TGFB1 Mouse

Transforming Growth Factor-Beta 1 Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

TGFB1 Mouse Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 135 amino acids (279-390 a.a) and having a molecular mass of 15.2kDa.
TGFB1 is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction

Transforming growth factor betas (TGF-betas) are signaling molecules that play a crucial role in regulating cell growth and differentiation during embryonic development. Three main types of TGF-betas (TGF-beta1, TGF-beta2, and TGF-beta3) have been identified in mammals. These proteins are initially produced in a precursor form and undergo enzymatic cleavage to release a smaller active protein of 112 amino acids. The active TGF-beta remains associated with the remaining portion of the precursor molecule, forming a complex.

Description

Recombinant Mouse TGFB1, produced in E. coli, is a single polypeptide chain without any glycosylation modifications. It consists of 135 amino acids (corresponding to amino acid positions 279-390 of the native protein) and has a molecular weight of 15.2 kDa. For purification and detection purposes, a 23 amino acid Histidine tag is attached to the N-terminus of the TGFB1 protein. Purification is carried out using proprietary chromatographic methods.

Physical Appearance

The product is a clear, colorless solution that has been sterilized by filtration.

Formulation

The TGFB1 protein is supplied in a solution at a concentration of 0.25 mg/ml. The solution contains 20mM Tris-HCl buffer (pH 8.0) and 10% glycerol.

Stability

For short-term storage (up to 2-4 weeks), keep the product refrigerated at 4°C. For extended storage, freeze the product at -20°C. To ensure long-term stability during frozen storage, it is recommended to add a carrier protein such as albumin (0.1% HSA or BSA). Repeated freezing and thawing of the product should be avoided to maintain its integrity and activity.

Purity

The purity of the TGFB1 protein is determined to be greater than 85.0% using SDS-PAGE analysis.

Synonyms
Transforming growth factor beta-1, TGF-beta-1, Tgfb, Tgfb-1, TGFbeta1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSALDTNYC FSSTEKNCCV RQLYIDFRKD LGWKWIHEPK GYHANFCLGP CPYIWSLDTQ YSKVLALYNQ HNPGASASPC CVPQALEPLP IVYYVGRKPK VEQLSNMIVR SCKCS.

Q&A

What are the normal expression patterns of TGF-β isoforms across different mouse tissues?

TGF-β isoform expression varies significantly between tissues in mice. Research shows that tissue-specific differences are generally larger than strain-specific differences. Most tissues contain measurable levels of all three TGF-β isoforms (TGF-β1, TGF-β2, and TGF-β3), though in varying proportions.

When analyzing TGF-β distribution, researchers should note:

  • The mammary gland uniquely exhibits high levels of TGF-β3 protein

  • Sex-hormone responsive tissues (mammary gland, uterus, ovary) demonstrate similar TGF-β1 and TGF-β2 levels

  • Blood cells, particularly platelets, contain 40-100× more TGF-β1 than most cells, which can affect tissue measurements

Methodologically, researchers should consider whether tissue perfusion is necessary for their specific study. While perfusion significantly reduces measured TGF-β1 in highly vascularized organs like liver by removing blood-derived TGF-β1, it has minimal effect on tissues like spleen, lung, and mammary tumors .

How do TGF-β1 knockout mice differ phenotypically from wild-type mice?

TGF-β1 knockout (Tgfb1^-/-) mice develop a severe, lethal inflammatory disease that is T-cell dependent. This pathology is not caused by pathogens but represents a fundamental autoimmune process. Key phenotypic characteristics include:

  • Multifocal inflammatory disease affecting multiple organ systems

  • Early mortality (typically around 3 weeks of age) in conventional knockouts

  • Thymic developmental abnormalities with progressive cortical depletion as inflammation progresses

  • Reduction in CD4+CD8+ thymocytes and relative increase in CD4+CD8- thymocytes

Importantly, the severity of this phenotype may vary depending on genetic background. The autoimmune pathology in TGF-β1-deficient mice requires self-antigen recognition, as demonstrated by experiments with TCR transgenic Tgfb1^-/- mice. When the T cell repertoire is restricted to recognize only a non-present foreign antigen (using the OVA-specific TCR transgene DO11.10 on a Rag1^-/- background), the inflammatory disease does not develop .

What methodological approaches are recommended for quantifying TGF-β proteins in mouse tissues?

