Target Name: TFRC
NCBI ID: G7037
Review Report on TFRC Target / Biomarker Content of Review Report on TFRC Target / Biomarker
TFRC
Other Name(s): sTfR | TFRC variant 2 | IMD46 | TFR1 | TFRC variant 1 | Transferrin receptor protein 1, serum form | TRFR | P90 | TFR1_HUMAN | Transferrin receptor protein 1 | TR | Transferrin receptor | Trfr | p90 | TfR1 | transferrin receptor | TFR | Soluble transferrin receptor protein 1 | TfR | soluble serum form sTfR | sTfR_(HUMAN) | CD71 | T9 | Transferrin receptor protein 1 (isoform 1)

Introduction to TFRC, A Potential Drug Target

Drug discovery and development are complex processes that require identifying specific protein targets or biomarkers within the human body. One such promising drug target is TFRC (transferrin receptor protein), which plays a critical role in iron metabolism and cellular uptake. This article will delve into the intricacies of TFRC as a drug target, its potential applications in various diseases, and the ongoing research in this field.

The Role of TFRC in Iron Metabolism

TFRC is a cell surface glycoprotein that facilitates the uptake of transferrin-bound iron into cells. Iron is an essential mineral that serves numerous crucial functions in the body, including oxygen transport, DNA synthesis, and cellular respiration. To fulfill its diverse roles, iron must be properly regulated within cells, and TFRC plays a central role in this process.

When iron levels are low, TFRC expression increases on cell surfaces, contributing to enhanced iron uptake. This mechanism ensures that cells have an adequate supply of iron to meet their metabolic demands. Conversely, when iron levels are high, TFRC expression decreases, preventing excessive iron accumulation and potential toxicity.

TFRC as a Promising Drug Target

TFRC's pivotal role in iron metabolism makes it an attractive target for therapeutic interventions. Various diseases, such as cancer and neurodegenerative disorders, exhibit dysregulated iron metabolism, leading to adverse health outcomes. Targeting TFRC could potentially modulate iron levels within cells and restore proper iron homeostasis.

In cancer, TFRC is highly expressed on the cell surface, allowing cancer cells to uptake more iron than normal cells. High iron levels in cancer cells promote cell growth, DNA synthesis, and angiogenesis, facilitating tumor progression. By targeting TFRC, researchers aim to disrupt iron uptake in cancer cells, slowing down their growth and potentially enhancing the efficacy of current therapies.

In neurodegenerative disorders like Alzheimer's and Parkinson's diseases, accumulating evidence suggests altered iron metabolism as a contributing factor to disease pathology. Increased iron deposition has been observed in affected brain regions, leading to oxidative stress and neuronal damage. Targeting TFRC could potentially regulate iron accumulation in the brain, potentially slowing down disease progression and mitigating symptom severity.

Current Research and Future Applications

Researchers have been actively investigating TFRC as a drug target in various disease contexts. Several strategies are being explored to target TFRC:

1. Antibody-based therapies: Monoclonal antibodies specifically targeting TFRC have shown promising results in preclinical studies. These antibodies bind to TFRC, blocking iron uptake and inhibiting cancer cell growth.

2. Nanoparticle-based drug delivery: Functionalized nanoparticles can be engineered to selectively target TFRC on cancer cells. By loading these nanoparticles with therapeutic agents, such as chemotherapeutic drugs or gene editing tools, researchers aim to deliver targeted therapies directly to cancer cells.

3. Small molecule inhibitors: Drug discovery efforts have identified small molecules that can inhibit TFRC-mediated iron uptake. These molecules prevent the binding of transferrin to TFRC, impairing iron uptake and interfering with cancer cell growth.

4. Imaging modalities: TFRC's overexpression on cancer cells can also be exploited for diagnostic purposes. Novel imaging techniques, such as positron emission tomography (PET) or magnetic resonance imaging (MRI), are being developed to detect TFRC expression levels. This information can aid clinicians in assessing disease progression and monitoring treatment response.

While the research on TFRC as a drug target is still predominantly in preclinical stages, the potential applications are substantial. However, challenges remain, such as achieving target selectivity, minimizing off-target effects, and optimizing drug delivery strategies. Future studies will likely focus on refining these approaches and evaluating their efficacy in clinical trials.

