TGFB3 (207 a.a.) Human

Transforming Growth Factor-Beta 3 (207 a.a.) Human Recombinant
Shipped with Ice Packs
In Stock

Description

Developmental Regulation

  • Embryogenesis: Essential for palate formation, lung development, and mammary gland morphogenesis . TGFB3 knockout mice exhibit cleft palate and neonatal lethality .

  • Cell Differentiation: Mediates stem cell differentiation by regulating extracellular matrix (ECM) adhesion molecules and cytoskeletal dynamics .

Immune Modulation

  • Anti-inflammatory: Suppresses IFN-γ production by Th1 cells and promotes regulatory T-cell (Treg) differentiation .

  • Pro-inflammatory Contexts: In autoimmune diseases like multiple sclerosis, TGFB3 induces pathogenic Th17 cells via T-bet-dependent pathways, contrasting TGF-β1’s anti-inflammatory roles .

Isoform-Specific Signaling

FeatureTGF-β3TGF-β1
EMT InductionCritical in cardiac morphogenesis Less potent in epithelial-mesenchymal transition (EMT)
Wound HealingReduces scarring via ECM remodeling Promotes fibrosis
Immune PolarizationDrives pathogenic Th17 cells Induces Tregs

Research Applications

TGFB3 (207 a.a.) Human is utilized in:

  • Stem Cell Studies: Directs differentiation of mesenchymal stem cells into chondrogenic lineages .

  • Immunoassays: Serves as a standard in ELISA and Western blotting for quantifying TGF-β3 in biological samples .

  • Therapeutic Development: Investigated for reducing post-surgical scarring and modulating autoimmune responses .

Primary Pathways

  1. SMAD-Dependent:

    • Binds TGF-β receptors (TβRII/TβRI), phosphorylating SMAD2/3 to regulate gene transcription .

    • Cross-talk with PI3K/AKT enhances survival signals via mTOR activation .

  2. Non-Canonical:

    • Activates Rho GTPases (RHO, RAC1) to reorganize actin cytoskeleton, influencing cell migration .

    • Synergizes with ERK pathways to amplify pro-fibrotic responses .

Key Downstream Targets

EffectorFunctionCitation
ROCK1/LIMK2Phosphorylates cofilin, stabilizing actin
PI3K/AKT/mTORPromotes cell survival and metabolic adaptation
FOXO Transcription FactorsLinks TGF-β3 to apoptosis regulation

Clinical and Pathological Relevance

  • Cancer: Elevated TGFB3 in late-stage tumors correlates with metastasis, though functional studies suggest it may inhibit tumorigenesis in breast and gastric tissues .

  • Cardiovascular Disease: Mutations in TGFB3 are linked to familial arrhythmogenic right ventricular dysplasia (ARVD1) .

  • Autoimmunity: Dual roles observed—TGFB3 deficiency exacerbates inflammation, while its overexpression drives Th17-mediated pathologies like EAE .

Product Specs

Introduction
The transforming growth factor beta (TGF-beta) family of cytokines plays a crucial role in embryonic development by mediating cell-to-cell interactions. In mammals, three TGF-beta isoforms (TGF-beta 1, TGF-beta 2, and TGF-beta 3) have been identified. These isoforms share a similar protein structure. Each is synthesized as a larger precursor protein that undergoes cleavage to release a 112-amino acid polypeptide. This active polypeptide remains associated with the remaining portion of the precursor molecule, forming a complex known as the latent TGF-beta complex.
Description
This product consists of the recombinant human TGF-beta 3 protein. It encompasses amino acids 644 to 850 of the full-length protein and has a total molecular weight of 50 kDa, including the GST tag.
Physical Appearance
The product is a clear solution that has been sterilized by filtration.
Formulation
This product is supplied as a 100 μl solution of purified human TGF-beta 3 protein at a concentration of 100 μg/ml. The buffer consists of 50 mM Tris-Acetate (pH 7.5), 1 mM EDTA, and 20% glycerol.
Stability
While the recombinant human TGF-beta 3 protein remains stable at 4°C for up to one week, it is recommended to store it at -20°C for optimal long-term stability. The addition of a carrier protein such as 0.1% HSA or BSA is advisable for extended storage. To maintain protein integrity, avoid repeated freeze-thaw cycles.
Applications
This product is suitable for various applications, including: (a) Enzyme-linked immunosorbent assay (ELISA), (b) Western blotting, (c) Inhibition assays.
Synonyms
Transforming Growth Factor-beta3, TGFB3, ARVD, FLJ16571, TGF-beta3.
Source
Escherichia Coli.

