Skip to main content
Erschienen in: European Journal of Medical Research 1/2014

Open Access 01.12.2014 | Meeting abstract

α5β1 integrins in hepatocytes act as receptors for bile acids with a (nor)ursodeoxycholane scaffold

verfasst von: Michele Bonus, Annika Sommerfeld, Dieter Häussinger, Holger Gohlke

Erschienen in: European Journal of Medical Research | Sonderheft 1/2014

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN
Ursodeoxycholic acid (UDCA) is a standard treatment in several cholestatic liver diseases (Figure 1A) [1]. In vivo, conjugation with taurine occurs rapidly and yields tauroursodeoxycholic acid (TUDC), which has been shown to promote choleresis by triggering the insertion of ATP-dependent transport proteins (e.g., the bile salt export pump (Bsep) and the multidrug resistance protein-2 (Mrp2)) into the canalicular membrane [2]. TUDC-induced recruitment of Bsep results from activation of focal adhesion kinase (FAK), phosphatidylinositol 3-kinase (PI3 kinase), and c-Src, which leads to downstream activation of extracellular signal-regulated kinases (Erks) and p38 mitogen-activated protein kinase (p38MAPK). Upon hepatocyte swelling, either induced by exposure to a hypoosmotic environment or insulin, α5β1 integrins become activated and trigger similar signaling events towards choleresis. α5β1 Integrins may also become activated by a swelling-independent way as previously shown by exposing hepatocytes to pathophysiological concentrations of urea [3]. Both TUDC-induced and swelling-dependent signaling were abolished in the presence of an antagonistic, RGD-motif containing hexapeptide (GRGD SP).
These findings led us to hypothesize that α5β1 integrin will act as a receptor for TUDC in hepatocytes. We tested this hypothesis in a combined experimental and computational study [4]. Immunofluorescence staining on cryosections of isolated perfused rat liver (IPRL) revealed the active conformation of β1 integrin within 1 min after addition of TUDC at a concentration of 20 µM. Furthermore, phosphorylation of Erk-1 and -2 as well as activation of the epidermal growth factor receptor were induced by TUDC within the same time span. These effects were sensitive to inhibition by GRGD SP but insensitive to the presence of an inactive control peptide (GRADSP). As TUDC does not affect hepatocyte volume, which excludes that TUDC triggers integrin activation osmotically, these findings demonstrated that TUDC directly activates α5β1 integrins and triggers signaling events towards choleresis. While swelling-induced β1 integrin activation occurs primarily in the plasma membrane, TUDC-induced β1 integrin activation occurs primarily in the cytosol of hepatocytes. We demonstrated that the presence of the Na+/taurocholate cotransporting polypeptide (Ntcp) is required for the latter. The need to uptake and/or concentrate TUDC inside the hepatocyte for β1 integrin activation to occur may explain why TUDC primarily acts in the liver.
In order to provide insights at a molecular level as to how TUDC activates α5β1-integrin, a complex structure of a homology model of the ectodomain of α5β1-integrin and TUDC was generated by molecular docking and subsequently subjected to molecular dynamics (MD) simulations of 200 ns length [4]. These simulations revealed pronounced conformational changes in three regions of the βA domain of the integrin (Figure 1B): I) Helix α1 straightens and becomes continuous; II) this leads to a tighter packing between the top of helix α7 and the center of α1, which has been characterized as “T-junction formation” in an X-ray structure of integrin αIIbβ3 bound to a ligand as well as in computational studies of agonist-bound integrins; III) as a result, helix α7 moves downwards and outwards, which imposes a torque on the hybrid domain. The induced rotational motion of the hybrid domain is a prerequisite for the unbending of the integrin