Recombinant TGFB1 is synthesized using diverse expression systems, each with distinct advantages:
TGFB1 Human Recombinant is utilized in:
Stimulates extracellular matrix production (collagen, fibronectin) .
Enhances mesenchymal stem cell differentiation into chondrocytes .
Plant-derived TGFB1 shows comparable activity to HEK293-produced protein (EC50: 0.5–1 ng/mL) .
Mutant variants (e.g., Cys33Ser) improve solubility without compromising receptor binding .
Recombinant TGFB1 supplementation restored immune tolerance in patient-derived T-cells (10 ng/mL, p < 0.001) .
Transforming growth factor betas (TGF-betas) are involved in cell-to-cell communication during embryonic development in mammals. There are three known types of TGF-betas: TGF-beta1, TGF-beta2, and TGF-beta3. All three TGF-betas are initially synthesized as larger precursor proteins. These precursor proteins undergo cleavage, resulting in a 112-amino acid polypeptide that remains connected to the inactive part of the molecule.
Recombinant human TGFB1, produced in CHO cells, is a glycosylated homodimeric polypeptide chain consisting of 2 chains of 112 amino acids each. The total molecular weight of the protein is 25.6kDa. TGFB1 is purified using proprietary chromatographic methods.
The product appears as a sterile, filtered, white, lyophilized (freeze-dried) powder.
The product is lyophilized from a sterile, filtered solution containing 0.1% trifluoroacetic acid (TFA) and trehalose at a protein-to-trehalose ratio of 1:20.
To reconstitute the lyophilized TGFB1, it is recommended to dissolve it in sterile 10mM HCl at a concentration of 0.1 mg/ml. This solution can then be further diluted in other aqueous solutions as needed.
Lyophilized TGFB1, while stable at room temperature for up to 3 weeks, should be stored in a dry environment below -18°C. Once reconstituted, the human TGFB1 should be stored at 4°C for 2-7 days. For long-term storage, it is recommended to store it below -18°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Repeated freezing and thawing cycles should be avoided.
The purity of the product is greater than 95.0% as assessed by SDS-PAGE analysis.
The ED50, determined by the dose-dependent inhibition of IL-4-induced proliferation of HT-2 cells, is 0.0149 ng/ml. This corresponds to a specific activity of 6.7 x 10^7 units/mg.
CHO cells.
Transforming Growth Factor Beta-1 (TGFB1) is a secreted protein belonging to the TGF-β family. The protein exists initially as a precursor that undergoes cleavage into two main components: the Latency-Associated Peptide (LAP) and the mature TGFB1 peptide. In its native state, these components remain non-covalently associated post-secretion, forming what is known as the small latent TGFB1 complex . Recombinant TGFB1 protein is typically produced using mammalian expression systems, such as HEK293 cells, to ensure proper folding and post-translational modifications essential for biological activity . The mature, active form corresponds to amino acids 279-390 of the full precursor protein, while the LAP component spans amino acids 30-278 .
The protein structure involves disulfide-linked homodimers that are critical for its functionality. Most commercially available recombinant proteins include specific mutations (like Cys33Ser) that enhance stability while maintaining biological activity . The molecular weight of the active form is approximately 25 kDa, while the LAP component has a theoretical molecular weight of around 31.4 kDa .
TGFB1 performs numerous cellular functions that make it a focus of diverse research areas. The protein regulates cell growth, proliferation, differentiation, and apoptosis across multiple tissue types . It is abundantly expressed in bone, articular cartilage, and chondrocytes, with increased expression observed in osteoarthritis (OA) . In neurological research, TGFB1 has been implicated in normal development, immune function, microglia function, and responses to neurodegeneration .
The protein's signaling pathways interact with numerous cellular processes including the cell cycle, MAPK signaling pathway, and cytokine-cytokine receptor interactions . TGFB1 is involved in both normal physiological processes and pathological conditions such as chronic myeloid leukemia, colorectal cancer, pancreatic cancer, dilated cardiomyopathy, and hypertrophic cardiomyopathy . Understanding these diverse functions is essential for researchers designing experiments to elucidate TGFB1's role in specific biological contexts.
Recombinant human TGFB1 is predominantly produced using mammalian expression systems, particularly HEK293 cells, to ensure proper folding and post-translational modifications essential for biological activity . The expression of TGFB1 involves encoding specific amino acid sequences (e.g., Leu30-Arg278 with Cys33Ser mutation for the LAP component) . The recombinant proteins are typically tagged (often with His-tags) to facilitate purification and detection in experimental settings .
