TFRC Human

Transferrin Receptor Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to TFRC Human

TFRC Human (Transferrin Receptor 1) is a transmembrane glycoprotein encoded by the TFRC gene. It facilitates iron uptake via receptor-mediated endocytosis, binding holo-transferrin and delivering iron into cells . This protein is critical for cellular iron homeostasis, erythropoiesis, and neurologic development .

Key Domains

  • Ectodomain: Binds transferrin and undergoes pH-dependent conformational changes during iron release .

  • Transmembrane Domain: Anchors the receptor in the plasma membrane .

Role in Iron Metabolism

TFRC regulates cellular iron uptake, compensating for iron deficiency and preventing toxicity . Dysregulation is linked to hereditary hemochromatosis and anemia .

Cancer Prognosis

High TFRC expression correlates with poor survival in pancreatic cancer (PAAD):

TFRC expression is associated with advanced tumor stages (T, N) and immune cell infiltration .

Immune Modulation

TFRC enhances ferroptosis in viral infections (e.g., CVB3) by increasing iron overload and lipid peroxidation . It also influences immune checkpoints (e.g., CD274, PDCD1) and chemokine-driven immune cell recruitment .

Tissue-Specific Expression Patterns

TissueExpression LevelClinical Relevance
Bone MarrowHighErythropoiesis regulation
LiverModerateIron storage and metabolism
PancreasHighCancer progression
Blood-Brain BarrierModerateDrug delivery applications

Data sourced from The Human Protein Atlas and TCGA .

Experimental Uses

  • Recombinant TFRC: Studied in E. coli for structural analysis and functional assays .

  • Antibody Therapies: Anti-TFRC antibodies (e.g., OX26) enable drug delivery across the blood-brain barrier .

Cancer Therapy

TFRC serves as a biomarker for:

  1. Pancreatic Cancer: Predicts immune infiltration and immunotherapy response .

  2. Leukemia/Lymphoma: Targeted by anti-CD71 antibodies in preclinical trials .

Ferroptosis Regulation

TFRC knockdown reduces iron-dependent lipid peroxidation, offering therapeutic potential in ischemia-reperfusion injury and viral infections .

Future Directions and Challenges

  1. Therapeutic Targeting: Developing TFRC-specific CAR-T therapies for pancreatic cancer .

  2. Iron Homeostasis Modeling: Leveraging TFRC to study anemia and hemochromatosis .

  3. Neurological Applications: Exploiting TfR1-mediated BBB transcytosis for neurodegenerative disease treatments .

Product Specs

Introduction
Transferrin receptor protein 1 (TFRC) is essential for cellular iron uptake from transferrin. This transmembrane glycoprotein consists of two disulfide-bonded monomers, each capable of binding a holo-transferrin molecule. This interaction forms an iron-Tf-TfR complex, which enters the cell via endocytosis.
Description
Recombinant human TFRC, expressed in E. coli, is a non-glycosylated polypeptide chain containing 696 amino acids (residues 89-760) with a molecular weight of 77.7 kDa. A 24 amino acid His-tag is fused to the N-terminus. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
The TFRC protein solution has a concentration of 1 mg/ml and is supplied in a buffer containing 20mM Tris-HCl (pH 8.0) and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, freeze the product at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 80% using SDS-PAGE analysis.
Synonyms
Transferrin receptor protein 1, TR, TfR, TfR1, Trfr, T9, p90, CD_antigen: CD71, Transferrin receptor, serum form, sTfR, TFRC, CD71.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMCKGVEP KTECERLAGT ESPVREEPGE DFPAARRLYW DDLKRKLSEK LDSTDFTGTI KLLNENSYVP REAGSQKDEN LALYVENQFR EFKLSKVWRD QHFVKIQVKD SAQNSVIIVD KNGRLVYLVE NPGGYVAYSK AATVTGKLVH ANFGTKKDFE DLYTPVNGSI VIVRAGKITF AEKVANAESL NAIGVLIYMD QTKFPIVNAE LSFFGHAHLG TGDPYTPGFP SFNHTQFPPS RSSGLPNIPV QTISRAAAEK LFGNMEGDCP SDWKTDSTCR MVTSESKNVK LTVSNVLKEI KILNIFGVIK GFVEPDHYVV VGAQRDAWGP GAAKSGVGTA LLLKLAQMFS DMVLKDGFQP SRSIIFASWS AGDFGSVGAT EWLEGYLSSL HLKAFTYINL DKAVLGTSNF KVSASPLLYT LIEKTMQNVK HPVTGQFLYQ DSNWASKVEK LTLDNAAFPF LAYSGIPAVS FCFCEDTDYP YLGTTMDTYK ELIERIPELN KVARAAAEVA GQFVIKLTHD VELNLDYERY NSQLLSFVRD LNQYRADIKE MGLSLQWLYS ARGDFFRATS RLTTDFGNAE KTDRFVMKKL NDRVMRVEYH FLSPYVSPKE SPFRHVFWGS GSHTLPALLE NLKLRKQNNG AFNETLFRNQ LALATWTIQG AANALSGDVW DIDNEF.

