Fact-checked by Grok 2 weeks ago

Transferrin

Transferrin is a monomeric found in that binds ferric iron (Fe³⁺) with high , facilitating its transport and delivery to cells throughout the body while maintaining iron and preventing toxicity from free iron. Structurally, human transferrin consists of a single polypeptide chain of approximately 679 , with a molecular weight of about 80 kDa, including two N-linked sites that contribute to its stability and solubility. The protein adopts a bilobal , comprising an amino-terminal lobe (N-lobe, residues 1–331) and a carboxyl-terminal lobe (C-lobe, residues 339–679), connected by a flexible seven-residue linker (residues 332–338). Each lobe is further divided into two subdomains—N1 and N2 for the N-lobe, and C1 and C2 for the C-lobe—forming a cleft in each that accommodates one Fe³⁺ ion, coordinated by specific residues including aspartate, , , and a synergistic anion. In its apo (iron-free) form, the lobes are in an open conformation, which closes upon iron binding to enhance , and this conformational change is critical for receptor interaction. Functionally, transferrin serves as the primary carrier of iron in circulation, binding up to two Fe³⁺ ions per molecule and delivering them via to tissues such as the , liver, and . It interacts with the transferrin receptor 1 (TfR1) on the cell surface, where the holotransferrin-receptor complex is internalized into endosomes; upon acidification ( ~5.5–6.0), iron is released, and the apotransferrin-receptor complex recycles to the plasma membrane for reuse, with a daily turnover rate of approximately 10 times. Beyond transport, transferrin helps sequester free iron to inhibit bacterial growth as part of innate immunity and mitigates by preventing the formation of from unbound iron. In physiological contexts, transferrin is synthesized mainly in the liver and secreted into , where its level (typically 20–45%) reflects systemic iron status: low indicates deficiency, prompting increased production, while high signals overload. Normal serum levels range from 204 to 360 mg/dL, and it is essential for by supplying iron for synthesis in erythroid precursors. Dysregulation of transferrin is associated with disorders like (elevated transferrin) and hereditary hemochromatosis (high ), underscoring its diagnostic value.

Structure

Molecular Composition

Transferrin is a encoded by the TF gene, which is located on the q21-q25 region of human chromosome 3. In humans, the mature protein comprises a single polypeptide chain of 679 , with a molecular weight ranging from 76 to 80 kDa when accounting for post-translational modifications. This weight includes two complex N-linked chains attached at residues 432 (Asn-432) and 630 (Asn-630), which are primarily biantennary structures often terminated with residues. The primary structure of transferrin folds into a bilobal consisting of two homologous domains: the N-terminal lobe (residues 1–331) and the C-terminal lobe (residues 339–679), connected by a flexible seven-residue linker (residues 332–338). Each lobe is further divided into two subdomains (N1/N2 and C1/C2), forming a deep cleft in which iron can bind, coordinated by specific residues including , , , and anion. This reflects an ancient event, resulting in the internal observed between the lobes. The N-linked glycosylation at Asn-432 and Asn-630 is critical for the protein's conformational stability, resistance to , and pharmacokinetic properties. These chains, which constitute about 5-6% of the total mass, shield hydrophobic regions and facilitate proper folding, while their sialylation prevents rapid clearance by hepatic asialoglycoprotein receptors, thereby extending the of transferrin to 7-10 days in circulation.

Iron-Binding Mechanism

Transferrin binds ferric iron (Fe³⁺) at two homologous sites, one in each of its N- and C-terminal lobes, enabling the transport of up to two iron atoms per molecule. The association constant for Fe³⁺ binding at neutral is exceptionally high, ranging from 10²⁰ to 10²² M⁻¹, reflecting the protein's role in sequestering iron with minimal free Fe³⁺ in circulation. This binding is cooperative but site-specific, with the C-lobe site exhibiting slightly higher affinity under physiological conditions. Effective Fe³⁺ coordination requires a synergistic anion, typically (HCO₃⁻), which occupies one coordination position and stabilizes the complex. The iron is bound in a distorted octahedral by four protein ligands per site: the phenolate oxygen atoms from two residues, the carboxylate oxygen from one aspartate residue, the imidazole nitrogen from one residue, and two oxygen atoms from the bidentate anion. Specific residues include Asp63, Tyr95, Tyr188, and His249 in the N-lobe, and Asp392, Tyr426, Tyr517, and His585 in the C-lobe. This arrangement ensures tight, reversible binding while preventing iron-catalyzed oxidative damage. The binding affinity is highly pH-dependent, with maximal stability at physiological 7.4 due to the deprotonated state of coordinating residues. At endosomal values of 5.5–6.0, of key ligands—such as the imidazole and phenolate groups—induces conformational changes that weaken the complex, facilitating iron release. These shifts involve opening of the interdomain cleft in each lobe, transitioning from the closed, iron-bound (holo) conformation to the more open, iron-free (apo) form. The affinity is quantified by the association constant K_a = \frac{[\ce{Tf-Fe}]}{[\ce{Tf}][\ce{Fe^3+}] }, where represents apotransferrin, and values differ markedly between apo- and holo-forms due to cooperative effects and . At 7.4, K_a for the first Fe³⁺ (to apo-Tf) is approximately 10²⁰.⁷ M⁻¹ for the C-lobe site, while the second site (monoferric to diferric) is around 10¹⁹.⁴ M⁻¹; these drop by orders of magnitude below 6. Spectroscopic studies confirm iron binding through characteristic ligand-to-metal charge transfer (LMCT) bands in the UV-Vis spectrum. Upon Fe³⁺ coordination, apotransferrin exhibits absorbance shifts with a broad peak at 430–470 nm (maximum ~465 nm), arising from phenolate-to-Fe³⁺ transitions, which are absent in the iron-free form. These spectral changes provide direct evidence of site occupancy and are used to monitor binding kinetics.

Receptor Interactions

Transferrin primarily interacts with two cell surface receptors: transferrin receptor 1 (TfR1) and transferrin receptor 2 (TfR2), both of which are type II transmembrane glycoproteins that function as disulfide-linked homodimers. TfR1 is ubiquitously expressed across cell types, with particularly high levels in iron-demanding tissues such as erythroid precursors, while TfR2 expression is predominantly restricted to hepatocytes and erythroid precursors. These receptors facilitate the specific recognition and uptake of holo-transferrin (iron-loaded transferrin), with TfR1 exhibiting a higher for holo-transferrin (K_d ≈ 1 at neutral ) compared to TfR2 (K_d ≈ 25-30 ). The binding of holo-transferrin to these receptors occurs with high specificity, primarily involving the C-terminal lobe of transferrin, which makes extensive contacts with the helical domain of the receptor ectodomain. This interaction is pH-dependent: at the cell surface (pH ≈ 7.4), holo-transferrin binds tightly, but decreases for apo-transferrin (iron-free), ensuring selective of iron-loaded forms. Structural studies, such as the cryo-EM-derived model of the TfR1-transferrin (PDB: 1SUV), reveal that upon binding, the N-lobe of transferrin shifts approximately 9 relative to the C-lobe, positioning the C-lobe to interact closely with the receptor and facilitating subsequent iron release. In contrast, TfR2 employs a distinct binding mechanism with reliance on non-conserved residues at the apical arm junction, resulting in weaker stabilization of holo-transferrin at neutral pH but enhanced in acidic environments. Following binding, the transferrin-receptor complex undergoes primarily through clathrin-coated pits, internalizing the complex into early s. Within the , the drops to approximately 5.5-6.0, triggering a conformational change in transferrin that promotes iron release from both lobes, while the apo-transferrin remains bound to the receptor with higher at this acidic . The released iron is then reduced (Fe³⁺ to Fe²⁺) and transported into the via transporters like DMT1, after which the apo-transferrin-receptor complex recycles back to the surface for and reuse. This process is highly efficient, with TfR1 supporting rapid cycling (half-time ≈ 7-10 minutes), whereas TfR2 exhibits slower internalization kinetics. TfR2's liver-specific expression positions it as a key sensor of systemic iron levels, where it interacts with holo-transferrin saturation to modulate expression through complexes involving HFE, thereby influencing overall iron homeostasis without direct involvement in bulk iron uptake. Mutations in TfR2, as seen in type III hereditary hemochromatosis, disrupt this sensing function, leading to dysregulated and .