ectodomain, which, in turn, is required for integrin activation according to current models. Neither did MD simulations of the ectodomain of α5β1 integrin bound to GRGD SP nor to taurocholic acid (TC) (Figure 1A) reveal such conformational changes, in line with results from immunofluorescence staining of IPRL that did not reveal an appearance of the active conformation of β1-integrin upon addition of TC either. The bile acids glycochenodeoxycholic acid (GCDC), taurochenodeoxycholic acid (TCDC), or taurolithocholic acid 3-sulfate (TLCS) were likewise ineffective with respect to β1 integrin activation according to immunofluorescence staining. All these bile acids differ from TUDC with respect to the configuration and/or presence or absence of functional groups in the cholane moiety.
In contrast, the taurine conjugate (Tnor UDCA) of nor UDCA (Figure 1A), a C23 homolog of UDCA that lacks a methylene group in the sidechain, is moderately effective in exerting anticholestatic effects in experimental hepatocellular cholestatis [5]. Preliminary results from immunofluorescence staining of IPRL indicate that Tnor UDCA and nor UDCA can activate β1 integrins, with stronger effects observed with nor UDCA. Another sidechain modification occurs if glycine rather than taurine is conjugated with the bile acid in the terminal synthesis step. Preliminary results from immunofluorescence staining indicate that the glyco-conjugated UDCA (GUDC; Figure 1A) does not activate β1 integrins although GUDC can be transported by the Ntcp [6]. In order to investigate the bile acids’ modes of action at a molecular level, we subjected nor UDCA and GUDC bound to the ectodomain of α5β1 integrin to MD simulations, employing the same setup as for the simulations above. In addition, we also performed MD simulations of the complex of α5β1 integrin with the unconjugated UDCA as well as of a ligand-free structure of the α5β1 ectodomain for reference. Together with the above results for TUDC and TC-bound α5β1 integrin, these simulations reveal a significant correlation between characteristic conformational changes in the βA domain and the potential of the bile acid to activate β1 integrins as observed in immunofluorescence staining: I) the higher this potential (TUDC, nor UDCA), the less is helix α1 kinked and the more is helix α7 tilted with respect to the ligand-free structure; II) changes in the opposite direction are observed for the non-activating bile acids (TC, GUDC); III) the MD simulations reveal that changes of helix α1 towards an activated integrin state are more pronounced than those of helix α7. According to these preliminary results, we predict that UDCA does not activate β1 integrins because the conformational characteristics of helices α1 and α7 observed with this bile acid do not differ much from those of the ligand-free structure. Finally, the MD simulations suggest that the cholane scaffolds of TUDC and nor UDCA adopt different binding modes in the cleft between the propeller and βA domains of α5β1 integrin; yet, the activating effects of both bile acids is funneled through helix α1 and from there leads to allosteric changes in the βA domain that propagate towards the hybrid domain.
In summary, in a combined computational and experimental study, we showed that TUDC directly activates α5β1 integrins inside hepatocytes and induces conformational changes in the β1 subunit that lead to integrin activation and swelling-independent signaling towards choleresis. A bile acid with a nor ursodeoxycholane scaffold (nor UDCA) was shown to activate β1 integrins even without conjugation. In contrast, bile acids modified in the cholane scaffold (TC, TCDC, TLCS) or conjugated to glycine (GCDC, GUDC) were shown to be non-activating. This suggests a unique role of the (nor)ursodeoxycholane scaffold for direct interaction with and activation of α5β1 integrins in connection with no or a taurine conjugation.

