Fibronectin Rat

Fibronectin Rat
Shipped with Ice Packs
In Stock

Description

Cell Adhesion and Migration

  • Schwann Cells: Fibronectin enhances adhesion, proliferation, and chemotaxis in rat Schwann cells, facilitating nerve repair .

  • Hepatic Stellate Cells: Activates ERK signaling via α5β1 integrins, increasing endothelin-1 synthesis and promoting cell contraction .

Tissue Remodeling

  • Cardiac Hypertrophy: Atrial fibronectin levels are 4–5× higher than ventricular. During hypertrophy, ventricular mRNA rises 4–6×, driven by thyroid hormone (T3) and mineralocorticoid pathways .

  • Wound Healing: Fibronectin deficiency impairs debris clearance via the reticuloendothelial system, delaying recovery .

Diabetic Retinopathy Model

ParameterControl RatsGalactosemic RatsAntisense-Treated Rats
Fibronectin mRNA100%179%130%
Fibronectin Protein100%204%144%
Basement Membrane Width85 nm139 nm105 nm
Antisense oligonucleotides reduced fibronectin overexpression and vascular pathology .

Hepatic Stellate Cell Activation

  • ERK Phosphorylation: Fibronectin induces ERK activation within 5 minutes via Src/Shc signaling .

  • Inhibition: RGD peptides and α5β1 antibodies block ERK phosphorylation, suppressing endothelin-1 synthesis .

Applications in Research

  • Cell Culture: Enhances adhesion for low-serum assays (10–50 µg/ml) .

  • Disease Models: Used to study liver fibrosis , diabetic retinopathy , and cardiac hypertrophy .

  • Therapeutic Targets: Antisense oligonucleotides against fibronectin mRNA mitigate ECM thickening .

Product Specs

Description
Plasma fibronectin is a glycoprotein that exists in two primary forms: a soluble dimer in plasma and an insoluble dimer in the extracellular matrix (ECM). Elevated levels of plasma fibronectin are linked to severe coronary artery disease and venous thromboembolism (VTE), particularly in males, suggesting a potential connection between biomarkers, arterial atherothrombosis risk factors, and VTE. This protein is involved in numerous cellular processes, including tissue repair, embryogenesis, blood clotting, and cell migration/adhesion. It functions as a general cell adhesion molecule, facilitating the binding of cells to collagen or proteoglycan substrates. Fibronectin plays a critical role in organizing cellular interaction with the ECM by binding to various ECM components and membrane-bound fibronectin receptors on cell surfaces. The plasma form is produced by hepatocytes, while fibroblasts, chondrocytes, endothelial cells, macrophages, and some epithelial cells synthesize the ECM form. This molecule has a molecular weight of 220kDa.
Physical Appearance
Sterile white powder, lyophilized (freeze-dried).
Formulation
Lyophilized from a 1 mg/ml solution in 20 mM Tris-Cl, pH 7.5.
Solubility
Reconstitute the lyophilized Rat Fibronectin in sterile 18M-cm H₂O to a concentration of at least 100 µg/ml. This solution can be further diluted in other aqueous solutions.
Stability
Store lyophilized fibronectin at 4°C. After reconstitution, store at 4°C for up to 2 weeks. For long-term storage, keep at -18°C. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 95.0% as determined by SDS-PAGE analysis.
Source
Rat Plasma.

Q&A

What is fibronectin and what are its primary functions in rat tissues?

Fibronectin is a large glycoprotein found in the extracellular matrix with numerous binding sites for other matrix components and cells. In rats, as in other mammals, fibronectin exists in two main forms: plasma fibronectin (p-FN), which is primarily secreted by hepatocytes into the circulation, and cellular fibronectin (c-FN), which is produced by fibroblasts, epithelial cells, and other cell types and deposited as fibrils in the extracellular matrix . Functionally, fibronectin plays crucial roles in cell adhesion, cell motility, wound healing, and maintenance of cell shape. It also participates in opsonization, which facilitates the clearance of particulate material from circulation, potentially contributing to the clearance of immune complexes or cellular debris .

How does rat fibronectin differ structurally from human fibronectin?

