1,007
Views
17
CrossRef citations to date
0
Altmetric
Review

Possible roles of LI-Cadherin in the formation and maintenance of the intestinal epithelial barrier

Article: e23815 | Received 14 Dec 2012, Accepted 29 Jan 2013, Published online: 01 Jan 2013

Abstract

LI-cadherin belongs to the so called 7D-cadherins, exceptional members of the cadherin superfamily which are characterized by seven extracellular cadherin repeats and a small cytosolic domain. Under physiological conditions LI-cadherin is expressed in the intestine and colon in human and mouse and in the rat also in hepatocytes. LI-cadherin was shown to act as a functional Ca2+-dependent adhesion molecule, linking neighboring cells and a lot of biophysical and biochemical parameters were determined in the last time. It is also known that dysregulated LI-cadherin expression can be found in a variety of diseases. Although there are several hypothesis and theoretical models concerning the function of LI-cadherin, the physiological role of LI-cadherin is still enigmatic.

The simple Epithelial Barrier and the Role of Cell-Cell-Adhesion

Epithelia cover inner and outer surfaces of the body of eumetazoic animals.Citation1-Citation3 Thus, epithelia represent the primary barrier for controlled transport of water or dissolved molecules into or out of the body.Citation4 For example, the intestinal mucosa contains a highly dynamic simple epithelium that fulfils the selective uptake of nutrients and ions, the resorption of water and the prevention of pathogen invasion. It has to be emphasized that controlled transport in this context means that on one side unwanted loss of water, ions or nutrients must be avoided.Citation1,Citation2,Citation5-Citation8 On the other side, necessary nutrients have to be absorbed and thus these molecules must be transported over the cellular barrier.Citation4,Citation7,Citation9-Citation11 For this process, there are two main transport pathways: transcellular and paracellular.Citation12 In the first case, the substances have to be taken up by the epithelial cells either directly by transporters in the membrane or by pinocytosis, i.e., by endozytosis (receptor mediated, constitutive or potocytosis). Then, eventually after modification by the epithelial cell the substances are secreted on the opposing side. This can be done by pumps (ATPases) or transporters, dependent on the electrochemical gradient, if the molecules are free in the cytosol. In case the substances are in vesicles, the release is performed by exocytosis. An uptake performed by vesicles (potocytosis or endocytosis), i.e., by uptake of caveolae or other vesicles, followed by transcellular transport of these vesicles, and fusion to the membrane of the opposing side, is called transcytosis. Besides the transcellular route, many substances are transported through the intercellular cleft (IC) at the lateral boarders of the cells.Citation5 The IC has to be sealed tight enough in order to avoid uncontrolled transport. For example, in the intestine undigested proteins or carbohydrates must not leave the intestinal lumen in significant amounts as they would act as antigens in the body. On the other hand, ions and water molecules shall be transported through the IC in a controlled fashion. Additionally, the epithelium undergoes continuous and rather rapid self-renewal while maintaining tight cell-cell adhesion. Enterocytes for example are derived from stem cells located in the crypts of Lieberkühn and differentiate as they migrate within three to five days toward the tip of the villus region.Citation13,Citation14 During this directional cell migration, the epithelial integrity of the intestine has to be ensured. This requires the adhesive contacts to rearrange and adapt to different conditions in a highly dynamic way. Thus, the diverse functions of the intestinal epithelium require multiple and differentially regulated adhesive systems.Citation1

The important biological and medical aspects of adhesion between adjoined cells are well established.Citation1,Citation8,Citation12,Citation15-Citation17 In the simple epithelial lining, which can be found for example in the intestine or the renal tubuli, adhesion between cells is mainly accomplished by the so called junctional complex. This junctional complex consists of tight junctions (TJ, zonula occludensCitation8), adherens junctions (AJ, zonula adherensCitation15,Citation18-Citation20) and desmosomes (macula adherensCitation15,Citation21). Besides the junctional complex, gap junctions allow for a molecular exchange between neighboring cells in some tissues.Citation22 The TJs are a branching network of sealing strands, where each strand is formed from a row of transmembrane proteins. These proteins are mainly claudins and occludins of both cell membranes with their extracellular domains joining directly.Citation5,Citation23 The TJ are responsible for sealing the intercellular cleft and for a selective paracellular transport of water or small dissolved molecules.

The AJs, which are mainly composed of cadherins, are responsible for the mechanical strength of the junctional complex.Citation15,Citation16,Citation19,Citation20,Citation24,Citation25 Moreover, desmosomes are also responsible for mechanical strength, forming spot-like interaction sites randomly arranged on the lateral sides of plasma membranes composed of a specialized form of cadherins, namely desmocadherins.Citation16,Citation18,Citation21,Citation26

The Cadherin Superfamily of Adhesion Molecules

Cadherins are Ca2+-dependent type I membrane proteins, i.e., they are single membrane spanning with the N-terminus localized extracellularly.Citation18,Citation27-Citation29 Cadherins are glycoproteins interacting with cadherins of adjoined cells in a mainly homophilic way. These adhesion molecules play an active role in tissue morphogenesis and patterningCitation27,Citation28,Citation30 as they are often expressed in a highly tissue-specific manner and found to be important for cell adhesion within a tissue, and therefore the integrity of that tissue.Citation31,Citation32 The extracellular part of cadherins generally consists of multiple repeats of a ~110 amino acid module, termed a cadherin ecto-domain (EC), that contains several cadherin-specific motifs.Citation33-Citation35 In the past the cadherin superfamily was subdivided into different subfamilies in different ways. This was typically done on the basis of sequence comparisons of the first cadherin domain EC1, on the number of EC-domains and on the functional properties of the cytoplasmic domain.Citation34-Citation36 Dependent on the textbook or review one might find different subdivisions into families, thus, the following categorisation is only one of many possibilities. Type I or ‘classical’ cadherins like E- or N-cadherin consist of 5 EC-domains which exhibit a highly conserved β-barrel conformation and contain four Ca2+-binding motifs. The large cytoplasic domain consisting of 150 to 160 amino acids binds to armadillo proteins and thereby interacts with the actin cytoskeleton. In the first extracellular domain (EC1) classical cadherins are harboring a HAV-motif in the cell adhesion recognition site. Type II or ‘atypical’ cadherins like VE-cadherin exhibit a highly similar structure like type I-cadherins but the HAV-motif is replaced by other sequences. The desmocadherins like desmocollins and desmogleins resemble the structure of classical cadherins in the extracellular part but the cytosolic portion is different, allowing these cadherins to interact with intermediate filaments instead of actin filaments. The flamingo cadherins and the protocadherins are distinguished from classical cadherins by additional EC-domains and a modified cytoplasmic portion. Cadherin-13 (T-cadherin of the chick) exhibits an extracellular part similar to classical cadherins but the extracellular part is attached to the membrane via a glycosylphosphatidylinositol- (GPI-) anchor. In addition, there are several molecules that contain cadherin motifs, but which are not included in any of the above categories, like cadherins-13, -16 and -17.Citation37-Citation42

