Fact-checked by Grok 2 weeks ago

Transferrin receptor

The transferrin receptor (TfR) is a transmembrane that mediates the cellular uptake of iron by binding to iron-loaded (holo-transferrin) and facilitating its internalization through , serving as the primary pathway for iron acquisition in vertebrates. This process is crucial for maintaining iron , supporting , and enabling iron transport across barriers like the blood-brain barrier. TfRs exist in multiple isoforms, with TfR1 (encoded by the TFRC gene) being the predominant form, a type II that forms a homodimer of two 90-97 kDa subunits linked by disulfide bonds, each containing cytoplasmic, transmembrane, and extracellular domains with a butterfly-like structure for high-affinity holo- binding (Kd ≈ 10⁻⁹ M at neutral pH). TfR2, encoded by TFR2, shares about 45% homology with TfR1 but exhibits lower affinity for (25- to 27-fold less) and is mainly expressed in hepatocytes and erythroid precursors, where it regulates expression to control systemic iron levels. Additional isoforms include TfR3 (glyceraldehyde-3-phosphate dehydrogenase, involved in rapid iron uptake under stress like ) and TfR4 (cubilin, for renal reabsorption), though TfR1 and TfR2 dominate physiological iron handling. The TFRC gene spans approximately 70 kb with 19 exons, and TfR expression is post-transcriptionally regulated by iron-responsive elements (IREs) in response to cellular iron needs. In function, TfR1 binds diferric at the cell surface, clusters into clathrin-coated pits, and undergoes ; within the , acidification ( ≈ 5.5) releases iron from transferrin, which is then reduced to Fe²⁺ and transported into the via divalent metal transporter 1 (DMT1), while the apo-transferrin-TfR1 complex recycles to the surface. This cycle allows efficient iron delivery without degrading transferrin, with TfR1 acting as a gatekeeper that adjusts uptake based on iron availability—high TfR1 levels in iron-deficient states enhance import, while iron excess suppresses expression via iron regulatory protein 2 (IRP2). TfR2 complements this by sensing holotransferrin levels to modulate , an iron-regulatory , thus preventing overload or deficiency. TfRs are ubiquitously expressed in nucleated s, with highest levels of TfR1 in hemoglobin-synthesizing erythroblasts (up to 1 million copies per ) and capillary endothelial cells for central nervous system iron supply, while TfR2 is primarily expressed in the liver and erythroid tissues. Dysregulation contributes to disorders: TfR1 mutations cause combined and , while TfR2 defects lead to hereditary hemochromatosis type 3; overexpression of TfR1 (often 100-fold higher than normal) in cancers like gliomas promotes tumor growth via excess iron and serves as a therapeutic target for antibody-drug conjugates or nanocarriers that exploit receptor-mediated delivery across the blood- barrier. Recent advances highlight TfR1's potential in targeted therapies for neurodegenerative diseases, such as Alzheimer's, by shuttling iron chelators or amyloid-clearing agents.

Overview

Definition and general function

The transferrin receptor (TfR) is a family of proteins that binds iron-loaded (Tf-Fe) to enable , serving as the primary mechanism for cellular iron uptake in vertebrates. TfR1 and TfR2 are transmembrane glycoproteins, while other members like TfR3 and TfR4 function through alternative surface-binding mechanisms. This process involves the receptor recognizing and internalizing the Tf-Fe complex at the cell surface, followed by acidification in endosomes to release iron for intracellular use. The general function of TfRs centers on regulating iron entry into cells to sustain systemic iron , which is crucial for fundamental biological processes including , mitochondrial respiration, oxygen transport via , and the activity of iron-dependent enzymes. By controlling iron availability, TfRs prevent both deficiency, which can lead to , and overload, which risks oxidative damage. TfRs exist in multiple forms that collectively support this role across diverse tissues, though their expression is modulated by cellular iron needs. TfRs were first identified in the late 1970s as the protein CD71 on reticulocytes, where high expression facilitates massive iron import for production during . This discovery highlighted TfRs' essential role in averting , particularly in rapidly dividing erythroid cells. While ubiquitously expressed at low levels in most cells, TfR levels are markedly upregulated in iron-demanding tissues like erythroid precursors to meet heightened requirements for proliferation and differentiation.

Types of transferrin receptors

Transferrin receptors (TfRs) are classified into four distinct types based on their molecular identities, genetic origins, and functional specializations in iron acquisition. TfR1, also known as CD71, is encoded by the located on 3q29 and serves as the primary receptor for cellular iron uptake across most tissues. TfR2 is encoded by the on 7q22.1, which produces two main isoforms: the full-length alpha isoform (TfR2α) responsible for membrane-bound iron sensing and a shorter isoform (TfR2β) with tissue-specific expression patterns. In contrast, TfR3 corresponds to glyceraldehyde-3-phosphate dehydrogenase (), a ubiquitously expressed glycolytic that exhibits transferrin-binding capability independent of its primary metabolic role. TfR4 is identified as cubilin, a large multiligand endocytic receptor encoded by the , which facilitates transferrin interactions through its multi-domain structure. The key distinctions among these receptors lie in their structural and operational characteristics. TfR1 and TfR2 are classical type II transmembrane glycoproteins that form homodimers and specifically bind iron-loaded (Tf-Fe) to mediate receptor-dependent , with TfR1 exhibiting higher affinity and broader expression than TfR2. TfR3, functioning as a protein, acts primarily as a stress-responsive transferrin binder that localizes to the cell surface under iron-depleting conditions, enabling rapid, low-affinity uptake via alternative endocytic pathways in select cell types such as macrophages. TfR4, through its cooperation with megalin, supports renal reabsorption of filtered transferrin in cells, relying on its extensive complement of domains for multivalent rather than dedicated transmembrane signaling. Evolutionarily, TfR1 and TfR2 share significant in their extracellular domains (approximately 45% sequence identity), reflecting a common ancestral origin within the transferrin receptor family that supports conserved iron-transport mechanisms across vertebrates. TfR3 and TfR4, however, represent repurposed proteins: GAPDH (TfR3) leverages its ancient enzymatic scaffold for secondary iron-binding roles, while cubilin (TfR4) evolved as part of a broader endocytic complex distinct from the TfR1/TfR2 lineage. Recent reviews up to 2025 have formalized TfR3 and TfR4 as bona fide transferrin receptors, based on demonstrated binding affinities and functional assays confirming their contributions to iron under physiological stress or tissue-specific contexts.

Molecular structure

Structure of TfR1

The transferrin receptor 1 (TfR1) is a 90 kDa type II transmembrane that functions as a homodimer on the cell surface. Each consists of a short cytoplasmic tail spanning residues 1–67, which includes a tyrosine-based motif (YTRF at positions 20–23), a single transmembrane from residues 68–88, and a large extracellular encompassing residues 89–760. The extracellular is responsible for binding and adopts a butterfly-like dimeric , with each comprising three globular subdomains: a protease-like , an apical , and a helical that together form a lateral cleft for interaction with (Tf). Key structural features of TfR1 include two inter-subunit bonds at residues 89 and 98, which covalently link the monomers and contribute to dimer stability, alongside non-covalent interactions that enhance overall integrity. The protein undergoes N-linked at three sites in the extracellular domain—Asn251, Asn317, and Asn727—which are crucial for proper folding, stability, and trafficking to the plasma membrane; at these sites, particularly Asn727, impair surface expression and . The Tf-binding site, located in the extracellular region at the interface of the helical and protease-like domains, exhibits sensitivity, facilitating high-affinity association with holo-Tf (iron-loaded ) at neutral on the cell surface. Insights into TfR1 were first provided by the of its ectodomain, resolved at 3.2 Å resolution in 1999 (PDB: 1CX8), revealing the dimeric butterfly shape and the positioning of the Tf-binding clefts. Subsequent structures, such as the TfR1-holo-Tf complex determined by cryo-EM at 7.5 Å resolution (PDB: 1SUV), illustrate how the receptor engages iron-loaded Tf with high affinity (Kd ≈ 1–5 nM) at physiological pH, while in acidic endosomes (pH ≈ 5.5) triggers conformational changes that promote iron release and receptor recycling. Dimerization via both covalent disulfides and non-covalent contacts is essential for this high-affinity binding, as monomeric forms exhibit reduced avidity. Compared to TfR2, TfR1 shares a similar transmembrane but possesses a shorter cytoplasmic tail.