Accurate quantification of TGF-β isoforms in mouse tissues requires careful consideration of several factors:

  • Sample preparation: Consider whether perfusion is necessary to eliminate blood contamination, particularly for highly vascularized tissues with low intrinsic TGF-β1. For most tissues, the decision depends on experimental goals, as perfusion is impractical in many settings .

  • Isoform-specific detection: Use isoform-specific antibodies or assays, as the three TGF-β isoforms have distinct expression patterns and functions. Commercial ELISA kits typically show minimal cross-reactivity between isoforms.

  • Latent versus active TGF-β: Most TGF-β exists in a latent form requiring activation. Researchers should clearly specify whether they are measuring total TGF-β (after acid activation) or only the active form.

  • Tissue processing protocols: For immunohistochemistry, in situ hybridization provides valuable spatial information about expression but may not directly correlate with protein levels due to post-transcriptional regulation and diffusion of secreted proteins .

  • Complementary approaches: Combining protein quantification (ELISA) with mRNA analysis (RT-qPCR) and in situ techniques provides a more complete picture of TGF-β biology.

How do strain-specific differences in TGF-β expression influence experimental outcomes?

While tissue-specific differences in TGF-β levels tend to be greater than strain-specific variations, important inter-strain differences exist that can affect experimental results:

Different mouse strains show distinct TGF-β expression profiles that may influence their susceptibility to certain pathologies:

These differences may explain strain-specific susceptibilities to various conditions. For example, strain-dependent variability in fibrosis development may reflect these intrinsic differences in TGF-β levels .

Methodological implications: Researchers should:

  • Choose mouse strains consistently within a study

  • Report strain information clearly in publications

  • Consider potential strain differences when comparing results across studies

  • Include appropriate strain-matched controls in experiments involving mutant mice on different genetic backgrounds

What is the mechanistic basis for T cell hyperactivation in TGF-β1-deficient mice?

The hyperactivation of T cells in TGF-β1-deficient mice results from a lowered threshold of activation and requires recognition of self-antigens. This process involves several mechanisms:

  • Self-antigen recognition requirement: Experiments using TCR-transgenic mice expressing the OVA-specific TCR (DO11.10) on a Tgfb1^-/- Rag1^-/- background demonstrate that T cell activation and inflammatory disease are eliminated when T cells cannot recognize self-antigens .

  • Calcium signaling alterations: Tgfb1^-/- T cells show increased intracellular calcium levels, contributing to their lower activation threshold. Unlike TGF-β1 function in T regulatory cells (which is SMAD3-dependent), this calcium/calcineurin-mediated function in conventional T cells appears to be SMAD3-independent .

  • Experimental demonstration: The critical role of self-antigen recognition can be demonstrated by comparing:

    • Tgfb1^-/- mice (severe inflammation, activated T cells)

    • Tgfb1^-/- DO11.10 Rag1^+/+ mice (milder inflammation, less activated T cells)

    • Tgfb1^-/- DO11.10 Rag1^-/- mice (no inflammation, no T cell activation)

The difference between the second and third groups is particularly informative. In Tgfb1^-/- DO11.10 Rag1^+/+ mice, some T cells express hybrid TCRs (transgenic TCRβ with endogenous TCRα), allowing self-antigen recognition and activation. In contrast, Tgfb1^-/- DO11.10 Rag1^-/- mice have T cells that exclusively express the transgenic TCR that cannot recognize self-antigens, preventing activation and disease .

How do TGF-β isoforms change in mammary tumors compared to normal tissue?

Research demonstrates reciprocal changes in TGF-β isoform expression during mammary tumorigenesis:

TGF-β1:

  • Protein levels are significantly increased in mammary tumors (3-8 fold higher than normal mammary tissue)

  • Increase varies by tumor model: ~3× higher in 4T1 and F311 tumors, ~8× higher in MVT1 tumors

  • mRNA expression is elevated in tumor cells compared to normal ductal epithelium

TGF-β2:

  • Increased in 4T1 and F3II tumors

  • Unchanged in MVT1 tumors

  • Varying mRNA expression patterns by tumor model

TGF-β3:

  • Consistently 2-3 fold lower in mammary tumors compared to normal mammary gland

  • Reduced mRNA expression in tumor cells while remaining high in normal mammary ducts

This reciprocal regulation of TGF-β1 (increased) and TGF-β3 (decreased) in mammary tumors suggests distinct roles for these isoforms in cancer progression. Methodologically, these differences can be demonstrated through:

  • Protein quantification (ELISA) of tissue extracts

  • In situ hybridization for mRNA localization

  • Immunohistochemistry (though protein diffusion may obscure cellular patterns)

What considerations are important when using humanized TGFB1 mouse models?

