1,983
Views
47
CrossRef citations to date
0
Altmetric
Review

Trends in drug delivery through tissue barriers containing tight junctions

, &
Article: e24565 | Received 22 Feb 2013, Accepted 04 Apr 2013, Published online: 01 Apr 2013

Abstract

A limitation in the uptake of many drugs is the restricted permeation through tissue barriers. There are two general ways to cross barriers formed by cell layers: by transcytosis or by diffusion through the intercellular space. In the latter, tight junctions (TJs) play the decisive role in the regulation of the barrier permeability. Thus, transient modulation of TJs is a potent strategy to improve drug delivery. There have been extensive studies on surfactant-like absorption enhancers. One of the most effective enhancers found is sodium caprate. However, this modulates TJs in an unspecific fashion. A novel approach would be the specific modulation of TJ-associated marvel proteins and claudins, which are the main structural components of the TJs. Recent studies have identified synthetic peptidomimetics and RNA interference techniques to downregulate the expression of targeted TJ proteins. This review summarizes current progress and discusses the impact on TJs' barrier function.

Introduction

Tissue barriers

To guarantee specific organ function, the body must maintain compartments with a special environment and gradients for various kinds of solutes and ions. To sustain these gradients, organs have layers of cells which form barriers between the environment and the organ tissue. Most of these are epithelial cells such as the brush border cells in the intestine or the cells in the lining of the kidney tubules. Others are the endothelial cells which line blood vessels, forming the blood-brain barrier or the blood-retina barrier, for instance. Other examples are the Sertoli cells forming the blood-testis barrier or Schwann cells which electrically seal the myelin sheaths of neurons. The common function of these barrier-forming cells is to regulate the transport of water, ions, nutrients and other solutes, in order to maintain the organ-specific environment in the steady-state. In addition, noxious agents and metabolic end products are removed from the organ tissue by means of efflux transporters. However, these functions may also impair the uptake of many drugs.

Barrier-forming cells are polarized. They possess an apical region facing the outer lumen and a basolateral plasma membrane region facing the organ side. Proteins (i.e., transport proteins, channels and receptors) within the apical plasma membrane cannot move to the basolateral side and vice versa. The protein patterns in these membrane compartments are different and they fulfil different functions, i.e., in the directed transport of molecules. This so-called fence function is due to a protein complex, the tight junctions (TJs), which are organized in strand networks. TJ-strands span the plasma membrane of adhering cells, like a belt at the interface between the apical and basolateral sites. TJs also establish a barrier in the extracellular space between neighboring cells by interactions between their TJ-strand networks. In consequence, these cells together with their TJs form a barrier which regulates transcellular flux indirectly and paracellular flux directly. Transcellular permeability is limited by the lipid surface of the cell and the outwards or inwards directed transporter activity (i.e., ABC transporters or Glut-1).Citation1 In general, only small lipophilic molecules are passively absorbed across barriers by the transcellular pathway if they are not a substrate of any of the transporters. Paracellular permeability can differ greatly between the diverse tissue barriers, depending on the composition of the expressed TJ proteins and on their expression level. Thus, TJs regulate paracellular passage of small hydrophilic substances, ions and water. However, large hydrophilic substances need an active permeation process, e.g., carrier-mediated transport.Citation2-Citation5

To improve the delivery of pharmaceutical agents through tissue barriers, drug enhancing methods have been developed. The largest body of information on a drug enhancer is available for caprate, as this is the best investigated agent for this purpose and has been used in clinical studies. Therefore, the main part of this review concerns caprate, as this is the standard approach in the field.

Tight junctions and claudins

TJ-complexes mainly consist of the transmembrane proteins occludin, tricellulin, marvelD3, as well as the claudin protein family and various scaffolding proteins, such as zonula occludens proteins 1–3 (ZO-1 to 3).Citation6-Citation9 Occludin was the first transmembrane TJ-protein discovered.Citation10 This is specifically located in TJs and plays a regulatory role. Nonetheless, since occludin-knockout mice are viable and fail to display defective barrier function, its physiological importance is still unclear. It may well be relevant to regulation under pathological circumstances, as it has multiple phosphorylation sites.Citation11,Citation12 In addition, occludin can be partially replaced by the other tight junction- associated marvel protein (TAMP) family members, tricellulin and/or marvelD3.Citation6 Tricellulin has recently been shown to be mainly localized at tricellular contacts.Citation13 Knockdown of tricellulin expression by RNA interference technology leads to impaired TJ organization, a decrease in transcellular electrical resistance (TER) and a size-selective increase in permeability.Citation14 Thus, tricellulin plays an important role in sealing tricellular contacts in epithelial barriers. However, a more essential TJ-protein family has been identified—the claudins.Citation15 The claudin family is currently thought to consist of up to ~27 members.Citation16 Claudins have a dominant influence on the properties of the TJs. They are expressed in a tissue-dependent combination, which results in tissue-specific barrier characteristics, since some claudins lead to a leakier barrier and others to a tighter barrier. Thus, the functional backbone of the TJ is formed by the claudins.Citation5

Claudins and TAMPs consist of two extracellular domains (ECDs) and four transmembrane domains, whereas the C- and N-terminal domains are located in the cytoplasm. Many of the claudins can bind via the C-terminus to the PDZ domain of the scaffolding protein ZO-1, which links them to the actin cytoskeleton. ZO-1 and -2 are essential for the spatial organization, but not for the formation of the claudin-based TJ-strands.Citation17 To form these strands, the claudins can interact in different ways, either trans (interaction between claudins in two neighboring cells) and cis (established between claudins along the same membrane).Citation18,Citation19 There are also homo- and heterophilic interactions between claudins and TAMPs.Citation20,Citation21

Although some claudins have very similar sequences, their expression can lead to quite different TJ characteristics. For example, claudin-2 is known to be involved in paracellular pore formation for cations, whereas claudin-5 seals the paracellular cleft for small molecules.Citation22,Citation23 Therefore, detailed information is necessary about the structural differences between claudins, especially their ECDs. In this context, some studies point to the importance of both ECDs for proper tightening of the paracellular cleft,Citation24 ion selectivityCitation25-Citation27 and the involvement in strand formation.Citation18,Citation28 In summary, claudins, particularly their ECDs, are potent targets for manipulating paracellular permeability.

Substances to Modulate Tight Junctions

In general, many organs are protected by a barrier to avoid contact with unwanted or even toxic substances. Regrettably, this means that drugs are also less able to reach their destination. This is true for the blood-brain barrier, which prevents the uptake of almost all large molecules and more than 98% of small molecule drugs into the brain.Citation29 Consequently, only small (< 500 Da) lipophilic drugs with fewer than 8–10 hydrogen bond to solvent water are believed to cross the blood-brain barrier.Citation30 Most designed drugs are only poorly absorbed, due to their size or hydrophilicity. To face this problem of low bioavailability, two major strategies are being pursued in pharmacological research. One is to modify and couple drugs so that they can cross a barrier. The other strategy is to use an absorption enhancer which increases barrier leakiness.

The first strategy is implemented, for example, by employing prodrugs. These are initially inactive compounds, which are modified to raise transcellular permeability, either by increased lipophilicity or by coupling to a ligand, which allow the drug to use a transporter.Citation31 After uptake, those drugs are activated, e.g., by enzymatic conversion. Other compounds achieve carrier-mediated uptake after modification, e.g., nanoparticles conjugated to aminosubstituted vitamin B12 derivatives.Citation32

Whereas the first strategy attempts to exploit transcellular uptake, the second strategy focuses on the paracellular pathway. For transient modification of the TJs, promising candidates have been described in the recent years: small molecules,Citation33-Citation35 peptidesCitation36 and small interfering ribonucleic acid (siRNA).Citation37 These are specifically directed against TJ molecules, especially against claudins, in contrast to indirect TJ modulation by the calcium chelators ethylenediamine tetraacetic acid/ethylene glycol tetraacetic acid (EDTA/EGTA)Citation38 or the clinically used hyperosmolar mannitol.Citation39 It has been demonstrated that hyperosmolar saline solution transiently enhances analgesic drug delivery through the perineural barrier in rats.Citation40

Since the claudins make the dominant contribution to the function of the TJs, specific targeting of the claudins is an obvious and promising approach. Additionally, the tissue-specific expression of the claudins allows tissue-barrier specific modulation, which should lead to fewer side effects.