Structure of TfR2

Transferrin receptor 2 (TfR2), specifically the predominant isoform TfR2α, is an 89 kDa type II composed of 801 that functions as a non-covalently associated homodimer. Its overall includes a short cytoplasmic spanning residues 1–80, which includes a tyrosine-based motif (YQRV); a hydrophobic from residues 81–104; and a large extracellular domain encompassing residues 105–801 responsible for interactions. This structural organization positions TfR2 primarily on the plasma membrane of hepatocytes and erythroid precursors, where it senses circulating iron levels without efficient . Key structural features of the extracellular domain include conserved residues that form bonds essential for maintaining the protein's folded conformation and stability. Additionally, the extracellular region contains two N-linked sites at Asn125 and Asn533, which contribute to proper folding, trafficking, and binding without directly affecting . The -binding pocket in the extracellular domain exhibits an altered configuration compared to its homolog, resulting in a lower for holotransferrin (Kd ≈ 20–30 at neutral ), which supports its role in detecting high-iron conditions rather than routine uptake. TfR2 exists in two main isoforms generated by : the full-length TfR2α (801 ), which includes all transmembrane and extracellular components for localization and function; and the truncated TfR2β (100 ), which lacks the transmembrane and extracellular domains, rendering it cytosolic and non-functional for iron uptake. In the cytoplasmic tail, residues 64–82 form a binding domain that interacts with the hereditary hemochromatosis protein HFE, facilitating downstream signaling for regulation. Like TfR1, TfR2 shares a homologous extracellular fold consisting of a butterfly-like dimer with helical and protease-like domains, though adapted for liver-specific iron sensing.

Structures of TfR3 and TfR4

Transferrin receptor 3 (TfR3), also known as glyceraldehyde-3-phosphate (GAPDH), is a multifunctional protein that serves as a non-classical binder. Unlike the homologous transmembrane TfR1 and TfR2, TfR3 and TfR4 represent moonlighting functions of multifunctional proteins with auxiliary roles in iron handling. It exists as a 37 that assembles into a homotetrameric with an approximate molecular weight of 150 kDa. TfR3 lacks a and functions as a peripheral or soluble protein. Its core features a classic NAD-binding Rossmann fold, characteristic of , with the -binding site located on the protein surface, enabling moonlighting functions beyond . TfR3 localizes to diverse cellular compartments, including the plasma membrane, , and , facilitated by post-translational modifications such as lipidation, which anchor it to membranes without integral embedding. Transferrin receptor 4 (TfR4), identified as cubilin, is a large peripheral membrane protein with a molecular weight of approximately 460 kDa. It comprises 27 complement C1r/C1s, Uegf, and BMP-1 (CUB) domains responsible for ligand binding, interspersed with 8 epidermal growth factor (EGF)-like domains that provide structural spacing and flexibility. Similar to TfR3, TfR4 lacks a transmembrane helix, relying instead on interactions with accessory proteins like megalin and amnionless (AMN) for membrane association and endocytic trafficking. The protein is heavily glycosylated, featuring both N-linked and O-linked modifications that contribute to its stability, solubility, and ligand affinity. Structural studies, including electron microscopy reconstructions, reveal an extended, chain-like architecture that forms a multi-domain "basket" suited for capturing diverse ligands during renal reabsorption. Recent cryo-EM studies of related complexes (as of 2024) further delineate the modular arrangement of CUB and EGF domains in complex with partners. In contrast to the transmembrane, dimerizing TfR1 and TfR2, both TfR3 and TfR4 operate as peripheral entities without direct integration, emphasizing auxiliary roles in transferrin handling. TfR3 repurposes the tetrameric glycolytic scaffold of GAPDH for surface transferrin interactions, while TfR4's expansive multi-domain setup supports broad endocytic capture, particularly in specialized tissues like the . These structural distinctions underscore their adaptation for context-specific iron management rather than constitutive uptake.

Biological functions

Iron uptake and homeostasis

The transferrin receptor (TfR), primarily TfR1, facilitates cellular iron uptake through a pathway that is essential for maintaining . Iron-loaded (holo-Tf or Tf-Fe) binds to TfR on the cell surface at neutral pH (approximately 7.4), where the complex is concentrated in clathrin-coated pits for . This process is energy-dependent, relying on by to drive vesicle scission from the plasma membrane, forming clathrin-coated vesicles that deliver the complex to early endosomes. Within the acidified (pH approximately 5.5), protonation induces the release of ferric iron (Fe³⁺) from , which is then reduced to ferrous iron (Fe²⁺) by the ferrireductase STEAP3. The Fe²⁺ is subsequently transported across the endosomal membrane into the via the divalent metal transporter 1 (DMT1), entering the labile iron pool for use in processes such as synthesis or storage. Meanwhile, the apo-transferrin (iron-free) remains bound to TfR and is recycled back to the cell surface via recycling endosomes, where at neutral pH it dissociates to allow TfR reuse. This cycle enables efficient iron delivery without accumulating free intracellularly. TfR expression and density play a critical role in systemic and cellular iron by inversely correlating with intracellular iron levels: high TfR density occurs during to enhance uptake, while low density in reduces acquisition to prevent toxicity from free iron, which can generate . This regulation supports safe iron storage in , buffering excess to avoid cellular damage while ensuring availability for essential functions. In humans, the TfR pathway accounts for the majority of cellular iron acquisition, with a daily systemic flux of approximately 20-25 mg to meet demands like .

Specific roles of each receptor type

Transferrin receptor 1 (TfR1) predominates in rapidly proliferating cells, such as erythroblasts during and various tumor cells, where it supports high iron demands for and . In addition to facilitating transferrin-bound iron uptake, TfR1 binds H-ferritin, enabling its and lysosomal , which modulates cellular iron availability and indirectly influences iron export pathways by altering intracellular iron pools. Furthermore, TfR1 serves as an entry receptor for pathogens, including merozoites, which exploit it for invasion of reticulocytes during infection. Transferrin receptor 2 (TfR2), primarily expressed in hepatocytes, functions as a key sensor of circulating iron levels, binding holo-transferrin to detect systemic iron status. It interacts with the to form a that, in low-iron states characterized by reduced holo-transferrin saturation, destabilizes and suppresses expression, thereby promoting intestinal iron absorption to restore iron balance. In hepatocytes, TfR2 also contributes to targeted iron delivery to mitochondria, supporting biosynthesis and by channeling transferrin-derived iron through endosomal pathways. Transferrin receptor 3 (TfR3), identified as the moonlighting function of glyceraldehyde-3-phosphate dehydrogenase (GAPDH), is induced under hypoxic conditions to enable rapid scavenging of transferrin-bound iron as an adaptive response to oxygen deprivation. This receptor is internalized via multiple routes, including and , particularly during cellular stress, allowing swift iron acquisition without relying on receptors. In cancer cells, TfR3 links glycolytic flux to , as GAPDH's enhances iron uptake to and proliferation under hypoxic tumor microenvironments. Transferrin receptor 4 (TfR4), corresponding to cubilin, plays a specialized role in the renal proximal tubules by mediating reabsorption of filtered transferrin-iron complexes, thereby preventing urinary loss of this essential resource. This process is crucial for conserving iron during filtration in the , maintaining systemic iron . of cubilin-bound transferrin-iron depends on its co-receptor megalin, which facilitates apical uptake and trafficking to lysosomes for iron release and recycling in cells.