While rat and human fibronectin share significant structural homology, there are species-specific differences that researchers should consider. Both contain similar functional domains, including the alternatively spliced regions EIIIA, EIIIB, and V. Cross-reactivity exists between certain antibodies that recognize both human and rat fibronectin, as evidenced by the Human/Rat Fibronectin Antibody (AF1918), which detects fibronectin in both species at approximately 300 kDa when analyzed by Western blotting . This cross-reactivity suggests conserved epitopes across species, though researchers should still validate antibody specificity when transitioning between human and rat models. When designing experiments, it's important to use species-specific detection tools whenever possible to ensure accurate results.

What are the major isoforms of fibronectin expressed in rat tissues?

Rat fibronectin exists in multiple isoforms resulting from alternative splicing of the single FN pre-mRNA, particularly in the EIIIA, EIIIB, and V regions. During development and in response to tissue injury, the expression pattern of these isoforms is tightly regulated. Plasma fibronectin typically lacks the EIIIA and EIIIB exons, while cellular fibronectin may contain various combinations of these regions . In rat fetal and postnatal development, the expression of FN-EIIIA and FN-EIIIB isoforms follows a specific temporal pattern, with higher expression during early developmental stages that progressively decreases with age . Additionally, the V region can be alternatively spliced into different forms, including the V120 form which has been shown to increase from 11% to 32% during liver regeneration .

What are the most reliable methods for detecting fibronectin in rat tissue samples?

For detecting fibronectin in rat tissue samples, several complementary methodologies offer reliable results. Immunohistochemistry (IHC) with specific antibodies allows visualization of fibronectin distribution within tissue sections. For example, fibronectin can be detected in paraffin-embedded sections using anti-fibronectin antibodies followed by appropriate detection systems, such as HRP-DAB staining with hematoxylin counterstaining . Western blotting provides quantitative assessment of fibronectin levels, with detection typically showing a band at approximately 300 kDa under reducing conditions . For more sensitive quantification, ELISA methods offer detection limits below 15 pg/ml in various sample types including serum, plasma, and cell culture supernatants . RT-PCR enables analysis of fibronectin mRNA expression and isoform distribution, while in situ hybridization can reveal the cellular sources of fibronectin synthesis within tissues . Each method has specific advantages, and combining multiple approaches provides the most comprehensive characterization.

How can I accurately quantify fibronectin levels in rat plasma and tissue samples?

Accurate quantification of fibronectin in rat samples requires careful consideration of sample preparation and detection methods. For plasma and serum samples, ELISA kits specifically designed for rat fibronectin offer high sensitivity (detection limits <15 pg/ml) and specificity with no detectable cross-reactivity with other relevant proteins . These assays typically employ a sandwich format with capture and detection antibodies specific to rat fibronectin. The Rat Fibronectin Rapid ELISA Kit provides a working range of 156-10,000 pg/ml, making it suitable for most physiological concentrations . For tissue samples, protein extraction should be optimized based on tissue type, with consideration for soluble versus matrix-bound fibronectin. Western blotting with densitometric analysis offers semi-quantitative assessment, while mass spectrometry-based approaches can provide absolute quantification. When comparing samples, it's essential to normalize fibronectin levels to total protein content or use appropriate housekeeping proteins as references.

What protocols are recommended for immunohistochemical detection of fibronectin in rat tissues?

For immunohistochemical detection of fibronectin in rat tissues, the following protocol is recommended based on published methodologies: Begin with immersion fixation of tissue samples, followed by paraffin embedding and sectioning. For optimal detection in rat liver sections, incubate with anti-fibronectin antibodies (such as AF1918) at a concentration of approximately 10 μg/mL overnight at 4°C . Signal detection can be performed using HRP-DAB systems (such as Anti-Sheep HRP-DAB Cell & Tissue Staining Kit) followed by hematoxylin counterstaining to visualize tissue architecture . For immunofluorescence detection in cultured rat cells, fix cells on coverslips and incubate with primary antibodies at 10 μg/mL for 3 hours at room temperature, followed by fluorophore-conjugated secondary antibodies such as NorthernLights 557-conjugated Anti-Sheep IgG . DAPI counterstaining helps visualize nuclei. For both methods, include appropriate negative controls (primary antibody omission) and positive controls (tissues known to express fibronectin) to validate staining specificity.

What role does fibronectin play in rat liver regeneration models?

