442
Views
48
CrossRef citations to date
0
Altmetric
Review Article

Modulation of UDP-glucuronosyltransferase activity by protein-protein association

, &
Pages 145-158 | Received 30 Jun 2009, Published online: 12 Oct 2009

References

  • Alston, K., Robinson, R. C., Park, S. S., Gelboin, H. V., Friedman, F. K. (1991). Interactions among cytochromes P450 in the endoplasmic reticulum. Detection of chemically cross-linked complexes with monoclonal antibodies. J Biol Chem 266:735–739.
  • Baulieu, M., Lévesque E., Hum, D. W., Bélanger, A. (1998). Isolation and characterization of a human orphan UDP-glucuronosyltransferase, UGT2B11. Biochem Biophys Res Commun 248:44–50.
  • Bock, K. W., Josting, D., Lilienblum, W., Pfeil, H. (1979). Purification of rat-liver microsomal UDP-glucuronyltransferase. Separation of two enzyme forms inducible by 3-methylcholanthrene or phenobarbital. Eur J Biochem 98:19–26.
  • Bock, K. W., Köhle, C. (2009). Topological aspects of oligomeric UDP-glucuronosyltransferases in endoplasmic reticulum membranes: advances and open questions. Biochem Pharmacol 77:1458–1465.
  • Bock, K. W., Lilienblum, W. (1979). Activation and induction of rat-liver microsomal UDP-glucuronyltransferase with 6-hydroxybenzo(a)pyrene and N-hydroxy-2-naphthylamine as substrates. Biochem Pharmacol 28:695–700.
  • Coffman, B. L., Rios, G. R., King, C. D., Tephly, T. R. (1997). Human UGT2B7 catalyzes morphine glucuronidation. Drug Metab Dispos 25:1–4.
  • Cosme, J., Johnson, E. F. (2000). Engineering microsomal cytochrome P450 2C5 to be a soluble, monomeric enzyme. Mutations that alter aggregation, phospholipid dependence of catalysis, and membrane binding. J Biol Chem 275:2545–2553.
  • Court, M. H., Krishnaswamy, S., Hao, Q., Duan, S. X., Patten, C. J., Von Moltke, L. L., et al. (2003). Evaluation of 3’-azido-3’-deoxythymidine, morphine, and codeine as probe substrates for UDP-glucuronosyltransferase 2B7 (UGT2B7) in human liver microsomes: specificity and influence of the UGT2B7*2 polymorphism. Drug Metab Dispos 31:1125–1133.
  • Davydov, D. R., Knyushko, T. V., Hoa, G. H. (1992). High-pressure–induced inactivation of ferrous cytochrome P450 LM2 (IIB4) CO complex: evidence for the presence of two conformers in the oligomer. Biochem Biophys Res Commun 188:216–221.
  • Edwards, R. J., Murray, B. P., Singleton, A. M., Boobis, A. R. (1991). Orientation of cytochrome P450 in the endoplasmic reticulum. Biochemistry 30:71–76.
  • Evans, W. E., Relling, M. V. (1999). Pharmacogenomics: translating functional genomics into rational therapeutics, Science 286:487–491.
  • Falany, C. N., Green, M. D., Tephly, T. R. (1987). The enzymatic mechanism of glucuronidation catalyzed by two purified rat-liver steroid UDP-glucuronosyltransferases. J Biol Chem 262:1218–1222.
  • Falany, C. N., Tephly, T. R. (1983). Separation, purification, and characterization of three isoenzymes of UDP-glucuronyltransferase from rat-liver microsomes. Arch Biochem Biophys 227:248–258.
  • Finel, M., Kurkela, M. (2008). The UDP-glucuronosyltransferases as oligomeric enzymes. Curr Drug Metab 9:70–76.
  • Fremont, J. J., Wang, R. W., King, C. D. (2005). Coimmunoprecipitation of UDP-glucuronosyltransferase (UGT) isoforms and cytochrome P450 3A4. Mol Pharmacol 67:260–262.