Regulation

Transcriptional regulation

The transcriptional regulation of transferrin receptors (TfRs) is mediated by specific promoter elements and transcription factors that respond to environmental cues such as , cell type, and developmental stage. For TfR1 (encoded by TFRC), the promoter contains hypoxia-inducible factor-1α (HIF-1α) binding sites, enabling induction under hypoxic conditions to enhance iron uptake in low-oxygen environments. In erythroid cells, GATA-1 and GATA-2 transcription factors bind to regulatory regions of the TFRC promoter, driving expression during as part of the broader erythroid gene program. Iron excess can repress TFRC transcription through mechanisms involving upstream stimulatory factor (USF) and Sp1 sites in the promoter, although this is secondary to post-transcriptional controls. TfR2 (encoded by TFR2) expression is driven by GATA-1 binding to consensus sequences in its promoter during erythroid differentiation. In , TfR2 transcription is regulated by hepatocyte nuclear factor 4α (HNF4α), which maintains basal expression and responds to iron signals for systemic . Unlike TfR1, TfR2 transcription is linked to the , peaking during S-phase, and is not directly responsive to iron levels. TfR3, identified as glyceraldehyde-3-phosphate dehydrogenase (GAPDH) in contexts of iron acquisition, is upregulated by HIF-1 under via hypoxia-response elements in its promoter, promoting glycolytic adaptation and iron-related functions. Its expression is constitutive in glycolytic tissues, reflecting its housekeeping role. TfR4, corresponding to cubilin, exhibits proximal tubule-specific expression driven by hepatocyte nuclear factor-1α (HNF-1α) binding to its promoter, essential for endocytic functions in the . Cubilin transcription is modulated by vitamin D response elements, linking it to metabolism and reabsorption processes. In general, TfR gene expression is influenced by chromatin accessibility modulated by iron status, with iron deficiency promoting histone acetylation at TFRC regulatory regions to enhance accessibility for transcription factors. These transcriptional controls complement post-transcriptional mechanisms for fine-tuning receptor levels.

Post-transcriptional regulation

The post-transcriptional regulation of transferrin receptor 1 (TfR1), encoded by the TFRC gene, is predominantly controlled by the iron-responsive element (IRE)/iron regulatory protein (IRP) system at the mRNA level. The TFRC mRNA features five IREs in its 3' untranslated region (UTR), which serve as binding sites for IRP1 and IRP2 under low intracellular iron conditions. Binding of these IRPs to the IREs shields the mRNA from endonucleolytic cleavage, thereby stabilizing it and promoting TfR1 synthesis to facilitate increased iron import. In contrast, high iron levels prevent IRP binding—through Fe-S cluster assembly in IRP1 and IRP2 degradation—resulting in mRNA destabilization and rapid turnover. This dynamic control leads to a marked variation in TFRC mRNA half-life, typically ranging from about 45–60 minutes in iron-replete cells to over 10 hours in iron-deficient states. The underlying iron regulatory loop can be expressed as the IRP binding affinity being inversely proportional to Fe-S cluster assembly: [\text{IRP binding affinity}] \propto \frac{1}{[\text{Fe-S cluster assembly}]} This relationship directly governs the observed changes in mRNA and TfR1 expression. Transferrin receptor 2 (TfR2), encoded by the TFR2 , lacks IREs in its mRNA and thus escapes IRE/IRP-mediated , relying instead on protein-level post-transcriptional mechanisms for regulation. TfR2 undergoes lysosomal degradation via the multivesicular body pathway, facilitated by its interaction with the CD81, which promotes sorting and reduces surface receptor levels; siRNA-mediated knockdown of CD81 extends TfR2 half-life and elevates its expression. This degradation occurs independently of detectable ubiquitination, distinguishing it from other turnover pathways. Palmitoylation further supports TfR2 membrane anchoring and , though its precise role in iron-responsive dynamics requires additional investigation. TfR3, alternatively functioning through glyceraldehyde-3-phosphate (GAPDH), exhibits post-translational regulation involving iron-dependent membrane recruitment during depletion states, enabling it to bind and support auxiliary iron uptake when primary receptors are limited. events, such as those mediated by or AMPK, modulate GAPDH's subcellular localization, including facilitation of nuclear export to enhance its cytosolic and membrane availability under low-iron conditions. TfR4, represented by the cubilin-amnionless (AMN) complex, undergoes post-translational trafficking regulation in renal proximal tubules to mediate transferrin endocytosis. Phosphorylation of AMN influences the complex's membrane targeting and endocytic efficiency, with iron excess suppressing endocytosis to curtail uptake and maintain homeostasis. This mechanism ensures context-specific iron handling, particularly in epithelial tissues.

Physiological roles

Role in erythropoiesis

The transferrin receptor 1 (TfR1) plays a pivotal role in by mediating the uptake of iron-bound , which is essential for production in developing erythroid cells. In reticulocytes, TfR1 is expressed at approximately 400,000 copies per cell, enabling these cells to acquire the vast majority of iron required for synthesis. This high expression level underscores TfR1's dominance in coordinating iron delivery, as the majority of the iron in erythroid cells is directed toward assembly, the primary oxygen-carrying protein in red blood cells. Without sufficient TfR1-mediated iron import, erythroid maturation is severely impaired, highlighting its indispensable function in this process. The iron acquired through TfR1 supports heme biosynthesis by providing ferrous iron to the mitochondrial enzyme 5-aminolevulinate synthase 2 (ALAS2), the rate-limiting step in the erythroid-specific heme pathway. Upon endocytosis of the transferrin-TfR1 complex in early endosomes, iron is released under acidic conditions and transported to mitochondria via mechanisms such as the "kiss-and-run" pathway, where endosomes transiently fuse with the mitochondrial surface to deliver iron directly. TfR1 exhibits a high recycling efficiency in erythroid cells, allowing rapid and repeated iron acquisition to meet the intense demands of hemoglobin synthesis during a short developmental window. TfR1 expression varies across erythroid developmental stages, peaking in basophilic erythroblasts where iron requirements are maximal for early accumulation. As erythroblasts progress to polychromatophilic and orthochromatic stages, TfR1 levels gradually decline, and it is ultimately downregulated and shed during maturation into mature red blood cells, coinciding with enucleation and the cessation of protein synthesis. This downregulation is linked to apoptotic signals that facilitate nuclear extrusion and membrane remodeling, ensuring that mature erythrocytes, which lack organelles, no longer require active iron uptake. Genetic evidence confirms TfR1's critical role, as homozygous of the TFRC in mice results in embryonic around E12.5, primarily due to profound from failed erythrocyte development and insufficient production. These embryos exhibit arrested and neural defects, demonstrating that TfR1-mediated iron delivery is vital from early onward.

