Fact-checked by Grok 2 weeks ago

Transcytosis

Transcytosis is a cellular process involving the vesicular of macromolecules, such as proteins and , from one side of a polarized to the other, enabling the selective movement of substances across physiological barriers while preserving the distinct compositions of adjacent compartments. The was first proposed by George Palade in the 1950s to explain capillary permeability. This mechanism combines at one plasma membrane domain, intracellular trafficking through vesicles, and at the opposing domain, often mediated by specific pathways like clathrin-coated pits or caveolae. In endothelial and epithelial cells, transcytosis plays a in maintaining by facilitating the transport of essential molecules, including nutrients like iron via transferrin and immunoglobulins such as polymeric IgA through the polymeric immunoglobulin receptor. For instance, at the blood-brain barrier, receptor-mediated transcytosis allows limited passage of insulin and while restricting larger or non-specific cargos to protect the . Adsorptive transcytosis, driven by electrostatic interactions, further enables the uptake of cationic substances, though it is less selective and can contribute to pathological leakage in conditions like . The process is regulated by molecular components including SNARE proteins for vesicle fusion, Rab GTPases for trafficking, and lipids like phosphatidylinositol 3-phosphate, ensuring directional transport in polarized cells. Dysregulation of transcytosis contributes to various diseases, such as Alzheimer's, and serves as a target for strategies across barriers. Overall, transcytosis exemplifies an evolutionarily conserved strategy for transcellular exchange in multicellular organisms, balancing permeability and protection across key tissue interfaces.

Introduction

Definition

Transcytosis is a specialized form of that enables the movement of macromolecules across polarized cells, such as epithelial and endothelial cells, through a process involving at one plasma membrane, vesicular trafficking across the , and at the opposite membrane. This mechanism allows relatively large substances, typically exceeding 10 kDa, to be shuttled vectorially from one extracellular compartment to another without mixing with the cell's endolysosomal pathways. Unlike paracellular transport, which occurs between adjacent cells through tight junctions, or simple diffusion across the limited to small, non-polar molecules, transcytosis provides a regulated, energy-dependent route for selective macromolecular passage in a directional manner. This polarized nature is particularly prominent in barrier-forming tissues, where it maintains tissue by directing cargo from apical to basolateral surfaces or vice versa. Representative examples of transcytosed molecules include polymeric (IgA), which is transported across mucosal epithelia to provide immune protection at secretory surfaces; iron bound to , facilitating nutrient delivery across endothelial barriers; and (LDL), which crosses vascular to support distribution in tissues. These processes highlight transcytosis's role in immune defense, nutrient uptake, and , often involving specific vesicular pathways for efficient trafficking.

Historical Development

Early observations of vesicular transport across epithelial barriers emerged in the mid-20th century, particularly through electron microscopy studies of nutrient absorption in the . In 1959, Samuel L. Palay and Leonard J. Karlin provided seminal evidence for the pathway of fat absorption, demonstrating that dietary were internalized via pinocytotic vesicles at the apical surface of enterocytes, ed across the in membrane-bound compartments, and released at the basolateral side into lacteals. This work highlighted the role of vesicular mechanisms in macromolecular , initially termed "cytopempsis" or , and laid foundational insights into transcellular movement beyond simple diffusion. The concept of transcytosis as a directed, vectorial process was formalized in the late 1970s through studies on capillary . Drawing from George E. Palade's earlier electron microscopy observations of vesicular shuttling in endothelial cells during the 1950s and 1960s, Nicolae Simionescu coined the term "transcytosis" in 1979 to describe the specific apical-to-basolateral or vice versa transport of plasma macromolecules, such as and (LDL), via plasmalemmal vesicles in the arterial . This terminology distinguished transcytosis from non-directional , emphasizing its role in maintaining and nutrient delivery. Key milestones in the 1980s included the elucidation of receptor-mediated transcytosis for immunoglobulins. Keith E. Mostov and identified the polymeric immunoglobulin receptor (pIgR) as the mediator of dimeric IgA transport across epithelial barriers, such as in the liver and intestine, where it binds IgA at the basolateral surface, traffics it vectorially, and releases it apically as secretory IgA. This discovery, confirmed through models like MDCK cells, established transcytosis as a critical mechanism for mucosal immunity. Advances in the leveraged genetic tools to dissect vesicular pathways, particularly the involvement of caveolae. In 2001, Schubert et al. demonstrated using caveolin-1 knockout mice that ablation of this protein, essential for caveolae formation, significantly reduced transcytosis across endothelial barriers, confirming caveolae as a primary route for fluid-phase and receptor-mediated transport. These findings shifted understanding toward caveolae-dependent mechanisms in endothelial permeability. In the 2020s, has further solidified the pathological relevance of transcytosis, with 2025 studies confirming caveolae-mediated LDL in models. For instance, investigations using advanced imaging and genetic perturbations in mouse aortas revealed that caveolin-1 drives LDL transcytosis into the intima, promoting plaque formation, and identified myosin-9 as a regulator enhancing this process under hyperlipidemic conditions. These developments underscore transcytosis as a therapeutic target for .

Mechanisms

Vesicular Transport Process

Transcytosis involves the directed vesicular transport of macromolecules across polarized cells, such as epithelial and endothelial cells, from one plasma membrane domain to the opposite one, preserving cellular polarity through sequential endocytic, sorting, and exocytic events. This process enables selective passage of cargo like antibodies, nutrients, and proteins while maintaining barrier integrity, with vesicles typically measuring 50-100 nm in diameter to facilitate efficient intracellular movement. The process begins with at either the apical or basolateral , depending on the direction of . In clathrin-dependent , cargo binds receptors and is internalized via clathrin-coated pits forming early endosomes, as seen in the of polymeric (pIgA) across epithelial cells. Alternatively, caveolae-mediated predominates in endothelial cells, where flask-shaped invaginations enriched in caveolin-1 internalize molecules like through cholesterol-dependent mechanisms, rapidly forming vesicles within seconds. These initial endocytic vesicles, often 50-70 nm in size for caveolae, capture extracellular material without immediate degradation. Following , intracellular sorting and trafficking direct the vesicles across the while avoiding lysosomal fusion. Cargo is routed through recycling endosomes—such as early endosomes marked by Rab5 and EEA1, progressing to common or apical recycling endosomes with Rab11 and Rab25—to prevent , as exemplified by pIgA transcytosis in polarized MDCK cells. This sorting is coupled with long-range trafficking along , powered by motor proteins like (for minus-end directed apical movement) and (for plus-end directed basolateral transport), ensuring bidirectional polarity in cells like enterocytes. The process is temporally dynamic, with complete transcytosis of IgA occurring in about 30 minutes in model epithelial systems. The final step entails and at the opposing membrane, where vesicles fuse to release cargo extracellularly. This fusion is mediated by SNARE proteins, such as syntaxin-3 and SNAP-23 at the apical surface or syntaxin-4 at the basolateral, in an NSF-dependent manner that coordinates precise membrane insertion. In polarized cells, tight junctions and microtubule orientation maintain directional fidelity, preventing leakage.