Researchers should evaluate several quality parameters before using recombinant TGFB1:
Additionally, researchers should verify protein identity through sequence confirmation and assess for proper folding through functional assays appropriate to their experimental design .
Reconstitution of lyophilized TGFB1 recombinant protein requires careful handling to maintain structural integrity and biological activity. Based on manufacturer protocols, the following method is recommended:
Allow the lyophilized protein to equilibrate to room temperature (20-25°C) for approximately 30 minutes before opening the vial to prevent moisture condensation on the lyophilized material.
Reconstitute using sterile, buffered solutions such as PBS (pH 7.4). For optimal results, gently add the reconstitution buffer along the sides of the vial rather than directly onto the lyophilized cake .
Allow the protein to dissolve completely by gentle rotation or inversion of the vial. Avoid vigorous vortexing or pipetting, as this can cause protein denaturation and aggregation.
Once reconstituted, the solution should be allowed to stand for 5-10 minutes at room temperature before use to ensure complete solubilization .
For applications requiring higher protein concentrations, reconstitution volumes can be adjusted, but researchers should verify that the protein remains fully soluble at higher concentrations.
The reconstituted protein may be used immediately or aliquoted and stored according to recommended storage conditions to avoid repeated freeze-thaw cycles .
Validating the biological activity of TGFB1 is crucial for experimental reliability. Several complementary approaches are recommended:
Receptor Binding Assays: ELISA-based methods can measure binding to receptors or interaction partners. A typical approach involves immobilizing LAP (TGF beta 1) at 2 μg/mL and measuring binding to biotinylated integrin partners (ITGAV&ITGB6) . The EC50 value (approximately 0.46 μg/mL) provides a quantitative measure of binding affinity.
Cell-Based Functional Assays: Several cell lines respond to TGFB1 with measurable outcomes:
Growth inhibition of Mv1Lu mink lung epithelial cells
Induction of PAI-1 expression in HepG2 cells
SMAD2/3 phosphorylation in responsive cell lines
Signal Transduction Verification: Western blot analysis for SMAD2/3 phosphorylation directly demonstrates activation of the canonical TGFB1 signaling pathway.
Gene Expression Analysis: qRT-PCR for known TGFB1-responsive genes (e.g., PAI-1, CTGF, COL1A1) can confirm functional activity.
For all activity measurements, researchers should include a standard curve using a reference TGFB1 preparation with known activity to normalize results between experiments .
When comparing different TGFB1 isoforms or species variants, researchers should address several critical experimental considerations:
Sequence Homology Analysis: While TGFB1 is highly conserved across species, subtle amino acid differences may affect receptor binding affinities and downstream signaling. Researchers should perform sequence alignment analysis and consider evolutionary conservation patterns when interpreting cross-species experiments.
Receptor Interaction Profiles: Different TGFB isoforms (TGFB1, TGFB2, TGFB3) have varying affinities for the TGF-beta receptor complex, which affects signaling intensity and duration. When comparing isoforms, concentration-response curves should be established for each variant to identify differences in potency.
Activation Requirements: The LAP component of TGFB1 can complex with and inactivate other human TGFB isoforms . This cross-regulation capability must be considered in experimental designs involving multiple isoforms.
Cell Type-Specific Responses: Different cell types express varying levels of TGFB receptors and signaling components, leading to context-dependent responses. Researchers should validate findings across multiple relevant cell types.
Quantification Methods: When comparing different TGFB variants, consistent quantification methods should be employed, ideally using both mass-based (e.g., ng/mL) and activity-based (e.g., ED50) measurements to account for potential differences in specific activity.
Creating standardized experimental protocols that address these considerations will enhance the validity and reproducibility of comparative studies involving different TGFB1 variants.
Proper storage of TGFB1 recombinant protein is critical for maintaining its structural integrity and biological activity. Based on established protocols, the following storage conditions are recommended:
Lyophilized Protein: Store at -20°C or preferably at -80°C. Though the lyophilized form is relatively stable and can maintain integrity at room temperature for up to 3 weeks, long-term storage at lower temperatures significantly extends shelf-life .
Reconstituted Protein:
Aliquoting Strategy: To prevent protein degradation from repeated freeze-thaw cycles, reconstituted protein should be divided into single-use aliquots before freezing. Each freeze-thaw cycle can decrease biological activity by 10-30% .
The expected stability timeframe under optimal storage conditions is approximately 12 months from the date of preparation . Researchers should maintain detailed records of receipt dates, reconstitution dates, and storage conditions to track potential activity loss over time.
