TGFBR1 Human, Active

Transforming Growth Factor Beta Receptor 1 Human Recombinant, Active
Shipped with Ice Packs
In Stock

Description

Table 1: Key Molecular Features of TGFBR1

FeatureDetails
Gene LocationChromosome 9q22.33
Protein Length507 amino acids
Molecular Weight~53 kDa
Key DomainsExtracellular ligand-binding, GS domain, kinase domain
IsoformsALK5 (primary isoform), TGFBR1*6A (oncogenic variant)
Active FormRecombinant human TGFBR1 expressed in HEK 293 or Freestyle 293-F cells

Functional Mechanisms

TGFBR1 operates through a canonical SMAD-dependent pathway and non-canonical signaling routes:

  1. Ligand Binding: TGF-β ligands (e.g., TGF-β1) bind TGFBR2, which recruits and phosphorylates TGFBR1 at the GS domain .

  2. SMAD Activation: Phosphorylated TGFBR1 activates SMAD2/3, which complex with SMAD4 to regulate gene transcription .

  3. Non-Canonical Pathways: TGFBR1 interacts with MAPK, ERK, and Rho-GTPase pathways to influence cell migration and differentiation .

Key regulatory mechanisms include:

  • FKBP12 Interaction: Prevents ligand-independent activation of TGFBR1 .

  • Receptor Endocytosis: Modulates signal duration and specificity .

Developmental and Physiological Roles

  • Embryogenesis: TGFBR1 knockout in mice causes lethal cardiac, craniofacial, and immune defects .

  • Tissue Homeostasis: Regulates extracellular matrix (ECM) remodeling via interactions with fibrillin-1 and latent TGF-β binding proteins (LTBPs) .

Table 2: TGFBR1 Dysregulation in Human Diseases

ConditionMechanismOutcome
Loeys–Dietz SyndromeTGFBR1 mutations reduce SMAD signalingAortic aneurysms, skeletal defects
Breast CancerTGFBR16A variant enhances ERK/Rho-GTP signalingIncreased metastasis risk (OR 1.15)
Ovarian TumorsConstitutively active TGFBR1 drives granulosa cell tumorigenesis in miceElevated inhibin, estradiol levels
FibrosisOveractivation promotes ECM depositionRenal fibrosis, diabetes

Table 3: TGFBR1-Targeted Inhibitors

InhibitorMechanismClinical Application
Galunisertib (LY-2157299)Blocks kinase activitySolid tumors, fibrosis
SB-431542Competitive ATP antagonistExperimental fibrosis models
RepSoxReduces SMAD2/3 phosphorylationStem cell differentiation

Therapeutic strategies include:

  • Antagonists: Counteract TGFBR1 overexpression in cancers and fibrotic diseases .

  • Gene Therapy: Silencing TGFBR16A in breast cancer models reduces tumor aggressiveness .

Active Recombinant TGFBR1 in Research

Recombinant human TGFBR1 (e.g., ab50036, ab271757) is produced in HEK 293 cells with >98% purity and <1 EU/µg endotoxin . Applications include:

  • Signal Transduction Studies: Mapping SMAD-dependent vs. non-canonical pathways .

  • Drug Screening: Evaluating inhibitor efficacy in vitro .

  • Disease Modeling: Generating constitutively active TGFBR1 mutants to study tumorigenesis .

Product Specs

Description
TGFBR1, a protein produced in Sf9 insect cells, is a single, glycosylated polypeptide chain with a molecular weight of 38kDa. It consists of 342 amino acids, spanning from position 27 to 126a.a. This protein is expressed with a 242 amino acid hIgG-His-Tag at the C-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
A colorless solution that has been sterilized by filtration.
Formulation
TGFBR1 protein solution at a concentration of 0.5mg/ml in Phosphate Buffered Saline (pH 7.4) containing 10% glycerol.
Stability
For short-term storage (2-4 weeks), the entire vial can be stored at 4°C. For longer storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing should be avoided.
Purity
SDS-PAGE analysis indicates a purity greater than 90.0%.
Biological Activity
The binding ability, as determined by a functional ELISA with Mouse CD105, is ≤ 2 ug/ml.
Synonyms

TGFBR1, AAT5, ACVRLK4, ALK-5, ALK5, ESS1, LDS1, LDS1A, LDS2A, MSSE, SKR4, tbetaR-I, TGFR-1.