Q&A

What is the human transferrin receptor (TFRC) and what are its key functions?

Human transferrin receptor (TFRC) is a cell surface receptor that binds ferric-iron-loaded transferrin in the bloodstream to facilitate cellular iron uptake. TFRC functions through several key mechanisms:

  • Mediates receptor-mediated endocytosis of ligand-occupied transferrin into specialized endosomes

  • Facilitates iron release through endosomal acidification

  • Recycles the apotransferrin-receptor complex to the cell surface with return to neutral pH

  • Positively regulates T and B cell proliferation through iron uptake

  • Acts as a lipid sensor regulating mitochondrial fusion via the JNK pathway

  • Mediates uptake of NICOL1 into fibroblasts where it may regulate extracellular matrix production

  • Functions as a receptor for certain arenavirus infections

Where is human TFRC expressed and how is its expression regulated?

TFRC shows a tissue-specific expression pattern with notable expression in:

  • Brain capillary endothelial cells (forming the blood-brain barrier)

  • Erythrocytes and developing nervous system cells

  • T and B lymphocytes during proliferation

  • Cancer cells (often upregulated)

Expression regulation involves multiple factors:

  • Intracellular iron levels (inverse relationship)

  • Hypoxic conditions (increases expression)

  • CRE signaling pathways

  • Cellular proliferation status

How does TFRC dysfunction contribute to disease pathology?

TFRC dysfunction is implicated in multiple diseases through iron homeostasis disruption:

Disease CategoryExamplesTFRC-Related Mechanism
NeurodegenerativeParkinson's Disease, ALS, Huntington's DiseaseDeregulated iron homeostasis leading to oxygen radical formation and cellular dysfunction
Iron-Related DisordersIron deficiency, iron overload conditionsAltered TFRC expression affecting iron uptake
Multiple SclerosisInflammatory demyelinationDisrupted iron regulation in CNS cells
CancerMultiple typesUpregulation correlating with tumor progression

Research indicates iron surplus leads to oxygen radical formation and cellular dysfunction, while iron deficiency can cause rapid cell death—both contributing to pathological states .

What therapeutic strategies leverage TFRC for CNS drug delivery?

TFRC has emerged as a critical target for CNS drug delivery due to its expression on the blood-brain barrier:

  • AAV-based approaches: Engineered adeno-associated virus (AAV) capsids like BI-hTFR1 that specifically bind human transferrin receptor show enhanced CNS-specific tropism and therapeutic potential in delivering enzymes like glucocerebrosidase for conditions such as Gaucher disease

  • Nanoparticle delivery systems:

    • PEGylated liposomes coated with transferrin (TFRC ligand)

    • Anti-TFRC antibodies linked to nanoparticles (immunoliposomes) carrying therapeutic payloads

  • Receptor-mediated transport (RMT): Leverages TFRC's natural transcytosis mechanisms to shuttle therapeutic agents from bloodstream to CNS

How does TFRC function in the stearate-mediated regulation of mitochondrial dynamics?

TFRC acts as a lipid sensor modulating mitochondrial fusion through differential responses to stearate (C18:0) levels:

  • Low stearate conditions: TFRC promotes activation of the JNK pathway, resulting in HUWE1-mediated ubiquitination and degradation of the mitofusin MFN2, inhibiting mitochondrial fusion

  • High stearate conditions: TFRC stearoylation inhibits JNK pathway activation, preventing MFN2 degradation and promoting mitochondrial fusion

This represents a novel function beyond iron transport, connecting metabolic status to mitochondrial dynamics .

What experimental models are available for studying human TFRC?

Researchers can access several validated models to study human TFRC:

Model TypeExamplesKey FeaturesApplications
Cell LinesTFRC-expressing HEK293 cellsFull-length human TFRC expressionBinding assays, endocytosis studies
Mouse ModelsgenO-hTFRC mousePhysiological regulation and expression pattern, lacks murine TFRC, fully functional immune systemIn vivo efficacy assessment, PK profiling
Transgenic ModelsTFRC knockin miceHuman TFRC expressionCNS-specific tropism studies for AAV vectors
Antibody ToolsAnti-TFRC antibody [13E4]Suitable for Flow Cytometry and ICC/IFDetection and quantification in human samples

These models enable research across neuroscience, immuno-oncology, and inflammation fields .

What methodologies are recommended for detecting and quantifying human TFRC?

Multiple validated techniques exist for TFRC detection and quantification:

  • Immunological detection:

    • Flow cytometry for surface expression analysis

    • Immunocytochemistry/Immunofluorescence (ICC/IF) with validated antibodies

    • Western blotting for total protein quantification

  • Functional assays:

    • Transferrin binding and endocytosis studies

    • Iron uptake measurements

    • Receptor internalization and recycling assays

For ICC/IF applications, protocols typically include 4% formaldehyde fixation, BSA/serum blocking, and overnight primary antibody incubation at established concentrations (e.g., 5μg/ml for ab38171) .