Occurrence and Synthesis

In Humans and Mammals

In humans, transferrin is primarily synthesized by hepatocytes in the liver, which accounts for the majority of transferrin . Secondary sites of synthesis include the epithelial cells and Sertoli cells in the testes, contributing smaller amounts to local or systemic pools. These hepatic and extrahepatic sources ensure the protein's availability for iron transport throughout the body. Serum transferrin concentrations in healthy adults typically range from 2.0 to 3.6 g/L, with iron levels between 20% and 45% under normal physiological conditions. These values reflect the protein's role in maintaining balanced iron distribution, with variations influenced by nutritional status and liver function. Developmentally, transferrin levels are low in the and at birth, gradually increasing postnatally to reach adult concentrations by late infancy or . Sex-based differences in serum transferrin are minimal in non-pregnant individuals, though levels rise during due to estrogen-mediated hepatic stimulation. Across mammals, transferrin serves as the principal circulating iron-binding , with close homologs such as present in exocrine secretions and to support neonatal iron acquisition. Genetic variations in the , which encodes transferrin, significantly influence levels; for instance, common polymorphisms account for up to 40% of the variability in circulating transferrin concentrations among individuals.

In Other Vertebrates and Species

Transferrin is highly conserved across all vertebrates, serving as a key iron-transport protein with adaptations to specific physiological needs. In fish, such as Atlantic salmon (Salmo salar), multiple transferrin isoforms exist, with differential expression observed during smoltification—a process involving osmoregulatory transitions from freshwater to seawater environments—suggesting roles in supporting ion balance and iron homeostasis under varying salinity conditions. In birds, ovotransferrin, the avian homolog, is prominently expressed in egg white, where it binds iron to provide a protected supply for embryonic development, preventing microbial growth while ensuring nutrient availability during incubation. The evolutionary origins of transferrin trace back to a gene duplication event in a common ancestor approximately 500–700 million years ago, resulting in the characteristic bilobal structure capable of binding two iron ions. This duplication preceded the divergence of s, with sequence identity between and mammalian transferrins typically ranging from 50% to 60%, reflecting conserved iron-binding motifs amid species-specific divergences. Phylogenetic analyses indicate multiple internal duplications within the transferrin family, contributing to subfamilies like serotransferrin and ovotransferrin across vertebrate lineages. In non-vertebrate species, transferrin analogs exist but often lack the full iron-binding capacity of vertebrate forms. Insects, such as Drosophila melanogaster and Manduca sexta, express monolobal transferrin-1 proteins that bind only one ferric ion with high affinity (log K' ≈ 18), contrasting with the bilobal vertebrate transferrins that accommodate two ions; these insect variants support iron transport and nutritional immunity but with reduced capacity. Melanotransferrin, a related single-lobe homolog found in some invertebrates and vertebrate melanoma cells, similarly exhibits limited iron-binding functionality, primarily aiding in cellular iron sequestration rather than systemic transport. Species-specific adaptations further diversify transferrin function. In reptiles, such as crocodiles and , transferrins display conserved N-glycosylation sites—often more pronounced than in some mammals—which enhance protein stability and resistance to , aiding iron in diverse environmental conditions. These glycosylation patterns are evident in sequences from species like the (Crocodylus niloticus) and bearded dragon (Pogona vitticeps), where they support ovotransferrin roles in egg similar to .

Physiological Functions

Iron Homeostasis and Transport

Transferrin serves as the primary plasma protein responsible for transporting iron from sites of absorption, such as the duodenal enterocytes, to sites of storage in the liver or utilization in tissues like the bone marrow and muscles, ensuring iron is delivered in a safe, ferric (Fe³⁺) form to prevent the toxicity associated with free ferrous (Fe²⁺) ions. Free Fe²⁺ can catalyze Fenton reactions, generating highly reactive hydroxyl radicals that damage cellular components including lipids, proteins, and DNA, leading to oxidative stress and potential cell death. By binding up to two Fe³⁺ atoms per molecule with extremely high affinity, transferrin maintains iron in a non-toxic, soluble state during circulation, minimizing the risk of such deleterious reactions. The systemic balance of iron is tightly regulated by the hormone , which modulates levels through its action on , the iron exporter on enterocytes, macrophages, and hepatocytes. When is elevated due to high iron levels, expression increases in the liver, binding to and inducing its , thereby suppressing iron export into the to limit further increases in and prevent . This feedback mechanism prevents by limiting and while coordinating with transferrin to distribute existing iron pools efficiently. In adults, transferrin facilitates a daily iron flux of approximately 20-25 mg through from senescent red blood cells by macrophages, far exceeding the less than 1-2 mg typically absorbed from the diet to replace basal losses. Transferrin exists in equilibrium between its apo form (iron-free) and holo form (iron-bound), with normal saturation maintained at around 30% to balance availability and prevent deficiency or overload; this level ensures sufficient diferric transferrin (about 10%) for delivery while keeping excess apo-transferrin (about 50%) available for binding newly released iron. This equilibrium is supported by coordination with , a ferroxidase that oxidizes absorbed Fe²⁺ to Fe³⁺ at sites, enabling its loading onto transferrin for safe transport. Such integration underscores transferrin's central role in maintaining iron across the body.

Role in Erythropoiesis

Transferrin plays a critical role in by delivering iron to developing erythroblasts, where transferrin receptor 1 (TfR1) is highly expressed to facilitate rapid iron uptake essential for synthesis. Erythroblasts, the precursors to mature red blood cells, exhibit one of the highest levels of TfR1 expression among mammalian cells, enabling the efficient of diferric transferrin and subsequent iron release in endosomes for transport to mitochondria. This process supports the incorporation of iron into , which accounts for approximately 80% of the body's total iron content, ensuring adequate oxygen-carrying capacity in erythrocytes. During the proliferation and differentiation stages of erythropoiesis, transferrin-bound iron is vital for heme biosynthesis, particularly through the action of ferrochelatase, the terminal enzyme in the heme pathway that inserts ferrous iron into protoporphyrin IX to form heme. Iron acquired via TfR1-mediated uptake is directed to mitochondria, where it integrates into the heme production machinery, supporting erythroid cell maturation from proerythroblasts to reticulocytes. This iron delivery is tightly coordinated with erythropoietin signaling, which enhances TfR1 expression to meet the demands of expanding erythroid populations. Erythropoiesis requires about 20-25 mg of iron per day in adults to sustain daily red blood cell production, with transferrin serving as the primary carrier to meet this demand. In response to erythropoietin stimulation, which drives increased erythropoietic activity, transferrin saturation rises as hepcidin is suppressed, promoting iron mobilization from stores and enhancing diferric transferrin availability for erythroid uptake. This dynamic adjustment ensures sufficient iron flux to the bone marrow without depleting systemic reserves. Disruptions in transferrin function impair iron delivery to erythroblasts, limiting heme synthesis and resulting in characterized by small, pale red blood cells. Low transferrin levels, as seen in hypotransferrinemia, reduce iron transport to the , leading to ineffective and iron overload in non-erythroid tissues due to compensatory mechanisms. TfR1 genetic variants contribute to congenital dyserythropoietic anemias by disrupting iron uptake and erythroid maturation. In fetuses, iron demand for is markedly higher than in adults, reflecting rapid hematopoietic expansion, with placental transferrin receptors playing a key role in maternal-fetal iron transfer. The layer of the expresses abundant TfR1 on the maternal-facing surface to capture circulating diferric transferrin, facilitating iron export to the for hemoglobin production in developing erythroid cells. This specialized transport ensures fetal iron sufficiency despite limited stores.

Immune System Involvement

Innate Immunity and Iron Withholding

Transferrin contributes to innate immunity through nutritional immunity, a host defense strategy that sequesters essential trace metals, particularly iron, to starve invading pathogens of this critical nutrient. In the bloodstream, the apo form of transferrin—the iron-free variant—binds free ferric iron (Fe³⁺) with exceptionally high affinity, maintaining plasma free iron concentrations at approximately 10⁻²⁴ M, which is orders of magnitude below the femtomolar levels (around 10⁻¹⁵ M) that many require for growth even with their siderophore-mediated acquisition systems. This prevents pathogens from accessing iron, inhibiting their and ; for instance, early observations in the demonstrated that transferrin in human serum directly limits microbial growth by withholding iron. During , host mechanisms further enhance this effect by reducing transferrin iron saturation, increasing apo-transferrin availability to bind and neutralize any released iron. Transferrin's iron-withholding function synergizes with that of , another transferrin family member, to provide compartmentalized protection against . While transferrin predominates in serum and extracellular fluids to block access by circulating , is secreted at mucosal barriers, such as in respiratory and gastrointestinal tracts, where it binds iron under acidic conditions to further restrict microbial iron uptake and disrupt pathogen membranes. This division of labor ensures comprehensive iron deprivation across host environments, enhancing overall defense without overlapping redundantly. The of transferrin's high-affinity iron-binding sites represents an adaptive response to infectious pressures, with molecular evidence of positive selection in at residues interfacing with bacterial transferrin-binding (TbpA). This rapid counters strategies for iron , such as receptor-mediated iron extraction, embodying a Red Queen-like where host sequestration drives microbial adaptations and vice versa over millions of years. Such evolutionary dynamics underscore transferrin's role as a frontline innate immune effector tailored to limit iron during . Experimental models confirm transferrin's protective function, as deficiencies lead to heightened bacterial virulence. In , transferrin 1 mutants exhibit increased susceptibility to systemic bacterial infections, with elevated pathogen burdens and reduced survival rates due to impaired iron sequestration. Similarly, mammalian studies using transgenic mice expressing human transferrin reveal that disruptions in transferrin-mediated iron handling exacerbate sepsis-like conditions from pathogens like , demonstrating direct impacts on infection outcomes. Recent investigations into viral contexts, including 2023–2024 analyses of , show that low transferrin levels correlate with greater disease severity, higher inflammatory markers, and increased mortality risk, suggesting broadened relevance in iron withholding against non-bacterial threats.