The Liver-Intestine Cadherin (LI-Cadherin) molecule

In the group of cadherins not falling into the established subfamilies a distinct group within the cadherin superfamily denoted as 7D-cadherins (7 Domain cadherins)Citation36,Citation37,Citation43-Citation45 was found in recent years. The LI- (Liver Intestine-) cadherin, also termed Cadherin-17 (corresponding to CDH17), which is expressed in polarized epithelial cells of liver and intestine in the ratCitation37,Citation38 was the first identified member of this family. Later the Ksp-cadherin (kidney specific) was identified in the kidney (corresponding to the CDH16 gen).Citation42,Citation46,Citation47 In contrast to classical cadherins, 7D-cadherins have seven extracellular cadherin repeats. Human LI-cadherin is a protein of 832 amino acids (120 kD) and shares 20–30% overall homology with classical cadherins.Citation44,Citation48 Instead of the HAV-motif in the cell adhesion recognition site of the EC-1 an AAL-sequence motif is found, which is closely related to the RAL-sequence found in desmogleins.Citation49,Citation50 Another striking difference between LI-cadherin and classical cadherins is the very short (20 amino acids) cytoplasmic domain of LI-cadherin showing no similarity to the highly conserved cytoplasmic region of classical cadherins or to the cytoplasmic portion of any other cadherin subfamily. Especially no binding sites for armadillo proteins, which are necessary for the interaction with catenins and thus with the cytoskeleton, could be identified.

It is assumed that LI-cadherin may have originated from a five-repeat cadherin by a duplication of the first two N-terminal repeats.Citation44,Citation48,Citation51 There are several indications for this hypothesis. One has to keep in mind that the overall amino acid identity of LI- and E-cadherin is less than 25%. However, a detailed comparison of the different domains of LI-cadherin and classical cadherins using the unbiased matrix plot analysis revealed a homology of EC1–5 of classical cadherins to EC3–7 of LI-cadherin and a homology of EC1–2 of LI-cadherin and EC1–2 of classical cadherins. This homology could be verified by a domain-specific protein sequence analysis comparing every LI-cadherin repeat to every E-cadherin repeat. Another structural hint is the missing Ca2+-binding pocket between EC2 and EC3 of LI-cadherin which could be explained by a duplication of EC1 and EC2. Moreover, classical cadherins possess a conserved tryptophan at position 2 in the EC1.Citation18 In LI-cadherin a tryptophan is found at the corresponding position within EC3. In EC1 there is a phenylalanine at position 2 which might adopt a similar function as tryptophan.Citation37,Citation44,Citation48,Citation52

LI- and classical cadherins were excessively compared at the genomic level in the past. The LI-cadherin gene spans 59 kb, contains 18 exons and harbors large introns, especially in the 5′ region.Citation48 The intron-exon organization is perfectly conserved between repeats 3–7 of LI-cadherin and 1–5 of classical cadherins. Moreover, the genomic structure of the repeats 1–2 and 3–4 is identical for LI-cadherin and highly similar to that of the repeats 1–2 of classical cadherins.Citation48

To clarify the genetic and phylogenetic origin of LI-cadherin and the second 7D-cadherin Ksp-cadherin, it might be necessary to investigate the orthologous genes and proteins in other species. Recently, the cdh17 was investigated in zebrafish kidney development.Citation51 cdh17 is the orthologous gene of the mammalian CDH17. However, sequence comparison and a detailed analysis of the expression pattern of the cdh17-protein indicated that zebrafish cdh17 is derived from a gene ancestral to both CDH16 and CDH17. Zebrafish cdh17 expression pattern comprises a combination of both mammalian cadherins, because cdh17 is expressed in both kidney and liver/intestinal tissues. It is tempting to assume that subsequent to divergence of the mammalian lineage, a gene duplication took place resulting in the closely related CDH16 and CDH17 genes. Till now, no zebrafish ortholog of the mammalian CDH16 was found.

Physiological and Pathophysiological Distribution of LI-Cadherin

As explained above, LI-cadherin was initially discovered in rat liver and intestine and thus named LI-cadherin.Citation37 Under physiological conditions, in human and mice, LI-cadherin is exclusively expressed in the small and large intestine but missing in the upper gastric tract and also in the liver.Citation43,Citation44 Here and in the following when using the term expression it is ment that the protein can be detected with standard techniques. In many cases where LI-cadherin is not expressed in a specific tissue it cannot be completely ruled out that the mRNA and even the protein are produced but then degraded in a very fast way.

In zebrafish, as explained above, the LI-cadherin ortholog is also expressed in the kindney.Citation51 The species specific expression pattern and its biological relevance is still unresolved.

During mouse embryogenesis LI-cadherin expression begins at embryonic day 12.5 as shown by Nothern- and Western-blot analysis.Citation43 A transient expression was found in the urogenital sinus and the common bile duct on day 13.5. Furthermore, under physiological conditions LI-cadherin was found exclusively in the intestinal epithelium. The expression onset coincides with the formation of intestinal villi. This developmental stage is characterized by excessive tissue remodeling, growth, and differentiation. It should be emphasized that LI-cadherin was found to be always coexpressed with E-cadherin in polarized cells.

LI-cadherin is uniformly distributed along the lateral cell membranes but is excluded from adherens junctions or desmosomes,Citation37 whereas the coexpressed classical cadherins or desmocadherins are concentrated in these specialized membrane regions. This was investigated by immuno-gold electron microscopy of rat tissue revealing that LI-cadherin is located solely on the basolateral surface of hepatocytes, enterocytes and goblet cells but is absent on the apical plasma membrane.

Under pathological conditions the distribution of LI-cadherin expression can be changed. Especially in the case of metaplasia and tumor development the LI-cadherin expression is impaired. This is in principal concordance with the function of other cadherins which inhibit tumor growth. Reduced cadherin mediated adhesion is one of the factors that may induce metastasis of cancer cells. Reduced expression or adhesive activity of classical cadherins can be a marker of breast, prostate, colon, and stomach cancer cells.Citation31,Citation53-Citation55 The observations for LI-cadherin are not that simple to interpret. A study of human colorectal cancer showed that reduced LI-cadherin expression is associated with high tumor grade, lymphatic invasion and lymph node metastasis.Citation56 Thus analysis of reduced LI-cadherin expression may help to indicate the biological aggressiveness of malignancy of this tumor.Citation57 Also in pancreatic tumors LI-cadherin expression can be impaired. In some tumors LI-cadherin is expressed in significant amounts although under physiological conditions LI-cadherin is not found in pancreas cells.Citation58 LI-cadherin is also found to be upregulated in Barrett’s carcinogenesis and Barrett's metaplasia of the esophagus,Citation59-Citation61 gastric cancer and intestinal metaplasia.Citation57,Citation62-Citation66 Changed LI-cadherin expressions are found as well in intrahepatic cholangiocarcinoma and hepatic carcinoma.Citation67-Citation71 It is presently not understood if the upregulation of LI-cadherin respectively its expression in cells normally lacking LI-cadherin is a cause for the tumor progression or if LI-cadherin is upregulated to compensate e.g., for the impaired E-cadherin mediated adhesion in the corresponding tumor-cells.

