期刊论文详细信息
BMC Gastroenterology
Cholestasis induces reversible accumulation of periplakin in mouse liver
Frank J Gonzalez1  Jeffrey M Peters6  Weiwei Shan3  Cherie R Anderson6  Sung-hoon Ahn5  Yatrik M Shah4  Tsutomu Matsubara2  Junko Satoh7  Shinji Ito7 
[1] Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 3106, 37 Convent Drive, Bethesda, Maryland 20892, USA;Department of Anatomy and Regenerative Biology, Graduate School of Medicine, Osaka City University, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan;Department of Pathology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, Texas 77030, USA;Department of Molecular and Integrative Physiology, University of Michigan, 7712B Med Sci II, 1137 E. Catherine St., Ann Arbor, MI 48109-5822, USA;Drug Discovery Platform Technology Team, Korea Research Institute of Chemical Technology, Sinseongno 19, Yoosung, Daejeon 305-343, South Korea;Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, Pennsylvania State University, 312 Life Sciences Building, University Park 16802, Pennsylvania, USA;Biofrontier Platform, Graduate School of Medicine, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
关键词: Urinary stasis;    Farnesoid X receptor;    Bile acids;    Cholestasis;    Periplakin;   
Others  :  857793
DOI  :  10.1186/1471-230X-13-116
 received in 2013-02-04, accepted in 2013-07-12,  发布年份 2013
PDF
【 摘 要 】

Background

Periplakin (PPL) is a rod-shaped cytolinker protein thought to connect cellular adhesion junctional complexes to cytoskeletal filaments. PPL serves as a structural component of the cornified envelope in the skin and interacts with various types of proteins in cultured cells; its level decreases dramatically during tumorigenic progression in human epithelial tissues. Despite these intriguing observations, the physiological roles of PPL, especially in non-cutaneous tissues, are still largely unknown. Because we observed a marked fluctuation of PPL expression in mouse liver in association with the bile acid receptor farnesoid X receptor (FXR) and cholestasis, we sought to characterize the role of PPL in the liver and determine its contributions to the etiology and pathogenesis of cholestasis.

Methods

Time- and context-dependent expression of PPL in various mouse models of hepatic and renal disorders were examined by immunohistochemistry, western blotting, and quantitative real-time polymerase chain reactions.

Results

The hepatic expression of PPL was significantly decreased in Fxr−/− mice. In contrast, the expression was dramatically increased during cholestasis, with massive PPL accumulation observed at the boundaries of hepatocytes in wild-type mice. Interestingly, the hepatic accumulation of PPL resulting from cholestasis was reversible. In addition, similar accumulation of PPL at cellular boundaries was found in epithelial cells around renal tubules upon ureteral obstruction.

Conclusions

PPL may be involved in the temporal accommodation to fluid stasis in different tissues. Further examination of the roles for PPL may lead to the discovery of a novel mechanism for cellular protection by cytolinkers that is applicable to many tissues and in many contexts.

【 授权许可】

   
2013 Ito et al.; licensee BioMed Central Ltd.

【 预 览 】
附件列表
Files Size Format View
20140723084546366.pdf 2745KB PDF download
156KB Image download
210KB Image download
186KB Image download
168KB Image download
137KB Image download
194KB Image download
248KB Image download
【 图 表 】