Response to Inflammation

Transferrin functions as a negative , exhibiting reduced serum levels during inflammatory responses primarily due to suppression of its hepatic by pro-inflammatory cytokines, including interleukin-6 (IL-6). This downregulation leads to a rapid decrease in circulating transferrin concentrations by 20-30% within hours of onset, limiting iron availability in the . Inflammatory conditions trigger iron redistribution from circulation to macrophages via hepcidin, a induced by IL-6, which promotes ferroportin degradation and sequesters iron intracellularly, thereby inducing hypoferremia to inhibit microbial proliferation. Serum transferrin levels generally recover and normalize following resolution of acute inflammation. In contrast, chronic inflammatory disorders, such as , maintain suppressed transferrin due to sustained cytokine signaling, contributing to prolonged hypoferremia and . Transferrin serves as a for inflammatory status, with its concentrations inversely correlated to levels of (CRP), enabling clinical assessment of severity alongside other acute-phase indicators. Post-2023 cohort studies have identified persistent iron dysregulation in patients, with low and observed up to 9 months post-infection in those with ongoing symptoms, linking this pattern to enduring inflammatory stress .

Role in Diseases

Iron Deficiency and Overload Disorders

In , low systemic iron levels trigger compensatory mechanisms that upregulate hepatic transferrin synthesis to maximize iron capture from the bloodstream, resulting in elevated transferrin concentrations often exceeding 400 mg/dL alongside reduced below 15%. This adaptation enhances the protein's capacity to bind and transport scant iron resources to erythropoietic tissues, though it fails to fully mitigate the resulting microcytic due to insufficient iron delivery. Secondary alterations in transferrin levels arise from various non-genetic iron imbalances; for instance, or chronic blood loss exacerbates iron scarcity, further elevating transferrin production as the liver responds to depleted stores. In contrast, conditions like thalassemia major, characterized by ineffective , often feature decreased serum transferrin levels due to chronic inflammation and suppressing synthesis, despite high from repeated transfusions. Hereditary hemochromatosis disrupts iron through mutations in genes like HFE, leading to unchecked intestinal absorption and exceeding 45%, which saturates the protein's binding sites and promotes non-transferrin-bound iron release, culminating in parenchymal deposition in the liver, heart, and . This overload overwhelms transferrin's regulatory role, accelerating oxidative damage and organ dysfunction as excess iron evades safe transport. Atransferrinemia, a rare congenital disorder caused by biallelic mutations in the TF gene, manifests as near-absent serum transferrin, provoking severe from impaired iron delivery to erythroblasts and paradoxical in the liver and heart due to unregulated absorption and . First described in , fewer than 20 cases have been reported worldwide, highlighting its extreme rarity and autosomal recessive inheritance. Treatment typically involves regular infusions to supply functional transferrin, which replenishes iron transport capacity and alleviates , while manages overload; emerging production of recombinant human transferrin offers promise for more targeted, animal-free supplementation to stabilize and prevent complications.

Associations with Cancer

Cancer cells, particularly in breast and ovarian malignancies, exhibit upregulated expression of transferrin receptor 1 (TfR1) to enhance iron acquisition, which supports rapid proliferation and tumor growth. In , TfR1 overexpression has been documented since the 1980s and correlates with increased iron uptake essential for . Similarly, in , TfR1 is highly expressed in high-grade serous tumors and tumor-initiating cells, promoting an "iron-addicted" phenotype that drives malignancy. This upregulation allows cancer cells to meet elevated iron demands for and metabolic processes, distinguishing them from normal tissues. Epidemiological evidence links high transferrin saturation levels to increased cancer risk, with meta-analyses indicating a 1.5-fold odds ratio for any cancer when saturation exceeds 60% compared to lower levels. In ovarian cancer, elevated transferrin saturation has been associated with higher risk and poorer outcomes, particularly in cases involving iron overload linked to HFE gene variants. As a prognostic marker, low serum transferrin levels are common in advanced cancers due to cachexia, serving as an independent predictor of reduced survival (hazard ratio 1.50) and higher mortality, especially in stages III and IV. Additionally, alterations in transferrin glycosylation, such as increased carbohydrate-deficient transferrin (CDT), act as tumor markers; for instance, serum CDT rises 2.5-fold in hepatocellular carcinoma, correlating with tumor size and severity. Therapeutic strategies targeting transferrin pathways include anti-TfR antibodies that induce lethal iron deprivation by sequestering and degrading TfR1, reducing iron uptake and triggering in malignant cells without blocking transferrin binding. complexes, which mimic iron and bind transferrin for cellular uptake, disrupt iron in tumors, inhibiting growth by interfering with iron-dependent enzymes and . These approaches exploit cancer cells' reliance on iron, showing promise in preclinical models for hematopoietic and solid tumors. Recent post-2023 advances highlight transferrin-targeted nanoparticles for enhanced chemotherapy delivery; a 2024 study demonstrated that transferrin-modified Fe₃O₄ nanoparticles loaded with miR-15a-5p, combined with photothermal therapy, significantly inhibited lung cancer proliferation and promoted apoptosis in vivo by downregulating YAP1. In immunotherapy contexts, high TfR1 expression correlates with increased immune effector infiltration in tumors, including melanoma, though it also elevates checkpoint molecule levels, influencing response to checkpoint blockade therapies. TfR1-targeted immunostimulants have further shown efficacy in enhancing photodynamic immunotherapy against metastatic tumors by boosting antitumor immunity.

Role in Neurodegenerative Diseases

Transferrin plays a in brain iron homeostasis, and dysregulation is implicated in neurodegenerative disorders such as (AD), (PD), and (MS). (CSF) transferrin levels serve as a potential , with studies showing altered concentrations reflecting iron metabolism disruptions. For instance, in PD and MS, CSF transferrin levels differ significantly from controls, potentially aiding diagnosis of proteinopathies. As of 2025, research indicates transferrin's utility in tracking disease progression, particularly in relation to iron accumulation contributing to and neuronal damage. Therapeutic strategies targeting transferrin receptor 1 (TfR1) at the blood-brain barrier are being explored to enhance for treating these conditions.

Clinical and Diagnostic Applications

Reference Ranges and Measurements

Transferrin levels in are typically measured to assess iron transport capacity and are reported in milligrams per deciliter (mg/dL) or grams per liter (g/L). In adults, the normal is 200-360 mg/dL (2.0-3.6 g/L), with variations by sex and age; levels are generally higher in women (250-380 mg/dL) than in men (215-365 mg/dL) due to physiological differences in iron stores and menstrual losses. During , transferrin concentrations increase progressively, peaking in the third trimester at 280-400 mg/dL, reflecting expanded volume and heightened iron demands for fetal development. In pediatric populations, ranges are lower in neonates (100-200 mg/dL), gradually rising to adult levels by approximately 1 year of age as iron metabolism matures. Transferrin is commonly quantified using immunological methods such as immunoturbidimetry or nephelometry, which detect antigen-antibody complexes formed with specific anti-transferrin antibodies; these automated assays provide high sensitivity and are widely used in clinical laboratories. As a proxy for transferrin concentration, (TIBC) is often measured, with a normal range of 250-450 μg/dL, since TIBC correlates directly with transferrin levels (approximately TIBC = transferrin in mg/dL × 1.25). Interpretation of results involves calculating , defined as ( / TIBC) × 100, with normal values of 20-50%; elevated TIBC (>450 μg/dL) typically signals , prompting further evaluation. Quality control in transferrin measurements is crucial, as interferences from lipemia, , or certain medications can falsely alter results by affecting light scattering in turbidimetric or nephelometric assays. Recent 2025 harmonization guidelines from the International Federation of Clinical Chemistry and Medicine (IFCC) emphasize standardized reference measurement procedures, such as optimized immunoturbidimetry, to ensure comparability across global laboratories and reduce variability in reported values. These efforts support consistent diagnostic thresholds, particularly in monitoring iron status across diverse populations.

