binding partners like TGF-β, VEGF, or specific integrins. Machine learning approaches trained on existing protein interaction data could predict novel binding partners and functional relationships. Network analysis incorporating transcriptomic and proteomic data can place CTGF in the context of broader signaling networks across different physiological and pathological conditions. These computational predictions should guide experimental designs, particularly for structural studies and the development of domain-specific inhibitors, creating an iterative process that accelerates research progress and therapeutic development for CTGF-related diseases.
CTGF is a cysteine-rich protein composed of four distinct modules:
The recombinant form of CTGF (182-250 a.a.) is produced in E. coli and is a fusion protein with a His Tag, having a total molecular mass of 15 kDa .
CTGF is involved in various cellular functions and developmental processes:
Studies have shown that CTGF is crucial for normal growth and development. For instance, mice lacking the CTGF gene exhibit severe defects in angiogenesis and skeletal development, leading to respiratory stress and chondrodysplasia . Additionally, CTGF is involved in the development of pancreatic beta cells and normal ovulation .