Verifying TGFB1 activity after storage periods is essential for experimental validity. A comprehensive approach involves:
Physical Inspection: Before use, visually inspect reconstituted TGFB1 solutions for any signs of precipitation, turbidity, or discoloration that may indicate denaturation or contamination.
Activity Verification Assays:
ELISA-Based Binding Assay: Compare binding efficiency to known TGFB1 interaction partners like ITGAV&ITGB6 integrins. Shifts in EC50 values (from the expected ~0.46 μg/mL) may indicate activity loss .
Cell-Based Functional Assay: Compare the dose-response curve in a standard bioassay (e.g., SMAD phosphorylation in responsive cells) to a freshly reconstituted reference standard.
Protein Integrity Analysis:
SEC-HPLC: To detect potential aggregation or degradation products
SDS-PAGE: To verify molecular weight and absence of degradation bands
Researchers should establish their own internal reference standards and acceptance criteria based on their specific experimental requirements. As a general guideline, preparations showing greater than 30% reduction in activity compared to fresh standards should be replaced with new preparations to ensure experimental reliability.
Maintaining TGFB1 activity throughout experimental procedures requires attention to several factors:
Carrier Protein Addition: For dilute solutions (<10 μg/mL), adding carrier proteins such as bovine serum albumin (0.1-0.5%) can prevent activity loss due to adsorption to laboratory plasticware and increase stability during storage.
Temperature Management:
Keep reconstituted protein on ice when in use
Allow solutions to reach room temperature before adding to cells to prevent temperature shock
Avoid repeated temperature cycling between refrigeration and room temperature
Buffer Considerations:
Mechanical Stress Reduction:
Avoid vigorous vortexing or pipetting
Use gentle rotation or inversion for mixing
Minimize air-liquid interface exposure that can cause protein denaturation
Workflow Planning:
Minimize the number of handling steps between storage and experimental use
Prepare fresh working solutions for each experiment rather than storing diluted preparations
By implementing these protective measures, researchers can significantly reduce activity losses during experimental procedures and improve reproducibility across experiments.
Several common challenges arise in TGFB1 research, each requiring specific troubleshooting approaches:
Activation Status Inconsistency: TGFB1 exists in both latent and active forms, which can lead to variability in experimental outcomes.
Interference from Endogenous TGFB1: Many cell types produce TGFB1, which can confound exogenous TGFB1 experiments.
Solution: Consider using TGFB1-neutralizing antibodies as controls, or TGFB1-knockout cell models for cleaner experimental systems.
Binding to Experimental Surfaces: TGFB1 can adsorb to plastic and glass surfaces, reducing effective concentration.
Solution: Pre-coat surfaces with BSA (0.1%) or use low-binding laboratory plasticware. Include carrier proteins in dilute TGFB1 solutions.
Context-Dependent Cellular Responses: TGFB1 effects vary dramatically by cell type, density, and culture conditions.
Solution: Always include appropriate positive and negative controls specific to the cell type. Perform dose-response curves for each new experimental system.
Receptor Saturation/Desensitization: Prolonged or high-dose TGFB1 exposure can down-regulate receptor expression.
Solution: Consider pulse treatments rather than continuous exposure for long-term experiments. Monitor receptor expression levels when relevant.
Each of these challenges can be mitigated through careful experimental design and appropriate controls tailored to the specific research question being addressed.
The dual nature of TGFB1—existing in both latent and active forms—represents a significant experimental consideration requiring specific design strategies:
Activation Status Characterization: Before designing experiments, researchers should determine whether they need to study:
Active TGFB1 signaling (using the mature 25 kDa form)
Latent TGFB1 biology (using the LAP-complexed form)
Activation mechanisms (using the full latent complex)
Form-Specific Experimental Approaches:
For Active TGFB1 Studies: Use recombinant mature TGFB1 (amino acids 279-390) for direct signaling experiments .
For Latent TGFB1 Studies: Use recombinant LAP (TGF beta 1) to examine regulatory mechanisms .
For Activation Studies: Include physiological activators such as integrins (ITGAV&ITGB6), proteases (plasmin, MMP9), or thrombospondin 1 .
Control Strategies:
Include both active and latent forms as controls when studying activation mechanisms
Use neutralizing antibodies specific to either the LAP or mature domains to distinguish effects
Consider cell systems with impaired TGFB1 activation machinery to study latent form functions
Quantification Methods:
Use ELISAs that distinguish between active and total TGFB1
Employ bioassays (such as SMAD phosphorylation) that only detect active forms
Consider reporter cell lines specifically responsive to active TGFB1
Physiological Context Consideration:
By explicitly addressing the latent versus active nature of TGFB1 in experimental design, researchers can avoid misinterpretation of results and gain more precise insights into TGFB1 biology.