Pathological Conditions

Congenital atransferrinemia, also known as hypotransferrinemia in its partial form, is an extremely rare autosomal recessive disorder caused by mutations in the transferrin (TF) gene on chromosome 3q21-25, resulting in absent or severely reduced serum transferrin levels. This leads to microcytic hypochromic anemia due to impaired iron delivery to erythroid precursors, alongside paradoxical iron overload in tissues such as the liver and heart, manifesting as hemosiderosis. Clinical features include severe anemia from infancy, growth retardation, pallor, fatigue, recurrent infections, and potential cardiac complications from iron deposition. Treatment primarily involves regular plasma infusions or administration of purified apotransferrin to replenish transferrin and mobilize excess tissue iron, often combined with iron chelation therapy to prevent further overload. Fewer than 20 cases have been reported worldwide, highlighting its rarity and the critical role of transferrin in iron homeostasis. Acquired hypotransferrinemia arises from secondary reductions in transferrin synthesis or increased loss, distinct from genetic forms. In chronic liver diseases such as , hepatic dysfunction impairs transferrin production, a major site of its , contributing to low serum levels alongside other hypoalbuminemic states. Protein , common in advanced affecting 50-90% of patients, further exacerbates this by limiting availability for like transferrin. These conditions result in functional despite adequate stores, promoting and complicating recovery, with management focusing on nutritional support and addressing the underlying liver pathology. In , characterized by massive exceeding 3.5 g/day, transferrin is lost in the urine due to glomerular barrier damage, as its molecular weight (around 80 kDa) allows when capacity is overwhelmed. This urinary transferrin loss contributes to by depleting serum proteins, reducing oncotic pressure and leading to , , and pleural effusions. Additionally, transferrin depletion promotes resistant to oral supplementation, as urinary iron losses compound the issue, often requiring intravenous iron or in affected patients. Treatment targets the underlying renal disease with immunosuppressants or ACE inhibitors, alongside diuretics and infusions to manage .

Therapeutic and Nanomedical Uses

Drug Delivery Systems

Transferrin has been extensively engineered as a targeting ligand in nanoparticle-based drug delivery systems, leveraging its natural affinity for the transferrin receptor (TfR) to facilitate receptor-mediated endocytosis and enhance cellular uptake, particularly across the blood-brain barrier (BBB). Common conjugation strategies involve attaching transferrin to various nanoparticle platforms, such as liposomes, gold nanoparticles, and polymeric carriers like chitosan or poly(lactic-co-glycolic acid) (PLGA), enabling selective delivery to TfR-overexpressing cells. For instance, transferrin-modified porous silicon nanoparticles have demonstrated improved BBB penetration and targeted accumulation in brain tissues compared to non-targeted counterparts. These systems often incorporate polyethylene glycol (PEG) coatings to prolong circulation time and shield against opsonization, while pH-sensitive linkers or polymers mimic the endosomal acidification process of natural iron release, promoting triggered drug payload liberation within acidic tumor microenvironments (pH ~6.5) or endosomes. In therapeutic applications, transferrin-conjugated nanoparticles have shown promise for delivering chemotherapeutic agents and nucleic acids to (CNS) tumors, notably . Transferrin-functionalized nanoparticles loaded with have exhibited enhanced against cells, with preclinical studies reporting up to 2.5-fold greater cellular uptake and reduced tumor cell viability by 70% relative to free drug or non-targeted nanoparticles. Similarly, transferrin receptor-targeted core-shell nanoparticles delivering siRNA have achieved significant in models, with uptake increases of 5-10-fold over non-targeted systems, leading to suppressed tumor growth in orthotopic xenografts. These approaches capitalize on the elevated TfR expression in malignant cells, including stem cells, to improve drug localization at the tumor site. The primary advantages of transferrin-mediated drug delivery include high specificity for TfR-rich tissues, such as tumors and the BBB endothelium, which minimizes off-target effects and systemic toxicity associated with free drugs like doxorubicin. Preclinical data indicate that these systems can enhance therapeutic indices by 2-5 times in terms of efficacy metrics, such as tumor regression rates, without proportionally increasing adverse effects. Challenges persist, including potential immunogenicity from repeated dosing and suboptimal stability in vivo; however, advances using human-derived transferrin variants have mitigated immune responses, with PEGylation further reducing anti-transferrin antibody formation. As of 2025, several transferrin-conjugated prototypes remain in preclinical optimization stages for CNS oncology.

Emerging Therapies

Recombinant human apotransferrin has emerged as a promising therapy for hypotransferrinemia, a rare genetic disorder characterized by absent or severely reduced transferrin levels leading to iron overload and anemia. Intravenous infusions of apo-transferrin (iron-free transferrin) aim to restore iron transport and balance by binding excess non-transferrin-bound iron, thereby alleviating hepatic iron accumulation and improving erythropoiesis. A clinical trial (NCT01797055) is investigating the pharmacokinetics, safety, and efficacy of apotransferrin infusions in patients with atransferrinemia. Treatment with human apotransferrin in patients with congenital hypotransferrinemia has demonstrated reductions in serum ferritin levels (82–97%) and improvements in markers of iron metabolism, including normalized hemoglobin and liver iron levels via MRI, without significant adverse events in 5 patients followed for 1.2–7.3 years. This offers a targeted replacement therapy where traditional iron chelation alone is insufficient. Transferrin receptor (TfR)-targeted immunotoxins represent an innovative approach to selectively eliminate cancer cells overexpressing TfR, particularly in hematologic malignancies like . These fusion proteins consist of an anti-TfR or conjugated to a cytotoxic toxin, such as A chain, which exploits the high TfR on malignant cells for and subsequent intracellular toxin release, inhibiting protein synthesis and inducing . For instance, immunotoxins using anti-TfR antibodies linked to A have shown potent against cell lines in preclinical models, with selective killing of TfR-positive blasts while sparing normal hematopoietic cells. Phase I trials of TfR- A constructs have demonstrated feasibility in refractory . Ongoing developments focus on humanized versions to minimize and enhance therapeutic windows. Iron chelators designed to mimic transferrin's iron-binding properties, such as analogs of desferrioxamine (DFO), offer enhanced therapies for disorders by facilitating iron mobilization from transferrin-bound pools. DFO and its derivatives form stable complexes with ferric iron, promoting its dissociation from transferrin through ternary complex formation in physiological conditions, thereby increasing urinary iron excretion and reducing tissue deposition. Recent analogs incorporate structural enhancements for better transferrin interaction and prioritize oral and reduced requirements to broaden in managing chronic , such as in transfusion-dependent anemias like . In adoptive immunotherapies, modulation of (TfR/CD71) expression on CAR-T cells has been investigated to optimize iron handling and enhance antitumor efficacy. Activated CAR-T cells upregulate TfR to support proliferation via increased iron uptake, but excessive TfR can lead to , impairing mitochondrial function and T cell persistence. Strategies to downregulate TfR through or small-molecule inhibitors may improve iron , reducing and boosting metabolic fitness in the . This approach links transferrin biology to , potentially extending CAR-T applicability beyond hematologic cancers.

Molecular Interactions and Regulation

Protein-Protein Interactions

Transferrin forms a complex with binding protein 3 (IGFBP-3) and (IGF-II), which enhances the affinity of IGFBP-3 for IGF-II by approximately fivefold, modulating the activity of this in circulation. The (K_d) for the transferrin-IGF-II interaction is approximately 831 nM (8.31 × 10^{-7} M), indicating moderate binding strength that supports this regulatory role. Post-translational modification of transferrin involves interactions with glycosyltransferases, such as N-acetylglucosaminyltransferase II (GnT-II), which add residues to generate fully glycosylated forms; defects in these enzymes lead to (CDT) variants. Transferrin's binding to its receptor, transferrin receptor 1 (TfR1), is essential for cellular iron uptake but is distinct from these serum-based interactions.