Humanized TGFB1 mouse models (B-hTGFB1) represent an important tool for translational research. Key characteristics and considerations include:

Model design and validation:

  • The human TGFB1 gene encoding the full-length protein is inserted following part of exon 2 of the mouse Tgfb1 gene

  • Expression analysis shows similar mRNA levels between human TGFB1 in the model and mouse Tgfb1 in wild-type mice

  • Flow cytometry confirms species-specific expression: human TGFB1 is exclusively detected in homozygous B-hTGFB1 mice, while mouse TGFB1 is detected in wild-type mice

Immune system integrity:

  • Analysis of spleen leukocyte subpopulations shows comparable percentages of T cells, B cells, NK cells, dendritic cells, granulocytes, monocytes, and macrophages between B-hTGFB1 mice and wild-type C57BL/6 mice

  • This indicates that substituting human TGFB1 for mouse Tgfb1 does not disrupt normal immune development and differentiation

Research applications:

  • These models enable more accurate evaluation of human-specific TGFB1-targeting therapeutics

  • They can be particularly valuable for testing antibodies or other biologics that may have species-specific binding properties

  • They provide a platform for studying human TGFB1 biology in an in vivo context

What are the species differences in microglial responses to TGF-β1 between rats and mice?

Understanding species differences in TGF-β1 responses is crucial for translational research. While the search results provide limited information on this specific comparison, they indicate that:

  • TGF-β1 is upregulated after central nervous system injuries and diseases involving microglial activation

  • The TGF-β1 receptor is highly expressed on microglia/macrophages within hematomas after intracerebral hemorrhage in rat striatum

  • Species similarities and differences exist in microglial responses to various stimuli, including pro-inflammatory (IFN-γ+TNF-α) and anti-inflammatory (IL-4) signals

For TGF-β1 specifically, both rat and mouse microglia can produce and respond to this cytokine, but they may exhibit differences in:

  • Expression of TGF-β receptors (e.g., Tgfbr2)

  • Downstream signaling pathways

  • Expression of inflammatory mediators, immune receptors and modulators in response to TGF-β1

Methodologically, researchers should:

  • Avoid generalizing findings between species without direct comparison

  • Consider using both rat and mouse models for preclinical studies of neuroinflammation

  • Quantify changes in multiple parameters, including gene expression, morphology, and functional responses when comparing species differences

What are the optimal methods for investigating TGF-β1's role in T cell regulation?

Investigating TGF-β1's role in T cell regulation requires multiple complementary approaches:

Genetic models:

  • Complete TGF-β1 knockout mice (Tgfb1^-/-) to study systemic loss of TGF-β1

  • Conditional knockout models (Tgfb1^fl/fl^ with cell-specific Cre) to examine cell-specific TGF-β1 requirements

  • TCR transgenic models on TGF-β1-deficient backgrounds to study antigen-specific responses

Experimental design strategies:

  • Use of TCR transgenic mice (e.g., DO11.10) crossed to Tgfb1^-/- mice to restrict the T cell repertoire

  • Further crossing to Rag1^-/- background to eliminate endogenous TCR rearrangement

  • Flow cytometric analysis of activation markers (CD44, CD62L, CD11a, CD69, CD25) to assess T cell activation status

  • Comparison of different genetic backgrounds to determine the requirement for self-antigen recognition

Analytical considerations:

  • Distinguish between thymic development effects and peripheral T cell activation

  • Assess both phenotypic (surface markers) and functional (cytokine production, proliferation) T cell parameters

  • Consider the influence of inflammatory environment on observed phenotypes

What strategies can researchers use to distinguish the functions of different TGF-β isoforms?

Distinguishing the functions of different TGF-β isoforms presents challenges due to their structural similarities but is essential given their distinct biological roles. Effective strategies include:

Genetic approaches:

  • Isoform-specific knockout mice (Tgfb1^-/-, Tgfb2^-/-, Tgfb3^-/-) display distinct phenotypes:

    • TGF-β1 null: autoimmune-like inflammatory disease or embryonic lethality due to angiogenic defects

    • TGF-β2 null: developmental defects in multiple organs, death before birth

    • TGF-β3 null: death immediately after birth due to cleft palate and inability to nurse

  • Knockin experiments: replacing one isoform with another while maintaining original regulatory elements (e.g., TGF-β3 coding sequence knocked into TGF-β1 locus with retention of TGF-β1 LAP)

Analytical methods:

  • Isoform-specific antibodies for immunohistochemistry and ELISA

  • In situ hybridization for spatial localization of isoform-specific mRNA expression

  • Comparing expression patterns across tissues and disease states (e.g., reciprocal regulation in tumors)

Experimental design considerations:

  • Comprehensive profiling of all three isoforms simultaneously in experimental models

  • Analysis of multiple tissues to capture tissue-specific differences

  • Inclusion of different mouse strains to account for strain-specific variations

  • Integration of protein and mRNA data to account for post-transcriptional regulation

Through these approaches, researchers can better understand the unique and overlapping functions of TGF-β isoforms in normal physiology and disease.