Role in tissue-specific iron distribution

Transferrin receptor 2 (TfR2), predominantly expressed in hepatocytes, plays a critical role in hepatic iron sensing by detecting plasma transferrin-iron (Tf-Fe) saturation levels to modulate expression through interactions with HFE protein. This sensing mechanism ensures appropriate hepcidin upregulation in response to elevated iron availability, thereby regulating systemic iron export from enterocytes and macrophages. Additionally, TfR2 facilitates the delivery of iron to hepatocytes, where it is stored primarily as , maintaining the liver's role as a central iron reservoir. In the , TfR4, identified as cubilin in with megalin, is essential for the of nearly 99% of filtered Tf-Fe in the proximal tubules, preventing urinary iron and averting hypoferremia. This process involves , allowing efficient recycling of transferrin-bound iron back into the circulation and supporting overall iron without significant contribution to cellular storage in renal tissue. At the blood-brain barrier, TfR1 enables the transport of iron into the , providing neurons with essential iron for processes such as synthesis in and production via . TfR1 expression on endothelial cells is high, supporting efficient yet controlled iron uptake to meet precise neuronal demands and avoid potential oxidative damage from excess iron. In other tissues, such as , TfR3—associated with (GAPDH)—facilitates rapid Tf-Fe uptake during hypoxic conditions, enabling a swift metabolic response to oxygen deprivation without altering baseline iron distribution. Collectively, these receptor-specific functions establish iron concentration gradients across organs, ensuring targeted allocation for in the liver, in the , controlled entry in the , and adaptive responses in muscle.

Role in diseases

Iron overload and deficiency disorders

The transferrin receptor 2 (TfR2) plays a critical role in hepatic iron sensing and regulation, and its mutations are associated with type 3 hereditary hemochromatosis, a subtype characterized by progressive primarily in the liver. For instance, the Y250X truncation mutation in the TFR2 gene disrupts TfR2 function, leading to impaired expression and excessive intestinal iron absorption, resulting in hepatic iron accumulation and potential . This mutation has been identified in families with early-onset hemochromatosis, confirming its causal role through mouse models that recapitulate the phenotype. Similarly, defects in the interaction between HFE protein and transferrin receptor 1 (TfR1) contribute to the more common type 1 hereditary hemochromatosis; HFE normally competes with for TfR1 binding to modulate iron uptake signaling, but common mutations like C282Y prevent this interaction, reducing levels and promoting systemic . These disruptions highlight how TfR1 and TfR2 facilitate by linking to regulatory pathways. In (ACD), also known as , inflammatory cytokines such as IFN-γ and LPS downregulate TfR1 expression on macrophages, contributing to iron sequestration within these cells and limiting iron availability for . This downregulation, combined with cytokine-induced upregulation (primarily via IL-6), degrades on macrophages, further trapping recycled iron and exacerbating the hypoferremic state typical of ACD. The resulting functional impairs synthesis despite adequate total body iron stores, distinguishing ACD from absolute . Cubilin, a multi-ligand endocytic receptor in renal proximal tubules, mediates the of filtered ; its deficiency underlies Imerslund-Gräsbeck syndrome, a rare autosomal recessive disorder featuring selective , , and due to impaired of low-molecular-weight proteins including . The involves low-molecular-weight proteins including , but the primary hematologic manifestation is due to . Studies have linked rare germline variants in the (encoding TfR1) to congenital , characterized by hypochromic red cells and partial resistance to iron therapy, expanding the spectrum of transferrin receptor-related iron disorders.

Involvement in cancer and infections

The transferrin receptor 1 (TfR1) is frequently overexpressed in malignant cells to meet the elevated iron demands associated with rapid proliferation. This upregulation occurs in a wide range of solid tumors, including and cancers, where TfR1 expression supports and survival. High levels of TfR1 have been linked to poor clinical outcomes, such as reduced overall survival in patients with and . In cancer, TfR1 contributes to tumor progression beyond iron transport by influencing vascularization. Elevated TfR1 expression correlates with increased (VEGF) levels, promoting and in aggressive tumors like . This association underscores TfR1's role in creating a supportive microenvironment for cancer expansion. TfR1 also serves as an entry receptor exploited by certain pathogens during infections. For , the viral (HA) protein binds to sialic acids on TfR1, facilitating and uncoating within cells. Bacterial pathogens such as Neisseria meningitidis and Neisseria gonorrhoeae bind directly to using their own transferrin-binding proteins (TbpA and TbpB) to acquire iron essential for survival. Similarly, the parasite Plasmodium vivax (closely related to P. falciparum) invades reticulocytes via a TfR1- bridge, where the parasite protein PvRBP2b binds TfR1 to enable cell entry. However, recent studies indicate that this interaction may not be essential for invasion by all strains, suggesting alternative pathways. Pathogens hijack the TfR1 machinery to enhance nutrient acquisition and invasion efficiency. In , P. vivax exploits this pathway to increase iron uptake during targeting, thereby supporting parasite replication. These mechanisms allow infectious agents to subvert host iron for their benefit. Studies have highlighted the role of transferrin receptor 2 (TfR2) in inflammatory conditions, particularly in macrophages during . Deletion of TfR2 in macrophages promotes pro-inflammatory M1-like , exacerbating joint inflammation and bone erosion in models of , indicating that TfR2 normally suppresses excessive immune activation in these cells.

Therapeutic targeting

Applications in drug delivery

The 1 (TfR1), highly expressed on brain endothelial cells forming the (), serves as a key target for receptor-mediated (RMT) to facilitate the delivery of biologics into the . This endogenous pathway, which normally transports iron-bound across the , can be hijacked by conjugating therapeutic molecules to anti-TfR1 antibodies or itself, enabling enhanced penetration without disrupting iron homeostasis. Pioneering work utilized the OX26 monoclonal antibody, which binds rat TfR1 without competing with transferrin, to mediate transcytosis of fused therapeutics in preclinical models. Similarly, transferrin conjugates have been employed to shuttle enzymes and antibodies across the BBB. Recent 2025 advances incorporate bispecific antibodies targeting both TfR1 and CD98hc, a BBB-enriched transmembrane protein, resulting in faster initial uptake via TfR1 and prolonged brain retention through CD98hc-mediated stabilization, achieving up to several-fold higher parenchymal exposure compared to monovalent TfR1 binders. The underlying mechanism involves saturable , reflecting the limited receptor capacity at the . To evade lysosomal degradation during endosomal trafficking, pH-insensitive mutants of anti-TfR1 antibodies have been engineered, maintaining binding stability in acidic compartments and promoting apical release into the rather than recycling or degradation. Representative examples include the ch128.1 IgG3 anti-TfR1 conjugated to anti-amyloid-β therapeutics, which demonstrated a 10-fold increase in uptake for models, enhancing amyloid clearance without peripheral toxicity. For , H-ferritin nanocages, which naturally bind TfR1 via their ferroxidase domain, have encapsulated siRNA or components, enabling efficient BBB crossing and targeted delivery and . Key challenges include sink" effect, where high-affinity binders sequester on TfR1-expressing erythrocytes and other peripheral tissues, reducing BBB availability. 2025 studies emphasize optimizing affinity with dissociation constants (Kd) of 10–100 nM to balance brain uptake efficiency and minimize off-target binding, as affinities below 10 nM exacerbate peripheral sequestration while higher values diminish .