Biophysical Properties of LI-Cadherin

LI-cadherin leads to cell aggregation

Soon after the identification of LI-cadherin it was found that LI-cadherin is a functional cell-adhesion molecule. This was mainly found out by cell-adhesion assays. Especially the hanging drop assay proved to be highly valuable.Citation37,Citation46,Citation52,Citation72-Citation74 For this cells expressing the cadherin under investigation are trypsinized and resuspended in culture medium. Droplets of the cell suspension are then placed on the inner side of an Petri dish lid. After defined time steps the number of particles in the hanging drop are counted. Counting is done typically some minutes ( = > number of particles N0) and several hours ( = > number of particles Nt) after droplet formation. The aggregation index is then calculated as (N0 - Nt)/N0.

In quantitative cell-cell adhesion assays based on stable transfected CHO cells or S2-cells, LI-cadherin and E-cadherin exhibited almost identical aggregation indices.Citation74 However, while E-cadherin induced a tight, spherical packing of the cells in hanging-drop experiments and a typical epithelial phenotype of plated cells, LI-cadherin expression resulted in less densely packed clusters in the hanging drop and an intermediate phenotype in the initially fibroblastoid growing CHO cells. LI-cadherin function is obviously sufficient to induce cell clustering at a comparable level to classical cadherins, but the missing link to the cytoskeleton seems to contradict a tight binding and the induction of a fully epithelial phenotype.

LI-cadherin molecules trans-interact directly and specifically

The homotypic, Ca2+-dependent LI-cadherin binding was further shown by single molecule atomic force microscopy (AFM), affinity shift chromatography and laser tweezer studies.Citation72,Citation74 These studies were mainly performed using a chimeric cadherin construct consisting of the full extracellular domain of the cadherin under investigation fused to the Fc-portion of an IgG (cadherin-Fc). The Fc-part was fused to the C-terminus of the LI-cadherin leaving the N-terminus free. This soluble protein can be purified and used for biophysical measurements.

For AFM force spectroscopy cadherin-Fc proteins are covalently attached to the tip of the AFM-cantilever and to the surface of a plate. The tip and plate are brought into interaction and separation by cyclic up- and downward movements of the tip at defined frequencies (force-distance cycles). During downward movement (approach) the tip of the cantilever will eventually hit the plate and, during further downward movement will be gradually deflected upwards until the end of the approach. During the following retrace movement the cantilever bends back with the same linear force slope until the tip separates from the plate to reach the unbent neutral position. During further progression of the retrace movement the cantilever will remain in the neutral position if no interaction between tip- and plate-bound cadherins occurs. Interaction between cadherins can proceed throughout the entire period during which plate and tip are in contact. If trans-interaction has taken place the cantilever will be pulled down below the neutral line, until a critical force is reached (unbinding force) at which the cadherin bond break. This unbinding event is characterized by an abrupt jump of the cantilever to the neutral position which can be detected. As the cadherins are attached to the plate and tip by flexible PEG-linkers, molecules can freely diffuse within the radius of the length of the linkers (~8 nm) allowing them to undergo unimpaired encounter reactions.Citation75,Citation76 Depending on the force loading rate (pulling velocity) the observed unbinding force changes and thus allows to determine the lifetime of the cadherin-cadherin bond. Trans-interacting LI-cadherin molecules exhibit rather weak unbinding forces of 17 to 51 pN at retrace velocities ranging from 150 to 3000 nm/s and a short lifetime of the LI-LI-bond of 1.41 sec. These findings are in line with those reported for classical cadherins like VE-cadherin (unbinding forces of 35 to 55 pN at 200 to 4000 nm/s) and a lifetime of only 0.55 sec.Citation77-Citation79 The unbinding force and lifetime of N-cadherin (≈40 pN, 1.1 sec)Citation80 determined with the same approach are in the same order of magnitude. Similar values were determined on living cells for the unbinding forces and lifetimes of VE-cadherin (50 pN; 2.2 sec), N-cadherin (30 pN; 1.0 sec) and E-cadherin (73 pN; 0.9 sec).Citation79-Citation81

Affinity shift chromatography allowed to determine the dissociation constant KD of the homophilic interaction of LI-cadherin molecules yielded an average KD of about 27 μM.Citation82 Using this approach the homotypic VE-cadherin binding exhibited an average KD of 78 (± 21) μM.Citation78

Specific trans-interaction of LI-cadherin in a cellular context was additionally shown using a laser tweezer approach.Citation82,Citation83 Polystyren micro-spheres were coated with a LI-cadherin-Fc and these spheres were allowed to settle onto LI-cadherin expressing CHO-cells. Within 15 min tight adhesive contacts were formed between the cells and the spheres. These contacts were probed by a laser tweezer. The percentage of spheres resisting displacement by the laser can be taken as a measure for the total binding activity.

Besides characterization of the binding parameters of the homophilic interaction of LI-cadherin molecules, the techniques described above also allow the identification of the heterotypic interaction of LI-cadherin and E-cadherin. The heterotypic trans-interaction of LI- and E-cadherin exhibits unbinding forces, dissociation constants, and lifetimes similar to those found for the homotypic trans-interaction of LI- and E-cadherin respectively.Citation82 The interaction of LI-Fc-coated micro spheres with E-cadherin-expressing CHO cells was not only associated with the accumulation of E-cadherin at the cell–sphere contact sites but also resulted in a cytoplasmic recruitment of catenins to the contact area. The recruitment indicates that the binding of cellular E-cadherin to micro-sphere bound LI-cadherin induces a cytoplasmic assembly of catenins similar to the homotypic E-cadherin interaction.