Key Molecular Components

Transcytosis relies on specialized molecular components that facilitate the formation, trafficking, and fusion of vesicles across cellular barriers, particularly in endothelial and epithelial cells. Among these, caveolae represent a prominent structural feature involved in non-clathrin-mediated . Caveolae are flask-shaped plasma membrane invaginations, typically 50-80 nm in diameter, formed by the oligomerization of caveolin proteins, which create cholesterol-rich domains essential for vesicle budding and transport. Caveolin-1 (Cav-1), the primary isoform in endothelial cells, is a 21-24 kDa with a hydrophobic domain that inserts into the , enabling it to scaffold caveolae formation and interact with signaling molecules and cargoes. Cav-1 interacts with and to stabilize these structures, and its on residues regulates caveolae dynamics during transcytosis. Caveolin-2 and caveolin-3 are accessory proteins; Cav-2 co-assembles with Cav-1 in non-muscle cells to modulate caveolae stability, while Cav-3 is muscle-specific and less involved in endothelial transcytosis. In brain endothelial cells, Cav-1 knockout disrupts caveolae-mediated transport of molecules like and LDL, confirming its essential role in maintaining barrier permeability without lysosomal degradation. Recent studies as of 2025 have confirmed the dominant role of caveolar transcytosis mediated by Cav-1 in LDL transport across arterial . Receptors on the plasma membrane initiate ligand-specific transcytosis by binding cargoes and recruiting endocytic machinery. The transferrin receptor (TfR), a dimeric type II transmembrane glycoprotein, binds holo-transferrin to internalize iron-loaded complexes via receptor-mediated endocytosis, followed by sorting in early endosomes for basolateral exocytosis in polarized cells. TfR's extracellular domain features two homologous lobes that undergo pH-dependent conformational changes, releasing iron in acidic endosomes while recycling apo-transferrin; in blood-brain barrier (BBB) endothelial cells, monovalent TfR antibodies exploit this pathway for therapeutic delivery without lysosomal targeting. The polymeric immunoglobulin receptor (pIgR), a 90-120 kDa type I transmembrane protein expressed on epithelial cells, binds dimeric IgA (dIgA) or pentameric IgM via its extracellular Ig-like domains, facilitating their apical transcytosis and cleavage to release secretory IgA. pIgR's cytoplasmic tail contains tyrosine-based motifs for basolateral internalization and Rab11 recruitment, ensuring directional transport across mucosal barriers. In endothelial contexts, the LDL receptor (LDLR), a 160 kDa mosaic protein with ligand-binding repeats, mediates transcytosis of low-density lipoprotein (LDL) particles across arterial endothelium, bypassing degradation through caveolae-dependent or clathrin-independent routes. LDLR's interaction with LDL involves calcium-dependent binding, and its endocytosis is dynamin-sensitive in some vascular beds, contributing to lipid delivery without intracellular accumulation. Adaptors and coat proteins orchestrate vesicle formation and scission during transcytotic . Clathrin, a 190 kDa triskelion-shaped protein, assembles into polyhedral lattices on the cytosolic face of the plasma membrane, driving the invagination of receptor-bound cargoes like TfR in clathrin-mediated (CME), which accounts for a significant portion of transcytosis in endothelial cells. Clathrin interacts with adaptor protein 2 (AP-2), a heterotetrameric complex (α, β2, μ2, σ2 subunits) that bridges the membrane and clathrin, recognizing tyrosine- or dileucine-based sorting signals on receptors such as TfR and pIgR. AP-2's μ2 subunit binds cargo tails directly, while its α-appendage recruits accessory proteins for lattice assembly, ensuring efficient pit formation with ~100-200 clathrin molecules per vesicle. Dynamin, a 100 kDa , forms helical collars around vesicle necks, hydrolyzing GTP to constrict and sever clathrin-coated pits, releasing ~50-100 nm vesicles for transcytosis; in BBB endothelium, dynamin inhibition blocks TfR-mediated transport, highlighting its role in both clathrin- and caveolae-dependent pathways. These components collectively enable selective cargo uptake, as seen in the vesicular transport process where dynamin scission precedes Rab-mediated trafficking. Trafficking elements guide vesicles through intracellular compartments to their exocytic destinations. Rab GTPases, small monomeric GTP-binding proteins, cycle between GDP-bound inactive and GTP-bound active states to direct vesicle tethering and fusion; Rab11, localized to endosomes, promotes the sorting and apical-to-basolateral transcytosis of cargoes like TfR and IgA by recruiting effectors such as Rab11-FIP2, which links vesicles to in polarized endothelial cells. Rab11's geranylgeranylation anchors it to membranes, and its activation facilitates tubule formation from early endosomes, preventing lysosomal diversion. SNARE proteins mediate membrane fusion during ; VAMP3 (vesicle-associated membrane protein 3, or cellubrevin), a 12 kDa v-SNARE on vesicles, forms trans-SNARE complexes with plasma membrane t-SNAREs like syntaxin-4 and SNAP-23 to drive TfR vesicle fusion at the basolateral membrane in BBB endothelium. VAMP3's SNARE motif zipper-like assembly releases energy for bilayer mixing, underscoring its specificity for recycling pathways over bulk . Depletion of VAMP3 reduces transcytotic efficiency. These elements ensure vectorial transport, integrating with receptor sorting for efficient macromolecule delivery. In endothelium, CXCR2 has emerged as a mediator of transcytosis across endothelial and epithelial barriers.