Q&A

What is the basic structure and function of human TGFB3?

TGFB3 (Transforming growth factor beta-3) is a protein encoded by the TGFB3 gene in humans. It belongs to the Transforming growth factor beta superfamily, which includes TGF-β family proteins, Bone morphogenetic proteins (BMPs), growth and differentiation factors (GDFs), inhibins and activins . The protein is produced in a complex with Latency-associated peptide (LAP) and becomes activated after cleavage by plasmin, matrix metalloproteases (MMPs), thrombospondin-1, and certain integrins . TGFB3 binds to TGF-beta RII, a type II serine/threonine kinase receptor . Functionally, it regulates cell proliferation, differentiation, motility, and controlled cell death (apoptosis), playing crucial roles in cell adhesion and extracellular matrix formation during development .

How does the signaling pathway of TGFB3 differ from other TGF-β isoforms?

While TGFB3 shares the canonical TGF-β pathway with other isoforms, its biological effects can be distinctly different. Unlike TGF-β1, which has been extensively studied in tumorigenesis, TGFB3 may actually play a protective role against tumor formation in tissues including skin, breast, oral and gastric mucosa . Research has demonstrated that TGF-β3 and TGF-β1 display clear isoform-specific biology, making extrapolation of data from one isoform to another inappropriate . When TGFB3 binds to its receptor, it triggers intracellular signaling through SMAD proteins, particularly pSMAD 2/3 and pSMAD 1/5/9, which can be detected within the first day of exposure to TGFB3 .

What is the molecular weight and sequence information for the active form of human TGFB3?

The active form of human TGFB3 recombinant protein has a molecular weight of approximately 12.7 kDa, with an apparent molecular mass of 14 kDa on SDS-PAGE . The sequence typically used in recombinant protein production spans Ala301-Ser412 with a Tyr340Phe substitution . This sequence information is critical for researchers designing experiments with recombinant TGFB3 proteins to ensure they are working with the correctly processed, biologically active form of the protein.

ParameterValue
Molecular Weight12.7 kDa
Apparent Molecular Weight14 kDa
SequenceAla301-Ser412(Tyr340Phe)
Purity> 95% (by reducing SDS-PAGE)

What are the optimal conditions for reconstitution and storage of recombinant TGFB3?

Recombinant human TGFB3 is typically provided as a lyophilized powder formulated from a 0.2 μm filtered solution of 4 mM HCl . For reconstitution, the protein can be dissolved in deionized water at a concentration of 1 μg/μL . Based on stability data, lyophilized proteins remain stable for up to 12 months when stored at -20 to -80°C . After reconstitution, the protein solution can be stored at 4-8°C for 2-7 days, but for longer-term storage, it is recommended to prepare aliquots to avoid repeated freeze-thaw cycles that could compromise protein activity .

How can researchers quantify TGFB3 activity in experimental systems?

The bioactivity of TGFB3 can be measured by its ability to inhibit the IL-4-dependent proliferation of TF-1 mouse T cells . The ED50 (effective dose for 50% response) for this effect typically ranges from 10-80 pg/ml . Researchers can also assess TGFB3 concentration using ELISA (Enzyme-Linked Immunosorbent Assay) methods, which have been successfully employed in studies tracking TGFB3 release from biomaterial scaffolds . For downstream signaling effects, activation of phosphorylated SMAD (pSMAD) 2/3 and pSMAD 1/5/9 can be evaluated through whole-mount immunohistochemistry approximately 24 hours after TGFB3 exposure .

What delivery systems are most effective for controlled release of TGFB3 in tissue engineering?

Nanofiber-based delivery systems have shown promise for modulating local TGFB3 concentration in tissue engineering applications. Research has demonstrated that electrospun PCL-PLGA (polycaprolactone-poly(lactic-co-glycolic acid)) nanofibers containing TGFB3 and BSA (bovine serum albumin) can provide controlled release profiles . The incorporation of 10% BSA significantly enhances TGFB3 release compared to 5% BSA formulations, resulting in approximately three-fold higher release over a 15-day period . These systems typically show an initial burst release followed by sustained delivery, making them suitable for applications requiring prolonged TGFB3 signaling.

Research data on TGFB3 release from nanofiber scaffolds has shown:

ParameterLow-dose TGF-β3 ScaffoldHigh-dose TGF-β3 Scaffold
TGF-β3 Content6.0 ± 0.3 ng12.7 ± 0.4 ng
Incorporation Efficiency46.0 ± 2.5%41.7 ± 2.5%
Fiber DiameterNo significant difference between groupsNo significant difference between groups

What developmental processes are critically dependent on TGFB3 signaling?