Surfactant-like agents

Various agents, such as surface active agents (surfactants), lipids and polymers, have been tested for their efficiency in improving the systemic availability of drugs.Citation33 So far only capric acid (C10H20O2) has achieved clinical relevance.

Sodium caprate (C10), the sodium salt of capric acid, also known as sodium decanoate, has been approved (in Japan and Sweden) as an absorption enhancer in a rectal suppository for the antibiotic ampicillin, with the trade name Doktacillin®.Citation41 Lindmark and colleagues showed that 50 mg/person C10 significantly increased ampicillin concentration in the serum of adult humans.Citation42 This absorption enhancement C10 was also clinically demonstrated for oral applications.Citation42 Since capric acid is present in dairy products, e.g., milk and plant oils (i.e., coconut oil), C10 is accepted by the US American Food and Drug Administration for use in food (Sec. 172.863 Salts of fatty acids. http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfCFR/CFRSearch.cfm?CFRPart=172&showFR=1; 2013).

C10 is an amphiphilic substance, with a hydrophilic “head” (carboxylate ion) and a hydrophobic “tail” (aliphatic C10 chain) and has a critical micelle concentration of about 50 mM. At this concentration, C10 may improve the solubility of hydrophobic substances and acts like a detergent.Citation43 C10-micelles may enhance in vivo absorption via transcellular transport.Citation44 On the other hand, it is well established that C10 widens TJs, thus serving to open the barriers.Citation34 Several in vivo studies have shown that this enhances drug uptake ().

Table 1. Increased drug uptake by caprate in vivo

The drug berberine is normally poorly absorbed after oral administration. Two studies have found that its bioavailability is enhanced when it is orally administered together with C10. The intestinal mucosa is not damaged.Citation45,Citation46 The in vitro enhancement ratios were 2.08, 1.49 and 3.49 in the rat duodenum, jejunum and ileum, respectively (C10: 0.2% w/v).Citation46 C10 (100 mg/kg) also increases the permeability for nanoparticles across the intestinal epithelium of ratsCitation47 and for chemically modified antisense oligonucleotides (C10: 25, 50 and 100 mg/kg) in pigsCitation48 after enteral application. After intraduodenal administration, the uptake of nucleotide prodrugs into the liver was enhanced (C10: 86 mg/kg).Citation49 Intracarotid injections of C10 lead to a transient, reversible and molecular weight-dependent opening of the blood-brain barrier in the rat, beginning 5 min after injection.Citation50,Citation51 After epidural injection of C10 with the anesthetic ropivacaine, the maximal intrathecal concentration of ropivacaine was elevated.Citation52 This effect is transient and reversible and limited to the arachnoid barrier.

Cell culture experiments have been performed to characterize the detailed mode of action of C10. These studies have mostly used cells from the colonCitation53,Citation54 or kidney,Citation55,Citation56 but also freshly isolated brain capillariesCitation55 and excised rat intestinal mucosa.Citation57 In these experiments TER decreased and/or the permeability of molecules with a molecular weight between 182 and 10,000 Da increased after incubation with C10 ().

Table 2. Increased drug uptake by caprate in vitro

The decrease in TER depends on the C10 concentration, the incubation time and the cell line used. 7.5–10 mM C10 seems to be the optimal concentration for MDCK and HT-29/B6 cells. Lower concentrations are less effective and higher concentrations do not decrease TER further. The maximum effect of C10 on TER is already reached within 5–30 min (50% of control value) and lasts for up to 300 min. After removal of C10, the TER returns to control values if the initial C10 concentration is below 30 mM.Citation56 In Caco-2 cells, concentrations of 25 mM to 50 mM C10 are necessary to increase permeability.Citation53 In all studies, C10 caused much higher permeation ratios for small molecules (up to 1 kDa) than for large molecules (4 to 20 kDa).Citation53,Citation54,Citation57 This biphasic permeability corresponds to the size-dependent permeation of molecules through Caco-2 or human intestinal epithelial (T84) monolayers, as shown by Watson and colleagues. A permeation study was performed with 24 polyethylene glycols of different molecular weight (238 to 1250 Da; ~3.5 to 7.39 Å). Permeation decreased markedly between 238 and 326 Da, but remained at a constant low value with the larger oligomers. After C10 treatment, the permeation profile was still biphasic, but was enhanced 6-fold over the whole size range. EGTA treatment also caused increased permeability, but no size selectivity was observed.Citation58 However, molecules >20 kDa did not permeate after C10 treatment.Citation54

Thus, C10 increases the permeability of cell monolayers in a biphasic, size-selective manner. This observation points toward modulation of the paracellular, rather than the transcellular pathway after C10 treatment.Citation54

The mode of action of C10 was illustrated by Maher et al.Citation44 In their model, C10 at concentrations up to 50 mM induces contraction of the actomyosin perijunctional ring, dilating the paracellular space.Citation34 This effect is mediated by an increase in the intracellular Ca2+ level, caused by phospholipase C activation and the subsequent cleavage of phosphatidylinositol 4,5-bisphosphate (PIP2) into inositol triphosphate (IP3) and diacylglycerol (DAG).Citation59 Intracellular Ca2+ forms complexes with calmodulin (CaM) in response to IP3, which in turn activate CaM-dependent protein kinases and the myosin light chain kinase.Citation60 Phosphorylation of the regulatory light chain of myosin II induces the contraction of the actomyosin perijunctional ring.Citation61 Contraction of the actin myosin ring complex is also induced by another signaling pathway, mediated by DAG and protein kinase C.Citation62 The TJ complex linked by ZO-1 to the actin cytoskeleton is then redistributed from the TJs to the cytoplasm.Citation34,Citation60,Citation62-Citation64

However, recent reports have shown that this model cannot be completely true as a general mode of action. In HT-29/B6 cells, for example, no morphological change in the perijunctional actomyosin ring could be observed after incubation with 10 mM C10.Citation54 Furthermore, the effect of C10 could not be blocked with inhibitors of the actin-myosin interaction, phospholipase C, Rho-kinase or by Ca2+ chelators.Citation54 The redistribution of the TJ-complex is under discussion as well, since several studies have reported that the membrane localization of ZO-1 is unchanged (30 mM C10Citation65; 7.5 mM C10Citation55). However, claudin-1, claudin-4, junctional adhesion molecule and β-catenin in human airway epithelial cells and claudin-5 in brain endothelial cells were reduced after C10 treatment.Citation55,Citation65 In MDCK cells, claudins-4, -5 and occludin were displaced from lipid rafts to more fluid membrane domains, whereas claudins-1, -2 and -3 were unchanged after C10 treatment.Citation56 In this context, it should be noted that another group found that both occludin and claudin-1 were partially removed from TJs after C10 treatment in skin epithelial cells.Citation66 Slightly different observations have been made with HT-29/B6 cells. While no changes could be observed in claudins-1 to -5, -8, occludin and tricellulin in the total lysate or membrane fraction, there was a selective and reversible decrease in tricellulin and claudin-5 at tricellular and bicellular TJs, respectively.Citation54 The reported differences in the effect of C10 on TJ-protein localization could be related to the kind of cell line and their diverse TJ-protein expression patterns. However, claudins and occludin are connected via ZO-1 to the actin cytoskeleton and no changes in ZO-1 localization upon C10 treatment were observed in HEK-293 (human embryonic kidney 293), MDCK, bEND5 (mouse brain endothelioma) and mouse brain capillaries.Citation55 The conclusion from these findings would be that there is more specific modulation of TJ-proteins, rather than just the modulation of the actin-myosin ring complex alone. Interestingly, the decreases in TER and claudin-5 in the membrane are reached within 5 min, suggesting that these events are directly linked.Citation55,Citation56 Since claudin-5 tightens the TJ against molecules with a molecular weight less than 800 Da, this may explain why low molecular weight molecules permeate much more rapidly than higher molecular weight proteins.Citation23

