TGFBR1 Human

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

Description

Introduction to TGFBR1 Human

TGFBR1 (Transforming Growth Factor Beta Receptor 1) is a protein-coding gene critical for mediating cellular responses to TGF-β superfamily ligands. This receptor regulates processes such as cell proliferation, differentiation, apoptosis, and immune modulation . Mutations in TGFBR1 are linked to connective tissue disorders like Loeys-Dietz syndrome (LDS) and cancer progression .

Protein Structure

  • Domains:

    DomainAmino Acid RangeFunction
    Signal peptide1–24Guides receptor to cell membrane
    Extracellular domain25–125Binds TGF-β ligands
    Transmembrane domain126–147Anchors receptor in the membrane
    GS domain175–204Regulates kinase activation
    Kinase domain205–503Phosphorylates downstream targets
  • Molecular Weight:

    • Native protein: 53 kDa .

    • Recombinant variants (e.g., expressed in Sf9 cells): 38–57 kDa due to glycosylation and truncations .

Isoforms and Aliases

  • Key isoforms include ALK-5, ACVRLK4, and SKR4 .

  • Interacts with FKBP12 to maintain kinase inactivity until ligand binding .

Signaling Mechanisms and Biological Roles

TGFBR1 forms a heteromeric complex with TGFBR2 upon TGF-β ligand binding. This activates a signaling cascade:

  1. Canonical Pathway: Phosphorylates SMAD2/3, which complexes with SMAD4 to regulate gene transcription .

  2. Non-Canonical Pathways: Activates MAPK and PI3K/AKT pathways, influencing cell motility and survival .

Key Functions:

  • Suppresses tumorigenesis via growth inhibition in epithelial cells .

  • Modulates extracellular matrix production and immune responses .

Loeys-Dietz Syndrome (LDS)

  • Mechanism: Over 35 missense mutations (e.g., R244Q) cause hyperactivation of TGF-β signaling, leading to aortic aneurysms and skeletal abnormalities .

  • Animal Models:

    • TGFBR1 knockout mice exhibit uterine defects and infertility .

    • Constitutively active mutants cause hypermuscled uteri and reduced glands .

Cancer

  • Somatic Mutations: The *6A allele (a 3–alanine deletion) is somatically acquired in 29.5% of colorectal cancer metastases, conferring TGF-β resistance and growth advantages .

  • Polymorphisms: Meta-analyses associate TGFBR1 IVS7+24G>A with increased cancer risk (OR = 1.26) .

Inhibitors in Preclinical/Clinical Studies

InhibitorTarget PathwayApplication
GalunisertibTGF-β/SMADPancreatic cancer trials
SB-431542ALK5 kinaseFibrosis research
RepSoxSMAD2/3 phosphorylationStem cell differentiation

Animal Models Highlighting Regenerative Potential

  • R244Q Mutant Mice: Exhibit accelerated wound healing via enhanced chondrogenesis in bone marrow stromal cells .

  • Kinase-Deficient Models: Show disrupted spinal cord development, leading to ectopic limb structures .

Clinical Implications and Future Directions

TGFBR1’s dual role as a tumor suppressor and promoter underscores its therapeutic complexity. Current efforts focus on:

  • Targeting TGFBR1 mutations in LDS with kinase inhibitors .

  • Exploiting *6A allele-specific pathways in metastatic cancers .

Challenges: Balancing TGF-β’s anti-tumor and pro-metastatic effects remains a key hurdle .

Product Specs

Introduction

Transforming growth factor beta receptor 1 (TGFBR1) is a single-pass transmembrane protein belonging to the TGF-beta receptor subfamily and the protein kinase superfamily. This secreted protein is involved in a wide range of cellular processes, including the regulation of cell growth, differentiation, proliferation, and programmed cell death (apoptosis). TGFBR1 also plays a role in immune system modulation and exhibits varying effects on different cell types or cells at different developmental stages.

Description

Produced in Sf9 insect cells using a baculovirus expression system, this recombinant TGFBR1 protein is a single, glycosylated polypeptide chain consisting of 342 amino acids (27-126a.a.). It has a molecular weight of 38.0 kDa, although it may appear between 40-57 kDa on SDS-PAGE due to glycosylation. 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 clear, colorless solution that has been sterilized by filtration.
Formulation

The TGFBR1 protein is supplied as a 0.5 mg/mL solution in phosphate-buffered saline (PBS) at pH 7.4 containing 10% glycerol.

Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein such as 0.1% HSA or BSA is recommended for long-term storage. Repeated freezing and thawing of the product should be avoided.
Purity

The purity of this protein is greater than 90.0% as determined by SDS-PAGE analysis.

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 primary function in human cells?

TGFBR1, also known as Activin Receptor Like Kinase 5 (ALK5), is a transmembrane serine/threonine protein kinase that functions as a type I receptor in the TGF-β signaling pathway. The TGFBR1 gene is approximately 50 kb long and located on chromosome 9q22.33 . TGFBR1 forms a heteromeric complex with type II TGF-β receptors when bound to TGF-β ligands, transducing signals from the cell surface to the cytoplasm . This receptor spans the cell membrane with one end projecting from the outer surface (extracellular domain) and the other remaining inside the cell (intracellular domain) . The primary function of TGFBR1 is to transmit TGF-β signals that regulate critical cellular processes including cell proliferation, differentiation, motility, and controlled cell death (apoptosis) .

How does TGFBR1 participate in signal transduction pathways?

TGFBR1 participates in signal transduction through a well-defined mechanism. When TGF-β ligand binds to the extracellular domain of TGFBR1, it activates the receptor and enables it to bind to another receptor (TGFBR2) on the cell surface. These proteins form a complex that initiates signal transduction, activating other proteins in the TGF-β pathway . Specifically, activated TGFBR1 phosphorylates downstream effector molecules, particularly SMAD proteins (SMAD2 and SMAD3). Once phosphorylated, these SMADs form complexes with SMAD4 and translocate to the nucleus where they regulate gene expression . This signaling cascade results in various cellular responses that are context-dependent, impacting critical processes from development to tissue homeostasis.

What are the common synonyms and alternative names for the TGFBR1 gene?

The TGFBR1 gene is known by several alternative names and symbols in the scientific literature, which can sometimes lead to confusion when searching databases. Common synonyms include AAT5, ACVRLK4, ALK-5, ALK5, ESS1, LDS1, LDS1A, LDS2A, MSSE, SKR4, TBR-i, TBRI, TGFR-1, and tbetaR-I . Understanding these alternative designations is important for conducting comprehensive literature searches and ensuring that researchers are accessing all relevant information about this gene and its protein product.

What phenotypes are associated with TGFBR1 mutations in humans?

TGFBR1 mutations in humans are associated with several distinct phenotypes, most notably Loeys-Dietz syndrome type I (LDS1), which affects connective tissue. More than 35 mutations in the TGFBR1 gene have been identified in LDS1 patients, who exhibit blood vessel abnormalities and skeletal deformities . Most LDS-causing mutations are present in one copy of the gene in each cell (heterozygous) and typically alter single amino acids in the TGFBR1 protein . Additionally, TGFBR1 variants have been associated with multiple self-healing squamous epithelioma (MSSE) . Research also suggests relationships between certain TGFBR1 polymorphisms and cancer susceptibility, though results across studies have been inconsistent .

How does TGFBR1 function in female reproductive biology?

TGFBR1 plays a critical role in female reproductive tract development and function. Studies using conditional knockout (cKO) mouse models with Tgfbr1 deletion in the female reproductive tract have demonstrated that these mice are sterile . The primary defect observed was the development of oviductal diverticula that impair embryo development and transit to the uterus. Molecular analysis revealed dysregulation of several cell differentiation and migration genes, including Krt12, Ace2, and MyoR, that are associated with female reproductive tract development . Furthermore, defective smooth muscle development was observed in the uteri of Tgfbr1 cKO mice. These findings establish that TGFBR1 is essential for maintaining the structural integrity and function of the female reproductive tract .

What is the paradoxical role of TGFBR1 in cancer development?

TGFBR1 exhibits a paradoxical dual role in cancer development, functioning as both a tumor suppressor and a potential tumor promoter depending on the context and stage of disease. In early stages of cancer, TGFBR1-mediated signaling typically suppresses tumor formation by inhibiting cell proliferation and promoting apoptosis . This tumor-suppressive role is evidenced by the fact that TGFBR1 helps prevent cells from growing and dividing too rapidly or in an uncontrolled way .

Contrastingly, other research showed that Tgfbr1 haploinsufficient mice developed twice as many intestinal tumors as wild-type mice, and when crossed with Apc Min/+ mice, developed almost three times more tumors than controls (14.5 vs 5.4), indicating a tumor-suppressive role in this setting . These apparently contradictory findings highlight the context-dependent nature of TGFBR1 function in cancer biology.