Regulation

Intracellular Pathways

The PI3K-Akt signaling pathway is a key intracellular regulator of transcytosis, primarily by facilitating endosomal trafficking while diverting cargo away from lysosomal degradation pathways. Upon activation, PI3K generates phosphatidylinositol-3,4,5-trisphosphate (PIP3), which recruits Akt to the plasma , where it becomes phosphorylated and activated. This activation promotes the recruitment of effector proteins that enhance the motility and sorting of endocytic vesicles towards recycling or transcytotic compartments, thereby increasing the efficiency of transcytosis in epithelial and endothelial cells. In the context of polymeric immunoglobulin receptor (pIgR)-mediated transcytosis, PI3K-Akt signaling coordinates reorganization, ensuring proper vesicle progression from early endosomes to the basolateral . External ligands, such as factors, can initiate this pathway through receptor tyrosine kinases, linking upstream signals to intracellular control of transcytotic flux.00703-8.pdf) MAPK/ERK signaling further modulates transcytosis by controlling receptor and vesicle events, particularly in ligand-responsive systems. ERK1/2 , often downstream of receptor kinases, leads to of Rab11-family interacting protein 5 (FIP5), a key effector in the Rab11 pathway. This enhances the association of FIP5 with Rab11, promoting the directed of endosomes and facilitating vesicle at the target membrane during transcytosis. In pIgR transcytosis, this kinase cascade, involving Yes-Src and , ensures efficient apical-to-basolateral delivery of dimeric IgA by regulating the sorting and steps at common endosomes. Inhibition of ERK signaling disrupts these processes, leading to impaired receptor and reduced transcytotic throughput. Calcium-dependent pathways, mediated by and (PKC), are essential for the final stage of transcytosis, particularly in and of vesicles with the plasma . Intracellular calcium elevations bind to , inducing a conformational change that activates downstream targets involved in SNARE complex assembly and vesicle priming.62005-3/fulltext) Calmodulin antagonists like W-7 inhibit pIgR transcytosis by disrupting endosomal sorting and delivery, highlighting its role in maintaining the integrity of the transcytotic pathway. Similarly, PKC isoforms, activated by calcium and diacylglycerol, phosphorylate SNARE proteins and cytoskeletal elements to facilitate vesicle and during basolateral in endothelial transcytosis. This dual regulation ensures precise temporal control, with PKC enhancing the efficiency of caveolae-mediated transcytosis under conditions. Feedback loops involving autophagy-related proteins, such as LC3, provide adaptive modulation of transcytosis under cellular , particularly in endothelial cells. LC3 lipidation and incorporation into autophagosomal membranes can intersect with endocytic pathways, influencing vesicle fate by promoting selective of transcytotic components or cargo under high-glucose . In endothelial models, CAV1-CAVIN1-LC3B signaling drives autophagic degradation that regulates (LDL) transcytosis, reducing excessive lipid transport across the blood-brain barrier during metabolic . Recent studies emphasize that LC3-mediated acts as a mechanism, dampening transcytosis to prevent barrier dysfunction in inflammatory or hyperglycemic environments.

Environmental Influences

Ligand concentration plays a key role in modulating the rate and efficiency of transcytosis, particularly in mucosal epithelia where high levels of dimeric IgA can saturate the polymeric immunoglobulin receptor (pIgR), thereby upregulating the overall transcytotic flux across the epithelial barrier. In intestinal mucosa, elevated IgA concentrations promote increased pIgR-mediated transport, enhancing the delivery of secretory IgA to the luminal surface for immune defense, with the process showing dose-dependent kinetics up to receptor saturation. This mechanism ensures adaptive responses to varying loads without altering receptor expression directly. pH and ionic gradients within endosomal compartments critically influence transcytosis by facilitating receptor-ligand , which directs sorting toward transcytotic or pathways. In early endosomes, the acidic milieu (pH ≈ 5.3–6.0) protonates ligands and receptors, promoting their separation for many receptor-mediated systems, such as or (LDL) transport across endothelia. Ionic imbalances, including shifts in or calcium concentrations, further stabilize these gradients, ensuring efficient vesicle maturation and preventing premature fusion with lysosomes, thus preserving integrity during basolateral-to-apical or apical-to-basolateral transit.01570-5.pdf) In endothelial cells, exerted by blood dynamics modulates caveolae-mediated transcytosis, with physiological levels enhancing the formation and trafficking of caveolin-1-enriched vesicles that facilitate LDL across the vascular barrier. Recent studies indicate that laminar downregulates caveolar dynamics, decreasing LDL transcytosis rates in arterial endothelia compared to disturbed , as observed in a 2022 study modeling impacts on transport. This mechanosensitive response adapts endothelial permeability to hemodynamic conditions, balancing nutrient delivery with barrier integrity. Hormonal signals, such as insulin, influence nutrient absorption in absorptive tissues like the intestine by promoting the activity and expression of glucose transporters such as SGLT1, thereby enhancing overall in response to postprandial cues. This hormonal modulation integrates systemic metabolic needs with local transport efficiency, often linking to downstream signaling pathways for fine-tuned regulation.

Physiological Roles

In Epithelial and Endothelial Barriers

Transcytosis is essential for maintaining selective permeability at the , where it enables the receptor-mediated transport of vital nutrients while preserving the barrier's tightness against nonspecific leakage. The facilitates the endocytosis and transcytosis of iron-bound from the blood to the brain parenchyma, supplying iron for neuronal metabolism without compromising the integrity of tight junctions. Similarly, the mediates the transcytosis of insulin across BBB endothelial cells, supporting and in the . In the , transcytosis underpins mucosal immunity by directing the apical secretion of immunoglobulins. Dimeric IgA, secreted by plasma cells in the , binds to the polymeric immunoglobulin receptor (pIgR) on the basolateral of epithelial cells and undergoes transcytosis to the apical surface, where it is cleaved and released as secretory IgA to coat the gut lumen and neutralize pathogens. In neonates, the neonatal Fc receptor (FcRn) drives apical-to-basolateral transcytosis of maternal IgG from ingested milk across the , conferring during early development. Across general endothelial barriers in capillaries, transcytosis of via plasmalemmal vesicles allows controlled movement of this protein from the bloodstream to the , aiding in the maintenance of colloidal and . This vesicular pathway, observed in myocardial and other continuous endothelia, ensures that albumin transport occurs independently of paracellular routes, preserving vascular selectivity. In healthy epithelial and endothelial barriers, transcytosis contributes a small fraction to total flux, prioritizing specific macromolecular delivery over bulk permeation to uphold . This limited role, regulated by intracellular components such as those suppressing caveolae formation, underscores transcytosis's precision in supply and immune surveillance.