How can TFRC targeting improve gene therapy for neurological disorders?

TFRC targeting significantly enhances gene therapy approaches for neurological disorders:

  • Blood-brain barrier penetration: Engineered AAV capsids (e.g., BI-hTFR1) that bind human TFRC demonstrate enhanced CNS tropism compared to standard vectors like AAV9

  • Therapeutic enzyme delivery: When used to deliver GBA1 (glucocerebrosidase gene), TFRC-targeting vectors substantially increased brain and cerebrospinal fluid enzyme activity compared to conventional AAV9 vectors

  • Disease-specific applications: Particularly valuable for treating Gaucher disease and potentially Parkinson's disease through GBA1 delivery

  • Species-specificity considerations: Enhanced tropism is CNS-specific and requires human TFRC expression, necessitating humanized models for preclinical testing

What is the significance of dermatoglyphic patterns in studying TFRC-related brain function?

While not directly related to TFRC, dermatoglyphic patterns provide complementary insights into brain development and function:

  • Fingerprint patterns and brain lobes: Each finger potentially represents activity in different brain lobes, with different fingerprint patterns (whorls, loops, arches) potentially correlating with cognitive traits

  • Quantitative assessments: Total Finger Ridge Count (TFRC) correlates with learning preferences and cognitive traits:

    • TFRC < 100: Needs stable learning environments

    • TFRC 100-149: Performance depends on external stimuli and guidance

    • TFRC 150-199: Easily distracted, suitable for multidisciplinary studies

    • TFRC ≥ 200: Good at multitasking with high short-term memory

  • Research applications: Can potentially complement TFRC protein studies in understanding brain development and function, particularly in personalized medicine approaches

How does the competitive binding between transferrin and HFE affect TFRC function?

The hereditary hemochromatosis protein HFE competes with transferrin for binding to TFRC:

  • Both proteins bind to an overlapping C-terminal site on TFRC

  • Competition affects transferrin binding affinity and subsequent iron uptake

  • This interaction represents a key regulatory mechanism for controlling cellular iron uptake

  • Mutations in HFE can disrupt this regulatory mechanism, potentially contributing to iron overload conditions

  • Understanding this competitive binding is crucial for developing therapeutic strategies for iron disorders

What are promising approaches for TFRC-targeted cancer therapies?

TFRC's upregulation in multiple cancer types creates several therapeutic opportunities:

  • TFRC-targeted immunotherapies: Leveraging increased expression on tumor cell surfaces

  • Antibody-drug conjugates: Utilizing anti-TFRC antibodies to deliver cytotoxic payloads

  • Iron-dependent cell death pathways: Exploiting cancer cells' heightened iron dependence

  • Combination strategies: Pairing TFRC targeting with immune checkpoint inhibitors

Studies in genO-hTFRC mouse models enable assessment of both efficacy and toxicity profiles of TFRC-binding therapeutics in an immunocompetent context .

How can researchers optimize TFRC-mediated drug delivery across the BBB?

Optimization strategies for TFRC-mediated BBB crossing include:

  • Binding affinity engineering: Moderate-affinity TFRC binders may perform better than high-affinity ones by facilitating release after transcytosis

  • Payload considerations: Size, charge, and stability affect transcytosis efficiency

  • Vector design optimization: For AAV capsids, specific modifications to enhance TFRC binding without compromising other functions

  • Dosing strategy development: Determining optimal concentration to maximize BBB crossing while minimizing potential toxicity

  • Combination approaches: Using multiple receptors simultaneously for enhanced delivery

Research using humanized TFRC mouse models allows direct comparison of different targeting strategies and detailed pharmacokinetic profiling .

Product Science Overview

Introduction

The Transferrin Receptor (TfR), also known as TfR-1 or CD71, is a type II transmembrane glycoprotein that plays a crucial role in iron homeostasis. It is primarily responsible for the uptake of transferrin-bound iron into cells. The receptor is expressed on the surface of various cell types, including erythroid progenitors, muscle cells, and proliferating cells .

Structure and Function

The Transferrin Receptor is a homodimer, consisting of two 95 kDa monomers linked by disulfide bonds, forming a 188 kDa dimer . Each monomer has an extracellular domain that binds to transferrin, a plasma protein that transports iron throughout the body. The binding of transferrin to TfR facilitates the internalization of the complex via receptor-mediated endocytosis .

Recombinant Human Transferrin Receptor

Recombinant human transferrin receptor is produced using various expression systems, including mammalian cell lines such as HEK293 and NS0 cells . The recombinant protein is typically tagged with a polyhistidine (His) tag to facilitate purification. The recombinant form retains the functional properties of the native receptor, including its ability to bind transferrin and mediate iron uptake.

Applications

Recombinant human transferrin receptor is widely used in research to study iron metabolism, receptor-mediated endocytosis, and related cellular processes. It is also employed in the development of therapeutic strategies for diseases associated with iron dysregulation, such as anemia and hemochromatosis .

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.