But how is the permeability of macromolecules enhanced by C10? This issue was also addressed by Watson and colleagues, who showed that the unspecific absorption enhancer EGTA increased the permeability of 10 kDa fluorescein isothiocyanate dextran (FD10) 21-fold, whereas C10 only results in a 1.9-fold increase.Citation58 This might be explained if C10 acts on tricellulin. In this scenario, the decrease in tricellulin allows macromolecules up to a certain size (10 kDa) to pass via the paracellular pathway, since it is known that tricellulin acts as a barrier to macromolecules.Citation54,Citation67 As tricellular contacts are rarer than bicellular contacts and the possibilities of macromolecular permeation are correspondingly rare, this could explain why low molecular weight molecules are more permeable. However, the exact mechanism behind the modulation of specific membrane proteins is still unclear.

If C10 is to be used as a drug enhancer, its cytotoxic effects must be studied. 50 mM C10 is at least harmful in Caco-2 cells.Citation53 Caco-2 cell cultures are apparently more sensitive to the cytotoxic effects of C10 than the intestine in vivo, since there is no evidences of mucosal irritation or damage after exposure to twice the concentration used in the cell culture experiments.Citation53 Perhaps this could be explained by the dilution effect in vivo, by the protective intestinal mucus layer or by the high regeneration of mucosa cell.Citation41 Furthermore, while the tmax (time point at which the maximal concentration is reached) of a drug or marker administered with C10 in vivo is between 10 to 60 min,Citation43,Citation48,Citation57 the in vitro effect lasts at least 300 min.Citation56 After removal of C10, it takes 120 min until the TER is back to control values.Citation54 This may contribute to the stronger cytotoxic effects.

The mode of administration of C10 and its effective concentration also influence cytotoxicity. Possibilities include reincubation with C10 or coadministration in a liquid or solid form, e.g., tablets by spray freeze-drying.Citation43,Citation57,Citation68 Because the uptake of C10 in vivo is quite fast (tmax ~7 min,Citation48) after injection, the drug is injected before and after the application of C10.Citation52 C10 was coadministered with two different size markers combined in tablet or in liquid form. It was found that the solid form enhanced uptake much better than the liquid form. Additionally, the smaller marker is taken up to a greater extent than the larger marker.Citation57 An in situ drug delivery study showed that co-administration of C10 and FD4 achieved the optimal effect compared with pre-incubation with C10.Citation68 In summary, C10 enhances paracellular drug uptake for molecules with a molecular weight less than 20 kDa. No difference between hydrophobic and hydrophilic drug enhancement was reported. However, most of the drugs investigated tended to be hydrophilic.

In addition to C10, tartaric acid, sodium taurodeoxycholate, sodium dodecyl sulfate (SDS) or p-t-octyl phenol polyoxyethylene-9.5 (Triton X-100) were tested as penetration enhancers with surface activity.Citation69 They were tested at the rat jejunum and have potential as enhancing systems for oral delivery of poorly absorbed hydrophilic compounds such as protein or peptide drugs. These approaches have not been further developed.

A further group of surfactant-like drug enhancers are short chain alkylglycerols.Citation70 Intra-arterial injection leads to concentration-dependent enrichment of co-applied cytostatic drugs and antibiotics in the brain.Citation71 Even the penetration of large molecules like albumin could be significantly increased.Citation72 Opening of the blood-brain barrier in vivo is reversible and lasts from a few minutes to about one hour, depending on the concentration used.Citation71,Citation73 In vitro and in vivo analysis showed that alkylglycerols are nontoxic.Citation74 In freshly isolated brain capillaries, the small molecule fluorescein diffused into the capillary lumen after addition of alkylglycerols through the paracellular cleft, indicating opening of the paracellular diffusion barrier.Citation72 However, the exact mode of action remains unknown.

Peptides

Specific targeting of TJ proteins could also be achieved by using peptides which are thought to bind to extracellular parts of TJ transmembrane proteins. Occludin has been the first target for barrier modulation, using an 18 amino acid peptide derived from the ECD2. After calcium depletion of T84 cells, the ECD2 peptide prevents reformation of TJs and binds to claudin-1, occludin and the junctional adhesion molecule-A.Citation36 Peptides derived from the N-terminal half of the first ECD of human occludin increased the permeability of airway epithelia.Citation75 Moreover, a 22-amino acid peptide of the ECD1 corresponding to the residues 209–230 of rat occludin was injected into the testis of adult rats (1.5–10 mg/testis) and was able to open the blood-testis barrier.Citation76 This peptide fused to the follicle stimulating hormone (FSH) specifically opened the blood-testis barrier after intraperitoneal injection (40 µg/adult rat), without disturbing the epithelia of other organs.Citation77

A similar effect was shown in T84 cell monolayers for a peptide related to the amino acids 53–80 of the ECD1 of mouse claudin-1, whereas claudin-1_146–160 and claudin-1_31–54 did not increase TER or FD3 permeability. This peptide increased paracellular gastric permeability for sucrose 1 day after oral administration in rats (0.1 mg/kg body weight).Citation78 A similar peptide was used to improve the delivery of pharmaceutical agents through the perineural barrier. Beginning at 48 h after perineural injection of 400 µM peptide, there was an improved effect of opioid receptor agonists (e.g., DAMGO) or sodium channel blockers (tetrodotoxin) on raised the mechanical nociceptive threshold.Citation79 This demonstrated that the perineural barrier was opened by an induction mechanism carried by the peptide.

Another ligand for a subgroup of claudins is the Clostridium perfringens enterotoxin (CPE), a common source of many symptoms of Clostridum perfringens-related food borne diseases. Among other claudins (6, 8 and 14), the main binding partners for CPE are the ECDs2 of claudin-3 and claudin-4.Citation80 CPE could be separated into a cytotoxic N-terminal and a claudin binding C-terminal part at the very end of CPE (290–319).Citation81 The larger C-terminal fragment (amino acid 194–319, cCPE) is capable of modulating TJs and increases the permeability of CPE sensitive claudin expressing epithelial cells (3.5 μg/ml, 18 h).Citation82 In rats, it was demonstrated that 1 μg/μl cCPE enhanced the nasal mucosal absorption of dextran ≥4 kDa and jejunal absorption and also for 150 kDa dextrans one hour after administration.Citation83 Since claudin-4 is highly expressed in human epithelial ovarian carcinomas, but not in normal ovary tissue, cCPE was tested as drug enhancer for cancer treatment. After application of 5 μg/ml cCPE for 24 h, the claudin-4 expression level decreased and claudin-4 was removed from TJs in cell culture, without showing any cytotoxicity (0.01–15 μg/ml, 0–48 h).Citation84 In a human epithelial ovarian carcinoma xenograft model, Gao and coworkers also found suppression of tumor growth by 59% compared with the vehicle, when the mice were treated with taxol (20 mg/kg) combined with cCPE (0.1 mg/kg) twice a week for a period of four weeks.

A toxicological study in mice using intravenous administration (5 mg/kg) or nasal cCPE administration (2 mg/kg), did not increase biochemical markers of liver and kidney injury but, after six administrations once a week, the cCPE-specific serum immunoglobulin G rose.Citation85

Specific targeting of claudins and occludin is an innovative and promising approach modulating TJ expressing cells directly and avoiding effects of the drug enhancer on other cell types. Peptides directed against certain claudins interfere with claudin-interactions only in TJs containing the target claudins of the peptide. Therefore, the effect is tissue barrier specific. Since peptide-treated barriers open slowly, but stay open for a period of hours, long-term treatments with antibiotics or chemotherapy are conceivable.