LI-cadherin is localized in cholesterol rich membrane fractions and stays mobile in the plasma membrane

The short cytosolic portion of LI-cadherin exhibits no structural similarity to the cytosolic domains of classical cadherins. Especially no catenin binding sites can be identified. It is generally believed that LI-cadherin does not interact with the cytoskeleton.Citation52 This is in line with recent measurements of the lateral mobility of cadherin molecules in the cell membrane.Citation73 The lateral mobility was quantified by FRAP (fluorescence recovery after photo bleaching). For this technique LI-cadherin was fluorescently labeled with yellow-fluorescent-protein (LI-YFP). This chimeric protein was expressed in CHO-cells. Then the YFP was photobleached in a restricted area using a laser beam. The time course of the recovery of the fluorescence signal is observed. From the time course of the recovery of the fluorescence signal the diffusion coefficient can be determined. In a recent study LI-cadherin exhibited a diffusion coefficient of 0.124 ± 0.004 m2/s at the contact site in the presence of physiological Ca2+. Upon Ca2+-removal by EGTA the diffusion coefficient rose about 3-fold to 0.420 ± 0.033 m2/s. The authors of the corresponding study concluded that LI-cadherin constitutively forms dimers that freely diffuse in the plasma membrane due to the lack of cytoplasmic interactions. The significantly higher affinity of the homotypic LI-cadherin interaction compared with that of a classical cadherin (see above) is of particular interest in this context. So far it is believed that LI-cadherin does not interact via catenins to the cytoskeleton. Model calculations have shown that both, transmembrane binding to the cytoskeleton and lateral clustering of membrane diffusible adhesion molecules, enhance cell-cell adhesion.Citation17 Clustering could thus contribute in addition to the two to three times higher homotypic affinity of the ectodomain to the overall adhesiveness of LI-cadherin. In the case of the chimeric LI-cadherin molecules fused to a GPI-anchor clustering is very likely, since GPI- anchored proteins are associated with raft-like membrane microdomains. An interesting result of a recent study is that in contrast to the E-cadherin, LI-cadherin is exclusively localized in cholesterol-rich microdomains.Citation82 Furthermore, the homotypic LI-cadherin interaction is strictly dependent on cholesterol levels. It is yet unclear which kind of cholesterol-rich microdomains (caveolae, rafts, etc.) attracts LI- cadherin. These findings appear to be important with regard to the cellular distribution of LI- and E-cadherin in the intestinal epithelium. As described above LI-cadherin is evenly distributed along the basolateral surface of enterocytes,Citation37 the majority of E-cadherin is concentrated in the adherens junctions.Citation84 As the regulation of the lateral mobility e.g., by cytoskeletal tethering is important for controlling cellular adhesion,Citation17,Citation79,Citation80 preformed cadherin clusters could enhance trans-interactions. This preclustering in combination with the high mobility might give rise to a fast establishment of flexible, i.e., highly dynamic adhesive contacts.

LI-cadherin trans-interaction is Ca2+-dependent in a highly cooperative way

The laser tweezer technique as well as the single molecule force spectroscopy using the AFM described above allowed to determine the Ca2+-dependency of the LI-cadherin trans-interaction. The binding activity observed by these two techniques was determined in dependence on the Ca2+-concentration. Trans-interaction of the complete LI-cadherin ectodomain occurred at an apparent KD of 680 µM Ca2+ in the AFM-experiments and at a KD of 720 μM Ca2+ in the laser tweezer experiments.Citation83 In both cases, Ca2+-binding showed a very high degree of cooperativity with a Hill coefficient nH of about 12 corresponding to an all-or-nothing response of LI-cadherin-mediated cell adhesion in response to small changes of extracellular Ca2+ in the range of 600 to 800 mM. Those values are close to the hysiological free Ca2+-concentration of about 1.2 mM. Thus, LI-cadherin appears to act as a Ca2+ -dependent adhesion switch in the lateral contact zones of intestinal epithelial cells.

Calcium ions generally play a crucial role for cadherin structure and function. While it is clear that Ca2+ is vital for cadherin trans-interaction, the role of Ca2+ for cis-dimerization of classical cadherins is under debate. Some studies indicate a Ca2+-independent cis-dimerizationCitation85,Citation86 whereas other experiments suggest Ca2+ to be necessary for cis-dimerization.Citation87,Citation88 For LI-cadherin it was recently shown by FRET-experiments that LI-cadherin cis-dimerizes constitutively on the cell surface.Citation73 The observed FRET efficiency found for LI-CFP/LI-YFP within and outside of cell–cell contacts as well as in the presence and absence of Ca2+ was constant. Thus, while Ca2+ is necessary for trans-interaction of LI-cadherin molecules, cis-dimerization of LI-cadherin is clearly independent of Ca2+ as well as of trans-interactions.

Possible Physiological Functions of LI-Cadherin

The exact biological function of LI-cadherin is currently not known. It has been speculated that due to the high lateral mobility and the flexible preformed clusters in combination with the unusual low KD, LI-cadherin could be responsible for fast and flexible initial formation of adhesive contacts between cells in development and during tissue regeneration.Citation44,Citation72,Citation73 Due to the heterophilic interaction with E-cadherin it might also regulate the E-cadherin mediated adhesion. The non-uniform findings from pathological observations in which up- as well as downregulation of LI-cadherin expression might be correlated with tumor generation and methastasis might be in line with this assumption. However, it is presently not clear if the changed expression of LI-cadherin in pathological cases like carcinogenesis is causally involved or if this is a secondary phenomenon.

It is worth noting that the 7D-cadherins are expressed in epithelia which are involved in water resorption under various osmotic conditions. In the gut for example, where LI-cadherin is expressed, water has to be reabsorbed from the chymus to avoid water loss. The luminal content of the gut shows osmolarities from almost pure water to the high osmolarity of the faeces, far above the physiological osmolarity of the interstitium of about 300 mM.Citation4 A similar situation can be found in the kidney where the urine is produced and where KSP can be found in the epithelium of the tubules. It is tempting to assume the involvement of 7D-cadherins in the regulation of water absorption, as in all these organs water transport plays a major role. It was speculated in the past that LI-cadherin might be involved in regulating the width of the intercellular cleft and thereby influencing the water transport.Citation44,Citation72,Citation89

Another noteworthy fact is the unusual Ca2+-dependency of LI-cadherin binding.Citation72 As we could show recently, LI-cadherin mediated adhesion becomes insufficient at Ca2+-levels just below the physiological concentration of about 1.5mM. This is in contrast to classical cadherins which can tolerate Ca2+-levels down to 0.3mM.Citation77,Citation79,Citation80,Citation90

The facts that (1) 7D-cadherins are expressed in epithelia where water transport under different osmotic conditions takes place and (2) LI-Cadherin displays an extreme sensitivity toward decreased Ca2+-levels led us to the development of a model for the water resorption in epithelia.Citation89 Taking into account that during water transport (due to viscous friction) a small pressure gradient will be built up in the intercellular cleft (IC), the width of the IC b becomes important. In the case of hypotonic medium in the lumen of the resorbing organ (e.g., the gut), a wide cleft (large b) facilitates water transport because of friction minimisation. On the other hand, if the medium is hypertonic, a narrow intercellular cleft favors water resorption since in the small volume, an osmotic gradient between the lumen and the IC can be built up by ATPases. This allows for water uptake from the lumen even if the content, e.g., the faeces, exhibits osmolarity far above the isotonic electrolyte concentration. The derived simple theoretical model shows interesting effects in support of the above hypothesis and suggests a role for 7D-cadherins in the regulation of osmotically driven water transport. Changing the width of the intercellular cleft (IC) between neighboring epithelial cells can regulate the direction and efficiency of water transport through a simple epithelium. In a narrow cleft the cells can increase the concentration of osmotic active substances easily by active transport, but the friction of the transported water is high. If the cleft is wide, friction is reduced but the cells can hardly built up high osmotic gradients. As the Ca2+-concentration is principally coupled to the overall electrolyte concentration, the activity of 7D-cadherins is presumably strictly coupled to the osmotic conditions in the water absorbing organs. Clearly the width has a dramatic effect on the electrolyte concentration and on the water flux. As expected for hypotonic conditions in the lumen, a wide intercellular cleft (b = 400 nm) leads to a higher water flux compared with a narrow cleft (b = 40 nm). In the case of a wide cleft the friction is reduced and the osmotic gradient can be maintained by diffusion of the electrolyte from the interstitium into the cleft.Citation89 The concentration in the IC follows very much the luminal concentration. However, under hypertonic conditions the water flux is inverted, i.e., water flows from the interstitium into the lumen if the cleft is 400 nm wide. Notably this is not the case if the cleft is narrow. For b = 40 nm the volume of the intercellular cleft is small, leading to a concentration in the IC significantly higher than the luminal concentration due to the electrolyte flux maintained by the ATPases. Under these conditions the osmotic gradient is still directed from the lumen into the cleft allowing to further increase the osmolarity of the luminal content.