In Nutrient and Macromolecule Transport

Transcytosis plays a crucial role in iron by facilitating the delivery of iron-bound transferrin across endothelial barriers to support cellular functions in protected compartments. In the blood- (BBB), receptor-mediated transcytosis of transferrin-iron complexes occurs through endothelial cells, where transferrin binds to transferrin receptor 1 (TfR1) on the luminal surface, is internalized via clathrin-coated vesicles, and is transported to the abluminal side for release into the parenchyma, thereby maintaining neural iron levels essential for synthesis and production. This process is saturable and energy-dependent, with studies demonstrating that holotransferrin (iron-loaded) is preferentially transcytosed compared to apotransferrin, ensuring efficient iron supply without compromising integrity. Similarly, in the , transferrin-mediated iron supports fetal development by enabling maternal iron delivery against a concentration gradient; this involves receptor-mediated endocytosis of transferrin-iron via TfR1 on the syncytiotrophoblast apical membrane, followed by release of iron into the and basal export via , rather than classical vesicular transcytosis of the intact complex. Beyond iron, transcytosis contributes to distribution, particularly through the of apolipoprotein B (ApoB)-containing low-density lipoproteins (LDL) across arterial , which delivers to subendothelial tissues. In arterial walls, LDL particles bind to receptors or proteoglycans on endothelial cells, undergoing caveolae-mediated transcytosis to the intima, where accumulation promotes homeostasis but can initiate atherogenesis if excessive. This is regulated by endothelial uptake pathways, with ApoB serving as the primary ligand, and quantitative models indicate that transcytotic flux rates correlate with LDL levels, influencing systemic partitioning. Representative studies using human aortic endothelial cells have shown directed LDL transcytosis, underscoring its role in nutrient delivery. The neonatal Fc receptor (FcRn)-mediated handling of (IgG) in extends IgG serum through protective , distinct from transcytosis. In vascular , IgG is internalized into endosomes at neutral , where it binds FcRn at acidic (~6.0), diverting it from lysosomal degradation back to the surface for re-release into circulation, thereby prolonging to about 21 days compared to hours for non-FcRn-bound proteins. This pathway maintains high IgG levels for immune surveillance, with mutations disrupting FcRn binding reducing by over 80% in animal models. Efficiency varies by IgG subclass, with IgG1 showing optimal FcRn affinity for sustained transport. In maternal-fetal exchange, placental transcytosis is vital for transferring IgG and select nutrients, providing and developmental support to the . FcRn on cells mediates directional transcytosis of maternal IgG from the apical (maternal) to basal () side via vesicular trafficking, with peak transfer in the third ensuring fetal IgG levels reach 100-120% of maternal concentrations for protection. This process involves pH-dependent binding in endosomes, similar to endothelial , and is selective for IgG over other immunoglobulins. For nutrients, transcytotic mechanisms complement carrier-mediated transport; for instance, receptor-mediated vesicular pathways deliver macromolecules like vitamin B12-transferrin conjugates, though small nutrients like glucose primarily use —transcytosis here emphasizes macromolecular supply critical for fetal immunity and growth. Disruptions in FcRn expression reduce IgG transfer by up to 90%, highlighting its essentiality.

Role in Pathogenesis

In Vascular and Inflammatory Diseases

Dysregulated transcytosis in vascular contributes to the of various diseases by promoting excessive leakage of components into tissues, exacerbating and tissue damage. In , enhanced caveolae-mediated transcytosis of (LDL) across endothelial cells facilitates subendothelial accumulation of lipids, a critical early event in plaque formation. Caveolin-1, the principal structural protein of caveolae, drives this process; its deficiency in mouse models reduces LDL transcytosis by up to threefold in aortic segments, thereby attenuating development and formation from oxidized LDL uptake by macrophages. Recent evidence from 2025 underscores that pro-inflammatory factors like (AGEs) and interleukin-1β amplify caveolin-1 expression via signaling, increasing LDL flux and promoting -laden plaques. In , elevated transcytosis disrupts the glomerular filtration barrier, leading to as an early marker of renal dysfunction. endocytose and transcytose through caveolae and neonatal (FcRn) pathways, but in pathological states such as , this process intensifies, with angiotensin II enhancing transport of up to 86% of endocytosed to the urinary space. This abnormal transcytosis precedes structural barrier damage, increasing glomerular permeability and contributing to progressive proteinuric kidney injury. During systemic inflammation, such as in sepsis, cytokines and damage-associated molecular patterns induce endothelial transcytosis of plasma proteins, resulting in vascular leakage and edema formation. Lipopolysaccharide (LPS), a bacterial endotoxin that triggers cytokine release including TNF-α and IL-1β, phosphorylates caveolin-1 within hours, boosting albumin transcytosis across lung endothelium and causing protein-rich interstitial edema via the Starling principle. High mobility group box 1 (HMGB1), a pro-inflammatory mediator elevated in sepsis, further enhances this via the RAGE/Src/caveolin-1 axis, promoting hyperpermeability in inflamed vessels. In inflamed endothelium, transcytosis flux can increase up to 10-fold, amplifying tissue swelling and inflammatory cascades.