【 参考文献 】
  • [1]Trauner M, Jansen PLM: Molecular pathogenesis of cholestasis. New York, USA: Eurekah.com and Kluwer Academic / Plenum Publishers; 2004.
  • [2]O'Leary JG, Pratt DS: Cholestasis and cholestatic syndromes. Curr Opin Gastroenterol 2007, 23(3):232-236.
  • [3]Yang JI, Yoon JH, Myung SJ, Gwak GY, Kim W, Chung GE, Lee SH, Lee SM, Kim CY, Lee HS: Bile acid-induced TGR5-dependent c-Jun-N terminal kinase activation leads to enhanced caspase 8 activation in hepatocytes. Biochem Biophys Res Commun 2007, 361(1):156-161.
  • [4]Wagner M, Fickert P, Zollner G, Fuchsbichler A, Silbert D, Tsybrovskyy O, Zatloukal K, Guo GL, Schuetz JD, Gonzalez FJ, et al.: Role of farnesoid X receptor in determining hepatic ABC transporter expression and liver injury in bile duct-ligated mice. Gastroenterology 2003, 125(3):825-838.
  • [5]Marschall HU, Wagner M, Bodin K, Zollner G, Fickert P, Gumhold J, Silbert D, Fuchsbichler A, Sjovall J, Trauner M: Fxr(−/−) mice adapt to biliary obstruction by enhanced phase I detoxification and renal elimination of bile acids. J Lipid Res 2006, 47(3):582-592.
  • [6]Stedman C, Liddle C, Coulter S, Sonoda J, Alvarez JG, Evans RM, Downes M: Benefit of farnesoid X receptor inhibition in obstructive cholestasis. Proc Natl Acad Sci USA 2006, 103(30):11323-11328.
  • [7]Chen F, Ananthanarayanan M, Emre S, Neimark E, Bull LN, Knisely AS, Strautnieks SS, Thompson RJ, Magid MS, Gordon R, et al.: Progressive familial intrahepatic cholestasis, type 1, is associated with decreased farnesoid X receptor activity. Gastroenterology 2004, 126(3):756-764.
  • [8]Sinal CJ, Tohkin M, Miyata M, Ward JM, Lambert G, Gonzalez FJ: Targeted disruption of the nuclear receptor FXR/BAR impairs bile acid and lipid homeostasis. Cell 2000, 102(6):731-744.
  • [9]Leung CL, Green KJ, Liem RK: Plakins: a family of versatile cytolinker proteins. Trends Cell Biol 2002, 12(1):37-45.
  • [10]Jefferson JJ, Leung CL, Liem RK: Plakins: goliaths that link cell junctions and the cytoskeleton. Nat Rev Mol Cell Biol 2004, 5(7):542-553.
  • [11]Sonnenberg A, Liem RK: Plakins in development and disease. Exp Cell Res 2007, 313(10):2189-2203.
  • [12]Simon M, Green H: Participation of membrane-associated proteins in the formation of the cross-linked envelope of the keratinocyte. Cell 1984, 36(4):827-834.
  • [13]Ruhrberg C, Hajibagheri MA, Parry DA, Watt FM: Periplakin, a novel component of cornified envelopes and desmosomes that belongs to the plakin family and forms complexes with envoplakin. J Cell Biol 1997, 139(7):1835-1849.
  • [14]Aho S, McLean WH, Li K, Uitto J: cDNA cloning, mRNA expression, and chromosomal mapping of human and mouse periplakin genes. Genomics 1998, 48(2):242-247.
  • [15]Kazerounian S, Uitto J, Aho S: Unique role for the periplakin tail in intermediate filament association: specific binding to keratin 8 and vimentin. Exp Dermatol 2002, 11(5):428-438.
  • [16]Candi E, Schmidt R, Melino G: The cornified envelope: a model of cell death in the skin. Nat Rev Mol Cell Biol 2005, 6(4):328-340.
  • [17]Ruhrberg C, Hajibagheri MA, Simon M, Dooley TP, Watt FM: Envoplakin, a novel precursor of the cornified envelope that has homology to desmoplakin. J Cell Biol 1996, 134(3):715-729.
  • [18]Boczonadi V, McInroy L, Maatta A: Cytolinker cross-talk: periplakin N-terminus interacts with plectin to regulate keratin organisation and epithelial migration. Exp Cell Res 2007, 313(16):3579-3591.
  • [19]Yoon KH, FitzGerald PG: Periplakin interactions with lens intermediate and beaded filaments. Invest Ophthalmol Vis Sci 2009, 50(3):1283-1289.
  • [20]Milligan G, Murdoch H, Kellett E, White JH, Feng GJ: Interactions between G-protein-coupled receptors and periplakin: a selective means to regulate G-protein activation. Biochem Soc Trans 2004, 32(Pt 5):878-880.
  • [21]Murdoch H, Feng GJ, Bachner D, Ormiston L, White JH, Richter D, Milligan G: Periplakin interferes with G protein activation by the melanin-concentrating hormone receptor-1 by binding to the proximal segment of the receptor C-terminal tail. J Biol Chem 2005, 280(9):8208-8220.
  • [22]Ward RJ, Jenkins L, Milligan G: Selectivity and functional consequences of interactions of family A G protein-coupled receptors with neurochondrin and periplakin. J Neurochem 2009, 109(1):182-192.