Robust TGFB1 signaling studies require comprehensive control experiments to ensure valid interpretation of results:
Dose-Response Controls:
Include a full dose-response curve (typically ranging from 0.01-10 ng/mL for active TGFB1)
Determine both threshold and saturation concentrations for your specific cell system
Include a time-course analysis to capture both early (SMAD-dependent) and late (SMAD-independent) signaling events
Specificity Controls:
Neutralizing Antibody Control: Include anti-TGFB1 neutralizing antibodies to confirm observed effects are specifically due to TGFB1
Receptor Inhibition Control: Use TGFBR1 kinase inhibitors (e.g., SB431542) to verify canonical pathway involvement
Isoform Controls: When possible, compare with other TGFB isoforms (TGFB2, TGFB3) to identify isoform-specific versus shared effects
Pathway Validation Controls:
Positive Control: Include a well-characterized TGFB1-responsive cell line or readout system
SMAD Pathway Control: Monitor SMAD2/3 phosphorylation as a direct indicator of canonical pathway activation
Non-SMAD Pathway Control: Assess involvement of parallel pathways (MAPK, PI3K) using specific inhibitors
Cell System Controls:
Receptor Expression Verification: Confirm expression of TGFB receptors in your experimental system
Cell Density Control: Maintain consistent cell densities, as this affects TGFB1 responsiveness
Serum Controls: Use serum-free or serum-reduced conditions to minimize interference from serum-derived TGFB1
Technical Controls:
Vehicle Control: Ensure that the buffer used for TGFB1 reconstitution doesn't affect the readout
Heat-Inactivated Control: Use heat-denatured TGFB1 to confirm that biological activity rather than contaminants drives observed effects
This comprehensive control framework helps distinguish specific TGFB1 effects from non-specific observations, improving data reliability and interpretability.
Implementing TGFB1 in advanced 3D culture and organoid systems requires considerations beyond traditional 2D approaches:
Delivery Optimization:
Gradient Formation: Consider using hydrogel or matrix systems that allow for controlled release of TGFB1, creating physiological gradients
Matrix Incorporation: Pre-incorporate TGFB1 into extracellular matrix components (e.g., Matrigel, collagen) to mimic natural sequestration
Timed Release Systems: Utilize biodegradable microspheres or nanoparticles for sustained, controlled TGFB1 release over extended periods
Concentration Adjustments:
3D systems typically require 2-5 fold higher TGFB1 concentrations than 2D cultures due to diffusion limitations and matrix binding
Establish specific dose-response curves for each 3D system rather than transferring 2D protocols
Consider that TGFB1 concentration gradients in 3D may create heterogeneous cellular responses
Activation Considerations:
Analysis Adaptations:
Develop imaging protocols for visualizing TGFB1 signaling in intact 3D structures (e.g., phospho-SMAD immunofluorescence)
Consider single-cell analysis techniques to capture heterogeneous responses within 3D structures
Implement computational modeling to understand TGFB1 diffusion and gradient formation
Specific Applications:
Organoid Differentiation: Pulse TGFB1 treatment at specific developmental timepoints
Tissue Engineering: Use TGFB1 gradients to guide spatial organization
Disease Modeling: Compare TGFB1 responses in normal versus disease-derived organoids
These specialized approaches enable researchers to more accurately model the complex TGFB1 biology that occurs in three-dimensional tissue environments.
Investigating TGFB1 cross-talk with other signaling pathways requires sophisticated experimental approaches:
Sequential Stimulation Protocols:
Use timed administration of TGFB1 and other pathway agonists to distinguish between priming effects and true cross-talk
Implement wash-out periods between stimulations to prevent direct ligand interaction
Compare simultaneous versus sequential stimulation to identify temporal aspects of cross-talk
Pathway-Specific Inhibition Matrix:
Create an inhibitor matrix combining TGFB1 pathway inhibitors (e.g., SB431542) with inhibitors of potential cross-talk pathways
Analyze both canonical outcomes (e.g., SMAD phosphorylation) and non-canonical outcomes (e.g., ERK activation)
Identify synergistic, additive, or antagonistic effects through statistical interaction analysis
Multi-Parameter Readout Systems:
Implement phospho-flow cytometry to simultaneously track multiple phosphorylation events at the single-cell level
Use multiplexed transcriptomics to identify gene sets regulated by pathway cross-talk rather than individual pathways
Apply proteomic approaches to map signaling network changes comprehensively
Genetic Modification Strategies:
Use CRISPR-Cas9 to knockout or modify key nodes in potential cross-talk pathways
Implement inducible expression systems to control timing and magnitude of signaling component expression
Develop fluorescent reporter systems for real-time visualization of multiple pathway activities
Mathematical Modeling Integration:
Develop computational models incorporating known signaling kinetics of TGFB1 and cross-talk pathways
Predict and experimentally validate points of pathway convergence and divergence
Use sensitivity analysis to identify critical nodes in the integrated signaling network
This systematic approach allows researchers to move beyond observational studies of pathway cross-talk toward mechanistic understanding of signaling integration.