A more rapidly acting but less specific peptide is a 6-mer, FCIGRL, called AT1002. This short, synthetic peptide corresponds to the amino acids 288–293 of the zonula occludens toxin from Vibrio cholera.Citation86 Both the toxin and the peptide reversibly open TJs and increase paracellular permeability in intestinal cells by binding to proteinase activating receptor (PAR) 2 and, subsequently, to actin polymerization mediated by protein kinase Cα activation.Citation87 In male Sprague-Dawley rats, intestinal uptake of cyclosporin A was increased by co-treatment with 10–40 mg/kg AT1002 (1.5 to 2.5-fold, 0–120 min after application).Citation88 Nasal administration of the low bioavailability agent ritonavir (Abbott Laboratories), an antiretroviral drug to treat HIV infections, was increased 2.55-fold up to 240 min with AT1002 when co-administered with the bioadhesive polymer carrageenan.Citation89 One problem with the use of AT1002 is the instability of the peptide in neutral to basic pH conditions. It has been recently shown that the stability of AT1002 could be increased up to 6 h in 5% dextrose solution.Citation90,Citation91 Another approach to stabilize AT1002 is the systematic exchange of amino acids, especially, the cysteine at position two.Citation92

siRNA and antibodies

An even more specific new approach to modulate tissue barrier functions is the administration of siRNA found to knockdown the expression of TJ proteins in barrier-forming epithelialCitation93,Citation94 and endothelial cells.Citation95 This affects ZO-1,Citation96 occludinCitation97 or claudins.Citation98

By selectively decreasing claudin-5 at the TJs, the chance of drugs reaching their target in the brain increases, as claudin-5 tightens the blood-brain barrier for small molecules.Citation23 Campbell et al. injected siRNA into mice specifically to modulate claudin-5 expression in blood vessels in the brain.Citation99 20 μg siRNA per mouse suppresses the expression of a protein by degrading its mRNA. Claudin-5 selective siRNA reduced claudin-5 protein content and size-selective opening (up to 1 kDa) of the blood-brain barrier between 24 and 72 h post injection. In a mouse model of traumatic brain injury, they also showed that water efflux to the blood from the brain—caused by edema—is increased.Citation99 siRNA in complex with in vivo-jetPeiTM, an in vivo delivery agent, is now being used in a phase I clinical trial (http://www.polyplus-transfection.com/transfection-reagents/5_in-vivodelivery/1_dna_sirna_delivery/dna-sirna-delivery-in-vivo-jetpei/).

Selective targetting of claudins is also the goal of several therapeutic anti-claudin antibodies since some types of tumors, such as breast, pancreatic or ovarian cancers, exhibit increased expression of claudins-3 and -4.Citation100,Citation101 Therefore, a monoclonal antibody, KM3907, with in vivo antitumor activity (10 mg/kg) was developed. KM3907 recognized claudins-3 and -4, but not claudins-5, -6 and -9.Citation102 Another monoclonal antibody against claudin-4 (KM3934) has also shown promising antitumor efficacy in preclinical models.Citation103 A monoclonal anti-claudin-1 antibody, which binds to the first ECD of claudin-1, was proven to efficiently inhibit infection of primary human hepatocytes with all major genotypes of hepatitis C virus. Fofana and coworkers preincubated cell cultures in vitro with 10 μg/ml for one hour without observing cytoxic effects (up to 1000 μg/ml) or effects on TJ integrity.Citation104

This demonstrates that the development of antibodies against ECDs of TJ proteins is possible. The design of non-cytotoxic antibodies targeting single TJ components could raise new options for the development of drug enhancers. Since TJ protein binding proteins and peptides take a short time to bind to their target, but several hours to show an effect on permeability, these antibodies should be analyzed for their potential as drug enhancers.

Conclusion

Different drug enhancing approaches, with very different properties, are available in experimental and clinical investigations. Hyperosmolar solutions have been used since the 1970s. These drastic procedures immediately and transiently disrupt the TJs. As yet, mannitol is the only compound with any clinical relevance. Nevertheless, hyperosmolar solutions give limited reproducibility, strong side-effects are common and a molar dosage is used.

Milder approaches have since been developed, based on surface active agents. They also show rather unspecific modes of action. However, there is evidence that some TJ proteins can be selectively targeted. The activity appears directly after administration and lasts for a short time. Millimolar concentrations are effective.

During the last decade, specific peptide ligands of ECDs of TJ proteins have been designed. They affect single TJ proteins, allowing selective targeting of specific barriers, depending on the expression pattern of the TJ proteins. Micromolar amounts of the peptides were found to be effective. However, the transient action has a lag phase of several hours and lasts for many hours.

The most advanced approach is to administer siRNA for downregulation of selected TJ proteins. This method is highly specific. Submicromolar dosage is required; drug delivery is improved after one day and lasts for a second day.

In summary, unspecific approaches need higher dosages, have a shorter duration of action and may cause more side effects. Specific procedures require lower doses, may cause fewer side effects and longer lasting effects. Longer opening times are disadvantageous. Consequently, one should prefer immediately acting agents, such as caprate, for the time necessary to deliver the drug, i.e., opening for some hours. On the other hand, procedures selectively targeting single TJ proteins are advantageous and, specific for defined tissue barriers. Moreover, fewer secondary effects would be expected. However, even more specific approaches, e.g., siRNA, have a longer duration of action which, in turn, may raise the risk of side effects.

Abbreviations:
TJ=

tight junctions

ABC=

ATP binding cassette

EDTA=

ethylene diamine tetraacetic acid

EGTA=

ethylene glycol tetraacetic acid

ZO=

zonula occludens protein

TAMP=

tight junction-associated marvel protein

marvel=

MAL and related proteins for vesicle trafficking and membrane link

TER=

transepithelial electrical resistance

ECD=

extracellular domains

C10=

sodium caprate

n.a.=

not accessible

FD=

fluorescein isothiocyanate dextran

CPE=

Clostridium perfringens enterotoxin

cCPE=

C-terminal fragment Clostridium perfringens enterotoxin

siRNA=

small interfering ribonucleic acid

Acknowledgments

The work was supported by the DFG [Grant BL308/7–4] and EU Project JUSTBRAIN. We thank Jessica Thi Hien Tran-Winkler for her help to prepare the manuscript.

Disclosure of Potential Conflicts of Interest

No potential conflict of interest was disclosed.