Gene Expression and Regulation

The transferrin gene () promoter features specific elements that drive basal expression, including binding sites for nuclear factor (HNF) family members, such as HNF-4 and HNF-3α, and CCAAT/enhancer-binding protein alpha (C/EBPα), which interact with proximal promoter regions (PRI and PRII) essential for high-level hepatic transcription. These factors cooperate to maintain constitutive expression in s, with mutations in these sites reducing promoter activity by up to 85%. Additionally, the TF enhancer contains two response elements (HREs) that bind hypoxia-inducible factor-1 (HIF-1), enabling transcriptional activation under hypoxic conditions often associated with low iron availability; this oxygen-sensing mechanism increases TF mRNA levels in liver cells exposed to . Iron deficiency indirectly upregulates TF gene expression through the iron-responsive element (IRE)/iron regulatory protein (IRP) system, which primarily acts post-transcriptionally on related iron homeostasis genes but influences transcriptional feedback by altering cellular iron sensing and HIF stabilization. In animal models, nutritional iron deficiency leads to elevated TF mRNA transcription in the liver, with early-phase increases observed during hypoxia-induced iron restriction. Retinoic acid further enhances TF transcription in hepatocytes, stimulating a 5-fold increase in transcriptional rate and up to 10-fold rise in steady-state mRNA levels via indirect mechanisms requiring protein synthesis. Epigenetic modifications, including histone acetylation at the TF locus, contribute to its regulation in hepatocytes by promoting an open chromatin structure conducive to liver-specific transcription factor access. Genetic polymorphisms, such as rs1049296 (Asp/His variant) in the TF gene, significantly affect serum transferrin levels and glycosylation patterns, accounting for a substantial portion of inter-individual variation in protein abundance. Tissue-specific control of TF expression involves alternative promoters; the liver utilizes a proximal promoter rich in HNF and C/EBP sites for plasma protein production, whereas a distinct brain-specific promoter, located upstream, drives localized expression in oligodendrocytes and neurons to support neural iron transport.