Source

Sf9, Baculovirus cells.

Amino Acid Sequence

ADLLLPGATA LQCFCHLCTK DNFTCVTDGL CFVSVTETTD KVIHNSMCIA EIDLIPRDRP FVCAPSSKTG SVTTTYCCNQ DHCNKIELPT TVKSSPGLGP VELVEPKSCD KTHTCPPCPA PELLGGPSVF LFPPKPKDTL MISRTPEVTC VVVDVSHEDP EVKFNWYVDG VEVHNAKTKP REEQYNSTYR VVSVLTVLHQ DWLNGKEYKC KVSNKALPAP IEKTISKAKG QPREPQVYTL
PPSRDELTKN QVSLTCLVKG FYPSDIAVEW ESNGQPENNY KTTPPVLDSD GSFFLYSKLT VDKSRWQQGN VFSCSVMHEA LHNHYTQKSL SLSPGKHHHH HH.

Q&A

What is TGFBR1 and what is its fundamental role in cellular signaling?

TGFBR1 (Transforming Growth Factor Beta Receptor 1) is a protein-coding gene that encodes a serine/threonine protein kinase receptor. It forms a heteromeric complex with type II TGF-beta receptors when bound to TGF-beta ligands, transducing signals from the cell surface to the cytoplasm . The formation of this receptor complex, composed of 2 TGFBR1 and 2 TGFBR2 molecules symmetrically bound to the cytokine dimer, results in phosphorylation and activation of TGFBR1 by the constitutively active TGFBR2 .

Once activated, TGFBR1 phosphorylates downstream effectors, particularly SMAD2, which then dissociates from the receptor and interacts with SMAD4. This SMAD2-SMAD4 complex translocates to the nucleus where it modulates transcription of TGF-beta-regulated genes, constituting the canonical SMAD-dependent TGF-beta signaling cascade . Beyond this canonical pathway, TGFBR1 also participates in non-canonical, SMAD-independent signaling pathways, highlighting its versatility in cellular communication networks.

How do structural features of TGFBR1 relate to its activation mechanism?

The activation of TGFBR1 follows a precise structural mechanism. When TGF-β ligands bind to TGFBR2, this binding induces the formation of a heteromeric complex with TGFBR1 . A critical structural feature of TGFBR1 is its Gly-Ser (GS) domain, which serves as the phosphorylation target for TGFBR2 .

In the signaling process:

  • TGF-β ligands first bind to TGFBR2

  • TGFBR1 is recruited to form the receptor complex

  • TGFBR2 phosphorylates the GS domain of TGFBR1

  • Activated TGFBR1 subsequently phosphorylates SMAD2/3

  • Phosphorylated SMAD2/3 associates with SMAD4 and translocates to the nucleus

The proper structural conformation of TGFBR1, particularly the accessibility of its GS domain, is essential for its activation. Mutations affecting this domain can lead to constitutive activation or inactivation of the receptor, resulting in various pathological conditions including Loeys-Dietz syndrome and Multiple Self-Healing Squamous Epithelioma .

What genetic models have proven most effective for studying TGFBR1 function?

Several genetic models have been developed to study TGFBR1 function, with conditional knockout (cKO) and constitutively active models being particularly informative. Since Tgfbr1 null mice die embryonically, conditional gene inactivation strategies using the Cre-loxP system have become essential for studying its post-developmental functions .

The most effective models include:

  • Conditional knockout models: Using tissue-specific Cre expression (e.g., Amhr2-Cre for female reproductive tract) to delete Tgfbr1 in specific tissues . These models have revealed critical roles for TGFBR1 in maintaining structural integrity of the female reproductive tract.

  • Constitutively active TGFBR1 models: These gain-of-function models express a constitutively active form of TGFBR1 in specific tissues . Remarkably, expression of constitutively active TGFBR1 in ovarian somatic cells leads to the development of sex cord-stromal tumors with complete penetrance .

  • Reporter models: The Tgfbr1-bgal allele, where a β-galactosidase reporter is inserted into the Tgfbr1 locus, allows monitoring of Tgfbr1 expression patterns while creating a null allele .