Use in cancer therapies

The transferrin receptor 1 (TfR1), often overexpressed on cancer cells to meet elevated iron demands, serves as a promising target for antibody-drug conjugates (ADCs) in . ADCs leveraging anti-TfR1 monoclonal , such as CX-2029—a probody drug conjugate linking an anti-CD71 to the auristatin derivative (MMAE)—exploit to deliver cytotoxic payloads selectively to tumor cells. In phase I clinical trials, CX-2029 demonstrated antitumor activity in patients with advanced solid tumors, with objective responses observed at doses up to 3 mg/kg; however, the program was terminated in 2023 with no further development. The trial showed a manageable safety profile characterized by primarily hematologic toxicities. Similarly, the humanized anti-TfR1 3TF12, originally developed as a (scFv), has been adapted into bivalent formats and conjugated to payloads for enhanced cytotoxicity against iron-dependent leukemias and lymphomas, inhibiting by blocking binding and promoting receptor internalization. Immunotoxins represent another strategy harnessing TfR1 for cancer , particularly in CD71-positive hematologic malignancies like . These agents fuse anti-TfR1 antibodies or ligands to bacterial toxins, such as truncated , enabling receptor-mediated uptake via the endocytic pathway, followed by toxin translocation to the to inhibit protein synthesis and induce . For instance, TfR-directed immunotoxins have shown potent against primary lymphoid malignant cells, including Burkitt's lymphoma lines, with values in the nanomolar range due to efficient and lysosomal escape. Preclinical studies highlight their specificity for proliferating cells overexpressing TfR1, minimizing off-target effects on normal tissues with lower receptor density. Nanocarrier systems, including TfR1-targeted liposomes, enhance the delivery of chemotherapeutic agents like to tumors while reducing systemic toxicity. These liposomes are surface-modified with or anti-TfR1 antibodies to facilitate , leading to 5–10-fold greater accumulation in tumor tissues compared to non-targeted formulations in preclinical models of and cancers. In multidrug-resistant cell lines, such as and SBC-3/ADR, TfR-targeted -loaded liposomes achieved up to 7-fold higher cellular uptake and restored to the , correlating with improved tumor in xenograft models. This approach capitalizes on TfR1's role in rapid , allowing sustained payload release within acidic endosomes. Recent advances in TfR1-targeted therapies include the development of single-domain antibodies (sdAbs) with high affinity and cross-species reactivity, enabling more stable and versatile formats for cancer applications. In 2025 studies, novel sdAbs targeting a unique TfR1 demonstrated superior and delivery in preclinical tumor models, offering potential for bispecific constructs that engage immune effectors. Furthermore, anlotinib, which modulates TfR1-dependent pathways, combined with inhibitors such as anti-PD-1 antibodies, has shown synergistic effects by boosting T-cell infiltration in models, as evidenced by preclinical data where this combination amplified responses. As of 2025, TfR1-targeted therapies remain investigational, with no FDA-approved options for . These innovations underscore TfR1's evolving role in precision , with ongoing trials exploring multimodal regimens.