References

  • Kinne RK. Endothelial and epithelial cells: general principles of selective vectorial transport. Int J Microcirc Clin Exp 1997; 17:223 - 30; http://dx.doi.org/10.1159/000179234; PMID: 9370122
  • Abbott NJ. Dynamics of CNS barriers: evolution, differentiation, and modulation. Cell Mol Neurobiol 2005; 25:5 - 23; http://dx.doi.org/10.1007/s10571-004-1374-y; PMID: 15962506
  • Brandner JM, Kief S, Wladykowski E, Houdek P, Moll I. Tight junction proteins in the skin. Skin Pharmacol Physiol 2006; 19:71 - 7; http://dx.doi.org/10.1159/000091973; PMID: 16685145
  • Camilleri M, Madsen K, Spiller R, Greenwood-Van Meerveld B, Verne GN. Intestinal barrier function in health and gastrointestinal disease. Neurogastroenterol Motil 2012; 24:503 - 12; http://dx.doi.org/10.1111/j.1365-2982.2012.01921.x; PMID: 22583600
  • Krause G, Winkler L, Mueller SL, Haseloff RF, Piontek J, Blasig IE. Structure and function of claudins. Biochim Biophys Acta 2008; 1778:631 - 45; http://dx.doi.org/10.1016/j.bbamem.2007.10.018; PMID: 18036336
  • Raleigh DR, Marchiando AM, Zhang Y, Shen L, Sasaki H, Wang Y, et al. Tight junction-associated MARVEL proteins marveld3, tricellulin, and occludin have distinct but overlapping functions. Mol Biol Cell 2010; 21:1200 - 13; http://dx.doi.org/10.1091/mbc.E09-08-0734; PMID: 20164257
  • Steed E, Rodrigues NT, Balda MS, Matter K. Identification of MarvelD3 as a tight junction-associated transmembrane protein of the occludin family. BMC Cell Biol 2009; 10:95; http://dx.doi.org/10.1186/1471-2121-10-95; PMID: 20028514
  • Furuse M. Knockout animals and natural mutations as experimental and diagnostic tool for studying tight junction functions in vivo. Biochim Biophys Acta 2009; 1788:813 - 9; http://dx.doi.org/10.1016/j.bbamem.2008.07.017; PMID: 18706387
  • Furuse M. Molecular basis of the core structure of tight junctions. Cold Spring Harb Perspect Biol 2010; 2:a002907; http://dx.doi.org/10.1101/cshperspect.a002907; PMID: 20182608
  • Furuse M, Hirase T, Itoh M, Nagafuchi A, Yonemura S, Tsukita S, et al. Occludin: a novel integral membrane protein localizing at tight junctions. J Cell Biol 1993; 123:1777 - 88; http://dx.doi.org/10.1083/jcb.123.6.1777; PMID: 8276896
  • Dörfel MJ, Huber O. Modulation of tight junction structure and function by kinases and phosphatases targeting occludin. J Biomed Biotechnol 2012; 2012:807356; http://dx.doi.org/10.1155/2012/807356; PMID: 22315516
  • Blasig IE, Bellmann C, Cording J, Del Vecchio G, Zwanziger D, Huber O, et al. Occludin protein family: oxidative stress and reducing conditions. Antioxid Redox Signal 2011; 15:1195 - 219; http://dx.doi.org/10.1089/ars.2010.3542; PMID: 21235353
  • Ikenouchi J, Furuse M, Furuse K, Sasaki H, Tsukita S, Tsukita S. Tricellulin constitutes a novel barrier at tricellular contacts of epithelial cells. J Cell Biol 2005; 171:939 - 45; http://dx.doi.org/10.1083/jcb.200510043; PMID: 16365161
  • Mariano C, Sasaki H, Brites D, Brito MA. A look at tricellulin and its role in tight junction formation and maintenance. Eur J Cell Biol 2011; 90:787 - 96; http://dx.doi.org/10.1016/j.ejcb.2011.06.005; PMID: 21868126
  • Furuse M, Fujita K, Hiiragi T, Fujimoto K, Tsukita S. Claudin-1 and -2: novel integral membrane proteins localizing at tight junctions with no sequence similarity to occludin. J Cell Biol 1998; 141:1539 - 50; http://dx.doi.org/10.1083/jcb.141.7.1539; PMID: 9647647
  • Mineta K, Yamamoto Y, Yamazaki Y, Tanaka H, Tada Y, Saito K, et al. Predicted expansion of the claudin multigene family. FEBS Lett 2011; 585:606 - 12; http://dx.doi.org/10.1016/j.febslet.2011.01.028; PMID: 21276448
  • Umeda K, Ikenouchi J, Katahira-Tayama S, Furuse K, Sasaki H, Nakayama M, et al. ZO-1 and ZO-2 independently determine where claudins are polymerized in tight-junction strand formation. Cell 2006; 126:741 - 54; http://dx.doi.org/10.1016/j.cell.2006.06.043; PMID: 16923393
  • Piontek J, Winkler L, Wolburg H, Müller SL, Zuleger N, Piehl C, et al. Formation of tight junction: determinants of homophilic interaction between classic claudins. FASEB J 2008; a 22:146 - 58; http://dx.doi.org/10.1096/fj.07-8319com; PMID: 17761522
  • Piontek J, Fritzsche S, Cording J, Richter S, Hartwig J, Walter M, et al. Elucidating the principles of the molecular organization of heteropolymeric tight junction strands. Cell Mol Life Sci 2011; 68:3903 - 18; http://dx.doi.org/10.1007/s00018-011-0680-z; PMID: 21533891
  • Cording J, Berg J, Kading N, Bellmann C, Tscheik C, Westphal JK, et al. Tight junctions: Claudins regulate the interactions between occludin, tricellulin and marvelD3, which, inversely, modulate claudin oligomerization. J Cell Sci 2012; 126:554 - 64; http://dx.doi.org/10.1242/jcs.114306; PMID: 23203797
  • Furuse M, Sasaki H, Tsukita S. Manner of interaction of heterogeneous claudin species within and between tight junction strands. J Cell Biol 1999; 147:891 - 903; http://dx.doi.org/10.1083/jcb.147.4.891; PMID: 10562289
  • Amasheh S, Meiri N, Gitter AH, Schöneberg T, Mankertz J, Schulzke JD, et al. Claudin-2 expression induces cation-selective channels in tight junctions of epithelial cells. J Cell Sci 2002; 115:4969 - 76; http://dx.doi.org/10.1242/jcs.00165; PMID: 12432083
  • Nitta T, Hata M, Gotoh S, Seo Y, Sasaki H, Hashimoto N, et al. Size-selective loosening of the blood-brain barrier in claudin-5-deficient mice. J Cell Biol 2003; 161:653 - 60; http://dx.doi.org/10.1083/jcb.200302070; PMID: 12743111
  • Wen H, Watry DD, Marcondes MC, Fox HS. Selective decrease in paracellular conductance of tight junctions: role of the first extracellular domain of claudin-5. Mol Cell Biol 2004; 24:8408 - 17; http://dx.doi.org/10.1128/MCB.24.19.8408-8417.2004; PMID: 15367662
  • Colegio OR, Van Itallie CM, McCrea HJ, Rahner C, Anderson JM. Claudins create charge-selective channels in the paracellular pathway between epithelial cells. Am J Physiol Cell Physiol 2002; 283:C142 - 7; PMID: 12055082
  • Piehl C, Piontek J, Cording J, Wolburg H, Blasig IE. Participation of the second extracellular loop of claudin-5 in paracellular tightening against ions, small and large molecules. Cell Mol Life Sci 2010; 67:2131 - 40; http://dx.doi.org/10.1007/s00018-010-0332-8; PMID: 20333434
  • Veshnyakova A, Krug SM, Mueller SL, Piontek J, Protze J, Fromm M, et al. Determinants contributing to claudin ion channel formation. Ann N Y Acad Sci 2012; a 1257:45 - 53; http://dx.doi.org/10.1111/j.1749-6632.2012.06566.x; PMID: 22671588
  • Blasig IE, Winkler L, Lassowski B, Mueller SL, Zuleger N, Krause E, et al. On the self-association potential of transmembrane tight junction proteins. Cell Mol Life Sci 2006; 63:505 - 14; http://dx.doi.org/10.1007/s00018-005-5472-x; PMID: 16456617