In Neurological and Barrier Dysfunction

In (AD), the breakdown of the (BBB) is exacerbated by the transcytosis of amyloid-β (Aβ) peptides across endothelial cells via the (RAGE). This process facilitates the influx of circulating Aβ into the brain parenchyma, promoting the accumulation of and contributing to neurodegeneration. Specifically, Aβ binding to RAGE on BBB endothelial cells triggers intracellular calcium influx and signaling, which disrupts tight junctions such as claudin-5 and , thereby increasing paracellular permeability alongside enhanced transcytosis. Experimental evidence from models of microvascular endothelial cells demonstrates that RAGE inhibition reduces Aβ transcytosis by up to 60% and preserves BBB integrity, highlighting the receptor's central role in AD . Furthermore, multimodal RAGE inhibitors have been shown to limit Aβ-mediated brain influx and monocyte adhesion in mouse models of AD, underscoring the therapeutic potential of targeting this pathway to mitigate plaque exacerbation. Dysfunction of the blood-retina barrier (BRB) in retinopathies, such as and , involves heightened VEGF-driven transcytosis, leading to vascular leakage and retinal . (VEGF) upregulates plasmalemma vesicle-associated protein (PLVAP) in retinal endothelial cells, promoting caveolae-mediated vesicular transport that allows fluid and macromolecules to cross the barrier. A 2025 review synthesizes evidence from preclinical models, including oxygen-induced in mice, where VEGF-induced PLVAP expression increases of 70 kDa tracers by enhancing transcytotic vesicles, without altering tight junctions. Inhibition of PLVAP or related pathways, such as P2X7 receptors, reduces this VEGF-mediated leakage by 40-50% in models, preserving BRB integrity and suggesting a shift from traditional therapies to more targeted transcytosis modulators. In (MS), dysregulated transcytosis across the enables the transport of inflammatory immune molecules, such as , into the , fostering and promoting demyelination. Upregulation of caveolae-mediated transcytosis in microvascular endothelial cells, driven by increased caveolin-1 expression, facilitates the shuttling of pro-inflammatory like via the Duffy antigen receptor for (DARC), amplifying T-cell infiltration and microglial activation. This enhanced vesicular transport contributes to BBB leakiness, allowing immune mediators to reach and axons, thereby accelerating demyelination in active MS lesions. Studies in experimental autoimmune models, a proxy for MS, show that inhibiting caveolin-1 reduces chemokine transcytosis and attenuates demyelination severity by limiting immune cell diapedesis. HIV-1 neuroinfection relies on transcytosis across the to establish viral reservoirs in the brain, bypassing immune surveillance and leading to chronic . The , primarily in free or cell-associated forms, undergoes in endothelial cells, followed by vesicular trafficking to the abluminal side, with approximately 17% of inoculum crossing within 4 hours via gp160 envelope-dependent mechanisms. This process involves interactions with proteoglycans and mannose-6-phosphate receptors, enabling HIV-1 to infect perivascular macrophages and without disrupting tight junctions. In models, HIV-1 transcytosis correlates with increased brain viral loads and , emphasizing its role in HIV-associated neurocognitive disorders.

Clinical Applications

Drug Delivery Across Barriers

Transcytosis serves as a key mechanism for overcoming impermeable biological barriers, such as the , to enable targeted delivery of therapeutics that cannot passively diffuse. Strategies leveraging bind ligands to endothelial receptors, triggering and vesicular transport across the cell, while adsorptive transcytosis exploits electrostatic interactions for non-specific uptake. These approaches are particularly vital for delivering large molecules like monoclonal antibodies (mAbs) to the , where the BBB restricts access in healthy states but can be exploited therapeutically. One prominent strategy targets the (TfR), highly expressed on brain endothelial cells, using conjugated nanoparticles to ferry mAbs across the BBB for brain tumor treatment. TfR-conjugated liposomes or gold nanoparticles bind the receptor on the luminal surface, initiating clathrin-independent and subsequent transcytosis to the abluminal side, allowing payload release into the parenchyma. For instance, low-affinity anti-TfR antibodies on gold nanoparticles achieved 0.23% injected dose per gram (ID/g) accumulation in models, enabling targeted delivery of chemotherapeutic mAbs while minimizing peripheral off-target effects. This approach has shown nearly two-fold higher uptake in brain capillaries compared to non-targeted carriers, enhancing tumor-specific in preclinical studies. Nasal-to-brain delivery via adsorptive transcytosis offers a non-invasive alternative, particularly for therapeutics, by administering drugs through the to bypass the . Polymeric micelles, such as those loaded with , leverage positive surface charges or cell-penetrating peptides to promote electrostatic adsorption and paracellular transport along olfactory nerves, facilitating direct CNS entry. In rat models, rotigotine-encapsulated micelles in thermosensitive gels demonstrated significantly enhanced brain bioavailability compared to , improving motor function restoration with reduced systemic exposure. This route exploits natural transcytotic pathways in nasal epithelia, achieving up to several-fold higher striatal drug levels relevant for therapies. Overall, RMT-based strategies have demonstrated uptake improvements of up to 5-10 fold over non-targeted methods in select models, though absolute parenchymal remains below 1% ID/g, underscoring the need for affinity optimization to enhance therapeutic indices. As of 2025, advancements include Phase III trials for TfR-targeted bispecific antibodies (e.g., for ), showing promising CNS penetration in human studies.

Therapeutic Modulation Strategies

Therapeutic modulation of transcytosis involves targeted interventions to either inhibit or enhance the process, aiming to mitigate progression by altering the of macromolecules across endothelial or epithelial barriers. Inhibitors primarily focus on reducing pathological transcytosis, such as that of (LDL) in , where excessive LDL entry into the arterial intima initiates plaque formation. blockers, which disrupt dynamin-dependent essential for caveolae-mediated transcytosis, have shown promise in preclinical models; for instance, Dyngo-4a at 30 μmol/L nearly eliminates LDL transcytosis in coronary artery endothelial cells, while dynasore halves transcytosis of LDL-sized tracers in human aortic endothelial cells. Similarly, inhibition of caveolin-1, a key structural protein in caveolae, reduces endothelial permeability to LDL; genetic knockout of caveolin-1 in mice results in approximately 50% less LDL uptake in aortas, and pharmacological agents like statins decrease caveolin-1 expression by about 75% at 10 nmol/L concentrations. Anti-ALK1 antibodies, targeting activin receptor-like kinase 1 involved in LDL binding and transcytosis, reduce LDL transcytosis by around 50% and area by 50% in LDL receptor-deficient mice, highlighting a potential antibody-based strategy. Enhancement strategies aim to prolong the half-life of therapeutic immunoglobulins in conditions like autoimmunity, where engineered interactions with the neonatal Fc receptor (FcRn) can extend IgG circulation without upregulating endogenous FcRn via . FcRn binds IgG at acidic pH in endosomes, it to extend serum half-life; modifications to the Fc region of therapeutic antibodies improve pH-dependent FcRn binding kinetics, dramatically prolonging half-life—for example, optimized variants achieve up to threefold extension compared to wild-type IgG in preclinical models. This approach is particularly relevant for autoimmunity treatments, where extended half-life of anti-pathogenic IgG (e.g., in or ) enhances efficacy without broadly affecting endogenous autoantibodies, as opposed to FcRn blockade strategies that reduce pathogenic IgG levels. Although direct for FcRn upregulation remains exploratory and lacks clinical validation, antibody engineering serves as a practical enhancer for transcytosis-mediated IgG . Clinical trials have advanced modulation strategies, particularly for renal and ocular diseases linked to dysregulated transcytosis. In diabetic nephropathy, where albumin transcytosis across glomerular endothelial cells contributes to albuminuria, phosphoinositide 3-kinase (PI3K) pathway inhibition shows potential; preclinical data indicate that PI3K/AKT suppression reduces albumin endocytosis and transcytosis, alleviating proteinuria in models of diabetic kidney disease. For retinopathy, anti-vascular endothelial growth factor (anti-VEGF) therapies control VEGF-induced transcytosis and vascular leakage; intravitreal aflibercept (VEGF-trap) reduces endothelial barrier disruption in proliferative diabetic retinopathy models, with Phase III trials (e.g., VISTA and VIVID) demonstrating sustained visual acuity improvements through decreased macular edema via transcytosis modulation. Safety considerations in transcytosis modulation emphasize avoiding unintended barrier disruption, which could exacerbate leakage or . Inhibiting caveolae-mediated pathways, as with dynamin blockers, risks off-target effects on general , potentially impairing nutrient transport or causing endothelial instability, as observed in caveolin-1 knockout models with altered . Enhancing FcRn interactions requires careful dosing to prevent excessive IgG accumulation, which might promote immune complex deposition in . In clinical contexts like therapy, repeated intravitreal administration can lead to transient barrier weakening or , necessitating monitoring for long-term endothelial integrity. Overall, balancing efficacy with barrier preservation involves preclinical validation of selectivity, as non-specific modulation may heighten risks of vascular dysfunction in diseases already involving pathogenic transcytosis.