Thus, it is tempting to assume that active 7D-cadherins, due to their trans-interaction with cadherins of neighboring cells, will cause a narrowing of the intercellular cleft. 7D-cadherins due to their location and their Ca2+-dependence could thus provide a way to passively adapt the direction and efficiency of water transport through epithelia. It has to be emphasized that no experimental proof for this hypothesis could be given so far. Thus, the exact physiological role of LI-cadherin remains enigmatic.

Acknowledgments

The author thanks I. Scholz, B. Vihar and F. Hischen for valuable comments on the manuscript. The DFG (project BA2272/10-1) is acknowledged for financial support

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

References

  • Koch S, Nusrat A. Dynamic regulation of epithelial cell fate and barrier function by intercellular junctions. Ann N Y Acad Sci 2009; 1165:220 - 7; http://dx.doi.org/10.1111/j.1749-6632.2009.04025.x; PMID: 19538310
  • Takashima S, Gold D, Hartenstein V. Stem cells and lineages of the intestine: a developmental and evolutionary perspective. Dev Genes Evol 2012; http://dx.doi.org/10.1007/s00427-012-0422-8; PMID: 23179635
  • Karam SM. Lineage commitment and maturation of epithelial cells in the gut. Front Biosci 1999; 4:D286 - 98; http://dx.doi.org/10.2741/Karam; PMID: 10077541
  • Spring KR. Routes and mechanism of fluid transport by epithelia. Annu Rev Physiol 1998; 60:105 - 19; http://dx.doi.org/10.1146/annurev.physiol.60.1.105; PMID: 9558456
  • Larsen EH, Willumsen NJ, Møbjerg N, Sørensen JN. The lateral intercellular space as osmotic coupling compartment in isotonic transport. Acta Physiol (Oxf) 2009; 195:171 - 86; http://dx.doi.org/10.1111/j.1748-1716.2008.01930.x; PMID: 18983444
  • Singh J, Mlodzik M. Planar Cell Polarity Signaling: Coordination of cellular orientation across tissues. Wiley Interdiscip Rev Dev Biol 2012; 1:479 - 99; http://dx.doi.org/10.1002/wdev.32; PMID: 23066429
  • Devriendt B, De Geest BG, Goddeeris BM, Cox E. Crossing the barrier: Targeting epithelial receptors for enhanced oral vaccine delivery. J Control Release 2012; 160:431 - 9; http://dx.doi.org/10.1016/j.jconrel.2012.02.006; PMID: 22353618
  • Assimakopoulos SF, Papageorgiou I, Charonis A. Enterocytes’ tight junctions: From molecules to diseases. World J Gastrointest Pathophysiol 2011; 2:123 - 37; http://dx.doi.org/10.4291/wjgp.v2.i6.123; PMID: 22184542
  • Kiil F. Mechanisms of transjunctional transport of NaCl and water in proximal tubules of mammalian kidneys. Acta Physiol Scand 2002; 175:55 - 70; http://dx.doi.org/10.1046/j.1365-201X.2002.00967.x; PMID: 11982505
  • Madara JL. Regulation of the movement of solutes across tight junctions. Annu Rev Physiol 1998; 60:143 - 59; http://dx.doi.org/10.1146/annurev.physiol.60.1.143; PMID: 9558458
  • van Heeswijk MP, van Os CH. Osmotic water permeabilities of brush border and basolateral membrane vesicles from rat renal cortex and small intestine. J Membr Biol 1986; 92:183 - 93; http://dx.doi.org/10.1007/BF01870707; PMID: 3761362
  • Komarova Y, Malik AB. Regulation of endothelial permeability via paracellular and transcellular transport pathways. Annu Rev Physiol 2010; 72:463 - 93; http://dx.doi.org/10.1146/annurev-physiol-021909-135833; PMID: 20148685
  • Gordon JI. Intestinal epithelial differentiation: new insights from chimeric and transgenic mice. J Cell Biol 1989; 108:1187 - 94; http://dx.doi.org/10.1083/jcb.108.4.1187; PMID: 2647764
  • Potten CS, Loeffler M. Stem cells: attributes, cycles, spirals, pitfalls and uncertainties. Lessons for and from the crypt. Development 1990; 110:1001 - 20; PMID: 2100251
  • Brieher WM, Yap AS. Cadherin junctions and their cytoskeleton(s). Curr Opin Cell Biol 2012; http://dx.doi.org/10.1016/j.ceb.2012.10.010; PMID: 23127608
  • Le Bras GF, Taubenslag KJ, Andl CD. The regulation of cell-cell adhesion during epithelial-mesenchymal transition, motility and tumor progression. Cell Adh Migr 2012; 6:365 - 73; http://dx.doi.org/10.4161/cam.21326; PMID: 22796940
  • Baumgartner W, Drenckhahn D. Transmembrane cooperative linkage in cellular adhesion. Eur J Cell Biol 2002; 81:161 - 8; http://dx.doi.org/10.1078/0171-9335-00233; PMID: 11998868
  • Angst BD, Marcozzi C, Magee AI. The cadherin superfamily: diversity in form and function. J Cell Sci 2001; 114:629 - 41; PMID: 11171368
  • Lechler T. Adherens junctions and stem cells. Subcell Biochem 2012; 60:359 - 77; http://dx.doi.org/10.1007/978-94-007-4186-7_15; PMID: 22674079
  • Levayer R. [Regulation of intercellular adhesion during epithelial morphogenesis]. Biol Aujourdhui 2012; 206:219 - 36; http://dx.doi.org/10.1051/jbio/2012021; PMID: 23171844
  • Brooke MA, Nitoiu D, Kelsell DP. Cell-cell connectivity: desmosomes and disease. J Pathol 2012; 226:158 - 71; http://dx.doi.org/10.1002/path.3027; PMID: 21989576
  • Wei C-J, Xu X, Lo CW. Connexins and cell signaling in development and disease. Annu Rev Cell Dev Biol 2004; 20:811 - 38; http://dx.doi.org/10.1146/annurev.cellbio.19.111301.144309; PMID: 15473861
  • Guo P, Weinstein AM, Weinbaum S. A dual-pathway ultrastructural model for the tight junction of rat proximal tubule epithelium. Am J Physiol Renal Physiol 2003; 285:F241 - 57; PMID: 12670832