References

  1. [1]
    Transferrin receptors | Experimental & Molecular Medicine - Nature
    Apr 22, 2025 · The transferrin receptor (TfR) is one of the key proteins involved in cellular iron uptake. TfR-mediated endocytosis of transferrin-bound ...
  2. [2]
    Transferrin: Biology and Use in Receptor-Targeted Nanotherapy of ...
    The main function of Tf is to transport iron in the body and, for the purpose of this review, to transport iron into the brain. Tf is the major source of iron ...
  3. [3]
    Transferrin Receptor - an overview | ScienceDirect Topics
    Transferrin is a vital serum glycoprotein, which is involved in iron homeostasis and the regulation of cell growth. Iron loaded transferrin traverses the cell ...
  4. [4]
    Transferrin receptor 1 - PubMed - NIH
    In succeeding years transferrin receptor 1 was established as a gatekeeper for regulating iron uptake by most cells, and the transferrin-to-cell endocytic ...
  5. [5]
    7037 - Gene ResultTFRC transferrin receptor [ (human)] - NCBI
    Sep 14, 2025 · Authors identified the transferrin receptor-1 (CD71) to be differentially expressed in neural stem cells and differentiated neurons. In this ...
  6. [6]
    7036 - Gene ResultTFR2 transferrin receptor 2 [ (human)] - NCBI
    Aug 19, 2025 · TFR2 maps to chromosome 7. It is an alternately spliced protein and lacks the entire transmembrane and cytoplasmic domains. Has a role ...
  7. [7]
    Characterization of glyceraldehyde-3-phosphate dehydrogenase as ...
    These findings demonstrate that upon iron depletion, GAPDH functions as a preferred receptor for transferrin rather than TfR1 in some but not all cell types.
  8. [8]
    Transferrin receptors - PubMed
    Apr 22, 2025 · Here we focus on recent advances in the studies on TfR and a brief discussion of the structures and functions of four different types of TfR, ...
  9. [9]
    The macrophage cell surface glyceraldehyde-3-phosphate ...
    Mammalian cells utilize transferrin-dependent mechanisms to acquire iron via transferrin receptors 1 and 2 (TfR1 and TfR2) by receptor-mediated endocytosis.
  10. [10]
    Megalin-dependent cubilin-mediated endocytosis is a major ... - PNAS
    In the present study, we report the identification of cubilin as a novel transferrin (Tf) receptor involved in catabolism of Tf. Consistent with a cubilin- ...Missing: TfR4 | Show results with:TfR4
  11. [11]
    Protein moonlighting in iron metabolism: glyceraldehyde-3 ...
    Nov 17, 2014 · Two is company three is a crowd: GAPDH is transferrin-receptor 3 ... transferrin, transferrin receptor 3 (TfR3) · View full article. 558 ...
  12. [12]
  13. [13]
    TFRC - Transferrin receptor protein 1 - Homo sapiens (Human)
    May 30, 2006 · Serum transferrin receptor (sTfR) is used as a means of detecting erythropoietin (EPO) misuse by athletes and as a diagnostic test for anemia.
  14. [14]
    Structure prediction of transferrin receptor protein 1 (TfR1) by ... - NIH
    Jan 29, 2020 · In the present study, we predicted the whole structure of TfR1 using homology modelling, docking, and molecular dynamics simulations.
  15. [15]
    Crystal structure of the ectodomain of human transferrin receptor
    The crystal structure of the dimeric ectodomain of the human TfR, determined here to 3.2 angstroms resolution, reveals a three-domain subunit.Missing: 1A3B seminal paper
  16. [16]
    Antibodies Targeting the Transferrin Receptor 1 (TfR1) as Direct Anti ...
    Mar 16, 2021 · There are two types of transferrin receptors: TfR1, also known as cluster of differentiation 71 (CD71), which is widely expressed and binds Tf ...
  17. [17]
    The critical glycosylation site of human transferrin receptor contains ...
    The critical glycosylation site of human transferrin receptor, at Asn-727, contains high-mannose oligosaccharides, vital for proper function.Missing: 45 65
  18. [18]
    Structure of the Human Transferrin Receptor-Transferrin Complex
    The structure of TfR-Tf complex helps account for known differences in the iron-release properties of free and receptor bound Tf.
  19. [19]
    1CX8: CRYSTAL STRUCTURE OF THE ECTODOMAIN OF HUMAN ...
    Sep 9, 1999 · The transferrin receptor (TfR) undergoes multiple rounds of clathrin-mediated endocytosis and reemergence at the cell surface, ...Missing: 1A3B seminal paper
  20. [20]
    1SUV: Structure of Human Transferrin Receptor-Transferrin Complex
    The structure of TfR-Tf complex helps account for known differences in the iron-release properties of free and receptor bound Tf.Missing: glycosylation seminal papers
  21. [21]
    Structure of the human transferrin receptor-transferrin complex
    The structure of TfR-Tf complex helps account for known differences in the iron-release properties of free and receptor bound Tf.
  22. [22]
    Transferrin receptors - PMC - NIH
    Apr 22, 2025 · Transferrin receptors (TfR) are key proteins involved in cellular iron uptake, with four types: TfR1, TfR2, TfR3, and TfR4.
  23. [23]
    The Functional Versatility of Transferrin Receptor 2 and Its ... - MDPI
    Tfr2α cDNA is 2.3 kb long (AF067864), and the Tfr2α is a protein of about 89 kDa encompassing 801 amino acids [2]. As Tfr1, Tfr2α has a short cytoplasmic tail ( ...Missing: truncated | Show results with:truncated
  24. [24]
    Protein moonlighting in iron metabolism: Glyceraldehyde-3 ...
    Aug 5, 2025 · Mammalian GAPDH is a~150 kDa glycolytic enzyme composed of four identical 37 kDa subunits 88 with approximately 2,000,000 molecules per cell and ...
  25. [25]
    1U8F: Crystal Structure Of Human Placental Glyceraldehyde-3 ...
    Aug 16, 2005 · 1U8F is the crystal structure of human placental Glyceraldehyde-3-Phosphate Dehydrogenase (GAPDH) at 1.75 Å resolution, determined by X-RAY ...
  26. [26]
    The Macrophage Cell Surface Glyceraldehyde-3-phosphate ...
    Mammalian cells utilize transferrin-dependent mechanisms to acquire iron via transferrin receptors 1 and 2 (TfR1 and TfR2) by receptor-mediated endocytosis.
  27. [27]
    Cubilin, the Intrinsic Factor-Vitamin B12 Receptor in Development ...
    Cubilin is a peripheral membrane protein consisting of 8 Epidermal Growth Factor (EGF)- ... CUB (defined as Complement C1r/C1s, Uegf, BMP1) domains. This ...
  28. [28]
    Structural assembly of the megadalton-sized receptor for intestinal ...
    Dec 6, 2018 · In cubilin, consensus Ca2+-binding sites are found in 13 of the 27 CUB-domains (domains 2, 5–9, 11–14, 20, 24 and 26). Several of these domains ...Missing: weight | Show results with:weight<|separator|>
  29. [29]
    Structural assembly of the megadalton-sized receptor for intestinal ...
    Dec 6, 2018 · Cubilin and the transmembrane protein amnionless (AMN) form the endocytic receptor cubam that is essential for intestinal vitamin B12 uptake.
  30. [30]
    The Intracellular Trafficking Pathway of Transferrin - PMC
    Iron-loaded Tf first binds to the Tf receptor (TfR) and enters the cell through clathrin-mediated endocytosis. Inside the cell, Tf is trafficked to early ...
  31. [31]
    Transcriptional regulation by iron of the gene for the transferrin ... - NIH
    Increasing intracellular iron levels by treatment of cells with either human diferric transferrin or hemin lowered the level of the transferrin receptors.
  32. [32]
    Biochemistry, Transferrin - StatPearls - NCBI Bookshelf - NIH
    Jul 7, 2025 · Transferrin is a blood plasma glycoprotein that plays a central role in iron metabolism by delivering ferric ions to various tissues, such as ...
  33. [33]
    Iron metabolism: current facts and future directions - PMC
    Transferrin receptor 2 is crucial for iron sensing in human hepatocytes. Am J Physiol Gastroin-test Liver Physiol. 2010;299:G778–83. doi: 10.1152/ajpgi ...
  34. [34]
    Transferrin receptor 1 in cancer: a new sight for cancer therapy - PMC
    This paper reviewed the research progresses of the roles of TFR1 in the tumorigenesis and cancer progression, the regulations of TFR1, and the therapeutic ...<|control11|><|separator|>
  35. [35]
    Binding and uptake of H-ferritin are mediated by human transferrin ...
    TfR1 accounts for most of the binding of HFt to all cell lines that we have studied, as well as to mitogen-stimulated blood lymphocytes and fresh reticulocytes.
  36. [36]
    Transferrin receptor 1 is a reticulocyte-specific receptor for ... - Science
    Jan 5, 2018 · We identified transferrin receptor 1 (TfR1) as the receptor for P. vivax reticulocyte-binding protein 2b (PvRBP2b).Transferrin Receptor 1 Is A... · Vivax Malaria Host Receptor · Deletions In Tfr1 Generated...