References

  1. [1]
  2. [2]
    Transcytosis - an overview | ScienceDirect Topics
    Transcytosis is defined as a mechanism of material transport through a cell that involves endocytosis and exocytosis, allowing relatively large substances, ...
  3. [3]
    Transcytosis at the Blood-Brain Barrier - PMC - PubMed Central
    Jan 30, 2019 · Transcytosis is the transcellular transport of molecules via vesicles. Macromolecules are first endocytosed or internalized by vesicles on one ...Missing: definition | Show results with:definition
  4. [4]
    Transcytosis to Cross the Blood Brain Barrier, New Advancements ...
    Jan 10, 2019 · Transcytosis is a phenomenon present in many different cell types, from neurons to intestinal cells, osteoclasts and endothelial cells. In ...
  5. [5]
    Transcytosis in the development and morphogenesis of epithelial ...
    Transcytosis is a form of specialized transport through which an extracellular cargo is endocytosed, shuttled across the cytoplasm in membrane‐bound vesicles, ...
  6. [6]
    Lung Endothelial Transcytosis - PMC - PubMed Central
    Introduction. Transcytosis, or transcellular vesicular transport, generally describes the intracellular movement of relatively large (>10 kDa) macromolecules ...
  7. [7]
    Channels across Endothelial Cells - NCBI - NIH
    At its simplest, transcytosis is the transport of macromolecular cargo from one side of a polarized cell to the other (e.g., apical to basolateral) within ...
  8. [8]
    A targeted RNAi screen identifies factors affecting diverse stages of ...
    Transcytosis has been best characterized for basolateral-to-apical transport of dimeric IgA (dIgA) by the polymeric immunoglobulin receptor (pIgR; Rojas and ...
  9. [9]
    The kinesin KIF16B mediates apical transcytosis of transferrin ... - NIH
    Jun 7, 2013 · LDL receptor undergoes apical transcytosis to ARE in AP-1B KD MDCK ... transcytosis of IgA in MDCK cells is via apical recycling endosomes.
  10. [10]
    A New Function for the LDL Receptor: Transcytosis of LDL across ...
    In the present study, we provide direct evidence that after binding to brain capillary ECs, there is a specific mechanism for the transport of LDL across the ...Missing: examples | Show results with:examples
  11. [11]
    Transcytosis - an overview | ScienceDirect Topics
    Transcytosis is a process that takes material from one side of a cell and transports it in the form of a membrane-coated vesicle through the cell for its ...
  12. [12]
    An electron microscopic study of the intestinal villus. II. The pathway ...
    The intestinal pathway for absorbed fat was traced in thin sections of intestinal villi from rats fed corn oil by stomach tube after a fast of 24 to 40 hours.Missing: early 1950s 1960s
  13. [13]
  14. [14]
    Caveolae-Mediated Transcytosis and Its Role in Neurological ...
    Mar 21, 2025 · Transcytosis is the transcellular transportation of molecules via vesicles, allowing macromolecules to pass between the circulation and the ...2. Caveolae-Mediated... · 2.1. Endocytosis · 3. Caveolae-Mediated...<|control11|><|separator|>
  15. [15]
    Molecular determinants of endothelial transcytosis and their role in ...
    Oct 1, 2007 · Transcytosis via caveolae is an important route for the regulation of endothelial barrier function and may participate in different vascular diseases.Missing: original | Show results with:original
  16. [16]
    Caveolae and caveolin-1 mediate endocytosis and transcytosis of ...
    Sep 13, 2010 · To explore the mechanisms involved in ox-LDL transcytosis across endothelial cells and the role of caveolae in this process.Ox-Ldl Binding And... · Caveolae Inhibitors... · Caveolae Play A Crucial Role...
  17. [17]
    Molecular architecture determines brain delivery of a transferrin ...
    Feb 28, 2022 · The precise molecular mechanisms underlying TfR-mediated transcytosis remain to be fully elucidated (Villasenor et al., 2019). Bivalent receptor ...
  18. [18]
    Role of Polymeric Immunoglobulin Receptor in IgA and IgM ...
    Since its function was first discovered in the 1980s, J chain, along with the molecular details of Ig polymerization, has largely been overlooked in the field ...
  19. [19]
    Transcytosis of LDL across the Blood–Brain Barrier | Journal of Cell ...
    The nondegradation of the LDL during the transcytosis indicates that the transcytotic pathway in brain capillary endothelial cells is different from the LDL ...
  20. [20]
    Intracellular transport and regulation of transcytosis across the blood ...
    Dec 6, 2018 · The blood–brain barrier is a dynamic multicellular interface that regulates the transport of molecules between the blood circulation and the brain parenchyma.
  21. [21]
    The β2-adrenergic receptor/βarrestin complex recruits the clathrin ...
    These findings point to a role for AP-2 in GPCR endocytosis, and they suggest that AP-2 functions as a clathrin adaptor for the endocytosis of diverse classes ...
  22. [22]
    Endosomal trafficking regulates receptor-mediated transcytosis of ...
    Peptides identified for Rab11 are common between Rab11a and Rab11b. ... transcytosis through brain capillary endothelial cells in vitro. Endothelium ...
  23. [23]
    Vamp3/syntaxin 4 mediates the basolateral membrane fusion of TfR ...
    Jul 2, 2025 · VAMP3 Contributes to the Transcytosis of Tf Across the BBB. VAMP1, VAMP2, VAMP3, VAMP4, VAMP7, and VAMP8 are major v-SNARE proteins involved in ...
  24. [24]
    Intracellular transport and regulation of transcytosis across the blood ...
    Dec 6, 2018 · Genetic knock-out of Mfsd2a in mice led to a marked increase in both brain endothelial caveolae and transcellular permeability in the brain ...<|control11|><|separator|>
  25. [25]
    Cdc42 and the Phosphatidylinositol 3-Kinase-Akt Pathway Are ...
    The dIgA-stimulated pIgR transcytosis is regulated by Rho family GTPases, phosphatidylinositol-3-kinase (PI3K), and requires the production of secondary ...