What experimental models are most effective for studying TGFBR1 function in vivo?

Research on TGFBR1 function has employed several complementary experimental models, each with specific advantages for addressing particular research questions:

  • Conventional knockout models: Complete Tgfbr1 knockout mice die embryonically, highlighting the essential developmental role of this receptor but limiting postnatal studies .

  • Conditional knockout models: These have proven particularly valuable for studying tissue-specific TGFBR1 functions. For example, reproductive tract-specific deletion using anti-Müllerian hormone receptor type 2 promoter-driven Cre recombinase revealed essential roles in female reproductive tract integrity .

  • Haploinsufficient models: Tgfbr1+/− mice have been crucial for understanding TGFBR1's role in cancer susceptibility. These models show that partial reduction in TGFBR1 signaling can significantly affect tumor development in various contexts .

  • Combined genetic models: Crossing Tgfbr1 haploinsufficient mice with cancer-predisposed models (e.g., EL-Kras mice for pancreatic cancer or Apc Min/+ mice for intestinal tumors) has provided insights into how altered TGFBR1 signaling modifies cancer risk in different tissues .

When designing experiments, researchers should consider that the effects of altered TGFBR1 signaling are highly context-dependent, varying by tissue type, developmental stage, and disease state. The choice of model should align with the specific biological question being addressed.

How should researchers interpret contradictory findings regarding TGFBR1 in cancer?

The contradictory findings regarding TGFBR1's role in cancer reflect its complex, context-dependent functions. To properly interpret such contradictions, researchers should consider:

  • Tissue-specific effects: TGFBR1 signaling appears to have different, sometimes opposite effects depending on the tissue. For instance, reduced TGFBR1 signaling inhibits precancerous lesion development in pancreatic tissue but promotes intestinal tumor formation .

  • Disease stage: TGF-β signaling generally acts as a tumor suppressor in early cancer stages but may promote progression in advanced disease. Experimental timepoints should be carefully considered when evaluating results.

  • Genetic context: The effects of altered TGFBR1 signaling can vary dramatically depending on co-occurring genetic alterations. For example, Tgfbr1 haploinsufficiency produces different outcomes in wild-type versus Kras-mutant or Apc-mutant backgrounds .

  • Quantitative aspects: The degree of TGFBR1 signaling reduction matters. Complete loss versus partial reduction may produce qualitatively different outcomes.

  • Methodological differences: Variations in experimental approaches, including how TGFBR1 alterations are generated and measured, can influence results.

When encountering contradictory findings, researchers should avoid overgeneralizing and instead carefully consider these contextual factors. Comprehensive experimental designs that examine multiple timepoints, tissues, and genetic backgrounds may help resolve apparent contradictions.

What methodological approaches can detect allele-specific expression of TGFBR1?

Allele-specific expression (ASE) of TGFBR1 has been studied as a potential cancer risk factor, though findings have been inconsistent. Several methodological approaches have been employed to detect ASE:

  • SNaPshot technology: This has been used to quantify allele-specific expression in blood and other tissues of colorectal cancer patients and controls. This method relies on naturally occurring transcribed SNPs to differentiate expression from the two alleles .

  • Analysis of SNP markers: Different studies have used various SNP markers to assess ASE, which may contribute to variability in results. The choice of SNP markers is crucial as it determines which individuals are informative for the analysis (typically 25-60% of individuals) .

  • Tissue selection considerations: ASE may vary between tissue types, so the source of nucleic acids is an important methodological consideration. Though some studies found consistent ASE patterns across different uncultured tissues from the same individuals, systematic analysis of multiple tissue types is generally lacking .

Research indicates that ASE differences between cases and controls are subtle, potentially too subtle to reliably assess cancer risk. Current technological limitations, particularly the reliance on naturally occurring transcribed SNPs, represent a significant constraint. Future advances that allow direct quantification of allele-specific transcription without relying on polymorphic markers may provide more definitive answers regarding TGFBR1 ASE .

How might TGFBR1 variants influence individual cancer susceptibility?

TGFBR1 variants can influence individual cancer susceptibility through several mechanisms, though the evidence remains complex and sometimes contradictory:

The translational potential of these findings remains uncertain. Current evidence suggests that constitutively decreased TGFBR1 signaling could be a modifier of cancer susceptibility and progression , but the clinical utility of TGFBR1 variation for cancer risk assessment requires further investigation through larger, more definitive studies.

