TGFB2 Human

Transforming Growth Factor Beta 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of TGFB2 Human

Transforming Growth Factor Beta 2 (TGFB2 Human) is a cytokine belonging to the TGF-β superfamily, playing critical roles in embryonic development, tissue remodeling, immune regulation, and cellular processes such as proliferation, differentiation, and apoptosis . It is encoded by the TGFB2 gene and exists as a secreted, glycosylated protein that forms homodimers stabilized by disulfide bonds . Recombinant human TGFB2 is widely used in research to study its signaling mechanisms and therapeutic potential.

Molecular Structure and Isoforms

  • Primary Structure: Mature human TGFB2 is derived from a 414-amino-acid precursor, cleaved into a 112-amino-acid active peptide (monomer) and a latency-associated peptide (LAP) .

  • Quaternary Structure: Functions as a homodimer with a molecular mass of ~25–27 kDa .

  • Isoforms: Two splice variants exist, though functional differences remain under investigation .

Biological Functions and Signaling Pathways

TGFB2 signals through transmembrane serine/threonine kinase receptors (TGFBR1 and TGFBR2), activating SMAD proteins and non-canonical pathways (e.g., MAPK) . Key roles include:

  • Embryogenesis: Critical for cardiac, lung, and urogenital system development .

  • Immune Regulation: Suppresses T-cell activity and modulates macrophage differentiation .

  • Tissue Repair: Promotes extracellular matrix synthesis and smooth muscle cell differentiation .

  • Tumor Suppression/Aggression: Dual role in cancer—inhibits early-stage tumors but promotes metastasis in advanced stages .

Recombinant TGFB2 Production and Applications

Recombinant human TGFB2 is produced in systems like HEK293 cells or plants (Nicotiana benthamiana) and purified for research use .

5.1. Role in Disease Pathogenesis

  • Cancer:

    • High TGFB2 mRNA correlates with poor survival in lower-grade gliomas (HR = 4.07, p < 0.001) .

    • In triple-negative breast cancer (TNBC), TGFB2-AS1 lncRNA inhibits TGFB2-driven metastasis, improving patient survival .

  • Cardiovascular Disease: Reduced TGFB2 activity in type 2 diabetes impairs vascular repair, worsening atherosclerosis .

  • Gastric Cancer: Elevated TGFB2 expression and hypomethylation predict poor prognosis (HR = 2.02, p = 0.036) .

5.2. Therapeutic Targeting

  • Preclinical Models: Blocking TGFB2 in TNBC reduces orthotopic tumor growth and lung metastasis (p < 0.05) .

  • Biomarker Potential: TGFB2 levels in plaques correlate with smooth muscle cell differentiation, suggesting diagnostic utility .

Clinical and Industrial Relevance

  • Drug Development: TGFB2 inhibitors (e.g., monoclonal antibodies) are under investigation for fibrosis and cancer .

  • Cell Culture: Used to maintain stem cells or induce epithelial-mesenchymal transition in vitro .

Product Specs

Introduction
Transforming growth factor beta-2 (TGFB2) is a protein with a molecular weight of 27.08 kDa. It consists of two identical chains, each composed of 118 amino acids, joined by a disulfide bond. TGFB2 belongs to a family of five structurally related cytokines that exhibit diverse effects on both normal and cancerous cells. These homodimeric proteins play crucial roles as multifaceted regulators of cellular functions. The three mammalian isoforms of TGF-β (TGFβ1, TGFβ2, and TGFβ3) share a common signaling pathway through the same receptor and elicit comparable biological responses. They participate in essential physiological processes such as embryonic development, tissue remodeling, and wound healing.
Description
Recombinant human TGFB2, produced in plants, is a homodimeric protein comprising two identical polypeptide chains, each consisting of 118 amino acids. Its total molecular weight is 27.08 kDa. The TGFB2 protein has a 6xHis tag fused at its N-terminus and undergoes purification using proprietary chromatographic methods.
Physical Appearance
White powder, sterile and lyophilized (freeze-dried).
Formulation
Lyophilized from a solution at a concentration of 1 mg/ml in 50 mM Tris-HCl buffer at pH 7.4.
Solubility
To reconstitute the lyophilized TGFB2, it is advised to dissolve it in sterile 18 MΩ·cm H₂O at a minimum concentration of 1 µg per 40 µl. This solution can be further diluted as needed in other aqueous solutions.
Stability
Lyophilized TGFB2 remains stable at room temperature for up to 3 weeks. However, it is recommended to store it in a dry environment below -18°C. After reconstitution, human TGFB2 should be kept at 4°C for 2-7 days. For long-term storage, it is advisable to add a carrier protein (0.1% HSA or BSA) and store it below -18°C. Repeated freeze-thaw cycles should be avoided.
Purity
Determined by SDS-PAGE analysis, the purity is greater than 97.0%.
Biological Activity
The biological activity of TGFB2 is assessed in cell culture by evaluating its ability to suppress the growth of mink lung epithelial (Mv1Lu) cells. The ED50 is less than 40 ng/ml, which corresponds to a specific activity of 25,000 units/mg.
Synonyms
Transforming growth factor, beta 2, cetermin, Glioblastoma-derived T-cell suppressor factor, polyergin, G-TSF, TGF-beta2, TGF-beta-2, transforming growth factor beta-2, BSC-1 cell growth inhibitor, TGFB-2.
Source
Nicotiana benthamiana.
Amino Acid Sequence
HHHHHHALDAAYCFRNVQDNCCLRPLYIDFKRDLGWKWIH
EPKGYNANFCAGACPYLWSSDTQHSRVLSLYNTINPEASAS
PCCVSQDLEPLTI LYYIGKTPKIEQLSNMIVKSCKCS.