  • Pardridge WM. The blood-brain barrier: bottleneck in brain drug development. NeuroRx 2005; 2:3 - 14; http://dx.doi.org/10.1602/neurorx.2.1.3; PMID: 15717053
  • Pardridge WM. Brain Drug Targeting: The Future of Brain Drug Development. Cambridge, U.K.: 2001.
  • Brandsch M, Knütter I, Bosse-Doenecke E. Pharmaceutical and pharmacological importance of peptide transporters. J Pharm Pharmacol 2008; 60:543 - 85; http://dx.doi.org/10.1211/jpp.60.5.0002; PMID: 18416933
  • Petrus AK, Allis DG, Smith RP, Fairchild TJ, Doyle RP. Exploring the implications of vitamin B12 conjugation to insulin on insulin receptor binding. ChemMedChem 2009; 4:421 - 6; http://dx.doi.org/10.1002/cmdc.200800346; PMID: 19101970
  • Hussain A, Arnold JJ, Khan MA, Ahsan F. Absorption enhancers in pulmonary protein delivery. J Control Release 2004; 94:15 - 24; http://dx.doi.org/10.1016/j.jconrel.2003.10.001; PMID: 14684268
  • Lindmark T, Kimura Y, Artursson P. Absorption enhancement through intracellular regulation of tight junction permeability by medium chain fatty acids in Caco-2 cells. J Pharmacol Exp Ther 1998; 284:362 - 9; PMID: 9435199
  • Walsh EG, Adamczyk BE, Chalasani KB, Maher S, O’Toole EB, Fox JS, et al. Oral delivery of macromolecules: rationale underpinning Gastrointestinal Permeation Enhancement Technology (GIPET). Ther Deliv 2011; 2:1595 - 610; http://dx.doi.org/10.4155/tde.11.132; PMID: 22833984
  • Nusrat A, Brown GT, Tom J, Drake A, Bui TT, Quan C, et al. Multiple protein interactions involving proposed extracellular loop domains of the tight junction protein occludin. Mol Biol Cell 2005; 16:1725 - 34; http://dx.doi.org/10.1091/mbc.E04-06-0465; PMID: 15659655
  • Campbell M, Nguyen AT, Kiang AS, Tam LC, Gobbo OL, Kerskens C, et al. An experimental platform for systemic drug delivery to the retina. Proc Natl Acad Sci U S A 2009; 106:17817 - 22; http://dx.doi.org/10.1073/pnas.0908561106; PMID: 19822744
  • Sousa SM, Bramante CM, Taga EM. Biocompatibility of EDTA, EGTA and citric acid. Braz Dent J 2005; 16:3 - 8; http://dx.doi.org/10.1590/S0103-64402005000100001; PMID: 16113926
  • Kanner AA, Marchi N, Fazio V, Mayberg MR, Koltz MT, Siomin V, et al. Serum S100beta: a noninvasive marker of blood-brain barrier function and brain lesions. Cancer 2003; 97:2806 - 13; http://dx.doi.org/10.1002/cncr.11409; PMID: 12767094
  • Hackel D, Krug SM, Sauer RS, Mousa SA, Böcker A, Pflücke D, et al. Transient opening of the perineurial barrier for analgesic drug delivery. Proc Natl Acad Sci U S A 2012; 109:E2018 - 27; http://dx.doi.org/10.1073/pnas.1120800109; PMID: 22733753
  • Lindmark T, Söderholm JD, Olaison G, Alván G, Ocklind G, Artursson P. Mechanism of absorption enhancement in humans after rectal administration of ampicillin in suppositories containing sodium caprate. Pharm Res 1997; 14:930 - 5; http://dx.doi.org/10.1023/A:1012112219578; PMID: 9244152
  • Leonard TW, Lynch J, McKenna MJ, Brayden DJ. Promoting absorption of drugs in humans using medium-chain fatty acid-based solid dosage forms: GIPET. Expert Opin Drug Deliv 2006; 3:685 - 92; http://dx.doi.org/10.1517/17425247.3.5.685; PMID: 16948563
  • Tong HH, Du Z, Wang GN, Chan HM, Chang Q, Lai LC, et al. Spray freeze drying with polyvinylpyrrolidone and sodium caprate for improved dissolution and oral bioavailability of oleanolic acid, a BCS Class IV compound. Int J Pharm 2011; 404:148 - 58; http://dx.doi.org/10.1016/j.ijpharm.2010.11.027; PMID: 21094233
  • Maher S, Leonard TW, Jacobsen J, Brayden DJ. Safety and efficacy of sodium caprate in promoting oral drug absorption: from in vitro to the clinic. Adv Drug Deliv Rev 2009; 61:1427 - 49; http://dx.doi.org/10.1016/j.addr.2009.09.006; PMID: 19800376
  • Fan D, Wu X, Dong W, Sun W, Li J, Tang X. Enhancement by sodium caprate and sodium deoxycholate of the gastrointestinal absorption of berberine chloride in rats. Drug Dev Ind Pharm 2012; http://dx.doi.org/10.3109/03639045.2012.723219; PMID: 23020091
  • Lv XY, Li J, Zhang M, Wang CM, Fan Z, Wang CY, et al. Enhancement of sodium caprate on intestine absorption and antidiabetic action of berberine. AAPS PharmSciTech 2010; 11:372 - 82; http://dx.doi.org/10.1208/s12249-010-9386-z; PMID: 20237966
  • Ma Y, Zhang L, Zhao X, Shen Q. Analysis of daidzein in nanoparticles after oral co-administration with sodium caprate to rats by ultra-performance liquid chromatography-quadrupole-time-of-flight mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2012; 907:21 - 6; http://dx.doi.org/10.1016/j.jchromb.2012.08.026; PMID: 23010479
  • Raoof AA, Ramtoola Z, McKenna B, Yu RZ, Hardee G, Geary RS. Effect of sodium caprate on the intestinal absorption of two modified antisense oligonucleotides in pigs. Eur J Pharm Sci 2002; 17:131 - 8; http://dx.doi.org/10.1016/S0928-0987(02)00162-8; PMID: 12393140
  • Cianetti S, Cooper VB, Attenni B, Pucci V, Fiore F, Giuliano C, et al. Enhancement of intestinal absorption of 2-methyl cytidine prodrugs. Drug Deliv 2010; 17:214 - 22; http://dx.doi.org/10.3109/10717541003667814; PMID: 20233089
  • Ohnishi T, Aida K, Awazu S. Enhancement of blood-brain barrier permeability by sodium caprate. J Pharm Pharmacol 1999; 51:1015 - 8; http://dx.doi.org/10.1211/0022357991773483; PMID: 10528983
  • Preston E, Slinn J, Vinokourov I, Stanimirovic D. Graded reversible opening of the rat blood-brain barrier by intracarotid infusion of sodium caprate. J Neurosci Methods 2008; 168:443 - 9; http://dx.doi.org/10.1016/j.jneumeth.2007.11.004; PMID: 18155299
  • Brandhonneur N, Dollo G, Ratajczak-Enselme M, Deniau AL, Chevanne F, Estèbe JP, et al. Ex vivo and in vivo diffusion of ropivacaine through spinal meninges: influence of absorption enhancers. Int J Pharm 2011; 404:36 - 41; http://dx.doi.org/10.1016/j.ijpharm.2010.10.049; PMID: 21056096
  • Chao AC, Nguyen JV, Broughall M, Griffin A, Fix JA, Daddona PE. In vitro and in vivo evaluation of effects of sodium caprate on enteral peptide absorption and on mucosal morphology. Int J Pharm 1999; 191:15 - 24; http://dx.doi.org/10.1016/S0378-5173(99)00213-6; PMID: 10556736
  • Krug SM, Amasheh M, Dittmann I, Christoffel I, Fromm M, Amasheh S. Sodium caprate as an enhancer of macromolecule permeation across tricellular tight junctions of intestinal cells. Biomaterials 2013; 34:275 - 82; http://dx.doi.org/10.1016/j.biomaterials.2012.09.051; PMID: 23069717
  • Del Vecchio G, Tscheik C, Tenz K, Helms HC, Winkler L, Blasig R, et al. Sodium caprate transiently opens claudin-5-containing barriers at tight junctions of epithelial and endothelial cells. Mol Pharm 2012; 9:2523 - 33; http://dx.doi.org/10.1021/mp3001414; PMID: 22827574