Can TGFBR1 be targeted therapeutically in human diseases?

The potential for therapeutic targeting of TGFBR1 in human diseases depends on the specific context:

Any therapeutic targeting of TGFBR1 would need to carefully consider tissue specificity, disease stage, and potential systemic effects given the receptor's widespread expression and diverse functions. Combination approaches that modulate rather than completely block signaling may offer the best balance of efficacy and safety.

What are the most significant unanswered questions about TGFBR1 in human biology?

Despite decades of research, several critical questions about TGFBR1 remain unanswered:

Addressing these questions will require integration of advanced genomic, transcriptomic, and proteomic approaches with sophisticated in vivo models that recapitulate human disease contexts.

How might emerging technologies enhance our understanding of TGFBR1 biology?

Emerging technologies offer promising avenues to advance TGFBR1 research:

  • CRISPR-Cas9 genome editing: This technology enables precise modification of TGFBR1 in cellular and animal models, allowing creation of specific mutations mimicking human diseases and facilitating functional studies of regulatory elements controlling TGFBR1 expression.

  • Single-cell technologies: Single-cell RNA sequencing and spatial transcriptomics can reveal cell-specific responses to TGFBR1 signaling within complex tissues, potentially clarifying the basis for context-dependent effects.

  • Advanced allele-specific expression analysis: Emerging methods that circumvent the need for naturally occurring transcribed SNPs could provide more comprehensive assessment of allele-specific TGFBR1 expression, addressing current technological limitations .

  • Organoid models: Patient-derived organoids offer systems to study TGFBR1 function in near-physiological human tissue contexts, potentially bridging gaps between animal models and human disease.

  • Systems biology approaches: Integration of multi-omics data with computational modeling may help untangle the complex signaling networks downstream of TGFBR1 and predict context-specific outcomes of pathway modulation.

These technological advances, particularly when applied in combination, hold promise for resolving longstanding questions about TGFBR1 biology and its role in human disease.

Product Science Overview

Definition and Classification

Transforming Growth Factor Beta Receptor I (TGFBR1), also known as Activin Receptor-Like Kinase 5 (ALK-5), is a serine/threonine kinase receptor that belongs to the protein kinase superfamily and the TGF-beta receptor subfamily . It is a single-pass type I membrane protein that plays a crucial role in the TGF-beta signaling pathway .

Biological Properties

TGFBR1 is expressed in all tissues examined, with the highest abundance in the placenta and the lowest in the brain and heart . The receptor is involved in various cellular processes, including cell growth, differentiation, apoptosis, and immune responses .

Expression Patterns and Tissue Distribution

TGFBR1 is ubiquitously expressed across different tissues. Its expression is particularly high in the placenta, indicating its significant role in reproductive and developmental processes . The receptor’s expression is relatively lower in the brain and heart .

Biological Functions

TGFBR1 functions as a tumor suppressor by inhibiting the cell cycle in the G1 phase . It is involved in the regulation of cellular growth, differentiation, apoptosis, motility, invasion, extracellular matrix production, angiogenesis, and immune responses . The receptor forms a complex with type II TGF-beta receptors upon ligand binding, leading to the phosphorylation and activation of SMAD transcriptional regulators .

Modes of Action

Upon ligand binding, TGFBR1 forms a receptor complex consisting of two type I and two type II transmembrane serine/threonine kinases . The type II receptors phosphorylate and activate the type I receptors, which then auto-phosphorylate and bind to SMAD proteins . This activation leads to the translocation of SMAD proteins to the nucleus, where they regulate the transcription of target genes .

Regulatory Mechanisms

The TGF-beta/SMAD pathway is tightly regulated to ensure proper cellular responses. Disruption of this pathway has been implicated in various human cancers, with the majority of colon and gastric cancers being caused by inactivating mutations in TGF-beta RII . The administration of TGF-beta has been shown to protect against mammary tumor development in transgenic mouse models .

Recombinant Human TGFBR1

Recombinant human TGFBR1 is produced using baculovirus expression systems in insect cells . The recombinant protein is often tagged with GST (Glutathione S-transferase) to facilitate purification and detection . It is supplied in a buffer containing Tris-HCl, NaCl, glutathione, EDTA, DTT, PMSF, and glycerol to maintain stability and activity .

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.