References

  1. [1]
    Biochemistry, Transferrin - StatPearls - NCBI Bookshelf - NIH
    Jul 7, 2025 · Transferrin is a blood protein that binds to and transports iron. When iron levels are low, the body increases transferrin production to enhance ...Missing: biology | Show results with:biology
  2. [2]
  3. [3]
    7018 - Gene ResultTF transferrin [ (human)] - NCBI
    This gene encodes a glycoprotein with an approximate molecular weight of 76.5 kDa. It is thought to have been created as a result of an ...
  4. [4]
    Human transferrin: cDNA characterization and chromosomal ... - PNAS
    Chromosomal mapping by in situ hybridization and somatic cell hybrid analysis indicate that the Tf gene is located at q21-25 on human chromosome 3, consistent ...Missing: glycoprotein | Show results with:glycoprotein
  5. [5]
    Transferrin mutations at the glycosylation site complicate diagnosis ...
    Mar 23, 2011 · In its most abundant form, this serum glycoprotein carries two complex N-glycans attached to Asn432 and Asn630. Each N-glycan is terminated ...
  6. [6]
    Transferrin - an overview | ScienceDirect Topics
    Human transferrin (hTf) is composed of alternating α-helices and β-sheets separated into an N-lobe and a C-lobe, each further sub-divided into domains I and II, ...
  7. [7]
    Structure of the Human Transferrin Receptor-Transferrin Complex: Cell
    The Tf polypeptide chain is arranged in two homologous halves (∼40% sequence identity), termed N- and C-lobe. Each lobe contains two domains (N1, N2 and C1, C2) ...
  8. [8]
    Transferrin: Internal Homology in the Amino Acid Sequence - Science
    When these are suitably aligned by placing two gaps, 40 percent of the amino acids in corresponding positions are identical. This indicates that the doubling of ...Missing: mammals | Show results with:mammals
  9. [9]
    Evolutionary Diversification of the Vertebrate Transferrin Multi-gene ...
    In a phylogenetic analysis of vertebrate transferrins (TFs), six major clades (subfamilies) were identified: (1) S, the mammalian serotransferrins; (2) ICA, ...
  10. [10]
    Comparison of Transferrin Sequences From Different Species
    Amino acid sequences of transferrins from eight species, from human to tobacco hornworm, have been compared. Eighty-four amino acids (12%) are invariant.Missing: identity | Show results with:identity
  11. [11]
    Comparison of transferrin sequences from different species
    Amino acid sequences of transferrins from eight species, from human to tobacco hornworm, have been compared. Eighty-four amino acids (12%) are invariant.Missing: identity | Show results with:identity
  12. [12]
    Transferrin Fusion Technology: A Novel Approach to Prolonging ...
    Transferrin naturally has a long circulating half-life (7–10 days for glycosylated transferrin and 14–17 days for the nonglycosylated form). Nonglycosylated ...
  13. [13]
    Normal transferrin glycosylation does not rule out severe ALG1 ...
    Apr 16, 2024 · N-glycosylation has a key role in functionality, stability, and half-life of most proteins. Therefore, congenital defects of glycosylation ...
  14. [14]
    Comparative analysis of transferrin and IgG N-glycosylation in two ...
    Mar 23, 2023 · However, changes in Tf N-glycome can alter its circulation half-life and affect iron transport, which might be an important element of the ...
  15. [15]
  16. [16]
  17. [17]
    Transferrin receptors | Experimental & Molecular Medicine - Nature
    Apr 22, 2025 · It is encoded by a 2,471-bp-long gene located on the long arm of human chromosome 7 (7q22.1) that consists of 18 exons. This gene ...
  18. [18]
    The Transferrin Receptors, TfR1 and TfR2 Bind ... - PubMed Central
    Circulating, holo-Tf binds to the cell-surface transferrin receptor-1 (TfR1) and is endocytosed. Through an endocytic process known as the Tf cycle, the iron is ...
  19. [19]
    Mysteries of the Transferrin-Transferrin Receptor 1 Interaction ...
    Binding of Tf to the TfR1 leads to receptor-mediated endocytosis and the endosome is the site where Fe is released from Tf. A conformational change in Tf is ...
  20. [20]
    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.Missing: PDB 1A3B
  21. [21]
    Transferrin - an overview | ScienceDirect Topics
    The gene transcribing human transferrin is located at the q21–25 region of chromosome 3. From a structural point of view, transferrin is a monomeric ...
  22. [22]
    The protective role of transferrin in Müller glial cells after iron ... - NIH
    In adults, Tf synthesis is also found in other cell types including oligodendrocytes [18], choroid plexus epithelial [19], Sertoli [20], mammary epithelial [21] ...<|control11|><|separator|>
  23. [23]
    Iron, transferrin, ferritin and total iron-binding capacity - Uniprix
    Women: 11-307 mcg/L; Men: 24-336 mcg/L. Transferrin: 2.0-3.6 g/L; Transferrin saturation: 0.20-0.50; Total iron-binding capacity (TIBC): 45-81 mcmol/L.
  24. [24]
    Developmental changes in serum iron, total iron-binding ... - PubMed
    Developmental changes in serum iron, total iron-binding capacity, and transferrin saturation in infancy. J Pediatr. 1977 Dec;91(6):875-7. doi: ...
  25. [25]
    Maternal iron status in early pregnancy and childhood body fat ...
    Maternal lower ferritin and higher transferrin in early pregnancy are associated with body fat accumulation and distribution but are not associated with ...
  26. [26]
    Lactoferrin and iron: structural and dynamic aspects of binding and ...
    Lactoferrin (Lf) has long been recognized as a member of the transferrin family of proteins and an important regulator of the levels of free iron in the ...
  27. [27]
    Genome-wide association study identifies two loci strongly affecting ...
    Up to 40% of genetic variation in total transferrin levels is due to variants in the transferrin (TF) and hemochromatosis (HFE) genes (6), but whether ...
  28. [28]
    Differential transferrin gene expression in Atlantic salmon (Salmo ...
    Jan 1, 1996 · Many of these cDNA clones encoded the iron binding protein transferrin. Transferrin mRNA levels were determined to be significantly higher ...
  29. [29]
    [PDF] Transcriptional profiling of the parr-smolt transformation in Atlantic ...
    salmon and expression of different alpha subunit isoforms changes dur- ... Differential transferrin gene expression in Atlantic salmon ... salmonids: osmoregulation ...
  30. [30]
    Ovotransferrin - an overview | ScienceDirect Topics
    Ovotransferrin is an iron-binding glycoprotein, found in avian egg white and in avian serum, belonging to the family of transferrin iron-binding glycoproteins.
  31. [31]
    (PDF) Physiological roles of ovotransferrin - ResearchGate
    Aug 6, 2025 · Ovotransferrin is an iron-binding glycoprotein, found in avian egg white and in avian serum, belonging to the family of transferrin iron ...
  32. [32]
    On the evolutionary significance and metal-binding characteristics of ...
    This time frame overlaps the one (200–500 million years ago) in which the common ancestor to all of the modern transferrins occurred (21). A monolobal ...
  33. [33]
    Functional Divergence of the N-Lobe and C-Lobe of Transferrin ...
    This structure is believed to originate from a gene duplication event about 670 million years ago [18,19]. Animals 12 03458 g003. Figure 3. Phylogenetic ...
  34. [34]
    Iron binding and release properties of transferrin-1 from Drosophila ...
    Jul 29, 2020 · The UV-Vis spectra for iron-bound DmTsf1 and MsTsf1 shows the characteristic absorption peaks in the UV region at ~295 nm (Figure S1). Thus, it ...Missing: 430-470 | Show results with:430-470
  35. [35]
    Iron binding and release properties of transferrin-1 from Drosophila ...
    Manduca sexta and Drosophila melanogaster transferrin 1 (Tsf1) bind one ferric ion. These transferrins have a very high affinity for iron (log K' = 18).Missing: melanotransferrin | Show results with:melanotransferrin
  36. [36]
    The membrane‐bound transferrin homologue melanotransferrin ...
    Oct 9, 2000 · MTf has many characteristics in common with serum Tf and previous studies have shown that it can bind Fe. This has led to speculation that MTf ...Missing: insects | Show results with:insects
  37. [37]
    Evolution of disulphide bridges and conservation of iron-binding ...
    Jul 1, 2007 · In birds and reptiles, one gene is responsible for the expression of serotransferrin in the liver and ovotransferrin in the oviduct (Thibodeau ...
  38. [38]
    A comparison of representative avian and reptilian transferrins...
    In the majority of reptilian transferrins, a putative N -glycosylation site corresponding to Asn473 in CtrF sequence is conserved (with the exception of the ...
  39. [39]
    Role of Transferrin in Iron Metabolism - IntechOpen
    Oct 5, 2022 · Transferrin is a glycoprotein that plays a central role in iron metabolism [3]. It is present at a concentration of 30-60 μM in blood [4].
  40. [40]
    Body iron metabolism and pathophysiology of iron overload - PMC
    The production of ROS by iron is mainly through the Fenton reaction, which eventually forms hydroxyl radicals from superoxide or hydrogen peroxide [35]. Among ...
  41. [41]
    Regulation of iron transport and the role of transferrin - ScienceDirect
    Transferrin exerts a crucial function in the maintenance of systemic iron homeostasis as component of a plasma iron sensing system that modulates hepcidin ...
  42. [42]
    Iron metabolism and iron disorders revisited in the hepcidin era
    Feb 1, 2020 · Only 1-2 mg iron are absorbed daily in the gut, compensating for an equal loss; most iron (20-25 mg/daily) is recycled by macrophages upon ...
  43. [43]
    Ceruloplasmin Ferroxidase Activity Stimulates Cellular Iron Uptake ...
    Jan 8, 1999 · A role for ceruloplasmin (Cp) in vertebrate iron metabolism is suggested by its potent ferroxidase activity catalyzing conversion of Fe2+ to Fe3+.
  44. [44]
    Transferrin Receptors in Erythropoiesis - PMC - PubMed Central - NIH
    Dec 19, 2020 · Erythropoiesis is the most iron-consuming process to support hemoglobin production. Iron delivery is mediated via transferrin internalization ...
  45. [45]
    Transferrin receptor 1-mediated iron uptake plays an essential role ...
    Transferrin receptor 1 (Tfr1) facilitates the uptake of iron at the cell surface by internalizing diferric transferrin. Tfr1 is ubiquitously expressed in ...Missing: synthesis | Show results with:synthesis
  46. [46]
    On Iron Metabolism and Its Regulation - PMC - PubMed Central
    Apr 27, 2021 · Most of the iron in the human body is associated with erythrocyte hemoglobin (~80%). The rest is stored in macrophages and hepatocytes or ...
  47. [47]
    Ferrochelatase forms an oligomeric complex with mitoferrin-1 ... - NIH
    Fech forms an oligomeric complex with Mfrn1 and Abcb10 to synergistically integrate mitochondrial iron importation and use for heme biosynthesis.