Fibronectin plays a significant role during rat liver regeneration, with notable reprogramming of its expression and alternative splicing patterns. Following partial hepatectomy, rat liver shows a 3-fold increase in fibronectin mRNA levels, which is specifically linked to the regeneration process rather than surgical stress . More remarkably, liver regeneration triggers significant alterations in fibronectin isoform expression. While normal rat liver primarily produces plasma fibronectin lacking EIIIA and EIIIB exons, regenerating liver tissue begins producing up to 17% of EIIIA+ fibronectin linked with all three V forms . Additionally, the V120 form increases from 11% to 32% during regeneration, being linked to both EIIIA+ and EIIIA- messengers . Interestingly, the EIIIB+ form remains completely absent in both normal and regenerating liver, indicating differential regulation of alternative splicing for the EIIIA and EIIIB regions during regeneration . These changes suggest that specific fibronectin isoforms may contribute to the extracellular matrix remodeling and cellular interactions needed for effective liver regeneration.

What methodologies are available for studying fibronectin isoform-specific functions in rat models?

Advanced methodologies for studying isoform-specific functions of rat fibronectin include selective targeting approaches combined with comprehensive phenotypic assessments. To differentiate between isoforms containing alternatively spliced EIIIA, EIIIB, and V regions, researchers can employ isoform-specific RT-PCR primers designed to amplify across splice junctions, allowing quantitative analysis of isoform ratios . For protein-level analysis, isoform-specific antibodies that recognize epitopes within the alternatively spliced regions enable selective detection by immunoblotting, immunohistochemistry, or flow cytometry. Functional studies benefit from recombinant expression systems producing specific fibronectin isoforms for in vitro cell behavior assays. More sophisticated approaches include adeno-associated virus (AAV) vectors encoding specific fibronectin isoforms for targeted overexpression, or CRISPR/Cas9-mediated genome editing to modify splice regulatory elements. RNA-based interventions using antisense oligonucleotides or siRNAs can selectively modulate isoform expression by interfering with alternative splicing machinery. These approaches help elucidate the distinct biological roles of fibronectin isoforms during development, tissue repair, and pathological conditions in rat models.

How can I design experiments to analyze the interaction between rat fibronectin and its cellular receptors?

Designing experiments to analyze rat fibronectin-receptor interactions requires multi-level approaches spanning from molecular to cellular analyses. First, solid-phase binding assays using purified rat fibronectin or specific domains (particularly the cell-binding RGD domain) with soluble integrin receptors can establish basic binding parameters. Surface plasmon resonance (SPR) or biolayer interferometry enable real-time kinetic measurements of these interactions. At the cellular level, adhesion assays using rat cells on fibronectin-coated surfaces, with or without function-blocking antibodies against specific integrins (particularly α5β1 and αvβ3), help identify the primary receptors mediating adhesion. Immunoprecipitation of fibronectin-receptor complexes from rat cells followed by mass spectrometry can reveal the complete interactome. For spatial analysis, proximity ligation assays or FRET microscopy visualize fibronectin-receptor interactions within native tissue contexts. Downstream signaling can be assessed through phosphoproteomic analysis following fibronectin engagement. For in vivo relevance, conditional knockout models targeting specific integrin receptors in rats, coupled with fibronectin challenge, can demonstrate the physiological importance of specific interactions in development or disease models.

What approaches can be used to study the role of fibronectin in extracellular matrix assembly in rat tissue engineering applications?

Studying fibronectin's role in extracellular matrix (ECM) assembly for rat tissue engineering applications requires integrated approaches spanning molecular, cellular, and tissue levels. At the molecular level, fluorescently labeled rat fibronectin allows visualization of fibril formation dynamics using time-lapse microscopy. Fibronectin fragments containing specific domains can determine which regions are essential for fibril assembly in rat cell cultures. For cellular analysis, decellularized ECM derived from rat tissues provides a native scaffold to study how cells remodel pre-existing fibronectin networks. Mechanical tension studies using flexible substrates demonstrate how cellular contractile forces influence fibronectin conformation and assembly. Advanced imaging techniques like second harmonic generation (SHG) microscopy or super-resolution microscopy reveal nanoscale organization of fibronectin within complex ECM networks. For tissue engineering applications, bioprinting technologies incorporating rat fibronectin into bioinks help create defined ECM environments. Bioreactors applying mechanical stimulation to rat cell-seeded scaffolds demonstrate how physical forces regulate fibronectin assembly in engineered tissues. Degradable fibronectin-based hydrogels with tunable mechanical properties serve as platforms to study how matrix stiffness influences rat cell behavior in three-dimensional environments, informing optimal scaffold design for specific tissue engineering applications.