  37. [37]
    The role of hepatic transferrin receptor 2 in the regulation of iron ...
    The binding sites of HFE and Tf do not appear to overlap in TfR2 (Chen et al., 2007). This lends itself to the hypothesis that Tf-binding to TfR1 releases HFE, ...Missing: bonds | Show results with:bonds
  38. [38]
    Iron Sensing as a Partnership: HFE and Transferrin Receptor 2
    Mar 4, 2009 · The discovery that HFE can also interact with TfR2 led to the proposal that an HFE-TfR2 complex may be involved in this hepcidin upregulation.Missing: hepatic | Show results with:hepatic
  39. [39]
    A novel transferrin/TfR2-mediated mitochondrial iron ... - PubMed - NIH
    Thus, we have discovered a novel mitochondrial iron transport system that goes awry in PD, and which may provide a new target for therapeutic intervention.Missing: liver | Show results with:liver
  40. [40]
    Moonlighting Protein Glyceraldehyde-3-Phosphate Dehydrogenase
    Our results suggest the role of GAPDH-mediated Tf uptake as a rapid response mechanism by which cells acquire iron during the early stages of hypoxia.Missing: TfR3 inducible scavenging
  41. [41]
    Characterization of glyceraldehyde-3-phosphate dehydrogenase as ...
    These findings demonstrate that upon iron depletion, GAPDH functions as a preferred receptor for transferrin rather than TfR1 in some but not all cell types. We ...<|separator|>
  42. [42]
    Proximal tubule transferrin uptake is modulated by cellular iron and ...
    Apr 26, 2019 · Proximal tubule transferrin uptake is modulated by cellular iron and mediated by apical membrane megalin-cubilin complex and transferrin ...
  43. [43]
    Transferrin receptor induction by hypoxia. HIF-1-mediated ... - PubMed
    The expression of transferrin receptor (TfR), which mediates cellular iron uptake, is regulated by hypoxia.
  44. [44]
    Iron deficiency can upregulate expression of transferrin receptor at ...
    Expression of TfR in cells placed in an iron-deficient milieu can increase by a mechanism which is independent of the IREs of TfR.Missing: chromatin accessibility histone acetylation
  45. [45]
    Regulation of expression of murine transferrin receptor 2
    During liver development, TfR2 was up-regulated and TfR1 was down-regulated. During erythrocytic differentiation of murine erythroleukemia (MEL) cells induced ...
  46. [46]
    Transferrin receptor 2 protein is not expressed in normal erythroid cells
    Unlike TFR1, TFR2 mRNA lacks iron regulatory elements, and TFR2 expression may be regulated by the cell cycle rather than by intracellular iron status [3].
  47. [47]
    Up-regulation of glyceraldehyde-3-phosphate dehydrogenase gene ...
    May 1, 2011 · Here, we report that hypoxia up-regulates the GAPDH gene expression through breast cancer-specific molecular mechanisms in MCF-7 cells.
  48. [48]
    HNF1α drives endocytosis in the proximal tubule - ScienceDirect.com
    Thus, HNF1α plays a key role in the constitutive expression of megalin and cubilin, hence regulating endocytosis in the proximal tubule of the kidney. These ...
  49. [49]
    Role of megalin and cubilin in the metabolism of vitamin D(3)
    Megalin and cubilin, endocytic receptors in proximal tubule cells, are involved in the reabsorption of vitamin D binding protein from glomerular filtrates.Missing: transcriptional HNF-
  50. [50]
    Characterization of K562 cells: uncovering novel chromosomes ...
    Aug 14, 2023 · Histone modifications related to the transferrin receptor gene (TFRC) using ChIP sequencing. To assess K562 and PBMC histone modifications ...<|control11|><|separator|>
  51. [51]
    Regulation of transferrin receptor-1 mRNA by the interplay between ...
    Intracellular iron is tightly regulated by coordinated expression of iron transport and storage genes, such as transferrin receptor-1 (TfR1) and ferritin.
  52. [52]
    Iron metabolism and the IRE/IRP regulatory system: an update
    The mechanism is posttranscriptional and involves two cytoplasmic iron regulatory proteins, IRP1 and IRP2. Under conditions of iron starvation, IRPs stabilize ...
  53. [53]
    Translation and the stability of mRNAs encoding the transferrin ...
    ing this element is iron-regulated (Fig. lA). In cells treated with the iron source hemin, an mRNA half-life of 45-60 min is observed, whereas in cells ...
  54. [54]
    The IRP/IRE system in vivo: insights from mouse models - Frontiers
    Iron regulatory proteins 1 and 2 (IRP1 and IRP2) post-transcriptionally control the expression of several mRNAs encoding proteins of iron, oxygen and energy ...
  55. [55]
    CD81 Promotes Both the Degradation of Transferrin Receptor 2 ...
    Knockdown of CD81 using siRNA significantly increased TfR2 levels by increasing the half-life of TfR2, indicating that CD81 promotes degradation of TfR2.Missing: IRE palmitoylation
  56. [56]
    Transferrin-directed internalization and cycling of transferrin receptor 2
    No ubiquitination of TfR2 by HA-ubiquitin was detected, indicating a lack of direct TfR2 ubiquitination involvement in its degradation. Publication types.
  57. [57]
    Characterization of glyceraldehyde-3-phosphate dehydrogenase as ...
    ... TfR3' 88 to mediate Tf-Fe uptake. The binding of ... Moonlighting Protein Glyceraldehyde-3-Phosphate Dehydrogenase ... Moonlighting glycolytic protein ...
  58. [58]
    Src‐mediated phosphorylation of GAPDH regulates its nuclear ...
    Jun 15, 2020 · Nuclear GAPDH promotes Pol β polymerase activity and increases base excision repair (BER) efficiency. Furthermore, GAPDH knockdown dramatically ...
  59. [59]
    Article AMPK-Dependent Phosphorylation of GAPDH Triggers Sirt1 ...
    Dec 17, 2015 · GAPDH is thus a pivotal and central regulator of autophagy under glucose deficiency, undergoing AMPK-dependent phosphorylation and nuclear translocation.
  60. [60]
    Contribution of Cubilin and Amnionless to Processing and ...
    Apr 24, 2025 · Amnionless (AMN) is a key protein for receptor-mediated endocytosis 13 . ... regulatory pathways, i.e., phosphorylation, glycosylation ...
  61. [61]
    Megalin-dependent cubilin-mediated endocytosis is a major ... - PMC
    Megalin-dependent cubilin-mediated endocytosis is a major pathway for the apical uptake of transferrin in polarized epithelia.Missing: AMN phosphorylation
  62. [62]
    Endocytic Receptors in the Renal Proximal Tubule | Physiology
    Aug 1, 2012 · Furthermore, megalin- and cubilin-mediated reabsorption of filtered 25-OH-vitamin D is essential for the renal hydroxylation of the vitamin into ...Endocytic Receptors In The... · Ligands · Megalin And Cubilin In...Missing: TfR4 | Show results with:TfR4
  63. [63]
    Transferrin receptors and iron uptake during erythroid cell ... - PubMed
    May 7, 1982 · During erythroid cell development the transferrin receptor level increased from about 300,000 per cell at the early normoblast stage to reach a ...
  64. [64]
    Regulation of Heme Synthesis by Mitochondrial Homeostasis Proteins
    ALAS translocation to the mitochondrial matrix is inhibited by high heme conditions and functions as a mechanism of feedback inhibition. ALAS2 turnover is ...<|separator|>
  65. [65]
    Transferrin Receptors in Erythropoiesis - PMC - NIH
    Dec 19, 2020 · TFR1-mediated Tf internalization is classically described as the canonical iron import pathway (Figure 1). TFR1 is ubiquitously expressed but ...
  66. [66]
    Evidence that the expression of transferrin receptor 1 on erythroid ...
    Expression is highest on proerythroblasts through basophilic erythroblasts, decreases with further erythroid differentiation [20], and is upregulated both ...
  67. [67]
    Hepcidin: regulation of the master iron regulator - PMC
    Hepcidin is a hormone that regulates iron by binding to ferroportin, limiting iron release. It is regulated by iron stores, hypoxia, inflammation, and ...
  68. [68]
    Magnetic Resonance Imaging Quantification of Liver Iron - PMC - NIH
    Iron overload is defined as iron that exceeds the upper limit of normal. In HH, the LIC may range up to 10 times the upper limit of normal (20 mg Fe/g dry ...
  69. [69]
    Proximal tubule transferrin uptake is modulated by cellular iron and ...
    Both the MCRC and TfR1 mediate hTf uptake across apical brush border membranes of PTECs and reciprocally respond to decreased cellular iron.
  70. [70]
    Transferrin and transferrin receptor function in brain barrier systems
    The interaction between transferrin and transferrin receptor appears to serve this function in the blood-brain, blood-CSF, and cellular-plasmalemma barriers.