  • Boggetti B, Niessen CM. Adherens junctions in Mammalian development, homeostasis and disease: lessons from mice. Subcell Biochem 2012; 60:321 - 55; http://dx.doi.org/10.1007/978-94-007-4186-7_14; PMID: 22674078
  • Meng W, Takeichi M. Adherens junction: molecular architecture and regulation. Cold Spring Harb Perspect Biol 2009; 1:a002899; http://dx.doi.org/10.1101/cshperspect.a002899; PMID: 20457565
  • Green KJ, Getsios S, Troyanovsky S, Godsel LM. Intercellular junction assembly, dynamics, and homeostasis. Cold Spring Harb Perspect Biol 2010; 2:a000125; http://dx.doi.org/10.1101/cshperspect.a000125; PMID: 20182611
  • Gumbiner BM. Regulation of cadherin-mediated adhesion in morphogenesis. Nat Rev Mol Cell Biol 2005; 6:622 - 34; http://dx.doi.org/10.1038/nrm1699; PMID: 16025097
  • Gumbiner BM. Regulation of cadherin adhesive activity. J Cell Biol 2000; 148:399 - 404; http://dx.doi.org/10.1083/jcb.148.3.399; PMID: 10662767
  • Yap AS, Brieher WM, Gumbiner BM. Molecular and functional analysis of cadherin-based adherens junctions. Annu Rev Cell Dev Biol 1997; 13:119 - 46; http://dx.doi.org/10.1146/annurev.cellbio.13.1.119; PMID: 9442870
  • Tepass U. Genetic analysis of cadherin function in animal morphogenesis. Curr Opin Cell Biol 1999; 11:540 - 8; http://dx.doi.org/10.1016/S0955-0674(99)00025-3; PMID: 10508657
  • Takeichi M. Morphogenetic roles of classic cadherins. Curr Opin Cell Biol 1995; 7:619 - 27; http://dx.doi.org/10.1016/0955-0674(95)80102-2; PMID: 8573335
  • Redies C, Takeichi M. Cadherins in the developing central nervous system: an adhesive code for segmental and functional subdivisions. Dev Biol 1996; 180:413 - 23; http://dx.doi.org/10.1006/dbio.1996.0315; PMID: 8954714
  • Shapiro L, Fannon AM, Kwong PD, Thompson A, Lehmann MS, Grübel G, et al. Structural basis of cell-cell adhesion by cadherins. Nature 1995; 374:327 - 37; http://dx.doi.org/10.1038/374327a0; PMID: 7885471
  • Suzuki ST. Structural and functional diversity of cadherin superfamily: are new members of cadherin superfamily involved in signal transduction pathway?. J Cell Biochem 1996; 61:531 - 42; http://dx.doi.org/10.1002/(SICI)1097-4644(19960616)61:4<531::AID-JCB6>3.0.CO;2-P; PMID: 8806076
  • Suzuki ST. Protocadherins and diversity of the cadherin superfamily. J Cell Sci 1996; 109:2609 - 11; PMID: 8937978
  • Nollet F, Kools P, van Roy F. Phylogenetic analysis of the cadherin superfamily allows identification of six major subfamilies besides several solitary members. J Mol Biol 2000; 299:551 - 72; http://dx.doi.org/10.1006/jmbi.2000.3777; PMID: 10835267
  • Berndorff D, Gessner R, Kreft B, Schnoy N, Lajous-Petter AM, Loch N, et al. Liver-intestine cadherin: molecular cloning and characterization of a novel Ca(2+)-dependent cell adhesion molecule expressed in liver and intestine. J Cell Biol 1994; 125:1353 - 69; http://dx.doi.org/10.1083/jcb.125.6.1353; PMID: 8207063
  • Dantzig AH, Hoskins JA, Tabas LB, Bright S, Shepard RL, Jenkins IL, et al. Association of intestinal peptide transport with a protein related to the cadherin superfamily. Science 1994; 264:430 - 3; http://dx.doi.org/10.1126/science.8153632; PMID: 8153632
  • Kremmidiotis G, Baker E, Crawford J, Eyre HJ, Nahmias J, Callen DF. Localization of human cadherin genes to chromosome regions exhibiting cancer-related loss of heterozygosity. Genomics 1998; 49:467 - 71; http://dx.doi.org/10.1006/geno.1998.5281; PMID: 9615235
  • Sato M, Mori Y, Sakurada A, Fujimura S, Horii A. The H-cadherin (CDH13) gene is inactivated in human lung cancer. Hum Genet 1998; 103:96 - 101; http://dx.doi.org/10.1007/s004390050790; PMID: 9737784
  • Thomson RB, Igarashi P, Biemesderfer D, Kim R, Abu-Alfa A, Soleimani M, et al. Isolation and cDNA cloning of Ksp-cadherin, a novel kidney-specific member of the cadherin multigene family. J Biol Chem 1995; 270:17594 - 601; http://dx.doi.org/10.1074/jbc.270.29.17594; PMID: 7615566
  • Thomson RB, Ward DC, Quaggin SE, Igarashi P, Muckler ZE, Aronson PS. cDNA cloning and chromosomal localization of the human and mouse isoforms of Ksp-cadherin. Genomics 1998; 51:445 - 51; http://dx.doi.org/10.1006/geno.1998.5402; PMID: 9721215
  • Angres B, Kim L, Jung R, Gessner R, Tauber R. LI-cadherin gene expression during mouse intestinal development. Dev Dyn 2001; 221:182 - 93; http://dx.doi.org/10.1002/dvdy.1146; PMID: 11376485
  • Gessner R, Tauber R. Intestinal cell adhesion molecules. Liver-intestine cadherin. Ann N Y Acad Sci 2000; 915:136 - 43; http://dx.doi.org/10.1111/j.1749-6632.2000.tb05236.x; PMID: 11193569
  • Wendeler MW, Jung R, Himmelbauer H, Gessner R. Unique gene structure and paralogy define the 7D-cadherin family. Cell Mol Life Sci 2006; 63:1564 - 73; http://dx.doi.org/10.1007/s00018-006-6014-x; PMID: 16791429
  • Wendeler MW, Praus M, Jung R, Hecking M, Metzig C, Gessner R. Ksp-cadherin is a functional cell-cell adhesion molecule related to LI-cadherin. Exp Cell Res 2004; 294:345 - 55; http://dx.doi.org/10.1016/j.yexcr.2003.11.022; PMID: 15023525
  • Thomson RB, Igarashi P, Biemesderfer D, Kim R, Abu-Alfa A, Soleimani M, et al. Isolation and cDNA cloning of Ksp-cadherin, a novel kidney-specific member of the cadherin multigene family. J Biol Chem 1995; 270:17594 - 601; http://dx.doi.org/10.1074/jbc.270.29.17594; PMID: 7615566