  • [23]van den Heuvel AP, De Vries-Smits AM, Van Weeren PC, Dijkers PF, De Bruyn KM, Riedl JA, Burgering BM: Binding of protein kinase B to the plakin family member periplakin. J Cell Sci 2002, 115(Pt 20):3957-3966.
  • [24]Beekman JM, Bakema JE, van de Winkel JG, Leusen JH: Direct interaction between FcgammaRI (CD64) and periplakin controls receptor endocytosis and ligand binding capacity. Proc Natl Acad Sci USA 2004, 101(28):10392-10397.
  • [25]Nishimori T, Tomonaga T, Matsushita K, Oh-Ishi M, Kodera Y, Maeda T, Nomura F, Matsubara H, Shimada H, Ochiai T: Proteomic analysis of primary esophageal squamous cell carcinoma reveals downregulation of a cell adhesion protein, periplakin. Proteomics 2006, 6(3):1011-1018.
  • [26]Hatakeyama H, Kondo T, Fujii K, Nakanishi Y, Kato H, Fukuda S, Hirohashi S: Protein clusters associated with carcinogenesis, histological differentiation and nodal metastasis in esophageal cancer. Proteomics 2006, 6(23):6300-6316.
  • [27]Matsumoto T, Kuruma H, Kamata Y, Satoh E, Ohkusa H, Takahashi K, Nishimori T, Tomonaga T, Satoh T, Iwamura MN F: Periplakin as a potential biological marker for bladder cancer. Proceedings of the 67th Annual Meeting of the Japanese Cancer Association 2008, 326.
  • [28]Aho S, Li K, Ryoo Y, McGee C, Ishida-Yamamoto A, Uitto J, Klement JF: Periplakin gene targeting reveals a constituent of the cornified cell envelope dispensable for normal mouse development. Mol Cell Biol 2004, 24(14):6410-6418.
  • [29]Sevilla LM, Nachat R, Groot KR, Klement JF, Uitto J, Djian P, Maatta A, Watt FM: Mice deficient in involucrin, envoplakin, and periplakin have a defective epidermal barrier. J Cell Biol 2007, 179(7):1599-1612.
  • [30]Shan W, Nicol CJ, Ito S, Bility MT, Kennett MJ, Ward JM, Gonzalez FJ, Peters JM: Peroxisome proliferator-activated receptor-beta/delta protects against chemically induced liver toxicity in mice. Hepatology 2008, 47(1):225-235.
  • [31]Trauner M, Fickert P, Halilbasic E, Moustafa T: Lessons from the toxic bile concept for the pathogenesis and treatment of cholestatic liver diseases. Wien Med Wochenschr 2008, 158(19–20):542-548.
  • [32]Woolbright BL, Jaeschke H: Novel insight into mechanisms of cholestatic liver injury. World J Gastroenterol 2012, 18(36):4985-4993.
  • [33]Schwabe RF, Brenner DA: Mechanisms of Liver Injury. I. TNF-alpha-induced liver injury: role of IKK, JNK, and ROS pathways. Am J Physiol Gastrointest Liver Physiol 2006, 290(4):G583-G589.
  • [34]Weber LW, Boll M, Stampfl A: Hepatotoxicity and mechanism of action of haloalkanes: carbon tetrachloride as a toxicological model. Crit Rev Toxicol 2003, 33(2):105-136.
  • [35]Plaa GL, Priestly BG: Intrahepatic cholestasis induced by drugs and chemicals. Pharmacol Rev 1976, 28(3):207-273.
  • [36]Goldfarb S, Singer EJ, Popper H: Experimental cholangitis due to alpha-naphthyl-isothiocyanate (ANIT). Am J Pathol 1962, 40:685-698.
  • [37]Fushimi K, Uchida S, Hara Y, Hirata Y, Marumo F, Sasaki S: Cloning and expression of apical membrane water channel of rat kidney collecting tubule. Nature 1993, 361(6412):549-552.
  • [38]Cao Y, Karsten U, Zerban H, Bannasch P: Expression of MUC1, Thomsen-Friedenreich-related antigens, and cytokeratin 19 in human renal cell carcinomas and tubular clear cell lesions. Virchows Arch 2000, 436(2):119-126.
  • [39]Pyle AL, Li B, Maupin AB, Guzman RJ, Crimmins DL, Olson S, Atkinson JB, Young PP: Biomechanical stress induces novel arterial intima-enriched genes: implications for vascular adaptation to stress. Cardiovasc Pathol 2010, 19(2):e13-e20.
  • [40]Aho S, Rothenberger K, Tan EM, Ryoo YW, Cho BH, McLean WH, Uitto J: Human periplakin: genomic organization in a clonally unstable region of chromosome 16p with an abundance of repetitive sequence elements. Genomics 1999, 56(2):160-168.
  • [41]Thomas AM, Hart SN, Kong B, Fang J, Zhong XB, Guo GL: Genome-wide tissue-specific farnesoid X receptor binding in mouse liver and intestine. Hepatology 2010, 51(4):1410-1419.
  • [42]Fujita PA, Rhead B, Zweig AS, Hinrichs AS, Karolchik D, Cline MS, Goldman M, Barber GP, Clawson H, Coelho A: The UCSC Genome Browser database: update 2011. Nucleic Acids Res 2011, 39(Database issue):D876-D882.
  文献评价指标  
  下载次数:30次 浏览次数:37次