  • Fretland, A. J., Omiecinski, C. J. (2000). Epoxide hydrolases: biochemistry and molecular biology. Chem Biol Interact 129:41–59.
  • Fujiwara, R., Nakajima, M., Oda, S., Yamanaka, H., Ikushiro, S. I., Sakaki, T., et al. (2009). Interactions between human UDP-glucuronosyltransferase (UGT) 2B7 and UGT1A enzymes. J Pharm Sci May 27. [Epub ahead of print]
  • Fujiwara, R., Nakajima, M., Yamanaka, H., Katoh, M., Yokoi, T. (2007a). Interactions between human UGT1A1, UGT1A4, and UGT1A6 affect their enzymatic activities. Drug Metab Dispos 35:1781–1787.
  • Fujiwara, R., Nakajima, M., Yamanaka, H., Nakamura, A., Katoh, M., Ikushiro, S., et al. (2007b). Effects of coexpression of UGT1A9 on enzymatic activities of human UGT1A isoforms. Drug Metab Dispos 35:747–757.
  • Ghosh, S. S., Sappal, B. S., Kalpana, G. V., Lee, S. W., Roy Chowdhury J., Roy Chowdhury, N. (2001). Homodimerization of human bilirubin-uridine-diphosphoglucuronate glucuronosyltransferase-1 (UGT1A1) and its functional implications. J Biol Chem 276:42108–42115.
  • Girard, H., Lévesque, E., Bellemare, J., Journault, K., Caillier, B., Guillemette, C. (2007). Genetic diversity at the UGT1 locus is amplified by a novel 3’ alternative splicing mechanism leading to nine additional UGT1A proteins that act as regulators of glucuronidation activity. Pharmacogenet Genom 17:1077–1089.
  • Gotoh, O. (1992). Substrate recognition sites in cytochrome P450 family 2 (CYP2) proteins inferred from comparative analyses of amino acid and coding nucleotide sequences. J Biol Chem 267:83–90.
  • Green, M. D., Bélanger, G., Hum, D. W., Bélanger, A., Tephly, T. R. (1997). Glucuronidation of opioids, carboxylic acid-containing drugs, and hydroxylated xenobiotics catalyzed by expressed monkey UDP-glucuronosyltransferase 2B9 protein. Drug Metab Dispos 25:1389–1394.
  • Grove, A. D., Kessler, F. K., Metz, R. P., Ritter, J. K. (1997). Identification of a rat oltipraz-inducible UDP-glucuronosyltransferase (UGT1A7) with activity towards benzo(a)pyrene-7,8-dihydrodiol. J Biol Chem 272:1621–1627.
  • Gschaidmeier, H., Bock, K. W. (1994). Radiation inactivation analysis of microsomal UDP-glucuronosyltransferases catalysing mono- and diglucuronide formation of 3,6-dihydroxybenzo(a)pyrene and 3,6-dihydroxychrysene. Biochem Pharmacol 48:1545–1549.
  • Guillemette, C., Lévesque, E., Bellemare, J., Harvey, M., Ménard, V., Girard, H. (2010). UDP-glucuronosyltransferase (UGT) enzyme diversity: beyond gene duplication. Drug Metab Rev. In press.
  • Hochman, Y., Kelley, M., Zakim, D. (1983). Modulation of the number of ligand binding sites of UDP-glucuronyltransferase by the gel to liquid-crystal phase transition of phosphatidylcholines. J Biol Chem 258:6509–6516.
  • Ikushiro, S., Emi, Y., Iyanagi, T. (1997). Protein-protein interactions between UDP-glucuronosyltransferase isozymes in rat hepatic microsomes. Biochemistry 36:7154–7161.
  • Ikushiro, S., Kominami, S., Takemori, S. (1992). Adrenal P-450scc modulates activity of P-45011 beta in liposomal and mitochondrial membranes. Implication of P-450scc in zone specificity of aldosterone biosynthesis in bovine adrenal. J Biol Chem 267:1464–1469.