Leveraging recombinant TGFB1 in disease modeling and therapeutic development requires specialized approaches tailored to pathological contexts:
Disease-Specific Dosing Paradigms:
Fibrotic Diseases: Use chronic, low-dose TGFB1 exposure (0.5-2 ng/mL) to mimic pathological states in fibrosis models
Cancer Models: Implement biphasic dosing protocols to reflect TGFB1's dual role as both tumor suppressor and promoter
Inflammatory Conditions: Coordinate TGFB1 administration with inflammatory stimuli to study its immunomodulatory functions
Therapeutic Intervention Strategies:
Neutralization Approaches: Test antibodies or trap proteins for efficacy in neutralizing excess TGFB1 activity
Activation Modulation: Use recombinant LAP components to study potential therapeutic targeting of TGFB1 activation mechanisms
Receptor Targeting: Combine recombinant TGFB1 with receptor modulators to identify optimal intervention points
Pathway Manipulation Assessment:
Use recombinant TGFB1 to establish baseline pathway activation in patient-derived cells
Compare responses between healthy and disease-state cells to identify altered signaling nodes
Test pathway-specific interventions in conjunction with recombinant TGFB1 challenge
Biomarker Development Applications:
Identify TGFB1-responsive genes or proteins that could serve as disease biomarkers
Develop cell-based reporter systems to measure active TGFB1 levels in patient samples
Correlate in vitro TGFB1 responsiveness with clinical parameters to establish predictive models
Multi-System Disease Modeling:
Implement co-culture systems incorporating multiple cell types relevant to disease pathology
Use organ-on-chip technologies with controlled TGFB1 administration to model complex disease environments
Develop in vivo releasing systems for spatiotemporally controlled TGFB1 delivery in animal models
This disease-focused approach facilitates translation of basic TGFB1 biology into clinically relevant applications and potential therapeutic developments for conditions where TGFB1 dysregulation plays a central role, such as fibrosis, cancer, and autoimmune disorders .
TGF-β1 was originally identified for its ability to induce phenotypic transformation of fibroblasts . It is the most abundant isoform and is secreted by almost every cell type . TGF-β1 is involved in various physiological processes, including embryogenesis, tissue remodeling, and wound healing . It is secreted predominantly as latent complexes, which are stored at the cell surface and in the extracellular matrix . The release of biologically active TGF-β1 from a latent complex involves proteolytic processing of the complex and/or induction of conformational changes by proteins such as thrombospondin-1 .
Human recombinant TGF-β1 is synthesized using genetic engineering techniques, enabling the production of large quantities of biologically active protein for research purposes . The recombinant form is typically derived from human embryonic kidney (HEK293) cells . The protein is a 25.0 kDa molecule with each subunit containing 112 amino acid residues, linked by a single disulfide bond . The recombinant TGF-β1 is purified to a high degree, with a purity of ≥ 98% as determined by SDS-PAGE gel and HPLC analyses .
The biological activity of TGF-β1 is measured by its ability to inhibit the IL-4-dependent proliferation of mouse HT-2 cells . The effective dose (ED50) for this activity is ≤ 0.05 ng/ml, corresponding to a specific activity of ≥ 2 x 10^7 units/mg . TGF-β1 has been implicated in various biological processes, including the formation of skin tumors . It is also involved in angiogenesis, immune regulation, and the promotion or inhibition of various immune events .
Due to its wide range of biological activities, TGF-β1 is a subject of extensive research. It has been studied in the context of cancer, fibrosis, and immune disorders . The recombinant form of TGF-β1 is used in various research applications to study its effects on cell proliferation, differentiation, and immune modulation . It is also used in tissue engineering and regenerative medicine due to its role in tissue remodeling and wound healing .