Table: Comparison of TGF-β Isoform Expression in Mouse Mammary Tumors vs. Normal Mammary Gland

TGF-β IsoformNormal Mammary Gland4T1 TumorsF311 TumorsMVT1 TumorsKey Observations
TGF-β1Baseline levels~3× higher~3× higher~8× higherConsistently upregulated in all tumor models
TGF-β2Baseline levelsIncreasedIncreasedUnchangedVariable response depending on tumor model
TGF-β3High expression~2-3× lower~2-3× lower~2-3× lowerConsistently downregulated in all tumor models

Data derived from protein quantification studies in mouse mammary tumor models compared to strain-matched normal mammary tissue

What are promising research areas for further understanding TGF-β1 function in mouse models?

Several research directions hold promise for advancing our understanding of TGF-β1 biology:

  • Isoform-specific functions in disease models: Further investigation of the reciprocal regulation of TGF-β1 (increased) and TGF-β3 (decreased) in cancer and other diseases.

  • Cell-specific TGF-β1 signaling: Development of more sophisticated conditional knockout and reporter models to study TGF-β1 production and response in specific cell types.

  • Strain-specific differences: Expanded analysis of strain differences in TGF-β biology to better understand genetic influences on TGF-β-mediated pathologies.

  • Translational applications: Continued refinement of humanized TGF-β1 mouse models to improve the predictive value of preclinical studies for human therapeutics.

  • Integration with other signaling pathways: Investigation of how TGF-β1 interacts with other regulatory systems to maintain immune homeostasis and tissue integrity.

Product Science Overview

Structure and Synthesis

TGF-β1 is a homodimeric protein, meaning it consists of two identical subunits linked by disulfide bonds. The precursor form of TGF-β1 contains 390 amino acids, including an N-terminal signal peptide, a latency-associated peptide (LAP), and a C-terminal region that becomes the active TGF-β1 upon activation . The mature form of TGF-β1 is produced through proteolytic cleavage, which removes the LAP and releases the active cytokine.

Biological Functions

TGF-β1 is involved in a wide range of biological processes:

  1. Cell Proliferation and Differentiation: TGF-β1 can either promote or inhibit cell proliferation depending on the cell type and context. It plays a critical role in the differentiation of various cell types, including immune cells, fibroblasts, and epithelial cells .

  2. Angiogenesis: TGF-β1 promotes the formation of new blood vessels, which is essential for tissue repair and regeneration .

  3. Immune Regulation: TGF-β1 has both immunosuppressive and immunostimulatory effects. It can inhibit the proliferation of T cells and the production of pro-inflammatory cytokines, while also promoting the differentiation of regulatory T cells (Tregs) and Th17 cells .

Mechanism of Action

TGF-β1 exerts its effects by binding to specific cell surface receptors, known as TGF-β receptors. The binding of TGF-β1 to the type II receptor (TβRII) leads to the recruitment and phosphorylation of the type I receptor (TβRI). This receptor complex then activates intracellular signaling pathways, including the Smad pathway, which regulates the expression of target genes involved in various cellular processes .

Applications in Research

Recombinant mouse TGF-β1 is extensively used in research to study its role in various biological processes and diseases. Some key applications include:

  1. Cancer Research: TGF-β1 is known to play a dual role in cancer, acting as a tumor suppressor in early stages and as a promoter of tumor progression in later stages. Researchers use recombinant TGF-β1 to investigate its effects on cancer cell proliferation, invasion, and metastasis .

  2. Immunology: TGF-β1 is a critical regulator of immune responses. It is used to study its effects on T cell differentiation, immune tolerance, and autoimmune diseases .

  3. Tissue Engineering and Regenerative Medicine: Due to its role in cell proliferation and differentiation, TGF-β1 is used in tissue engineering to promote the regeneration of damaged tissues and organs .

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.