These complementary approaches have significantly advanced our understanding of TGFBR1 function in various physiological and pathological contexts, particularly in reproductive biology and tumor development.

How should researchers design experiments to distinguish between canonical and non-canonical TGFBR1 signaling pathways?

Distinguishing between canonical (SMAD-dependent) and non-canonical (SMAD-independent) TGFBR1 signaling requires careful experimental design focused on specific pathway components and outputs.

Methodological approaches:

  • Phosphorylation analysis: Monitor phosphorylation of SMAD2/3 (canonical pathway) versus non-SMAD proteins such as TRAF6, MAP3K7, and PARD6A (non-canonical pathways) . Western blotting with phospho-specific antibodies allows quantitative assessment of each pathway's activation.

  • Genetic manipulation strategies:

    • Use SMAD2/3 knockdown/knockout in conjunction with TGFBR1 activation to determine SMAD-dependency

    • Employ TGFBR1 mutants that selectively activate canonical or non-canonical pathways

    • Utilize pathway-specific inhibitors alongside TGFBR1 activation

  • Transcriptional readouts:

    • Employ SMAD-responsive luciferase reporters for canonical signaling

    • Use pathway-specific gene expression profiles to distinguish between signaling modes

  • Time-course experiments: Canonical and non-canonical pathways often exhibit different activation kinetics, with non-canonical responses sometimes occurring more rapidly.

For robust interpretation, researchers should implement multiple complementary approaches and include appropriate controls, such as TGFBR1 kinase inhibitors and ligand-blocking antibodies, to confirm pathway specificity. Considering that TGFBR1 can induce TRAF6 autoubiquitination leading to MAP3K7 activation and apoptosis, or regulate epithelial-to-mesenchymal transition through PARD6A phosphorylation , experimental designs must account for these diverse signaling outcomes.

What are the molecular mechanisms by which TGFBR1 maintains female reproductive tract integrity?

TGFBR1 plays a crucial role in maintaining female reproductive tract integrity through multiple molecular mechanisms, as revealed by studies using conditional knockout mouse models .

Conditional deletion of Tgfbr1 in the female reproductive tract using Amhr2-Cre leads to striking phenotypes including the development of oviductal diverticula and defective uterine smooth muscle development . These structural abnormalities prevent embryo development and transit to the uterus, resulting in complete sterility.

At the molecular level, TGFBR1 maintains reproductive tract integrity through:

  • Regulation of cell differentiation genes: Tgfbr1 cKO mice show dysregulation of critical cell differentiation genes including Krt12, Ace2, and MyoR that are potentially associated with female reproductive tract development .

  • Smooth muscle development: TGFBR1 signaling is essential for proper development of smooth muscle layers in the uterus. Defective smooth muscle development is a prominent feature in the uteri of Tgfbr1 cKO mice .

  • Functional similarity with TGFBR2: Both TGFBR1 and TGFBR2 appear to play similar roles in maintaining structural integrity of the female reproductive tract. Conditional deletion of either receptor leads to comparable phenotypes, suggesting they function in the same pathway to maintain reproductive tract integrity .

These findings highlight how TGFBR1-mediated signaling controls essential developmental and maintenance processes in the female reproductive system through regulation of specific genetic programs governing cellular differentiation and tissue architecture.

How does TGFBR1 signaling interact with oocyte-derived factors in regulating ovarian function?

The interaction between TGFBR1 signaling and oocyte-derived factors presents an intriguing area of reproductive biology research. In vitro studies have suggested that TGFBR1 can mediate signaling of growth differentiation factor 9 (GDF9), an oocyte-secreted protein required for early ovarian folliculogenesis, cumulus cell functions, and oocyte developmental competence .

Additional insights into TGFBR1 and oocyte factor interactions include:

  • Regulation of TGFBR1 expression: Recombinant BMP15 or GDF9 can suppress Tgfbr1 mRNA expression in mouse granulosa cells after 5-hour treatment, suggesting a regulatory feedback mechanism between oocyte factors and receptor expression .

  • Cumulus expansion: Despite its role in other reproductive tissues, Tgfbr1 cKO mice show normal cumulus expansion, indicating that TGFBR1 is not essential for this GDF9-regulated process in vivo .