  • Ikushiro, S., Sahara, M., Emi. Y., Yabusaki, Y., Iyanagi, T. (2004). Functional coexpression of xenobiotic metabolizing enzymes, rat cytochrome P450 1A1 and UDP-glucuronosyltransferase 1A6, in yeast microsomes. Biochim Biophys Acta 1672:86–92.
  • Ishii, Y., Iwanaga, M., Nishimura, Y., Takeda, S., Ikushiro, S., Nagata, K., et al. (2007). Protein-protein interactions between rat hepatic cytochromes P450 (P450s) and UDP-glucuronosyltransferases (UGTs): evidence for the functionally active UGT in P450-UGT complex. Drug Metab Pharmacokinet 22:367–376.
  • Ishii, Y., Miyoshi, A., Maji, D., Yamada, H., Oguri, K. (2004). Simultaneous expression of guinea pig UDP-glucuronosyltransferase 2B21 and 2B22 in COS-7 cells enhances UDP-glucuronosyltransferase 2B21-catalyzed chloramphenicol glucuronidation. Drug Metab Dispos 32:1057–1060.
  • Ishii, Y., Miyoshi, A., Watanabe, R., Tsuruda, K., Tsuda, M., Yamaguchi-Nagamatsu, Y., et al. (2001). Simultaneous expression of guinea pig UDP-glucuronosyltransferase 2B21 and 2B22 in COS-7 cells enhances UDP-glucuronosyltransferase 2B21-catalyzed morphine-6-glucuronide formation. Mol Pharmacol 60:1040–1048.
  • Ishii, Y., Oguri, K., Yoshimura, H. (1993). Purification and characterization of a morphine UDP-glucuronyltransferase isoform from untreated rat liver. Biol Pharm Bull 16:754–758.
  • Ishii, Y., Takeda, S., Yamada, H., Oguri, K. (2005). Functional protein-protein interaction of drug-metabolizing enzymes. Front Biosci 10:887–895.
  • Iyanagi, T., Haniu, M., Sogawa, K., Fujii-Kuriyama, Y., Watanabe, S., Shively, J. E., Anan, K.F. (1986) Cloning and characterization of cDNA encoding 3-methylcholanthrene inducible rat mRNA for UDP-glucuronosyltransferase. J Biol Chem 261:15607–15614.
  • Iyanagi, T. (2007). Molecular mechanism of phase I and phase II drug-metabolizing enzymes: implications for detoxication. Int Rev Cytol 260:35–112.
  • Jackson, M. R., Nilsson, T., Peterson, P. A. (1990). Identification of a consensus motif for retention of transmembrane proteins in the endoplasmic reticulum. EMBO J 9:3153–3162.
  • Kim, K. H., Ahn, T., Yun, C. H. (2003). Membrane properties induced by anionic phospholipids and phosphatidylethanolamine are critical for the membrane-binding and catalytic activity of human cytochrome P450 3A4. Biochemistry 42:15377–15387.
  • Koiwai, O., Aono, S., Adachi, Y., Kamisako, T., Yasui, Y., Nishizawa, M., et al. (1996). Crigler-Najjar syndrome type II is inherited both as a dominant and as a recessive trait. Hum Mol Genet 5:645–647.
  • Kominami, S., Harada, D., Takemori, S. (1994). Regulation mechanism of the catalytic activity of bovine adrenal cytochrome P-450 11 beta. Biochim Biophys Acta 1192:234–240.
  • Kuo, C. K., Hanioka, N., Hoshikawa, Y., Oguri, K., Yoshimura, H. (1991). Species difference of site-selective glucuronidation of morphine. J Pharmacobio-Dyn 14:187–193.
  • Kurkela, M., Garcia-Horsman, J. A., Luukkanen, L., Morsky, S., Taskinen, J., Baumann, M., et al. (2003). Expression and characterization of recombinant human UDP-glucuronosyltransferases (UGTs). UGT1A9 is more resistant to detergent inhibition than other UGTs and was purified as an active dimeric enzyme. J Biol Chem 278:3536–3544.