  • Sugibayashi K, Onuki Y, Takayama K. Displacement of tight junction proteins from detergent-resistant membrane domains by treatment with sodium caprate. Eur J Pharm Sci 2009; 36:246 - 53; http://dx.doi.org/10.1016/j.ejps.2008.09.011; PMID: 19013238
  • Perera G, Barthelmes J, Vetter A, Krieg C, Uhlschmied C, Bonn GK, et al. Thiolated polycarbophil/glutathione: defining its potential as a permeation enhancer for oral drug administration in comparison to sodium caprate. Drug Deliv 2011; 18:415 - 23; http://dx.doi.org/10.3109/10717544.2011.570807; PMID: 21554106
  • Watson CJ, Rowland M, Warhurst G. Functional modeling of tight junctions in intestinal cell monolayers using polyethylene glycol oligomers. Am J Physiol Cell Physiol 2001; 281:C388 - 97; PMID: 11443038
  • Tomita M, Hayashi M, Awazu S. Absorption-enhancing mechanism of sodium caprate and decanoylcarnitine in Caco-2 cells. J Pharmacol Exp Ther 1995; 272:739 - 43; PMID: 7853188
  • Turner JR. ‘Putting the squeeze’ on the tight junction: understanding cytoskeletal regulation. Semin Cell Dev Biol 2000; 11:301 - 8; http://dx.doi.org/10.1006/scdb.2000.0180; PMID: 10966864
  • Feighery LM, Cochrane SW, Quinn T, Baird AW, O’Toole D, Owens SE, et al. Myosin light chain kinase inhibition: correction of increased intestinal epithelial permeability in vitro. Pharm Res 2008; 25:1377 - 86; http://dx.doi.org/10.1007/s11095-007-9527-6; PMID: 18163202
  • Anderberg EK, Lindmark T, Artursson P. Sodium caprate elicits dilatations in human intestinal tight junctions and enhances drug absorption by the paracellular route. Pharm Res 1993; 10:857 - 64; http://dx.doi.org/10.1023/A:1018909210879; PMID: 8321854
  • Sakai M, Imai T, Ohtake H, Azuma H, Otagiri M. Effects of absorption enhancers on cytoskeletal actin filaments in Caco-2 cell monolayers. Life Sci 1998; 63:45 - 54; http://dx.doi.org/10.1016/S0024-3205(98)00235-5; PMID: 9667764
  • Suzuki T, Hara H. Difructose anhydride III and sodium caprate activate paracellular transport via different intracellular events in Caco-2 cells. Life Sci 2006; 79:401 - 10; http://dx.doi.org/10.1016/j.lfs.2006.01.044; PMID: 16566947
  • Coyne CB, Ribeiro CM, Boucher RC, Johnson LG. Acute mechanism of medium chain fatty acid-induced enhancement of airway epithelial permeability. J Pharmacol Exp Ther 2003; 305:440 - 50; http://dx.doi.org/10.1124/jpet.102.047654; PMID: 12606647
  • Kurasawa M, Kuroda S, Kida N, Murata M, Oba A, Yamamoto T, et al. Regulation of tight junction permeability by sodium caprate in human keratinocytes and reconstructed epidermis. Biochem Biophys Res Commun 2009; 381:171 - 5; http://dx.doi.org/10.1016/j.bbrc.2009.02.005; PMID: 19338770
  • Krug SM, Amasheh S, Richter JF, Milatz S, Günzel D, Westphal JK, et al. Tricellulin forms a barrier to macromolecules in tricellular tight junctions without affecting ion permeability. Mol Biol Cell 2009; 20:3713 - 24; http://dx.doi.org/10.1091/mbc.E09-01-0080; PMID: 19535456
  • Wang X, Maher S, Brayden DJ. Restoration of rat colonic epithelium after in situ intestinal instillation of the absorption promoter, sodium caprate. Ther Deliv 2010; 1:75 - 82; http://dx.doi.org/10.4155/tde.10.5; PMID: 22816121
  • Takatsuka S, Kitazawa T, Morita T, Horikiri Y, Yoshino H. Enhancement of intestinal absorption of poorly absorbed hydrophilic compounds by simultaneous use of mucolytic agent and non-ionic surfactant. Eur J Pharm Biopharm 2006; 62:52 - 8; http://dx.doi.org/10.1016/j.ejpb.2005.07.008; PMID: 16289777
  • Unger C, Eibl H, von Heyden HW, Krisch B, Nagel GA. [Blood-brain barrier and the penetration of cytostatic drugs]. Klin Wochenschr 1985; 63:565 - 71; http://dx.doi.org/10.1007/BF01733202; PMID: 2411995
  • Erdlenbruch B, Jendrossek V, Eibl H, Lakomek M. Transient and controllable opening of the blood-brain barrier to cytostatic and antibiotic agents by alkylglycerols in rats. Exp Brain Res 2000; 135:417 - 22; http://dx.doi.org/10.1007/s002210000553; PMID: 11146820
  • Erdlenbruch B, Alipour M, Fricker G, Miller DS, Kugler W, Eibl H, et al. Alkylglycerol opening of the blood-brain barrier to small and large fluorescence markers in normal and C6 glioma-bearing rats and isolated rat brain capillaries. Br J Pharmacol 2003; b 140:1201 - 10; http://dx.doi.org/10.1038/sj.bjp.0705554; PMID: 14597599
  • Erdlenbruch B, Jendrossek V, Kugler W, Eibl H, Lakomek M. Increased delivery of erucylphosphocholine to C6 gliomas by chemical opening of the blood-brain barrier using intracarotid pentylglycerol in rats. Cancer Chemother Pharmacol 2002; 50:299 - 304; http://dx.doi.org/10.1007/s00280-002-0497-4; PMID: 12357304
  • Erdlenbruch B, Schinkhof C, Kugler W, Heinemann DE, Herms J, Eibl H, et al. Intracarotid administration of short-chain alkylglycerols for increased delivery of methotrexate to the rat brain. Br J Pharmacol 2003; a 139:685 - 94; http://dx.doi.org/10.1038/sj.bjp.0705302; PMID: 12812991
  • Everett RS, Vanhook MK, Barozzi N, Toth I, Johnson LG. Specific modulation of airway epithelial tight junctions by apical application of an occludin peptide. Mol Pharmacol 2006; 69:492 - 500; http://dx.doi.org/10.1124/mol.105.017251; PMID: 16288084
  • Chung NP, Mruk D, Mo MY, Lee WM, Cheng CY. A 22-amino acid synthetic peptide corresponding to the second extracellular loop of rat occludin perturbs the blood-testis barrier and disrupts spermatogenesis reversibly in vivo. Biol Reprod 2001; 65:1340 - 51; http://dx.doi.org/10.1095/biolreprod65.5.1340; PMID: 11673248
  • Wong CH, Mruk DD, Lee WM, Cheng CY. Targeted and reversible disruption of the blood-testis barrier by an FSH mutant-occludin peptide conjugate. FASEB J 2007; 21:438 - 48; http://dx.doi.org/10.1096/fj.05-4144com; PMID: 17167075
  • Mrsny RJ, Brown GT, Gerner-Smidt K, Buret AG, Meddings JB, Quan C, et al. A key claudin extracellular loop domain is critical for epithelial barrier integrity. Am J Pathol 2008; 172:905 - 15; http://dx.doi.org/10.2353/ajpath.2008.070698; PMID: 18349130
  • Zwanziger D, Hackel D, Staat C, Böcker A, Brack A, Beyermann M, et al. A peptidomimetic tight junction modulator to improve regional analgesia. Mol Pharm 2012; 9:1785 - 94; http://dx.doi.org/10.1021/mp3000937; PMID: 22524793
  • Winkler L, Gehring C, Wenzel A, Müller SL, Piehl C, Krause G, et al. Molecular determinants of the interaction between Clostridium perfringens enterotoxin fragments and claudin-3. J Biol Chem 2009; 284:18863 - 72; http://dx.doi.org/10.1074/jbc.M109.008623; PMID: 19429681