Q&A

What is the molecular structure of human TGF-β2?

Human TGF-β2 is encoded by a cDNA that produces a 414 amino acid (aa) precursor containing a 19 aa signal peptide and a 395 aa proprotein. A furin-like convertase processes this proprotein to generate an N-terminal 232 aa latency-associated peptide (LAP) and a C-terminal 112 aa mature TGF-β2. After secretion, disulfide-linked homodimers of LAP and TGF-β2 remain non-covalently associated, forming the small latent TGF-β complex. The mature human TGF-β2 protein shows 100% amino acid identity with porcine, canine, equine, and bovine TGF-β2, and 97% identity with mouse and rat TGF-β2 .

How does TGF-β2 signaling work in human cells?

TGF-β2 signaling begins with binding to a complex of the accessory receptor betaglycan (TGF-β RIII) and a type II serine/threonine kinase receptor called TGF-β RII. This receptor then phosphorylates and activates another serine/threonine kinase receptor, TGF-β RI (also called activin receptor-like kinase (ALK)-5), or alternatively, ALK-1. The entire complex phosphorylates and activates Smad proteins that regulate transcription. TGF-β2 also utilizes Smad-independent pathways, which explains the diverse cellular responses observed in different contexts .

How is TGF-β2 activated from its latent form?

TGF-β2 is activated from latency through multiple pathways that include:

  • Proteolytic processing by plasmin

  • Action of matrix metalloproteases

  • Interaction with thrombospondin 1

  • Binding to specific integrins

The activation process is critical for making TGF-β2 available to interact with its receptors. Covalent linkage of LAP to latent TGF-β binding proteins (LTBPs) creates a large latent complex that can interact with the extracellular matrix, providing spatial control of TGF-β2 activity .

What role does TGF-β2 play in liver fibrosis?

TGF-β2 has been identified as a significant contributor to biliary-derived liver diseases. Research using MDR2 knockout mice (a model for human cholestatic liver disease) has shown that TGF-β2 induces expression of fibrotic genes in cholangiocytes and hepatic stellate cells. In human studies, upregulated TGFB2 was found in liver tissue of patients with primary sclerosing cholangitis (PSC) and primary biliary cholangitis (PBC). In situ hybridization studies have demonstrated localization of TGFB2 near portal tracts and in areas of fibrogenic rearrangements in PSC and PBC patients, as well as in MDR2-KO mice .

How does TGF-β2 expression correlate with disease severity in cholestatic liver diseases?

TGFB2 expression shows significant correlation with inflammatory grade in patients with cholestatic liver diseases. In a cohort study of PBC patients, TGFB2 was remarkably upregulated in "high risk" patients who eventually needed liver transplantation compared to "low risk" patients who responded to UDCA treatment. High TGFB2 expression was also significantly associated with higher Scheuer grades (III and IV), suggesting that TGFB2 expression may have predictive value regarding patients' treatment response .

What is the cellular localization of TGF-β2 in healthy versus diseased liver tissue?

In situ hybridization studies have revealed that in both human patients with PSC/PBC and in MDR2-KO mice, TGFB2/TgfB2 is predominantly localized near portal tracts and in areas of fibrogenic rearrangements. This matches previous findings describing TGFB2 expression in cirrhosis of undefined origin predominantly in biliary epithelial cells. This specific localization pattern suggests a role in the pathogenesis of biliary-derived liver diseases .

What are the optimal in vitro models for studying TGF-β2 functions?

For in vitro studies of TGF-β2 function, several cell models have proven effective:

  • Cholangiocyte cell lines: Studies have used murine (603B) and human (MMNK1) cholangiocyte cell lines to study TGF-β2 effects on fibrogenic marker gene expression. Treatment of these cells with TGF-β2 results in upregulation of fibrogenic genes such as Acta2 (alpha smooth muscle actin gene), Col1A1 (collagen type I alpha 1), fibronectin, and PdgfrB (platelet-derived growth factor receptor beta) .