  • Kurkela, M., Hirvonen, J., Kostiainen, R., Finel, M. (2004b). The interactions between the N-terminal and C-terminal domains of the human UDP-glucuronosyltransferases are partly isoform-specific, and may involve both monomers. Biochem Pharmacol 68:2443–2450.
  • Kurkela, M., Morsky, S., Hirvonen, J., Kostiainen, R., Finel, M. (2004a). An active and water-soluble truncation mutant of the human UDP-glucuronosyltransferase 1A9. Mol Pharmacol 65:826–831.
  • Kurkela, M., Patana, A. S., Mackenzie, P. I., Court, M. H., Tate, C. G., Hirvonen, J., et al. (2007). Interactions with other human UDP-glucuronosyltransferases attenuate the consequences of the Y485D mutation on the activity and substrate affinity of UGT1A6. Pharmacogenet Genom 17:115–126.
  • Lewis, D. F., Modi, S., Dickins, M. (2002). Structure-activity relationship for human cytochrome P450 substrates and inhibitors. Drug Metab Rev 34:69–82.
  • Lévesque, E., Girard, H., Journaultm K., Lépinem J., Guillemettem C. (2007). Regulation of the UGT1A1 bilirubin-conjugating pathway: role of a new splicing event at the UGT1A locus. Hepatology 45:128–138.
  • Mackenzie, P. I. (1986a). Rat-liver UDP-glucuronosyltransferase. Sequence and expression of a cDNA encoding a phenobarbital-inducible form. J Biol Chem 261:6119–6125.
  • Mackenzie, P. I. (1986b). Rat-liver UDP-glucuronosyltransferase. cDNA sequence and expression of a form glucuronidating 3-hydroxyandrogens. J Biol Chem 261:14112–14117.
  • Mackenzie, P. I. (1987). Rat-liver UDP-glucuronosyltransferase. Identification of cDNAs encoding two enzymes which glucuronidate testosterone, dihydrotestosterone, and beta-estradiol. J Biol Chem 262:9744–9749.
  • Mackenzie, P. I. (1990). The cDNA sequence and expression of a variant 17beta-hydroxysteroid UDP-glucuronosyltransferase. J Biol Chem 265:8699–8703.
  • Mackenzie, P. I., Bock, K. W., Burchell, B., Guillemette, C., Ikushiro, S., Iyanagi, T., et al. (2005). Nomenclature update for the mammalian UDP-glycosyltransferase (UGT) gene superfamily. Pharmacogenet Genom 15:677–685.
  • Mackenzie, P. I., Hjelmeland, L. M., Owens, I. S. (1984). Purification and immunochemical characterization of a low-pI form of UDP-glucuronosyltransferase from mouse liver. Arch Biochem Biophys 231: 487–497.
  • Mackenzie, P. I., Owens, I. S., Burchell, B., Bock, K. W., Bairoch, A., Bélanger, A., et al. (1997). The UDP-glycosyltransferase gene superfamily: recommended nomenclature update based on evolutionary divergence. Pharmacogenetics 7:255–269.
  • Mackenzie, P. I., Rodbourn, L., Iyanagi, T. (1993). Glucuronidation of carcinogen metabolites by complementary DNA-expressed uridine 5’-diphosphate glucuronosyltransferases. Cancer Res 53:1529–1533.
  • Matern, H., Matern, S., Gerok, W. (1982). Isolation and characterization of rat-liver microsomal UDP-glucuronosyltransferase activity toward chenodeoxycholic acid and testosterone as a single form of enzyme. J Biol Chem 257:7422–7429.
  • Matsui, M., Nagai, F. (1986). Genetic deficiency of androsterone UDP-glucuronosyltransferase activity in Wistar rats is due to the loss of enzyme protein. Biochem J 234:139–144.
  • Meech, R., Mackenzie, P. I. (1997a). Structure and function of uridine diphosphate glucuronosyltransferases. Clin Exp Pharmacol Physiol 24:907–915.
  • Meech, R., Mackenzie, P. I. (1997b). UDP-glucuronosyltransferase, the role of the amino terminus in dimerization. J Biol Chem 272:26913–26917.