What are common technical challenges when working with rat fibronectin and how can they be addressed?

Working with rat fibronectin presents several technical challenges that require specific strategies to overcome. One major issue is protein degradation during isolation and storage, as fibronectin is susceptible to proteolytic cleavage. This can be addressed by including multiple protease inhibitors (PMSF, EDTA, aprotinin, and leupeptin) in all buffers and maintaining samples at 4°C during processing. For isolation from rat plasma, gelatin-Sepharose affinity chromatography yields the highest purity, but competing proteins can reduce binding efficiency; sequential chromatography with ion exchange followed by gelatin affinity improves results. When working with tissue samples, the insolubility of matrix-incorporated fibronectin necessitates sequential extraction protocols using increasing concentrations of denaturing agents. For immunodetection, the large size of fibronectin (approximately 300 kDa) makes Western blot transfer inefficient; extended transfer times or specialized buffers for high molecular weight proteins improve results . Antibody cross-reactivity between rat and other species may lead to background signals in heterogeneous samples; careful antibody selection and validation with appropriate controls mitigates this issue. Finally, the complex alternative splicing of rat fibronectin challenges accurate isoform quantification; designing PCR primers spanning splice junctions and using digital PCR or RNA sequencing provides more precise measurements .

How should I optimize ELISA protocols for detecting rat fibronectin in different sample types?

Optimizing ELISA protocols for rat fibronectin detection across different sample types requires systematic adjustment of multiple parameters. For serum and plasma samples, dilution optimization is critical as fibronectin concentrations typically exceed the standard curve range; preliminary testing with serial dilutions (1:1000 to 1:10,000) helps identify the optimal range . Different anticoagulants affect fibronectin recovery, with heparin, EDTA, and citrate all being compatible but potentially yielding different absolute values; maintain consistency in anticoagulant choice throughout a study . For tissue extracts, optimization of extraction buffers is essential—use of mild detergents (0.5% NP-40 or Triton X-100) preserves fibronectin structure while enhancing solubility. Cell culture supernatants typically contain lower fibronectin concentrations, requiring minimal dilution or concentration procedures before analysis. For all sample types, the matrix effect can influence assay performance; prepare standards in a matrix matching the samples or use sample-specific standard curves when possible. Sandwich ELISA formats offer superior specificity and sensitivity (detection limits <15 pg/ml); optimize antibody concentrations and incubation times for each sample type . Finally, thorough plate washing (5-7 washes per step) minimizes background, while extended substrate development times may be needed for low-abundance samples. Validation with spike-and-recovery experiments confirms accuracy across different sample matrices.

What reference genes are recommended for normalizing fibronectin expression in qRT-PCR studies of rat tissues?

Selecting appropriate reference genes for normalizing fibronectin expression in qRT-PCR studies of rat tissues requires consideration of tissue type and experimental conditions. No single reference gene maintains perfect stability across all rat tissues and experimental scenarios. For liver regeneration studies, GAPDH shows variable expression and should be avoided as the sole reference gene; instead, a combination of beta-actin and 18S rRNA provides more reliable normalization . In cardiac development studies, HPRT1 (hypoxanthine phosphoribosyltransferase 1) and YWHAZ (tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta) demonstrate superior stability compared to traditional references . For studies involving multiple tissue types, PPIA (peptidylprolyl isomerase A) and RPL13A (ribosomal protein L13a) show consistent expression across diverse rat tissues. The geNorm or NormFinder algorithms should be employed to systematically evaluate reference gene stability within the specific experimental context. Best practice involves using at least three validated reference genes and calculating their geometric mean for normalization. Additionally, validation experiments comparing different reference genes should be performed for each experimental design, as factors like inflammation, hypoxia, or growth factor treatment can differentially affect reference gene expression. When reporting qRT-PCR data for rat fibronectin, explicit documentation of reference gene selection rationale enhances reproducibility and reliability of findings.

How should variations in fibronectin expression between different rat strains be interpreted?