  • Hepatic stellate cells: These cells are key in liver fibrosis development and respond to TGF-β2 signaling.

  • Recombinant protein assays: Using recombinant human TGF-β2 protein in bioactivity assays has been reported to produce reproducible results with over 97% purity .

When designing experiments, it's important to consider the temporal aspects of TGF-β2 signaling, as some effects may be time-dependent. For example, in MMNK1 cells, TGFB1 was significantly induced after 6 hours of TGF-β2 treatment .

What animal models are appropriate for TGF-β2 research in liver diseases?

The MDR2 knockout (KO) mouse model has been established as an effective model for studying TGF-β2 in biliary-derived liver diseases. This model mimics human cholestatic liver diseases such as PSC and PBC. In untreated MDR2-KO mice, TgfB2 expression is markedly elevated compared to wild-type animals (approximately 8.7-fold). These mice develop progressive liver fibrosis and inflammatory responses similar to human biliary diseases .

Other models used in TGF-β2 research include:

  • Bile duct ligation (BDL) models

  • TgfB2 knockout mice to study developmental aspects (though these show multiple developmental defects)

  • Conditional knockout models for tissue-specific deletion

When using these models, researchers should consider age-dependent effects, as disease progression changes over time. For instance, studies on TGF-β2 silencing in MDR2-KO mice have been conducted on 14-week-old animals to target early-stage biliary liver disease .

What techniques are most effective for measuring TGF-β2 expression and activity?

Multiple complementary techniques should be employed to comprehensively assess TGF-β2 expression and activity:

  • qRT-PCR: For quantifying TGFB2 mRNA levels in tissue or cells

  • Western blot: For protein expression, though distinguishing between latent and active forms is important

  • In situ hybridization: For localizing TGFB2 expression in tissue sections

  • Immunohistochemistry: For protein localization in tissues

  • Bioactivity assays: Using recombinant TGF-β2 protein to assess functional effects

  • Reporter assays: For measuring TGF-β signaling pathway activation

  • Phospho-SMAD analysis: To directly measure pathway activation

When interpreting results, it's important to assess both the expression levels and the activation status of TGF-β2, as the protein exists in latent forms that must be activated to exert biological effects .

What are the current approaches for therapeutically targeting TGF-β2?

Several approaches have been developed for targeting TGF-β2 in research and potential therapeutic applications:

  • Antisense oligonucleotides (AONs): TgfB2-directed AONs have been used to blunt TgfB2 expression in MDR2-KO mice. This approach significantly reduced TgfB2 expression (12.5-fold reduction) without negative impact on liver parameters or body weight. AON treatment reduced TgfB2 levels in MDR2-KO mice to levels comparable to healthy controls .

  • Neutralizing antibodies: Though less specific for TGF-β2 alone, pan-TGF-β neutralizing antibodies have been used in models of Marfan syndrome to attenuate multisystem disease manifestations .

  • Receptor inhibitors: Targeting TGF-β receptors with small molecule inhibitors can block signaling from all TGF-β ligands.

  • Angiotensin receptor blockers: These have been shown to attenuate TGF-β signaling indirectly in some disease models .

The choice of approach depends on the disease context and the need for isoform-specific targeting versus broader TGF-β pathway inhibition.

What are the effects of TGF-β2 silencing in fibrotic liver diseases?

TGF-β2 silencing using antisense oligonucleotides in MDR2-KO mice has shown multiple beneficial effects on fibrogenesis:

  • Reduced fibrosis markers: AON treatment resulted in reduced collagen deposition, decreased hydroxyproline content, and diminished αSMA expression.

  • Increased anti-fibrotic factors: Induced PparG (peroxisome proliferator-activated receptor) expression was observed, reflecting a significant reduction of fibrogenesis.

  • Modulation of inflammatory response: Expression analyses revealed AON-specific regulatory effects on Ccl3, Ccl4, Ccl5, Mki67, and Notch3 expression.

  • Altered immune cell infiltration: AON treatment increased tissue infiltration by F4/80-positive cells including eosinophils, while decreasing the number of CD45-positive inflammatory cells.

These effects occurred without adverse effects on healthy livers, suggesting therapeutic potential .

How do mutations in TGFB2 contribute to human diseases?

Loss-of-function mutations in TGFB2 have been associated with syndromic presentations. While the search results don't provide comprehensive details on these syndromes, they do mention that despite the loss-of-function nature of these mutations, there may be paradoxical activation of TGF-β signaling cascades, particularly through ERK1/2 pathways .

This paradoxical situation highlights the complexity of TGF-β signaling and suggests that both loss and gain of function in this pathway can lead to disease, depending on the context. In mouse models of Marfan syndrome (MFS), antagonism of TGF-β signaling using either TGF-β neutralizing antibodies or angiotensin receptor blockers attenuates multisystem disease manifestations, including aortic aneurysm .