Acknowledgements

This study was supported by the Deutsche Forschungsgemeinschaft through the Collaborative Research Centers SFB 575 (‘Experimental Hepatology’, Düsseldorf) and SFB 974 (‘Communication and Systems Relevance during Liver Damage and Regeneration’, Düsseldorf) and the Clinical Research Group KFO 217 (‘Hepatobiliary Transport in Health and Disease’, Düsseldorf), and by the initiative ‘Fit for Excellence’ at the Heinrich-Heine-University. The authors are grateful to the ‘Zentrum für Informations- und Medientechnologie’ (ZIM) at the Heinrich-Heine-University for computational support. We are grateful to Dr. Nadine Homeyer for fruitful discussions.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
The Creative Commons Public Domain Dedication waiver (https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
download
DOWNLOAD
print
DRUCKEN
Literatur
1.
Zurück zum Zitat Beuers U: Drug insight: mechanisms and sites of action of ursodeoxycholic acid in cholestasis. Nature Clin Pract Gastroenterol Hepatol 2006, 3: 318–328. 10.1038/ncpgasthep0521CrossRef Beuers U: Drug insight: mechanisms and sites of action of ursodeoxycholic acid in cholestasis. Nature Clin Pract Gastroenterol Hepatol 2006, 3: 318–328. 10.1038/ncpgasthep0521CrossRef
2.
Zurück zum Zitat Kurz AK, Graf D, Schmitt M, Vom Dahl S, Häussinger D: Tauroursodesoxycholate-induced choleresis involves p38(MAPK) activation and translocation of the bile salt export pump in rats. Gastroenterology 2001, 121: 407–419. 10.1053/gast.2001.26262PubMedCrossRef Kurz AK, Graf D, Schmitt M, Vom Dahl S, Häussinger D: Tauroursodesoxycholate-induced choleresis involves p38(MAPK) activation and translocation of the bile salt export pump in rats. Gastroenterology 2001, 121: 407–419. 10.1053/gast.2001.26262PubMedCrossRef
3.
Zurück zum Zitat Reinehr R, Gohlke H, Sommerfeld A, Vom Dahl S, Häussinger D: Activation of integrins by urea in perfused rat liver. J Biol Chem 2010, 285: 29348–29356. 10.1074/jbc.M110.155135PubMedCentralPubMedCrossRef Reinehr R, Gohlke H, Sommerfeld A, Vom Dahl S, Häussinger D: Activation of integrins by urea in perfused rat liver. J Biol Chem 2010, 285: 29348–29356. 10.1074/jbc.M110.155135PubMedCentralPubMedCrossRef
4.
Zurück zum Zitat Gohlke H, Schmitz B, Sommerfeld A, Reinehr R, Häussinger D: a5b1-Integrins are sensors for tauroursodeoxycholic acid in hepatocytes. Hepatology 2013, 57: 1117–1129. 10.1002/hep.25992PubMedCrossRef Gohlke H, Schmitz B, Sommerfeld A, Reinehr R, Häussinger D: a5b1-Integrins are sensors for tauroursodeoxycholic acid in hepatocytes. Hepatology 2013, 57: 1117–1129. 10.1002/hep.25992PubMedCrossRef
5.
Zurück zum Zitat Fickert P, Pollheimer MJ, Silbert D, Moustafa T, Halilbasic E, Krones E, Durchschein F, Thuringer A, Zollner G, Denk H, et al.: Differential effects of norUDCA and UDCA in obstructive cholestasis in mice. J Hepatol 2013, 58: 1201–1208. 10.1016/j.jhep.2013.01.026PubMedCentralPubMedCrossRef Fickert P, Pollheimer MJ, Silbert D, Moustafa T, Halilbasic E, Krones E, Durchschein F, Thuringer A, Zollner G, Denk H, et al.: Differential effects of norUDCA and UDCA in obstructive cholestasis in mice. J Hepatol 2013, 58: 1201–1208. 10.1016/j.jhep.2013.01.026PubMedCentralPubMedCrossRef
6.
Zurück zum Zitat Maeda K, Kambara M, Tian Y, Hofmann AF, Sugiyama Y: Uptake of ursodeoxycholate and its conjugates by human hepatocytes: role of Na(+)-taurocholate cotransporting polypeptide (NTCP), organic anion transporting polypeptide (OATP) 1B1 (OATP-C), and oatp1B3 (OATP8). Mol Pharm 2006, 3: 70–77. 10.1021/mp050063uPubMedCrossRef Maeda K, Kambara M, Tian Y, Hofmann AF, Sugiyama Y: Uptake of ursodeoxycholate and its conjugates by human hepatocytes: role of Na(+)-taurocholate cotransporting polypeptide (NTCP), organic anion transporting polypeptide (OATP) 1B1 (OATP-C), and oatp1B3 (OATP8). Mol Pharm 2006, 3: 70–77. 10.1021/mp050063uPubMedCrossRef
Metadaten
Titel
α5β1 integrins in hepatocytes act as receptors for bile acids with a (nor)ursodeoxycholane scaffold
verfasst von
Michele Bonus
Annika Sommerfeld
Dieter Häussinger
Holger Gohlke
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
European Journal of Medical Research / Ausgabe Sonderheft 1/2014
Elektronische ISSN: 2047-783X
DOI
https://doi.org/10.1186/2047-783X-19-S1-S13

Weitere Artikel der Sonderheft 1/2014

European Journal of Medical Research 1/2014 Zur Ausgabe