TGFB3 plays essential roles in multiple developmental processes. It is crucial for palate development, as it regulates molecules involved in cellular adhesion and extracellular matrix formation during palatal fusion . Without TGFB3, mammals develop cleft palate due to the failure of epithelial cells on both sides of the developing palate to fuse properly . Additionally, TGFB3 controls lung development by regulating cell adhesion and ECM formation in pulmonary tissues . It also directs wound healing processes by regulating the movements of epidermal and dermal cells in injured skin . The protein is particularly abundant in tissues that develop into skeletal muscles and plays a key role in their formation .

What genetic disorders are associated with TGFB3 mutations?

At least 11 mutations in the TGFB3 gene have been found to cause Loeys-Dietz syndrome type V, a connective tissue disorder characterized by blood vessel abnormalities, heart defects, and skeletal deformities . These mutations lead to the production of TGFB3 protein with little or no function, preventing it from binding to its receptors . Paradoxically, despite the inability of TGFB3 to bind to its receptors, TGF-β pathway signaling occurs at an even greater intensity than normal, possibly due to compensatory increases in the activity of other proteins in this signaling pathway . Understanding these pathological mechanisms is critical for researchers developing therapeutic strategies for TGFB3-related disorders.

What is the current understanding of TGFB3's role in cancer biology?

The role of TGFB3 in cancer appears to differ significantly from that of TGF-β1. While elevated levels of TGFB3 expression have been detected in late-stage tumors, suggesting a potential link to tumorigenesis, functional data supporting a causative role are lacking . In fact, studies indicate that TGFB3 may actually play a protective role against tumor formation in various tissues including skin, breast, oral and gastric mucosa . Researchers should be cautious about interpreting correlations between TGFB3 levels and cancer as causal relationships, as these observations might reflect TGFB3's normal biological roles in processes that are often dysregulated in cancer . Current evidence suggests that acute administration of low doses of exogenous TGFB3 is unlikely to influence tumor initiation or progression .

How can TGFB3 be utilized to direct stem cell differentiation in regenerative medicine?

TGFB3 has demonstrated significant potential for directing stem cell differentiation, particularly toward fibrochondrogenic lineages. Experimental data show that TGFB3 released from nanofiber scaffolds supports proteoglycan deposition by synovium-derived stem cells (SDSC) . A dose-dependent response has been observed, with consistently higher proteoglycan content detected on high-dose compared to low-dose TGFB3 scaffolds . Glycosaminoglycan (GAG) synthesis is significantly elevated in cells exposed to TGFB3 compared to controls, with effects detectable from day 7 onwards . For optimal results in regenerative medicine applications, researchers should consider both the concentration and delivery kinetics of TGFB3, as these parameters significantly impact cellular responses and differentiation outcomes.

What experimental approaches can distinguish between the effects of different TGF-β isoforms?

Given the demonstrated isoform-specific biology between TGF-β1 and TGF-β3, researchers need sophisticated approaches to distinguish their effects. These might include:

  • Isoform-specific neutralizing antibodies or receptor-blocking agents

  • Isoform-selective genetic knockdown using siRNA or CRISPR-Cas9 technologies

  • Parallel experiments with recombinant proteins of each isoform at equivalent concentrations

  • Analysis of downstream signaling pathways using phospho-specific antibodies against SMAD proteins

  • Transcriptomic or proteomic profiling following selective isoform stimulation

What are the challenges in translating TGFB3 research from preclinical models to clinical applications?

The translation of TGFB3 research to clinical applications faces several challenges. Human recombinant TGF-β3 (avotermin, planned trade name Juvista) successfully completed Phase I/II trials but failed in Phase III trials . This highlights the complexity of moving from controlled laboratory conditions to human subjects. Factors that may contribute to these translation challenges include:

  • Dose-dependent effects that may vary between preclinical models and humans

  • Context-dependent signaling that changes with tissue type and pathological state

  • Species-specific differences in TGFB3 signaling networks

  • Challenges in achieving targeted, controlled delivery in clinical settings

  • Potential immunogenicity of recombinant proteins in human subjects

Researchers must carefully consider these factors when designing studies aimed at clinical translation of TGFB3-based therapies.

How should researchers interpret conflicting results when studying TGFB3 in different experimental systems?