  • Ovulation and fertilization potential: Tgfbr1 cKO mice maintain normal ovulation and can produce fertilizable oocytes, further demonstrating that TGFBR1 is not required for these specific aspects of ovarian function .

These findings reveal subtle but important distinctions between in vitro and in vivo roles of TGFBR1, particularly in its interactions with oocyte-derived factors, underscoring the complexity of TGF-β superfamily signaling in reproductive biology.

What is the evidence linking TGFBR1 gene polymorphisms to human disease susceptibility?

TGFBR1 gene polymorphisms have been linked to various human diseases, with substantial evidence coming from genetic association studies. One significant association involves diabetic retinopathy (DR), where TGF-β1/TGFBR1 signaling may contribute to pathology through disrupted angiogenesis .

A meta-analysis examining TGF-β1 gene polymorphisms in relation to diabetic retinopathy found significant associations for the +869T/C(L10P) polymorphism . Specifically:

Genetic ModelOdds Ratio (OR)95% Confidence IntervalAssociation
Allele model (L vs P)1.341.03-1.73Significant
Recessive model (LL vs LP+PP)1.701.13-2.56Significant

These findings suggest that individuals carrying certain TGFBR1 pathway-related polymorphisms may have increased susceptibility to diabetic retinopathy . Other studies have implicated TGFBR1 mutations in:

  • Loeys-Dietz Syndrome: A connective tissue disorder characterized by aortic aneurysms and other cardiovascular abnormalities .

  • Multiple Self-Healing Squamous Epithelioma: A skin condition associated with TGFBR1 variations .

To properly evaluate these associations, researchers should employ:

  • Comprehensive genotyping of multiple polymorphisms

  • Adequately powered sample sizes

  • Proper controls for population stratification

  • Meta-analysis approaches when possible

  • Functional validation of associated variants

These methodological considerations help establish reliable disease associations and potential mechanistic insights for TGFBR1-related pathologies.

What is the dual role of TGFBR1 in tumor suppression versus tumor promotion?

TGFBR1 exhibits a remarkable dual role in cancer biology, functioning as both a tumor suppressor and tumor promoter depending on cellular context, timing, and signaling intensity. This duality creates one of the most intriguing paradoxes in cancer research.

Tumor Suppressive Functions:
TGF-β proteins are well-established tumor suppressors in many contexts, inhibiting cell proliferation and promoting apoptosis in normal epithelial cells . This tumor-suppressive role operates primarily through the canonical SMAD-dependent pathway.

Tumor Promoting Functions:
Strikingly, research has demonstrated that constitutive activation of TGFBR1 in ovarian somatic cells drives gonadal tumor development with complete penetrance . These tumors exhibit multiple granulosa cell markers and cause elevated serum inhibin and estradiol levels, resembling human granulosa cell tumors .

The mechanisms underlying TGFBR1's tumor-promoting effects include:

  • Altered tumor microenvironment: Overactivation of TGFBR1 promotes angiogenesis, creating a favorable environment for tumor growth .

  • Enhanced cell proliferation: Constitutively active TGFBR1 enhances ovarian cell proliferation while simultaneously impairing normal cell differentiation .

  • Dysregulated gene expression: TGFBR1 overactivation causes dysregulation of critical genes involved in ovarian function .

This context-dependent duality has important implications for therapeutic approaches. The constitutively active TGFBR1 mouse model phenocopies several features of human granulosa cell tumors and may serve as a valuable platform for preclinical testing of targeted therapies . Understanding the molecular switches that determine whether TGFBR1 functions as a tumor suppressor or promoter is crucial for developing effective therapeutic strategies.

What are the optimal approaches for detecting and measuring activated TGFBR1 in human tissues?

Detecting and measuring activated TGFBR1 in human tissues requires sophisticated methodological approaches that target specific aspects of receptor activation. The following techniques provide complementary information about TGFBR1 activation status:

Protein-based detection methods:

  • Phospho-specific immunohistochemistry/immunofluorescence:

    • Utilizes antibodies specifically recognizing phosphorylated residues in the GS domain of TGFBR1

    • Enables visualization of activated receptor in situ while preserving tissue architecture

    • Can be combined with cell-type specific markers for contextual analysis

  • Proximity ligation assays (PLA):

    • Detects protein-protein interactions between TGFBR1 and TGFBR2 or downstream effectors