Conclusion

TFRC represents a promising drug target with significant therapeutic potential in diseases characterized by dysregulated iron metabolism. Its critical role in iron uptake and cellular homeostasis makes it an attractive candidate for therapeutic interventions. Ongoing research efforts are exploring various strategies, such as antibody-based therapies, nanoparticle drug delivery, small molecule inhibitors, and imaging techniques, to target TFRC effectively.

While translating these promising preclinical findings into clinical applications poses challenges, the discovery of TFRC as a drug target opens up new avenues for the treatment of conditions like cancer and neurodegenerative disorders. By understanding and manipulating the intricate mechanisms of TFRC, researchers aim to revolutionize disease management and improve patient outcomes in the future.

Protein Name: Transferrin Receptor

Functions: Cellular uptake of iron occurs via receptor-mediated endocytosis of ligand-occupied transferrin receptor into specialized endosomes (PubMed:26214738). Endosomal acidification leads to iron release. The apotransferrin-receptor complex is then recycled to the cell surface with a return to neutral pH and the concomitant loss of affinity of apotransferrin for its receptor. Transferrin receptor is necessary for development of erythrocytes and the nervous system (By similarity). A second ligand, the heditary hemochromatosis protein HFE, competes for binding with transferrin for an overlapping C-terminal binding site. Positively regulates T and B cell proliferation through iron uptake (PubMed:26642240). Acts as a lipid sensor that regulates mitochondrial fusion by regulating activation of the JNK pathway (PubMed:26214738). When dietary levels of stearate (C18:0) are low, promotes activation of the JNK pathway, resulting in HUWE1-mediated ubiquitination and subsequent degradation of the mitofusin MFN2 and inhibition of mitochondrial fusion (PubMed:26214738). When dietary levels of stearate (C18:0) are high, TFRC stearoylation inhibits activation of the JNK pathway and thus degradation of the mitofusin MFN2 (PubMed:26214738)

The "TFRC Target / Biomarker Review Report" is a customizable review of hundreds up to thousends of related scientific research literature by AI technology, covering specific information about TFRC comprehensively, including but not limited to:
•   general information;
•   protein structure and compound binding;
•   protein biological mechanisms;
•   its importance;
•   the target screening and validation;
•   expression level;
•   disease relevance;
•   drug resistance;
•   related combination drugs;
•   pharmacochemistry experiments;
•   related patent analysis;
•   advantages and risks of development, etc.
The report is helpful for project application, drug molecule design, research progress updates, publication of research papers, patent applications, etc. If you are interested to get a full version of this report, please feel free to contact us at BD@silexon.ai

More Common Targets

TG | TGDS | TGFA | TGFA-IT1 | TGFB1 | TGFB1I1 | TGFB2 | TGFB2-AS1 | TGFB3 | TGFBI | TGFBR1 | TGFBR2 | TGFBR3 | TGFBR3L | TGFBRAP1 | TGIF1 | TGIF2 | TGIF2-RAB5IF | TGIF2LX | TGIF2LY | TGM1 | TGM2 | TGM3 | TGM4 | TGM5 | TGM6 | TGM7 | TGOLN2 | TGS1 | TH | TH2LCRR | THADA | THAP1 | THAP10 | THAP11 | THAP12 | THAP12P1 | THAP12P7 | THAP2 | THAP3 | THAP4 | THAP5 | THAP6 | THAP7 | THAP7-AS1 | THAP8 | THAP9 | THAP9-AS1 | THBD | THBS1 | THBS2 | THBS2-AS1 | THBS3 | THBS3-AS1 | THBS4 | THBS4-AS1 | THEG | THEG5 | THEGL | THEM4 | THEM5 | THEM6 | THEMIS | THEMIS2 | THG1L | Thioredoxin-disulfide reductase (TrxR) | THNSL1 | THNSL2 | THO complex | THOC1 | THOC2 | THOC3 | THOC5 | THOC6 | THOC7 | Thomsen-Friedenreich Antigen (CD176) | THOP1 | THORLNC | THPO | THRA | THRAP3 | THRB | Three amino acid loop extension transcription regulators | Threonine protease | THRIL | THRSP | THSD1 | THSD1P1 | THSD4 | THSD4-AS1 | THSD7A | THSD7B | THTPA | THUMPD1 | THUMPD2 | THUMPD3 | THUMPD3-AS1 | THY1 | Thymidine Kinase | THYN1