  26. [26]
    Overview of Crosstalk Between Multiple Factor of Transcytosis in ...
    Jan 21, 2020 · Activation of VEGF/PI3K/Akt pathway may induce actin reorganization in human angioma cells (Wang et al., 2011), a process known to be crucial ...
  27. [27]
    Article ERK1/2 Regulate Exocytosis through Direct Phosphorylation ...
    May 15, 2012 · At the sorting endosomes, ERK1/2 phosphorylation of Rab11-FIP5 controls transcytosis of the polymeric immunoglobulin receptor in epithelial ...
  28. [28]
    [PDF] A kinase cascade leading to Rab11-FIP5 controls transcytosis of the ...
    Our results reveal a novel Yes–EGFR–ERK–FIP5 signalling network for regulation of pIgA–pIgR transcytosis. Membrane traffic must be tightly, yet flexibly, ...
  29. [29]
    The calmodulin antagonist W-7 affects transcytosis, lysosomal ...
    Nov 18, 1994 · The present study provides evidence for a role of calmodulin in several steps of membrane transport along the endocytic pathway.
  30. [30]
    CAV1-CAVIN1-LC3B-mediated Autophagy Regulates High Glucose ...
    Our results reveal that high glucose stimulates LDL transcytosis by a novel CAV1-CAVIN1-LC3B signaling-mediated autophagic degradation pathway.
  31. [31]
    CAV1-CAVIN1-LC3B-mediated autophagy regulates high glucose ...
    In our study, we first evaluated the effects of high glucose on lipid transcytosis across endothelial cells and lipid retention. Our results demonstrated that ...
  32. [32]
    PI3KCIIα-Dependent Autophagy Program Protects From Endothelial ...
    Dec 28, 2023 · Autophagy activation is triggered by low shear stress and high shear stress in endothelial cell, but required different PI3K (phosphoinositide 3 ...
  33. [33]
    studies of diffusion, binding to the human polymeric Ig receptor, and ...
    Binding data showed that pIgR bound pIgA and pIgM with similar affinity. Internalization of both ligands was fast and took place at similar rates; transcytosis ...
  34. [34]
    Development of a primary mouse intestinal epithelial cell monolayer ...
    Nov 13, 2013 · An LPS dose–response curve was performed to determine whether (d) IgA transcytosis and (e) pIgR expression were dose-dependent. All LPS-treated ...
  35. [35]
    Receptor Cell Biology: Receptor-Mediated Endocytosis - Nature
    Many of the ligand- receptor complexes dissociate within this acidic environment, thus providing a mechanism for the sorting of ligand from receptor.
  36. [36]
    Caveolin-1 is a primary determinant of endothelial stiffening ... - Nature
    Oct 24, 2022 · This study demonstrates that Cav-1 plays a critical role in endothelial stiffening induced by oxLDL in vitro and by dyslipidemia, disturbed flow and ageing in ...
  37. [37]
    Mechanisms of Glucose Absorption in the Small Intestine in ... - MDPI
    In addition, insulin infused in the portal vein rapidly increased absorption of glucose in the small intestine. It has been suggested that this stimulation is ...Missing: transcytosis | Show results with:transcytosis<|control11|><|separator|>
  38. [38]
    Pancreatic endocrine and exocrine signaling and crosstalk ... - Nature
    Feb 14, 2025 · In its basal state, GLUT4 is stored in vesicles within the cell, and upon insulin binding to IR, the number of GLUT4-containing vesicles that ...
  39. [39]
    Transcytosis to Cross the Blood Brain Barrier, New Advancements ...
    Jan 11, 2019 · In polarized cells, unidirectional transcytosis refers to the transport of macromolecules from apical to basolateral plasma membranes.
  40. [40]
    Regulation of the polymeric immunoglobulin receptor and IgA ...
    Secretory IgA (SIgA) is generated by cooperation between two distinct cell types in mucous membranes: plasma cells in the lamina propria that secrete dimeric ...
  41. [41]
    FcRn-mediated antibody transport across epithelial cells revealed ...
    In newborn rodents, FcRn transfers IgG from milk to blood by apical-to-basolateral transcytosis across intestinal epithelial cells.
  42. [42]
    Transcytosis of albumin in capillary endothelium - PMC - NIH
    polymeric albumin transcytosis was obtained using albumin-gold complexes. The results are discussed in terms of vesicular transport of albumin across the ...
  43. [43]
    Receptor-mediated transcytosis of transferrin through blood-brain ...
    These results clearly show that the iron-Tf complex is transcytosed across brain capillary endothelial cells by a receptor-mediated pathway without any ...
  44. [44]
    The placenta: the forgotten essential organ of iron transport - NIH
    In addition to maternal transferrin-iron, other circulating forms of iron such as non-transferrin-bound iron and heme may also be taken up by the placenta.
  45. [45]
    Transcytosis of LDL Across Arterial Endothelium: Mechanisms and ...
    Feb 27, 2025 · Transport of LDL (low-density lipoprotein) from plasma to arterial intima is thought to be rate limiting in the development of atherosclerosis.
  46. [46]
    Transport of low-density lipoproteins (LDL) into the arterial wall - NIH
    The transcytosis of low-density lipoprotein (LDL) across the endothelium and its accumulation in the arterial wall is the initial step of atherosclerosis.
  47. [47]
    Endothelial Transcytosis of Lipoproteins in Atherosclerosis - Frontiers
    A number of studies have recently demonstrated that low density lipoprotein (LDL) transcytosis across the endothelium is dependent on the function of caveolae, ...
  48. [48]
    The recycling and transcytotic pathways for IgG transport by FcRn ...
    May 18, 2009 · Another hallmark of FcRn function is that the receptor sorts IgG away from lysosomes, explaining why IgG has the longest half-life of any ...
  49. [49]
    The Neonatal Fc Receptor (FcRn): A Misnomer? - Frontiers
    Albumin injected into hypoalbuminemic individuals shows a half-life 4–5-fold longer than normal, which is in line with the rate of albumin salvage being also ...