Interpreting variations in fibronectin expression between rat strains requires careful consideration of genetic background, physiological differences, and experimental context. Different rat strains (such as Wistar, Sprague-Dawley, and Fischer) may exhibit baseline differences in fibronectin levels and isoform distributions that reflect their distinct genetic backgrounds. These strain-specific variations can significantly impact experimental outcomes, especially in disease models where fibronectin plays a crucial role. When analyzing such differences, researchers should first establish strain-specific reference ranges through systematic measurement of fibronectin across multiple animals within each strain. Variations exceeding 20% between strains warrant further investigation into the underlying genetic or physiological mechanisms. Strain differences may reflect adaptations to different physiological demands rather than pathological states, necessitating careful interpretation in disease models. Additionally, fibronectin expression patterns may show strain-dependent responses to experimental interventions, requiring strain-matched controls. For translational relevance, researchers should consider how strain-specific fibronectin patterns align with human pathophysiology. Meta-analysis of published literature can help identify consistent strain-dependent effects across multiple studies, strengthening interpretations. Finally, complementary approaches such as genetic mapping or transcriptome analysis can help identify strain-specific regulatory mechanisms underlying observed variations in fibronectin expression.

What are the most significant findings regarding the role of fibronectin in rat models of tissue repair and regeneration?

Research on fibronectin in rat models has revealed several significant findings regarding tissue repair and regeneration. In liver regeneration, one of the most dramatic discoveries is the substantial reprogramming of fibronectin splicing that occurs following partial hepatectomy, with up to 17% of fibronectin incorporating the EIIIA domain that is normally absent from liver-derived plasma fibronectin . This alternative splicing is accompanied by a 3-fold increase in total fibronectin mRNA levels specifically linked to the regeneration process . In cardiac development and repair, the expression pattern of fibronectin shows a progressive decrease from 11 days postconception through adulthood, with temporal disconnection between mRNA and protein levels suggesting complex post-transcriptional regulation . Across multiple tissue types, the adhesive properties of fibronectin play crucial roles in cell migration and tissue organization during repair. Interestingly, despite variations in fibronectin-binding capacity between bacterial strains in models of endocarditis, these differences do not necessarily translate to different infection rates, highlighting the complex interplay between fibronectin and its binding partners in vivo . Together, these findings emphasize fibronectin's multifaceted roles in tissue repair, including providing adhesive scaffolds for cellular migration, regulating cellular phenotypes through receptor interactions, and undergoing dynamic remodeling through alternative splicing to meet tissue-specific regenerative needs.

What emerging technologies show promise for advancing our understanding of rat fibronectin biology?

Several emerging technologies show exceptional promise for advancing our understanding of rat fibronectin biology. Single-cell RNA sequencing now enables unprecedented resolution of cell-specific fibronectin expression patterns within heterogeneous tissues, revealing previously unrecognized cellular sources and isoform distributions. Spatial transcriptomics preserves the tissue context while providing transcriptome-wide data, allowing correlation of fibronectin expression with local microenvironmental features. CRISPR-Cas9 genome editing permits precise modification of fibronectin alternative splicing regulatory elements or domain-specific mutations in rats, creating models for studying domain-specific functions. For protein-level analysis, advanced mass spectrometry approaches including crosslinking mass spectrometry (XL-MS) and hydrogen-deuterium exchange mass spectrometry (HDX-MS) provide detailed structural information about fibronectin conformation and interaction partners in different physiological states. Super-resolution microscopy techniques like STORM and PALM enable visualization of fibronectin fibril assembly and organization at nanoscale resolution. Organ-on-chip technologies incorporating rat cells within defined extracellular matrix compositions allow controlled manipulation of fibronectin presentation while monitoring cellular responses under physiologically relevant conditions. Finally, computational approaches integrating multi-omics data through machine learning algorithms can identify novel regulatory networks controlling fibronectin expression and function across developmental stages and disease conditions, generating testable hypotheses for future experimental validation.

What are important unanswered questions about fibronectin function in rat disease models?