    • Generates fluorescent signals only when proteins are in close proximity

    • Provides spatial information about receptor complex formation

  • Phospho-flow cytometry:

    • Allows quantitative single-cell analysis of phosphorylated TGFBR1

    • Enables simultaneous assessment of multiple parameters

    • Particularly useful for heterogeneous tissues or blood samples

Downstream signaling assessment:

  • Phospho-SMAD2/3 detection:

    • Serves as a proximal readout of canonical TGFBR1 activation

    • Can be measured by western blot, immunohistochemistry, or flow cytometry

    • The nuclear localization of phospho-SMAD2/3 provides additional confirmation of pathway activation

  • Transcriptional profiling:

    • Analysis of TGFBR1-responsive gene signatures using RT-qPCR or RNA-seq

    • Provides functional readout of receptor activation

    • Can distinguish between canonical and non-canonical signaling outputs

For comprehensive analysis, researchers should employ multiple complementary techniques and include appropriate controls, such as samples treated with TGFBR1 kinase inhibitors. Establishing baseline activation levels in normal tissues is essential for interpreting pathological changes in receptor activation.

How should researchers interpret seemingly contradictory experimental findings about TGFBR1 function?

Interpreting contradictory findings about TGFBR1 function requires a systematic approach that considers biological context, experimental design, and methodological differences. TGFBR1 research presents particular challenges due to its complex signaling mechanisms and context-dependent functions.

Methodological framework for resolving contradictions:

  • Context-dependent functionality assessment:

    • TGFBR1 functions differently across tissues and developmental stages

    • The same genetic manipulation can produce opposite phenotypes in different contexts

    • For example, while TGFBR1 is crucial for female reproductive tract integrity , its role in other tissues may differ substantially

  • Experimental model evaluation:

    • Compare loss-of-function (knockout) versus gain-of-function (constitutively active) models

    • Different Cre drivers for conditional manipulation may have varying specificities and efficiencies

    • Consider temporal aspects of manipulation (developmental vs. adult)

    • For instance, constitutively active TGFBR1 promotes ovarian tumor development , while knockout models reveal roles in reproductive tract integrity

  • Signaling pathway analysis:

    • Distinguish between canonical (SMAD-dependent) and non-canonical pathways

    • Consider potential compensation by related receptors

    • Evaluate interaction with other signaling networks

  • Statistical approaches for data integration:

    • Meta-analysis for combining multiple studies, as demonstrated in TGF-β1 polymorphism research

    • Employ strict selection criteria and exclusion parameters

    • Consider both fixed and random effects models based on heterogeneity tests

    • Use forest plots to visualize effect sizes across studies

  • Experimental validation strategies:

    • Design experiments specifically targeting observed contradictions

    • Use multiple complementary approaches to test the same hypothesis

    • Consider in vitro versus in vivo differences, as seen with GDF9 signaling

By applying these systematic approaches, researchers can transform apparent contradictions into deeper insights about the complex and context-dependent functions of TGFBR1, ultimately advancing our understanding of this crucial signaling pathway in human biology and disease.

Product Science Overview

Structure and Function

TGF-βR1 is a serine/threonine kinase receptor that, upon binding with its ligand, TGF-β, forms a heteromeric complex with TGF-βR2. This complex then phosphorylates and activates downstream signaling molecules, primarily the SMAD proteins, which translocate to the nucleus to regulate gene expression .

Recombinant Human TGF-βR1

Recombinant human TGF-βR1 is produced using advanced biotechnological methods, often involving expression systems such as Escherichia coli or Chinese Hamster Ovary (CHO) cells. These systems allow for the production of high-purity, biologically active forms of the receptor, which are essential for research and therapeutic applications .

Applications

Recombinant TGF-βR1 is widely used in scientific research to study the TGF-β signaling pathway and its implications in various diseases, including cancer, fibrosis, and cardiovascular diseases. It is also utilized in drug development to screen for potential inhibitors or modulators of the TGF-β pathway .

Therapeutic Potential

The TGF-β signaling pathway is a target for therapeutic intervention in several diseases. Recombinant TGF-βR1 can be used to develop and test new drugs aimed at modulating this pathway. For instance, inhibitors of TGF-βR1 are being explored as potential treatments for cancer and fibrotic 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.