  • Jung R, Wendeler MW, Danevad M, Himmelbauer H, Gessner R. Phylogenetic origin of LI-cadherin revealed by protein and gene structure analysis. Cell Mol Life Sci 2004; 61:1157 - 66; http://dx.doi.org/10.1007/s00018-004-3470-z; PMID: 15141301
  • Koch PJ, Goldschmidt MD, Walsh MJ, Zimbelmann R, Franke WW. Complete amino acid sequence of the epidermal desmoglein precursor polypeptide and identification of a second type of desmoglein gene. Eur J Cell Biol 1991; 55:200 - 8; PMID: 1935985
  • Koch PJ, Walsh MJ, Schmelz M, Goldschmidt MD, Zimbelmann R, Franke WW. Identification of desmoglein, a constitutive desmosomal glycoprotein, as a member of the cadherin family of cell adhesion molecules. Eur J Cell Biol 1990; 53:1 - 12; PMID: 1706270
  • Horsfield J, Ramachandran A, Reuter K, LaVallie E, Collins-Racie L, Crosier K, et al. Cadherin-17 is required to maintain pronephric duct integrity during zebrafish development. Mech Dev 2002; 115:15 - 26; http://dx.doi.org/10.1016/S0925-4773(02)00094-1; PMID: 12049763
  • Kreft B, Berndorff D, Böttinger A, Finnemann S, Wedlich D, Hortsch M, et al. LI-cadherin-mediated cell-cell adhesion does not require cytoplasmic interactions. J Cell Biol 1997; 136:1109 - 21; http://dx.doi.org/10.1083/jcb.136.5.1109; PMID: 9060475
  • Hirohashi S. Inactivation of the E-cadherin-mediated cell adhesion system in human cancers. Am J Pathol 1998; 153:333 - 9; http://dx.doi.org/10.1016/S0002-9440(10)65575-7; PMID: 9708792
  • Hirohashi S. Molecular aspects of adhesion-epigenetic mechanisms for inactivation of the E-Cadherin-mediated cell adhesion system in cancers. Verh Dtsch Ges Pathol 2000; 84:28 - 32; PMID: 11217445
  • Hirohashi S, Kanai Y. Cell adhesion system and human cancer morphogenesis. Cancer Sci 2003; 94:575 - 81; http://dx.doi.org/10.1111/j.1349-7006.2003.tb01485.x; PMID: 12841864
  • Takamura M, Ichida T, Matsuda Y, Kobayashi M, Yamagiwa S, Genda T, et al. Reduced expression of liver-intestine cadherin is associated with progression and lymph node metastasis of human colorectal carcinoma. Cancer Lett 2004; 212:253 - 9; http://dx.doi.org/10.1016/j.canlet.2004.03.016; PMID: 15279905
  • Dong W-G, Yu Q-F, Xu Y, Fan L-F. Li-cadherin is inversely correlated with galectin-3 expression in gastric cancer. Dig Dis Sci 2008; 53:1811 - 7; http://dx.doi.org/10.1007/s10620-007-0080-2; PMID: 17999183
  • Takamura M, Sakamoto M, Ino Y, Shimamura T, Ichida T, Asakura H, et al. Expression of liver-intestine cadherin and its possible interaction with galectin-3 in ductal adenocarcinoma of the pancreas. Cancer Sci 2003; 94:425 - 30; http://dx.doi.org/10.1111/j.1349-7006.2003.tb01459.x; PMID: 12824888
  • Weimann A, Rieger A, Zimmermann M, Gross M, Hoffmann P, Slevogt H, et al. Comparison of six immunohistochemical markers for the histologic diagnosis of neoplasia in Barrett’s esophagus. Virchows Arch 2010; 457:537 - 45; http://dx.doi.org/10.1007/s00428-010-0972-y; PMID: 20844891
  • Weimann A, Zimmermann M, Gross M, Slevogt H, Rieger A, Morawietz L. CDX2 and LI-cadherin expression in esophageal mucosa: use of both markers can facilitate the histologic diagnosis of Barrett’s esophagus and carcinoma. Int J Surg Pathol 2010; 18:330 - 7; PMID: 20444732
  • Mokrowiecka A, Zonnur S, Veits L, Musial J, Kordek R, Lochowski M, et al. Liver-Intestine-Cadherin Is a Sensitive Marker of Intestinal Differentiation During Barrett’s Carcinogenesis. Dig Dis Sci 2012; http://dx.doi.org/10.1007/s10620-012-2425-8; PMID: 23053896
  • Ryu KH, Shim KN, Jung SA, Yoo K, Joo YH, Lee JH. Significance of preoperative tissue levels of vascular-endothelial cadherin, liver-intestine cadherin and vascular endothelial growth factor in gastric cancer. Korean J Gastroenterol 2012; 60:229 - 41; http://dx.doi.org/10.4166/kjg.2012.60.4.229; PMID: 23089909
  • Dong W, Yu Q, Xu Y. Altered expression of a Li-cadherin in gastric cancer and intestinal metaplasia. Dig Dis Sci 2007; 52:536 - 42; http://dx.doi.org/10.1007/s10620-006-9316-9; PMID: 17226075
  • Su M-C, Yuan R-H, Lin C-Y, Jeng Y-M. Cadherin-17 is a useful diagnostic marker for adenocarcinomas of the digestive system. Mod Pathol 2008; 21:1379 - 86; http://dx.doi.org/10.1038/modpathol.2008.107; PMID: 18552820
  • Wang XQ, Luk JM, Leung PP, Wong BW, Stanbridge EJ, Fan ST. Alternative mRNA splicing of liver intestine-cadherin in hepatocellular carcinoma. Clin Cancer Res 2005; 11:483 - 9; PMID: 15701831
  • Ge J, Chen Z, Wu S, Yuan W, Hu B, Chen Z. A clinicopathological study on the expression of cadherin-17 and caudal-related homeobox transcription factor (CDX2) in human gastric carcinoma. Clin Oncol (R Coll Radiol) 2008; 20:275 - 83; http://dx.doi.org/10.1016/j.clon.2008.01.013; PMID: 18353622
  • Liu LX, Lee NP, Chan VW, Xue W, Zender L, Zhang C, et al. Targeting cadherin-17 inactivates Wnt signaling and inhibits tumor growth in liver carcinoma. Hepatology 2009; 50:1453 - 63; http://dx.doi.org/10.1002/hep.23143; PMID: 19676131
  • Lee NP, Poon RTP, Shek FH, Ng IOL, Luk JM. Role of cadherin-17 in oncogenesis and potential therapeutic implications in hepatocellular carcinoma. Biochim Biophys Acta 2010; 1806:138 - 45; PMID: 20580775