Conflicting results when studying TGFB3 across different experimental systems may stem from context-dependent effects. Researchers should systematically evaluate:

  • Concentration effects: TGFB3 often shows dose-dependent responses, with different concentrations potentially yielding opposite effects

  • Temporal considerations: Short-term versus long-term TGFB3 exposure may activate different signaling pathways

  • Cell type specificity: TGFB3 may have different effects in epithelial versus mesenchymal cells

  • Interaction with other signaling pathways: Cross-talk with other growth factors may modify TGFB3 responses

  • Experimental conditions: Differences in matrix composition, oxygen tension, or mechanical forces might influence TGFB3 signaling

When analyzing contradictory data, researchers should consider these variables and design experiments with appropriate controls to isolate specific effects.

What are common pitfalls in experimental design when studying TGFB3 signaling?

Common experimental pitfalls when studying TGFB3 signaling include:

  • Inadequate controls for TGFB3 activation: Remember that TGFB3 is produced in a latent complex and requires activation

  • Cross-reactivity issues: Ensure antibodies and detection methods distinguish between TGF-β isoforms

  • Timing considerations: TGFB3 effects may vary with exposure duration; pSMAD activation occurs within 24 hours

  • Storage and handling issues: Improper reconstitution or storage can lead to loss of bioactivity

  • Overlooking context-dependent signaling: Cellular responses to TGFB3 may vary dramatically depending on the cellular microenvironment

To avoid these pitfalls, researchers should include appropriate positive and negative controls, validate reagents for isoform specificity, and carefully document experimental conditions.

How can researchers optimize TGFB3 delivery systems for specific tissue engineering applications?

Optimization of TGFB3 delivery systems for tissue engineering requires careful consideration of several parameters:

  • Release kinetics: Adjust polymer composition to control initial burst release versus sustained delivery. PCL-PLGA systems with 10% BSA show enhanced release profiles compared to 5% BSA formulations

  • Dose optimization: Titrate TGFB3 loading to achieve the desired biological effect. Research shows that scaffolds containing approximately 12.7 ng TGFB3 (high-dose) induce stronger responses than those with 6.0 ng (low-dose)

  • Scaffold architecture: Fiber alignment and diameter can influence cell-material interactions and subsequent TGFB3 signaling

  • Incorporation efficiency: Account for the fact that typical incorporation efficiency ranges from 41.7% to 46.0% when designing delivery systems

  • Biological validation: Confirm bioactivity of released TGFB3 through functional assays such as SMAD phosphorylation analysis or target cell responses

These considerations will help researchers develop delivery systems that provide appropriate TGFB3 concentrations at the right time and location for specific tissue engineering applications.

Product Science Overview

Introduction

Transforming Growth Factor-Beta 3 (TGF-β3) is a member of the TGF-β superfamily, which includes TGF-β1 and TGF-β2. These cytokines play crucial roles in cellular processes such as proliferation, differentiation, and apoptosis. TGF-β3, in particular, is known for its involvement in embryogenesis, tissue regeneration, and immune regulation.

Structure and Composition

The human recombinant TGF-β3 is a 207 amino acid protein. It is typically produced in an E. coli expression system and purified to a high degree of purity, often exceeding 98% as determined by SDS-PAGE and HPLC analyses . The recombinant form is a homodimer, consisting of two identical 112 amino acid subunits linked by a single disulfide bond .

Biological Activity

TGF-β3 is highly conserved across species, with human and mouse TGF-β3 demonstrating 100% sequence homology . This conservation underscores its fundamental role in biological processes. The protein is secreted in a precursor form, which includes a bioactive C-terminal domain and an N-terminal latency-associated peptide (LAP). The mature protein functions as a disulfide-linked homodimer .

Functions and Applications

TGF-β3 exerts pleiotropic effects, including:

  • Suppression of B- and T-cell effector activity: This makes it a critical player in immune regulation.
  • Mediation of tissue healing: TGF-β3 is involved in wound healing and tissue regeneration.
  • Suppression of tumor proliferation: It has been shown to inhibit the growth of certain cancer cells .

Additionally, TGF-β3 promotes the expansion of CD4+CD25+ T-cells, which are essential for maintaining immune tolerance and preventing autoimmunity .

Research and Clinical Implications

The versatility of TGF-β3 extends to various research applications, including:

  • Stem cell differentiation: It is used to guide the differentiation of stem cells into specific cell types.
  • T-cell regulation and differentiation: TGF-β3 is crucial for studying immune responses and developing immunotherapies.
  • Functional assays: It is employed in assays such as ELISA, Western Blotting, and inhibition assays to explore its biological activities .

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.