  71. [71]
    Targeting transferrin receptors at the blood-brain barrier improves ...
    Sep 4, 2017 · The transferrin receptor is responsible for the transport of iron ... iron transport at the blood-brain barrier, and neuronal and glial pathology.
  72. [72]
    Transferrin and transferrin receptors update - ScienceDirect.com
    Human TfR2, another receptor for Tf, is predominantly expressed in hepatocytes and erythroid precursors, and holo-Tf dramatically upregulates its expression.
  73. [73]
    Transferrin receptor in primary and metastatic breast cancer
    Dec 20, 2023 · A wide survey shows high expression of transferrin receptor not only in primary breast tumors, but also in refractory/relapsing breast cancer ...
  74. [74]
    Integrated analysis identifies TfR1 as a prognostic biomarker which ...
    Sep 13, 2021 · Emerging evidence indicates that transferrin receptor 1 (TfR1) plays vital roles in regulating cellular iron import. However, the distinct role ...
  75. [75]
    Transferrin receptor-1 and VEGF are prognostic factors for ...
    Sep 4, 2019 · Furthermore, high TfR1 and VEGF expression was significantly correlated to poor overall survival, and both TfR1 and VEGF were independent ...
  76. [76]
    Transferrin receptor-1 and VEGF are prognostic factors for ... - NIH
    TfR1 and VEGF expression was significantly correlated to histologic grade, Enneking stage, and distant metastasis. TfR1 expression was significantly correlated ...
  77. [77]
    Influenza A virus exploits transferrin receptor recycling to enter host ...
    May 16, 2023 · The binding of virions to TfR1 via sialic acids confirmed its role as a directly acting entry factor, but unexpectedly even headless TfR1 ...
  78. [78]
    Influenza A virus exploits transferrin receptor recycling to enter host ...
    This approach identified transferrin receptor 1 (TfR1) as a candidate entry protein. Genetic gain-of-function and loss-of-function experiments, as well as in ...Significance · Abstract · Supporting Information
  79. [79]
    Identification of Transferrin-Binding Domains in TbpB Expressed by ...
    The transferrin iron acquisition system of Neisseria gonorrhoeae is necessary for iron uptake from transferrin in the human host and requires the ...
  80. [80]
    Utility of Hybrid Transferrin Binding Protein Antigens for Protection ...
    Feb 18, 2019 · The surface transferrin receptor proteins from Neisseria gonorrhoeae have been recognized as ideal vaccine targets due to their critical role in survival.
  81. [81]
    Transferrin receptor 1 is a reticulocyte-specific ... - PubMed Central
    We identify transferrin receptor 1 (TfR1) as the receptor for P. vivax reticulocyte-binding protein 2b (PvRBP2b). The structure of the N-terminal domain of ...Missing: per | Show results with:per
  82. [82]
    Deletion of Transferrin Receptor 2 (Tfr2) aggravates inflammation ...
    Aug 9, 2025 · Deletion of Transferrin Receptor 2 (Tfr2) aggravates inflammation and bone erosion by promoting macrophage activation in inflammatory arthritis.
  83. [83]
    Fc-engineered large molecules targeting blood-brain barrier ...
    Feb 20, 2025 · Blood brain barrier-crossing molecules targeting transferrin receptor (TfR) and CD98 heavy chain (CD98hc) are widely reported to promote enhanced brain ...
  84. [84]
    High-affinity transferrin receptor binding improves brain delivery of ...
    Aug 21, 2025 · Transferrin receptor (TfR)-mediated transcytosis is a well-established method for delivering biologic therapeutics and diagnostics to the brain.Missing: TfR3 TfR4
  85. [85]
    Blood‐brain barrier transport using a high affinity, brain‐selective ...
    Nov 25, 2020 · Transferrin receptor 1 (TfR1), which mediates uptake of iron-loaded transferrin (Tf) with subsequent transfer of iron into the brain and the ...
  86. [86]
    Dual targeting of transferrin receptor and CD98hc enhances brain ...
    Aug 26, 2025 · Stronger TfR affinity drives faster brain uptake and catabolism, while stronger CD98hc affinity yields higher, more sustained concentrations, ...
  87. [87]
    Drug Transport across the Blood–Brain Barrier - Sage Journals
    Aug 29, 2012 · Certain small molecule drugs may cross the BBB via lipid-mediated free diffusion, providing the drug has a molecular weight <400 Da and forms <8 hydrogen bonds.Brain Drug Transport And... · Carrier-Mediated Transport... · Receptor-Mediated Transport...<|control11|><|separator|>
  88. [88]
    A pH-sensitive binding modality allows successful transferrin ... - PMC
    Sep 22, 2025 · ... TfR led to lysosomal degradation of the antibody and TfR. The work of Roche researchers went further, demonstrating that a bivalent ...
  89. [89]
    Rational design of engineered H-ferritin nanoparticles with improved ...
    HFn NPs have a unique hollow spherical structure that can encapsulate nucleic acid drugs (NADs) and specifically bind to cancer cells and BBB endothelial cells ...Missing: TfR | Show results with:TfR
  90. [90]
    The effects of dose, valency, and affinity on TfR-mediated brain ...
    Apr 8, 2025 · The aim was to evaluate the independent effects of valency and affinity on TfR-mediated brain delivery at different doses.Recombinant Antibody... · Biacore Affinity Analysis · Pharmacokinetics And Ex Vivo...Missing: sink Kd
  91. [91]
    Identification and optimization of trans-species reactive TfR1-binding ...
    May 23, 2025 · The transferrin receptor 1 (TfR1) is enriched in brain endothelial cells and is one of the most studied receptors for drug delivery to the ...
  92. [92]
    Phase I, First-in-Human Study of the Probody Therapeutic CX-2029 ...
    Aug 15, 2021 · PROCLAIM-CX-2029 is a phase I first-in-human study of CX-2029, a Probody–drug conjugate targeting CD71 (transferrin receptor 1) in adults with advanced solid ...
  93. [93]
    CX-2029, a PROBODY drug conjugate targeting CD71 (transferrin ...
    May 25, 2020 · Background: CX-2029 is a PROBODY drug conjugate (PDC) of MMAE, a potent microtubule inhibitor, directed against CD71 (transferrin receptor ...<|control11|><|separator|>
  94. [94]
    Development of human single-chain antibodies to the transferrin ...
    Jul 1, 2010 · We optimized this activity of 3TF12 and 3GH7 by engineering 55-kDa bivalent antibody formats, namely, F12CH and H7CH, which could block cell ...
  95. [95]
    Cytotoxic Activity of an Anti-Transferrin Receptor Immunotoxin on ...
    Transferrin receptor expression is closely related to the rate of cell proliferation, and conjugates between anti-transferrin receptor monoclonal antibodies and ...
  96. [96]
    Targeted Diphtheria Toxin-Based Therapy: A Review Article - Frontiers
    Of the available cytolethal toxins, diphtheria toxin (DT) is one of the most frequently used for these strategies. Of the many DT-based therapeutic strategies ...
  97. [97]
    Potent in Vitro and in Vivo Effects Against Human Tumors - PubMed
    5E9-gelonin was extremely toxic in vitro to human tumor cell lines expressing the 5E9 antigen, including a Burkitt's lymphoma, an adult T-cell acute lymphocytic ...
  98. [98]
    Anti-Cancer Activity of Doxorubicin-Loaded Liposomes Co-Modified ...
    Similarly, the TfR has been widely exploited for drug delivery into cancer cells as it is overexpressed in many cancer types due to its role in iron homeostasis ...
  99. [99]
    Effect of transferrin receptor-targeted liposomal doxorubicin in P ...
    In this study, we examined whether transferrin receptor (Tf-R) targeted liposomes can efficiently deliver encapsulated doxorubicin (DXR) into MDR cells (SBC-3/ ...
  100. [100]
    Anti-cancer activity of doxorubicin-loaded liposomes co-modified ...
    The dual-targeted liposomes showed a 7-fold increase in cell association compared to either of the single-ligand targeted ones in human cervical carcinoma (HeLa) ...
  101. [101]
    Novel single-domain antibodies targeting a unique transferrin ...
    Oct 17, 2025 · Optimal BBB crossing correlates with fast TfR1 dissociation and moderate affinity. Abstract. The blood-brain barrier (BBB) is a major obstacle ...
  102. [102]
    Anlotinib potentiates anti‐PD1 immunotherapy via transferrin ...
    Aug 2, 2024 · The combination of anlotinib with an anti‐PD‐1 antibody enhanced the immune response against HCC in preclinical models. Anlotinib remarkably ...
  103. [103]
    High transferrin receptor expression marks tumors with increased ...
    May 31, 2023 · We found TFRC-H tumors have increased infiltration by immune effector populations but also express significantly higher levels of immune checkpoint molecules.