  48. [48]
    Erythropoietin signaling regulates heme biosynthesis - PMC - NIH
    This integrated pathway culminates with the direct phosphorylation of the crucial heme biosynthetic enzyme, ferrochelatase (FECH) by protein kinase A (PKA).
  49. [49]
    Crosstalk between Iron Metabolism and Erythropoiesis - PMC
    Two to three million red blood cells (RBCs) are produced every second and require 30–40 mg of iron delivered to the erythron to make 30 pg of hemoglobin per ...
  50. [50]
    Erythropoietin administration in humans causes a marked and ... - NIH
    Profound hepcidin suppression appeared abruptly 24 hours after subcutaneous erythropoietin (P=0.003), and was near maximal at onset, with peak (mid-afternoon) ...
  51. [51]
    Evidence that the expression of transferrin receptor 1 on erythroid ...
    The human body requires ~20–25 mg of iron per day to maintain its daily red cell production. ... Humans and mice with iron-restrictive erythropoiesis caused by ...
  52. [52]
    Congenital Atypical Microcytic Anemia Accompanied by ... - NIH
    Congenital hypochromic microcytic anemia and hemochromatosis are present in a(hypo)transferrinemia cases wherein the transferrin levels are negligible/ ...
  53. [53]
    Microcytic Hypochromic Anemia - StatPearls - NCBI Bookshelf - NIH
    TIBC is usually increased in iron deficiency anemia, while transferrin saturation is markedly decreased in iron deficiency anemia. Ferritin levels below 12 ...
  54. [54]
    A c.726C>G (p.Tyr242Ter) nonsense mutation-associated with ... - NIH
    TFRC/TFR1 is the receptor for iron-bound Transferrin at the surface of plasma membrane responsible for transportation of iron. It has been reported that TFRC ...
  55. [55]
    The placenta: the forgotten essential organ of iron transport - NIH
    Transferrin-iron is the major, if not only, maternal iron source for placental transfer, and its uptake is mediated by transferrin receptor 1 (TFR1) (Figure 1).
  56. [56]
    Placental iron transport: The mechanism and regulatory circuits - NIH
    Transferrin-bound iron is thought to be the predominant iron source taken up by the placenta.
  57. [57]
    Maternal, Fetal and Placental Regulation of Placental Iron Trafficking
    Maternal diferric-transferrin (Tf) binds to transferrin receptor 1 (TfR1) on the apical side of the STB where it is internalized into clathrin coated endosomes.
  58. [58]
    Buried Treasure: Evolutionary Perspectives on Microbial Iron Piracy
    The sequestration of essential iron has emerged as a crucial innate defense system termed nutritional immunity, leading pathogens to evolve mechanisms of 'iron ...
  59. [59]
    Nutritional Immunity: Starving Pathogens of Trace Minerals - PMC
    Feb 4, 2016 · They found that transferrin bound iron, thereby sequestering iron from pathogens and preventing microbial growth. ... iron and zinc sequestration ...
  60. [60]
    Key players in the regulation of iron homeostasis at the host ...
    Oct 24, 2023 · For instance, serum Tsf and LF are two secreted transferrins known to be involved in nutritional immunity by hiding iron to help protect the ...
  61. [61]
    Escape from bacterial iron piracy through rapid evolution of transferrin
    Iron is a precious cellular metal, sequestered by hosts and scavenged by pathogens (1-3). Vertebrate iron transport is mediated by serum transferrin, a protein ...
  62. [62]
    Iron sequestration by transferrin 1 mediates nutritional immunity in ...
    Mar 31, 2020 · Iron sequestration is a recognized innate immune mechanism against invading pathogens mediated by iron-binding proteins called transferrins.
  63. [63]
    Iron sequestration by transferrin 1 mediates nutritional immunity in ...
    Mar 18, 2020 · Lactoferrin is found on mucosal surfaces, and in biological fluids including milk and saliva, indicating that it is part of the innate immune ...
  64. [64]
    Transgenic Mice Expressing Human Transferrin as a Model for ... - NIH
    A hypoferremic state is a defense mechanism that helps reduce the amount of free transferrin by as much as 70%, allowing control of meningococcal infection (30) ...Missing: knockout | Show results with:knockout
  65. [65]
    Iron and iron-related proteins in COVID-19 - PMC - PubMed Central
    Jul 18, 2022 · Transferrin and COVID-19 severity​​ In a COVID-19-related study, transferrin levels were low at the time of hospitalization and a decrease in ...
  66. [66]
    Acute-phase proteins as indicators of disease severity and mortality ...
    Of the newly tested acute-phase proteins, only transferrin shows significance as a marker of disease severity and mortality in COVID-19 disease.
  67. [67]
    Physiology, Acute Phase Reactants - StatPearls - NCBI Bookshelf
    Apr 24, 2023 · Interleukin-6 (IL-6) is the primary cytokine responsible for inducing the production in the liver. IL-1, tumor necrosis factor-alpha (TNF-alpha) ...Function · Pathophysiology · Clinical Significance
  68. [68]
    Acute phase proteins | eClinpath
    Negative acute phase proteins decrease in plasma concentration by greater than 25% in response to inflammation. This reduction can occur rapidly (within 24 ...General · Measurement · C-reactive proteinMissing: percentage | Show results with:percentage
  69. [69]
    IL-6 mediates hypoferremia of inflammation by inducing the ...
    IL-6 is the necessary and sufficient cytokine for the induction of hepcidin during inflammation and that the IL-6–hepcidin axis is responsible for the ...
  70. [70]
    How to Manage, Treat Anemia of Inflammation in Patients with ...
    Dec 17, 2017 · In anemia of inflammation, the concentration of ferritin is normal or increased and circulating transferrin is low. This contrasts with iron- ...
  71. [71]
    Diagnosis and management of iron deficiency in chronic ...
    Dec 4, 2020 · Transferrin saturation (TSAT) <6% in combination with low ferritin is diagnostic of ID, but in the presence of inflammation, a seemingly ...Abstract · Serum inflammatory markers... · Transferrin saturation in CIC: a...
  72. [72]
    Low transferrin levels predict heightened inflammation in patients ...
    Reduced serum transferrin and iron levels, along with the increased CRP and high ferritin, were strongly associated with the heightened inflammatory and immune ...
  73. [73]
    Iron dysregulation and inflammatory stress erythropoiesis associates ...
    Mar 1, 2024 · Iron dysregulation and inflammatory stress erythropoiesis associates with long-term outcome of COVID-19.
  74. [74]
    Investigation of iron deficiency anaemia - PMC - NIH
    The serum transferrin concentration (expressed in mg/dL) increases in IDA, as the body tries to compensate for low iron levels and this correlates positively ...Ferritin · Iron Studies · Causes Of Iron Deficiency...
  75. [75]
    A guide to diagnosis of iron deficiency and iron deficiency anemia in ...
    Diagnosis of ID is not always easy. Low serum levels of ferritin or transferrin saturation, imply a situation of absolute or functional ID. It is sometimes ...
  76. [76]
    Diagnosis and management of iron deficiency in chronic ... - NIH
    Dec 4, 2020 · Low iron also upregulates hepatic production of transferrin, resulting in high TIBC, contributing to low TSAT. Lack of iron available to the BM ...Ferritin In Absolute And... · Ferritin In Cic: Making The... · Transferrin Saturation In...
  77. [77]
    Correlation of T-regulatory Cells and Iron Status in β-Thalassemia ...
    Feb 16, 2023 · Result Significantly higher levels of serum iron, ferritin, transferrin saturation, and CD4+ cell percentage were observed in cases than in ...
  78. [78]
    Iron Overload in Beta Thalassaemia Major and Intermedia Patients
    Results: 87.4% of the beta thalassaemia major patients showed very high ferritin levels. The mean serum ferritin level was found to be 2767.52 ng/ml. 44.4% ...
  79. [79]
    HFE-Related Hemochromatosis - GeneReviews® - NCBI Bookshelf
    Apr 11, 2024 · Elevated transferrin saturation in persons with HFE HC (≥60% in males, ≥50% in females) is an indicator of increased recycling of iron and ...
  80. [80]
    Inherited iron overload disorders - PubMed
    Apr 5, 2020 · Atransferrinemia and DMT1 deficiency are characterized by iron deficient erythropoiesis, severe microcytic anemia with high transferrin ...
  81. [81]
    Hereditary Hemochromatosis and Iron Metabolism - PMC - NIH
    Transferrin saturation increases successively with age in adults with HH. People with TS > 45% in repeat test should be studied for iron overload using serum ...
  82. [82]
    Atransferrinemia | About the Disease | GARD
    Atransferrinemia is a very rare hematologic disease caused by a transferrin (TF) deficiency and characterized by microcytic, hypochromic anemia.
  83. [83]
    Entry - #209300 - ATRANSFERRINEMIA - OMIM - (OMIM.ORG)
    Atransferrinemia is characterized by microcytic anemia and by iron loading. It can be treated effectively by plasma infusions.
  84. [84]
    Congenital Hypotransferrinemia, an Unusual Cause of Iron ... - NIH
    Nov 16, 2016 · Hereditary hypotransferrinemia is a very rare cause of iron deficiency anemia in childhood characterized by microcytic hypochromic anemia ...
  85. [85]
    High-level production of animal-free recombinant transferrin from ...
    Treatment of the rare disease hereditary atransferrinemia is the most obvious, where the beneficial effects of transferrin infusion have been described [4].
  86. [86]
    Transferrin receptor 1 in cancer: a new sight for cancer therapy - PMC
    The TFR1 levels on cell surface, which modulate the uptake of TF binding iron, are associated with cell proliferation rate [96]. Since cancer cells have higher ...
  87. [87]
    The Role of Iron in Cancer Progression - PMC - PubMed Central - NIH
    As early as 1980, studies have shown that TFR1 is significantly up-regulated in breast cancer (27). Subsequent studies have confirmed that TFR1 is highly ...
  88. [88]
    Targeting iron to contrast cancer progression - ScienceDirect.com
    Cancer cell walls are generally characterized by upregulated expression of the transferrin receptor TfR1, leading to increased cellular uptake of the iron- ...
  89. [89]
    Risk of cancer by transferrin saturation levels and ... - PubMed
    From the meta-analysis, the odds ratio of any cancer for transferrin saturation ≥60% versus a reference group was 1.5 (95% CI: 1.2-1.8) for women and men ...
  90. [90]
    Hemochromatosis and Ovarian Cancer - David M Robertson, 2011
    In a large clinical study, C282Y heterozygotes were shown to have significantly elevated transferrin saturation values and serum ferritin levels [1] ...Abstract · Summary Of Methods & Results · Discussion
  91. [91]
    Comprehensive evaluation of serum hepatic proteins in ... - PubMed
    Mar 4, 2024 · Low albumin, prealbumin, and transferrin levels were all independent prognostic factors affecting patients with cancer cachexia, especially in patients in the ...