  50. [50]
    The therapeutic age of the neonatal Fc receptor - Nature
    Feb 1, 2023 · FcRn blockade is a novel and effective strategy to reduce circulating levels of pathogenic IgG autoantibodies and curtail IgG-mediated diseases.
  51. [51]
    Factors Affecting the FcRn-Mediated Transplacental Transfer of ...
    Oct 13, 2017 · To be successfully transferred across the placenta, maternal IgG must cross the synctiotrophoblast layer, the villous stroma, and the fetal ...
  52. [52]
    Transplacental delivery of therapeutic proteins by engineered ...
    Transplacental delivery of maternal immunoglobulin G (IgG) provides humoral protection during the first months of life until the newborn's immune system ...
  53. [53]
    IgG Placental Transfer in Healthy and Pathological Pregnancies - PMC
    Placental transfer of maternal IgG antibodies to the fetus is an important mechanism that provides protection to the infant while his/her humoral response ...Missing: exchange | Show results with:exchange
  54. [54]
  55. [55]
    A New Pathogenesis of Albuminuria: Role of Transcytosis
    Jun 15, 2018 · Subsequently, in 1979, Simionescu et al. [2] used the term “transcytosis” to describe the vectorial transport of macromolecules within ...Transcytosis By Gecs · Transcytosis By Podocytes · Transcytosis By Ptcs<|control11|><|separator|>
  56. [56]
    Endothelial Transcytosis in Acute Lung Injury - PubMed Central - NIH
    Inflammation likely indirectly increases endothelial transcytosis, as neutrophil binding to intercellular adhesion molecule 1 (ICAM1) in vivo increases ...<|control11|><|separator|>
  57. [57]
    High Mobility Group Box Protein 1 Boosts Endothelial Albumin ...
    Aug 30, 2016 · Albumin leakage via the paracellular pathway is a result of damage to cell-cell junctions, whereas albumin transcytosis results from endocytosis ...<|control11|><|separator|>
  58. [58]
    Apolipoprotein M-bound sphingosine-1-phosphate regulates blood ...
    Nov 25, 2019 · Furthermore, we report an up to 10–fold increase in adsorptive transcytosis in the endothelial cells of pial and penetrating arterioles, but not ...
  59. [59]
    Receptor-mediated transcytosis for brain delivery of therapeutics
    In this review, we summarize well-studied RMT pathways, and explore mechanisms engaged in BBB transport by various RMT receptors.
  60. [60]
    Blood–Brain Barrier Transport of Transferrin Receptor-Targeted ...
    The recent evidence for passing targeted nanoparticles through the BBB shows great promise for future drug delivery of biologics to the brain.
  61. [61]
    Antibody-conjugated polymer nanoparticles for brain cancer
    Aug 20, 2025 · Antibody-conjugated nanoparticles enable the selective targeting of brain cancer cells by exploiting surface receptors that are overexpressed ...
  62. [62]
    Polymeric nanocarriers for nose-to-brain drug delivery in ...
    This work reviews the use of polymeric nanocarriers for intranasal (nose-to-brain) drug delivery in neurodegenerative diseases and neurodevelopmental disorders.
  63. [63]
  64. [64]
    Caveolae-Mediated Transport at the Injured Blood-Brain Barrier as ...
    Liposomal nanocarriers have also been shown to traffic through caveolae following stroke and can deliver both hydrophobic and hydrophilic compounds. In each ...
  65. [65]
    Pathology-directed drug delivery strategies: How to overcome blood ...
    Oct 22, 2025 · Selective liposomal transport through blood brain barrier disruption in ischemic stroke reveals two distinct therapeutic opportunities. ACS ...
  66. [66]
    Selective Liposomal Transport Through Blood Brain Barrier ...
    Mar 9, 2019 · We show selective recruitment of clinically used liposomes into the ischaemic brain that correlates with biphasic blood brain barrier (BBB) breakdown.Missing: mimicking crossing
  67. [67]
    Increased brain uptake of targeted nanoparticles by adding an acid ...
    We show a method for increasing the ability of high-avidity, transferrin (Tf)-containing nanoparticles to enter the brain through transcytosis.Abstract · Sign Up For Pnas Alerts · Results
  68. [68]
    Engineering FcRn binding kinetics dramatically extends antibody ...
    Apr 18, 2025 · The prolonged serum half-life of IgG antibody is governed by its pH-dependent interaction with FcRn, enabling efficient binding at acidic ...
  69. [69]
    effects on albumin transcytosis across glomerular endothelial cells
    These studies provide the first evidence of interference with albumin transcytosis across GECs as a novel approach to the treatment of diabetic albuminuria.
  70. [70]
    PI3K Inhibitor Pipeline Outlook 2025: Clinical Trial Studies,
    Aug 26, 2025 · DelveInsight's, "PI3K Inhibitor Pipeline Insight 2025" report provides comprehensive insights about 20+ companies and 25+ pipeline drugs in ...
  71. [71]
    Dll4 Suppresses Transcytosis for Arterial Blood-Retinal Barrier ...
    Feb 12, 2020 · We revealed the association between VEGF signaling and transcytosis in HR and suggest that transcytosis is implicated in other retinal diseases ...
  72. [72]
    VEGF-targeting drugs for the treatment of retinal neovascularization ...
    Main clinical trials dedicated on the assessment of anti-VEGF molecules in diabetic retinopathy. ... The clinical status, including glycemic control and ...
  73. [73]
    The role of transcytosis in the blood-retina barrier - PubMed Central
    Transcytosis, also known as transcellular vesicular transport, refers to the intracellular transportation of large molecules (>10 kDa) within vesicles ...2 Transcytosis In Healthy... · 5 Transcytosis In... · Glossary
  74. [74]
    Modulation of the Blood–Brain Barrier for Drug Delivery to Brain - PMC
    This review describes conventional and emerging BBB modulation strategies and related mechanisms, and safety issues according to BBB structures and functions, ...Missing: considerations | Show results with:considerations