How can researchers address the paradoxical effects observed in TGF-β2 signaling?

The paradoxical effects of TGF-β2 signaling pose significant research challenges. Despite loss-of-function mutations in TGFB2, downstream signaling pathways may show activation rather than inhibition. To address these complexities:

  • Study context-specific effects: Examine TGF-β2 effects in different cell types and physiological states to understand context-dependent responses.

  • Investigate compensatory mechanisms: Explore how other TGF-β family members (TGF-β1, TGF-β3) may compensate for altered TGF-β2 function.

  • Analyze canonical vs. non-canonical pathways: Distinguish between Smad-dependent (canonical) and Smad-independent (non-canonical) signaling to understand divergent outcomes.

  • Develop conditional knockout models: Use tissue-specific and inducible knockout approaches to avoid developmental confounders and study adult physiology.

  • Employ systems biology approaches: Integrate transcriptomic, proteomic, and functional data to build comprehensive models of TGF-β2 signaling networks .

What are promising future directions for TGF-β2 research in human diseases?

Several promising research directions for TGF-β2 include:

  • Isoform-specific targeting: Developing more selective tools to target TGF-β2 without affecting TGF-β1 or TGF-β3 signaling.

  • Biomarker development: Using TGF-β2 expression levels as prognostic or predictive biomarkers in diseases like PBC, where high TGFB2 expression correlates with treatment response.

  • Cell-specific modulation: Creating approaches to target TGF-β2 in specific cell types (e.g., cholangiocytes in liver disease) while preserving beneficial functions in other tissues.

  • Combination therapies: Investigating how TGF-β2 targeting might synergize with other therapeutic approaches in fibrotic diseases.

  • Long-term safety assessment: Evaluating the long-term consequences of TGF-β2 inhibition, given its important roles in multiple physiological processes .

What are the best practices for reproducible TGF-β2 research?

To ensure reproducible TGF-β2 research:

  • Standardize protein sources: Use well-characterized recombinant TGF-β2 proteins with documented bioactivity. For example, recombinant human TGF-β2 protein with >97% purity has been cited in over 88 publications with reproducible results in bioactivity assays .

  • Validate antibody specificity: Ensure antibodies can distinguish between TGF-β2 and other TGF-β isoforms.

  • Consider activation status: Always account for the latent versus active forms of TGF-β2 when measuring protein levels or activity.

  • Use multiple detection methods: Combine mRNA quantification, protein detection, and functional assays to comprehensively assess TGF-β2 biology.

  • Document experimental conditions: Factors such as cell density, passage number, and serum conditions can significantly affect TGF-β2 responses.

  • Include appropriate controls: Use both negative controls and positive controls (such as known TGF-β2 responsive genes) to validate experimental systems .

Product Science Overview

Introduction

Transforming Growth Factor Beta 2 (TGF-β2) is a member of the Transforming Growth Factor Beta (TGF-β) family, which is a superfamily of growth factors involved in regulating various biological processes such as cell growth, proliferation, differentiation, and apoptosis . TGF-β2 is a secreted protein that plays a crucial role in immune homeostasis, balancing lymphocyte proliferation, apoptosis, hematopoiesis, and embryogenesis .

Biological Functions

TGF-β2 is known for its diverse biological functions. It acts as a strong growth inhibitor for normal and transformed epithelial, lymphoid, fibroblast, and keratinocyte cells . Additionally, TGF-β2 inhibits antigen-induced T cell proliferation, epithelial cell proliferation, and induces mesenchymal cell proliferation . It also has cell-specific effects on apoptosis, making it a key player in maintaining cellular homeostasis .

Expression and Activation

TGF-β2 is produced by various cell types, including eosinophils, astrocytes, glioblastoma, and other cancer-derived cell types . The TGF-β signaling cascade is initiated by the binding of mature ligands to corresponding receptors on the cell surface, forming a complex that activates Smad or non-Smad molecules to transduce signals into the nucleus and regulate the expression of target genes .

Recombinant Human TGF-β2

Recombinant Human TGF-β2 is produced using recombinant DNA technology, which involves inserting the gene encoding TGF-β2 into a suitable expression system, such as a mouse myeloma cell line . The recombinant protein is then purified to achieve high purity levels, typically greater than 97% . Recombinant TGF-β2 is used in various research applications, including studying its effects on cell proliferation, differentiation, and apoptosis.

Applications in Research

Recombinant Human TGF-β2 is widely used in research to study its role in various biological processes and diseases. It is particularly important in the development of cardiac, lung, craniofacial, limb, eye, ear, and urogenital systems . Additionally, dysregulation of TGF-β2 signaling has been associated with tumorigenesis, eye diseases, cardiovascular diseases, immune disorders, and motor system 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.