  92. [92]
    Serum carbohydrate-deficient transferrin in patients with ... - PubMed
    The mean serum %CDT value was increased 2.5-fold in hepatocellular carcinoma, and false-positive results were found in 31 (46%) of the 67 patients. The serum % ...
  93. [93]
    Lethal iron deprivation induced by non-neutralizing antibodies ... - NIH
    Aug 28, 2011 · A number of antibodies have been developed that induce lethal iron deprivation (LID) by targeting the transferrin receptor 1 (TfR1/CD71) and ...Tfr1 Targeting And Antibody... · Inhibition Of Tfr1 Function... · Effect On Tfr1 Trafficking...
  94. [94]
    Iron-Targeting Antitumor Activity of Gallium Compounds and Novel ...
    Recent studies show that gallium compounds and metal-thiosemicarbazone complexes inhibit tumor cell growth by targeting iron homeostasis.
  95. [95]
    Review Gallium and its competing roles with iron in biological systems
    Gallium can mimic iron and interfere with iron-dependent cellular function. Gallium-based compounds have therapeutic potential in cancer and infection.
  96. [96]
    Effect of Transferrin‐Modified Fe3O4 Nanoparticle Targeted Delivery ...
    Nov 27, 2024 · The aim of this study was to investigate the effect of the targeted delivery of Fe 3 O 4 NPs modified with transferrin (Tf) compared with photothermal ...
  97. [97]
    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.
  98. [98]
    Transferrin receptor-targeted immunostimulant for photodynamic ...
    Jun 3, 2024 · In this work, a transferrin receptor (TfR) targeted immunostimulant (PTI) is fabricated for photodynamic immunotherapy against metastatic tumors ...
  99. [99]
    Transferrin: What It's For, Normal Levels & High or Low Levels
    Jan 19, 2024 · Men: 215 - 365 mg/dL; Women: 250 - 380 mg/dL. Reference levels may vary depending on the laboratory, and therefore it is important to evaluate ...Missing: serum adults
  100. [100]
    Lab Values, Normal Adult - Medscape Reference
    Jul 28, 2025 · Total serum iron: 50-150 μg/dL. Transferrin: 215-365 mg/dL or 2.15-3.65 g/L. Urea: 1.2-3 mmol/L. Uric acid: Blood: Males: 4-8.5 mg/dL or 0.24 ...<|control11|><|separator|>
  101. [101]
    Transferrin (Serum) During Pregnancy - babyMed
    Negative Pregnancy Adult: 200 to 400 mg/dL or 2.5 to 5 µmol/L · Pregnancy Trimester One: 254 to 344 mg/dL or 3.1 to 4.2 µmol/L · Pregnancy Trimester Two: 220 to ...
  102. [102]
    [PDF] Reference Ranges Document - Children's Hospital of Philadelphia
    TRANSFERRIN (mg/dL). Age. Male. Female. All ages. 180-370. 180-370. TRANSFERRIN SATURATION (%). Age. Male. Female. All ages. 6.0-40.0. 6.0-40.0. TOTAL IRON ...
  103. [103]
    The Postnatal Hypotransferrinemia of Early Preterm Newborn Infants
    The early preterm newborn infant's transferrin levels rise slowly so that 7-8 weeks after birth they are 78% of the level found in normal term infant sera and ...Missing: reference range<|control11|><|separator|>
  104. [104]
    Measurement of soluble transferrin receptor by immunoturbidimetry ...
    Objective: To evaluate and compare two new commercially available immunoturbidimetric and immunonephelometric assays for measuring soluble transferrin receptor ...Missing: techniques | Show results with:techniques
  105. [105]
    Typical Normal Serum Values for Iron, Iron-Binding Capacity, Ferritin ...
    Typical Normal Serum Values for Iron, Iron-Binding Capacity, Ferritin, and Transferrin Saturation ; Men. 75–150 mcg/dL (13–27 micromol/L) ; Women. 60–140 mcg/dL ( ...
  106. [106]
    Transferrin Saturation: Reference Range, Interpretation, Collection ...
    Oct 27, 2025 · Transferrin saturations of less than 20% indicate iron deficiency, while transferrin saturations of more than 50% suggest iron overload.
  107. [107]
    TIBC (Total Iron-Binding Capacity) Test - Cleveland Clinic
    Generally, the normal TIBC range is 240 micrograms per deciliter (mcg/dL) to 450 mcg/dL. It's important to know that providers typically combine the results of ...
  108. [108]
    IFCC Committee on Plasma Proteins (C-PP) reference method for ...
    Aug 29, 2025 · Details : C1RMP_P14 - IFCC Committee on Plasma Proteins (C-PP) reference method for transferrin ; Optimized immunoturbidimetry/immunonephelometry.
  109. [109]
    Harmonization of clinical laboratory results | myadlm.org
    Harmonized laboratory test results are important for making effective, necessary and correct treatment decisions to provide appropriate patient care.
  110. [110]
    Atransferrinemia - Symptoms, Causes, Treatment | NORD
    Atransferrinemia is an extremely rare genetic disorder characterized by low levels of healthy, functional red cells in the blood (hypochromic, microcytic anemia) ...Missing: 1953 | Show results with:1953
  111. [111]
    Hereditary hypotransferrinemia can lead to elevated transferrin ...
    In all patients, conditions able to decrease transferrin concentration such as malnutrition, protein malabsorption, hepatocellular failure, and urinary protein ...
  112. [112]
    Malnutrition in Liver Cirrhosis:The Influence of Protein and Sodium
    Protein calorie malnutrition (PCM) is associated with an increased risk of morbidity and mortality in patients with cirrhosis and occurs in 50%-90% of these ...
  113. [113]
    Nephrotic Syndrome - StatPearls - NCBI Bookshelf
    Nephrotic syndrome (NS) is a clinical syndrome defined by massive proteinuria responsible for hypoalbuminemia, with resulting hyperlipidemia, edema, and various
  114. [114]
    Iron deficiency in proteinuric children with nephrotic syndrome
    Massive proteinuria in nephrotic syndrome causes depletion of various proteins. Iron deficiency can occur due to urinary loss of iron, transferrin, ...
  115. [115]
  116. [116]
    P1465: SAFETY AND EFFICACY OF APOTRANSFERRIN ... - NIH
    Aug 8, 2023 · The aim of our study was to investigate the safety and efficacy of apotransferrin administration on markers of iron metabolism, erythropoiesis, and spleen size
  117. [117]
    Resistance of myeloid leukaemia cell lines to ricin A-chain ... - PubMed
    Only two antibodies, MEM75 and 120-2A3, both directed against the transferrin receptor (TfR) were predicted to form immunotoxins that would inhibit protein ...Missing: fusion | Show results with:fusion
  118. [118]
    Molecular events contributing to cell death in malignant human ...
    Oct 15, 2006 · In fact, it has even been shown that an anti-murine TfR antibody conjugated to the potent plant toxin ricin is not cytotoxic to early mouse ...Antibodies And Antibody... · Results · Anti-Htfr Igg3-Av...
  119. [119]
    An anti-transferrin receptor-avidin fusion protein exhibits both strong ...
    Leukemia and lymphoma cells, which are of hematopoietic origin, overexpress the TfR compared with normal tissues (6, 9, 14, 15) and may be especially sensitive ...Antibodies And Antibody... · Proliferation Assays · Results
  120. [120]
    (PDF) AAV Gene Therapy for Liver Disease - ResearchGate
    Aug 6, 2025 · The field of adeno-associated virus (AAV) gene therapy has progressed rapidly over the past decade, with the advent of novel capsid ...
  121. [121]
    Directed evolution of liver-detargeted AAV vectors for systemic gene ...
    Aug 21, 2025 · We found that AAVhu.32 is liver-detargeted compared to AAV9, and AAV5 exhibits high skeletal muscle biodistribution albeit with low transgene ...
  122. [122]
    Next-generation human liver gene therapy - NIH RePORTER
    The limitations of current AAV vectors need to be overcome for broad application of AAV liver gene therapy because most liver diseases require more hepatocytes ...
  123. [123]
    Chelate mediated transfer of iron from transferrin to desferrioxamine
    Low molecular weight chelating agents, capable of forming ternary complexes with transferrin and ferric iron, can promote a rapid transfer of iron from ...
  124. [124]
    The assessment of serum nontransferrin-bound iron in chelation ...
    May 1, 2000 · This paper introduces a novel assay for NTBI and shows its application to monitoring chelator activity in patients with thalassemia and detecting NTBI in a ...
  125. [125]
    Iron Chelators in Treatment of Iron Overload - PMC - NIH
    In clinical use, deferoxamine, deferiprone, and deferasirox are the three United States Food and Drug Administration-approved iron chelators.
  126. [126]
    Elevated transferrin receptor impairs T cell metabolism and function ...
    T cells in systemic lupus erythematosus (SLE) exhibit multiple metabolic abnormalities. Excess iron can impair mitochondria and may contribute to SLE.
  127. [127]
    Reprogramming CAR-T cells: Targeting SLC transporters for tumor ...
    Additionally, the transferrin receptor (CD71) is upregulated upon T cell activation to enhance iron uptake. However, elevated CD71 expression can impair T ...
  128. [128]
  129. [129]
    Emergence of Ferrichelatase Activity in a Siderophore-Binding ...
    Mar 9, 2020 · We establish that human ferric holo-transferrin interacts directly with bacterial ... binds hydroxamate siderophores but with lower affinity ...
  130. [130]
    Complete replacement of serum by albumin, transferrin, and ... - NIH
    Albumin, transferrin, and lipids can replace serum entirely for support of LPS-stimulated murine B lymphocytes in culture.Missing: stability | Show results with:stability
  131. [131]
    Congenital disorders of glycosylation - PMC - NIH
    A type II serum transferrin CDT pattern indicates Golgi defects such as N-acetylglucosaminyltransferase (GnT) II deficiency (CDG type IIA, MGAT2-CDG).
  132. [132]
    Persistent complement dysregulation with signs of ... - Science
    Jan 19, 2024 · Low antithrombin III levels in Long Covid patients were accompanied by signs of increased cleavage by thrombin, a driver of TCC formation.<|control11|><|separator|>
  133. [133]
    Transcriptional regulation of transferrin and albumin genes by ...
    Apr 15, 1992 · Transferrin and albumin, which are both secreted from the human hepatoma cell line Hep3B, were regulated transcriptionally by retinoic acid ...Missing: enhances | Show results with:enhances
  134. [134]
    The Hepatocyte Nuclear Factor Network and Liver-Specific Gene ...
    This review provides an overview of our current knowledge about liver-enriched transcription factors and their role in liver function and development. We review ...
  135. [135]
    Genome-wide association study identifies two loci strongly affecting ...
    Up to 40% of genetic variation in total transferrin levels is due to variants in the transferrin (TF) and hemochromatosis (HFE) genes (6), but whether ...
  136. [136]
    Brain-specific expression of the human transferrin gene. Similar ...
    The -164/+1 promoter region was sufficient to confer cell type-specific transcription in oligodendrocytes, epithelial choroid plexus cells, and B103 cells.