  • Veshnyakova A, Protze J, Rossa J, Blasig IE, Krause G, Piontek J. On the interaction of Clostridium perfringens enterotoxin with claudins. Toxins (Basel) 2010; 2:1336 - 56; http://dx.doi.org/10.3390/toxins2061336; PMID: 22069641
  • Veshnyakova A, Piontek J, Protze J, Waziri N, Heise I, Krause G. Mechanism of Clostridium perfringens enterotoxin interaction with claudin-3/-4 protein suggests structural modifications of the toxin to target specific claudins. J Biol Chem 2012; b 287:1698 - 708; http://dx.doi.org/10.1074/jbc.M111.312165; PMID: 22128179
  • Matsuhisa K, Kondoh M, Suzuki H, Yagi K. Comparison of mucosal absorption-enhancing activity between a claudin-3/-4 binder and a broadly specific claudin binder. Biochem Biophys Res Commun 2012; 423:229 - 33; http://dx.doi.org/10.1016/j.bbrc.2012.05.060; PMID: 22659740
  • Gao Z, Xu X, McClane B, Zeng Q, Litkouhi B, Welch WR, et al. C terminus of Clostridium perfringens enterotoxin downregulates CLDN4 and sensitizes ovarian cancer cells to Taxol and Carboplatin. Clin Cancer Res 2011; 17:1065 - 74; http://dx.doi.org/10.1158/1078-0432.CCR-10-1644; PMID: 21123456
  • Suzuki H, Kondoh M, Kakutani H, Yamane S, Uchida H, Hamakubo T, et al. The application of an alanine-substituted mutant of the C-terminal fragment of Clostridium perfringens enterotoxin as a mucosal vaccine in mice. Biomaterials 2012; 33:317 - 24; http://dx.doi.org/10.1016/j.biomaterials.2011.09.048; PMID: 21983135
  • Di Pierro M, Lu R, Uzzau S, Wang W, Margaretten K, Pazzani C, et al. Zonula occludens toxin structure-function analysis. Identification of the fragment biologically active on tight junctions and of the zonulin receptor binding domain. J Biol Chem 2001; 276:19160 - 5; http://dx.doi.org/10.1074/jbc.M009674200; PMID: 11278543
  • Goldblum SE, Rai U, Tripathi A, Thakar M, De Leo L, Di Toro N, et al. The active Zot domain (aa 288-293) increases ZO-1 and myosin 1C serine/threonine phosphorylation, alters interaction between ZO-1 and its binding partners, and induces tight junction disassembly through proteinase activated receptor 2 activation. FASEB J 2011; 25:144 - 58; http://dx.doi.org/10.1096/fj.10-158972; PMID: 20852064
  • Song KH, Fasano A, Eddington ND. Effect of the six-mer synthetic peptide (AT1002) fragment of zonula occludens toxin on the intestinal absorption of cyclosporin A. Int J Pharm 2008; 351:8 - 14; http://dx.doi.org/10.1016/j.ijpharm.2007.09.011; PMID: 17954018
  • Song KH, Eddington ND. The impact of AT1002 on the delivery of ritonavir in the presence of bioadhesive polymer, carrageenan. Arch Pharm Res 2012; a 35:937 - 43; http://dx.doi.org/10.1007/s12272-012-0520-1; PMID: 22644862
  • Song KH, Eddington ND. The influence of stabilizer and bioadhesive polymer on the permeation-enhancing effect of AT1002 in the nasal delivery of a paracellular marker. Arch Pharm Res 2012; b 35:359 - 66; http://dx.doi.org/10.1007/s12272-012-0217-5; PMID: 22370791
  • Song KH, Eddington ND. The influence of AT1002 on the nasal absorption of molecular weight markers and therapeutic agents when co-administered with bioadhesive polymers and an AT1002 antagonist, AT1001. J Pharm Pharmacol 2012; c 64:30 - 9; http://dx.doi.org/10.1111/j.2042-7158.2011.01381.x; PMID: 22150669
  • Li M, Oliver E, Kitchens KM, Vere J, Alkan SS, Tamiz AP. Structure-activity relationship studies of permeability modulating peptide AT-1002. Bioorg Med Chem Lett 2008; 18:4584 - 6; http://dx.doi.org/10.1016/j.bmcl.2008.07.028; PMID: 18667315
  • Masuda S, Oda Y, Sasaki H, Ikenouchi J, Higashi T, Akashi M, et al. LSR defines cell corners for tricellular tight junction formation in epithelial cells. J Cell Sci 2011; 124:548 - 55; http://dx.doi.org/10.1242/jcs.072058; PMID: 21245199
  • Yamamoto S, Nakase H, Matsuura M, Masuda S, Inui K, Chiba T. Tacrolimus therapy as an alternative to thiopurines for maintaining remission in patients with refractory ulcerative colitis. J Clin Gastroenterol 2011; 45:526 - 30; http://dx.doi.org/10.1097/MCG.0b013e318209cdc4; PMID: 21336140
  • Kluger MS, Clark PR, Tellides G, Gerke V, Pober JS. Claudin-5 controls intercellular barriers of human dermal microvascular but not human umbilical vein endothelial cells. Arterioscler Thromb Vasc Biol 2013; 33:489 - 500; http://dx.doi.org/10.1161/ATVBAHA.112.300893; PMID: 23288152
  • Van Itallie CM, Fanning AS, Bridges A, Anderson JM. ZO-1 stabilizes the tight junction solute barrier through coupling to the perijunctional cytoskeleton. Mol Biol Cell 2009; 20:3930 - 40; http://dx.doi.org/10.1091/mbc.E09-04-0320; PMID: 19605556
  • Yu AS, McCarthy KM, Francis SA, McCormack JM, Lai J, Rogers RA, et al. Knockdown of occludin expression leads to diverse phenotypic alterations in epithelial cells. Am J Physiol Cell Physiol 2005; 288:C1231 - 41; http://dx.doi.org/10.1152/ajpcell.00581.2004; PMID: 15689410
  • Hou J, Gomes AS, Paul DL, Goodenough DA. Study of claudin function by RNA interference. J Biol Chem 2006; 281:36117 - 23; http://dx.doi.org/10.1074/jbc.M608853200; PMID: 17018523
  • Campbell M, Hanrahan F, Gobbo OL, Kelly ME, Kiang AS, Humphries MM, et al. Targeted suppression of claudin-5 decreases cerebral oedema and improves cognitive outcome following traumatic brain injury. Nat Commun 2012; 3:849; http://dx.doi.org/10.1038/ncomms1852; PMID: 22617289
  • Kominsky SL. Claudins: emerging targets for cancer therapy. Expert Rev Mol Med 2006; 8:1 - 11; http://dx.doi.org/10.1017/S1462399406000056; PMID: 16887048
  • Morin PJ. Claudin proteins in human cancer: promising new targets for diagnosis and therapy. Cancer Res 2005; 65:9603 - 6; http://dx.doi.org/10.1158/0008-5472.CAN-05-2782; PMID: 16266975
  • Kato-Nakano M, Suzuki M, Kawamoto S, Furuya A, Ohta S, Nakamura K, et al. Characterization and evaluation of the antitumour activity of a dual-targeting monoclonal antibody against claudin-3 and claudin-4. Anticancer Res 2010; 30:4555 - 62; PMID: 21115905
  • Suzuki M, Kato-Nakano M, Kawamoto S, Furuya A, Abe Y, Misaka H, et al. Therapeutic antitumor efficacy of monoclonal antibody against Claudin-4 for pancreatic and ovarian cancers. Cancer Sci 2009; 100:1623 - 30; http://dx.doi.org/10.1111/j.1349-7006.2009.01239.x; PMID: 19555390
  • Fofana I, Krieger SE, Grunert F, Glauben S, Xiao F, Fafi-Kremer S, et al. Monoclonal anti-claudin 1 antibodies prevent hepatitis C virus infection of primary human hepatocytes. Gastroenterology 2010; 139:953 - 64, e1-4; http://dx.doi.org/10.1053/j.gastro.2010.05.073; PMID: 20685314
  • Sharma P, Varma MV, Chawla HP, Panchagnula R. In situ and in vivo efficacy of peroral absorption enhancers in rats and correlation to in vitro mechanistic studies. Farmaco 2005; 60:874 - 83; http://dx.doi.org/10.1016/j.farmac.2005.08.007; PMID: 16243320