Despite extensive research, several important questions about fibronectin function in rat disease models remain unanswered. The precise signaling mechanisms by which specific fibronectin isoforms influence cell behavior during tissue repair and regeneration are not fully elucidated, particularly regarding how EIIIA+ fibronectin promotes liver regeneration . The relative contributions of locally produced cellular fibronectin versus plasma-derived fibronectin in tissue repair and fibrosis models remain unclear, with evidence suggesting that their roles may be tissue-specific and context-dependent . The temporal dynamics of fibronectin deposition and degradation during the progression of fibrotic diseases in rat models, and how these dynamics influence disease outcomes, warrant deeper investigation. The mechanistic relationship between fibronectin's mechanical properties and cellular responses in different tissue microenvironments remains poorly understood, particularly how matrix stiffness affects fibronectin conformation and subsequent cellular signaling. Additionally, the potential for targeting specific fibronectin isoforms or domains as therapeutic strategies in rat models of fibrosis, thrombosis, or impaired wound healing represents an important translational research direction. Finally, the interaction between fibronectin and other extracellular matrix components in determining tissue-specific responses to injury or disease requires systematic characterization, as these interactions likely influence cellular behavior in complex ways that cannot be predicted from studies of individual matrix components.

How might advanced computational modeling contribute to our understanding of fibronectin structure-function relationships in rat research?

Advanced computational modeling offers transformative potential for understanding fibronectin structure-function relationships in rat research through multiple approaches. Molecular dynamics simulations can predict conformational changes in rat fibronectin under different mechanical forces, revealing how tension exposure of cryptic binding sites might regulate cell behavior in mechanically dynamic environments. Homology modeling between rat and human fibronectin structures identifies conserved functional domains versus species-specific regions, guiding translational research. Protein-protein interaction modeling predicts binding interfaces between fibronectin and its numerous partners, including integrins, collagen, and growth factors, generating testable hypotheses about domain-specific functions. Machine learning algorithms integrating transcriptomic, proteomic, and phenotypic data can identify novel regulatory relationships in fibronectin expression networks across different physiological and pathological states. Agent-based modeling at the cellular scale simulates how fibronectin deposition patterns influence collective cell behaviors during development and wound healing. Multiscale modeling integrates molecular, cellular, and tissue-level simulations to predict how molecular-level changes in fibronectin structure propagate to tissue-level functional outcomes. Finite element modeling of fibronectin matrices with different architectural features predicts their mechanical properties and potential influence on cellular mechanotransduction. These computational approaches complement experimental studies by generating predictions across scales and conditions that would be challenging to test experimentally, accelerating discovery while reducing animal usage in accordance with 3Rs principles (Replacement, Reduction, Refinement).

Product Science Overview

Structure and Isoforms

Fibronectin is a dimer composed of two nearly identical subunits, each approximately 250 kDa in size . These subunits are linked covalently near their C-termini by a pair of disulfide bonds. Each monomer consists of three types of repeating units: type I, type II, and type III repeats . The protein can exist in multiple forms due to alternative splicing of a single pre-mRNA, which can generate as many as 20 variants in human fibronectin . This splicing occurs in several regions, including the EDA and EDB domains, which are highly expressed during fibrosis and cancer development .

Biological Functions

Fibronectin is involved in a wide variety of cellular interactions with the extracellular matrix (ECM) and plays important roles in cell adhesion, migration, growth, and differentiation . It is critically important in vertebrate development, as demonstrated by the early embryonic lethality of mice with targeted inactivation of the fibronectin gene . In rats, fibronectin is synthesized by various cell types, including fibroblasts, endothelial cells, and certain cancer cells .

Tissue Distribution

Fibronectin is widely expressed in multiple tissues and organs. Plasma fibronectin is synthesized by hepatocytes and exists in a compact, inactive conformation in the bloodstream . It is a major component of fibrin clots and plays a significant role in wound healing. Cellular fibronectin isoforms are synthesized by fibroblasts, endothelial cells, and many types of cancer cells .

Regulatory Mechanisms

The expression and function of fibronectin are regulated by various factors, including growth factors, cytokines, and mechanical stress. Fibronectin binds to distinct ECM components such as collagens, growth factors, and cell surface integrins to carry out its functions . The protein undergoes conformational changes upon binding to integrins or other cell surface receptors, exposing its functional domains and activating extracellular matrix assembly .

Clinical Implications

Fibronectin has been implicated in many essential biological processes, including tissue repair, fibrosis, and tumor development . The EDA and EDB isoforms of fibronectin are particularly highly expressed during fibrosis and cancer development, making them potential diagnostic and therapeutic targets . Understanding the role of fibronectin in these processes can provide insights into the development of new treatments for various diseases.

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.