  • Zhu R, Wong K-F, Lee NPY, Lee K-F, Luk JMC. HNF1α and CDX2 transcriptional factors bind to cadherin-17 (CDH17) gene promoter and modulate its expression in hepatocellular carcinoma. J Cell Biochem 2010; 111:618 - 26; http://dx.doi.org/10.1002/jcb.22742; PMID: 20568120
  • Yu Q-F, Dong W-G, Ren J-L. Knockdown of Li-cadherin increases metastatic behaviors of LoVo cells. J Cancer Res Clin Oncol 2010; 136:1641 - 9; http://dx.doi.org/10.1007/s00432-010-0822-y; PMID: 20204409
  • Takamura M, Yamagiwa S, Wakai T, Tamura Y, Kamimura H, Kato T, et al. Loss of liver-intestine cadherin in human intrahepatic cholangiocarcinoma promotes angiogenesis by up-regulating metal-responsive transcription factor-1 and placental growth factor. Int J Oncol 2010; 36:245 - 54; PMID: 19956853
  • Wendeler MW, Drenckhahn D, Gessner R, Baumgartner W. Intestinal LI-cadherin acts as a Ca2+-dependent adhesion switch. J Mol Biol 2007; 370:220 - 30; http://dx.doi.org/10.1016/j.jmb.2007.04.062; PMID: 17512947
  • Bartolmäs T, Hirschfeld-Ihlow C, Jonas S, Schaefer M, Geßner R. LI-cadherin cis-dimerizes in the plasma membrane Ca(2+) independently and forms highly dynamic trans-contacts. Cell Mol Life Sci 2012; 69:3851 - 62; http://dx.doi.org/10.1007/s00018-012-1053-y; PMID: 22842778
  • Baumgartner W, Wendeler MW, Weth A, Koob R, Drenckhahn D, Gessner R. Heterotypic trans-interaction of LI- and E-cadherin and their localization in plasmalemmal microdomains. J Mol Biol 2008; 378:44 - 54; http://dx.doi.org/10.1016/j.jmb.2008.02.023; PMID: 18342884
  • Baumgartner W, Hinterdorfer P, Schindler H. Data analysis of interaction forces measured with the atomic force microscope. Ultramicroscopy 2000; 82:85 - 95; http://dx.doi.org/10.1016/S0304-3991(99)00154-0; PMID: 10741656
  • Hinterdorfer P, Baumgartner W, Gruber HJ, Schilcher K, Schindler H. Detection and localization of individual antibody-antigen recognition events by atomic force microscopy. Proc Natl Acad Sci U S A 1996; 93:3477 - 81; http://dx.doi.org/10.1073/pnas.93.8.3477; PMID: 8622961
  • Baumgartner W, Hinterdorfer P, Ness W, Raab A, Vestweber D, Schindler H, et al. Cadherin interaction probed by atomic force microscopy. Proc Natl Acad Sci U S A 2000; 97:4005 - 10; http://dx.doi.org/10.1073/pnas.070052697; PMID: 10759550
  • Baumgartner W, Drenckhahn D. Plasmalemmal concentration and affinity of mouse vascular endothelial cadherin, VE-cadherin. Eur Biophys J 2002; 31:532 - 8; http://dx.doi.org/10.1007/s00249-002-0248-9; PMID: 12451422
  • Baumgartner W, Schütz GJ, Wiegand J, Golenhofen N, Drenckhahn D. Cadherin function probed by laser tweezer and single molecule fluorescence in vascular endothelial cells. J Cell Sci 2003; 116:1001 - 11; http://dx.doi.org/10.1242/jcs.00322; PMID: 12584244
  • Baumgartner W, Golenhofen N, Grundhöfer N, Wiegand J, Drenckhahn D. Ca2+ dependency of N-cadherin function probed by laser tweezer and atomic force microscopy. J Neurosci 2003; 23:11008 - 14; PMID: 14657157
  • Perret E, Benoliel AM, Nassoy P, Pierres A, Delmas V, Thiery JP, et al. Fast dissociation kinetics between individual E-cadherin fragments revealed by flow chamber analysis. EMBO J 2002; 21:2537 - 46; http://dx.doi.org/10.1093/emboj/21.11.2537; PMID: 12032067
  • Baumgartner W, Wendeler MW, Weth A, Koob R, Drenckhahn D, Gessner R. Heterotypic trans-interaction of LI- and E-cadherin and their localization in plasmalemmal microdomains. J Mol Biol 2008; 378:44 - 54; http://dx.doi.org/10.1016/j.jmb.2008.02.023; PMID: 18342884
  • Wendeler MW, Drenckhahn D, Gessner R, Baumgartner W. Intestinal LI-cadherin acts as a Ca2+-dependent adhesion switch. J Mol Biol 2007; 370:220 - 30; http://dx.doi.org/10.1016/j.jmb.2007.04.062; PMID: 17512947
  • Boller K, Vestweber D, Kemler R. Cell-adhesion molecule uvomorulin is localized in the intermediate junctions of adult intestinal epithelial cells. J Cell Biol 1985; 100:327 - 32; http://dx.doi.org/10.1083/jcb.100.1.327; PMID: 3880756
  • Chitaev NA, Troyanovsky SM. Adhesive but not lateral E-cadherin complexes require calcium and catenins for their formation. J Cell Biol 1998; 142:837 - 46; http://dx.doi.org/10.1083/jcb.142.3.837; PMID: 9700170
  • Brieher WM, Yap AS, Gumbiner BM. Lateral dimerization is required for the homophilic binding activity of C-cadherin. J Cell Biol 1996; 135:487 - 96; http://dx.doi.org/10.1083/jcb.135.2.487; PMID: 8896604
  • Pertz O, Bozic D, Koch AW, Fauser C, Brancaccio A, Engel J. A new crystal structure, Ca2+ dependence and mutational analysis reveal molecular details of E-cadherin homoassociation. EMBO J 1999; 18:1738 - 47; http://dx.doi.org/10.1093/emboj/18.7.1738; PMID: 10202138
  • Vunnam N, Pedigo S. Sequential binding of calcium leads to dimerization in neural cadherin. Biochemistry 2011; 50:2973 - 82; http://dx.doi.org/10.1021/bi101872b; PMID: 21366346
  • Ahl M, Weth A, Walcher S, Baumgartner W. The function of 7D-cadherins: a mathematical model predicts physiological importance for water transport through simple epithelia. Theor Biol Med Model 2011; 8:18; http://dx.doi.org/10.1186/1742-4682-8-18; PMID: 21663598
  • Heupel WM, Baumgartner W, Laymann B, Drenckhahn D, Golenhofen N. Different Ca2+ affinities and functional implications of the two synaptic adhesion molecules cadherin-11 and N-cadherin. Mol Cell Neurosci 2008; 37:548 - 58; http://dx.doi.org/10.1016/j.mcn.2007.12.003; PMID: 18201900