  • Meech, R., Mackenzie, P. I. (1998). Determinants of UDP-glucuronosyltransferase membrane association and residency in the endoplasmic reticulum. Arch Biochem Biophys 356:77–85.
  • Meech, R., Yogalingam, G., Mackenzie, P. I. (1996). Mutational analysis of the carboxy-terminal region of UDP-glucuronosyltransferase 2B1. DNA Cell Biol 15:489–494.
  • Myasoedova, K. N., Tsuprun, V. L. (1993). Cytochrome P450: hexameric structure of the purified LM4 form. FEBS Lett 325:251–254.
  • Nagar, S., Zalatoris, J. J., Blanchard, R. L. (2004). Human UGT1A6 pharmacogenetics: identification of a novel SNP, characterization of allele frequencies, and functional analysis of recombinant allozymes in human liver tissue and in cultured cells. Pharmacogenetics 14:487–499.
  • Nagata, K., Ozawa, S., Miyata, M., Shimada, M., Yamazoe, Y., Kato, R. (1994). Primary structure and molecular basis of polymorphic appearance of an acetyltransferase (AT-II)* in hamsters. Pharmacogenetics 4:91–100.
  • Nakajima, M., Yamanaka, H., Fujiwara, R., Katoh, M., Yokoi, T. (2007). Stereoselective glucuronidation of 5-(4’-hydroxyphenyl)-5-phenylhydantoin by human UDP-glucuronosyltransferase (UGT) 1A1, UGT1A9, and UGT2B15: effects of UGT-UGT interactions. Drug Metab Dispos 35:1679–1686.
  • Oguri, K., Kurogi, A., Yamabe, K., Tanaka, M., Yoshisue, K., Ishii, Y., et al. (1996). Purification of a phenobarbital-inducible UDP-glucuronosyltransferase isoform from dog liver which catalyzes morphine and testosterone glucuronidation. Arch Biochem Biophys 325:159–166.
  • Oguri, K., Yamada, H., Yoshimura, H. (1994). Regiochemistry of cytochrome P450 isozymes. Annu Rev Pharmacol Toxicol 34:251–279.
  • Okamura, K., Ishii, Y., Ikushiro, S., Mackenzie, P. I., Yamada, H. (2006). Fatty acyl-CoA as an endogenous activator of UDP-glucuronosyltransferases. Biochem Biophys Res Commun 345:1649–1656.
  • Operaña, T. N., Tukey, R. H. (2007). Oligomerization of the UDP-glucuronosyltransferase 1A proteins: homo- and heterodimerization analysis by fluorescence resonance energy transfer and coimmunoprecipitation. J Biol Chem 282:4821–4829.
  • Ouzzine, M., Magdalou, J., Burchell, B., Fournel-Gigleux, S. (1999). An internal signal sequence mediates the targeting and retention of the human UDP-glucuronosyltransferase 1A6 to the endoplasmic reticulum. J Biol Chem 274:31401–31409.
  • Peters, W. H., Jansen, P. L., Nauta, H. (1984). The molecular weights of UDP-glucuronyltransferase determined with radiation-inactivation analysis. A molecular model of bilirubin UDP-glucuronyltransferase. J Biol Chem 259:11701–11705.
  • Puig, J. F., Tephly, T. R. (1986). Isolation and purification of rat-liver morphine UDP-glucuronosyltransferase. Mol Pharmacol 30:558–565.
  • Radominska-Pandya, A., Bratton, S., Little, J. M. (2005). A historical overview of the heterologous expression of mammalian UDP-glucuronosyltransferase isoforms over the past twenty years. Curr Drug Metab 6:141–160.
  • Satoh, T., Hosokawa, M. (1998). The mammalian carboxylesterases: from molecules to functions. Annu Rev Pharmacol Toxicol 38:257–288.
  • Seidegård, J., DePierre, J. W. (1983). Microsomal epoxide hydrolase. Properties, regulation, and function. Biochim Biophys Acta 695:251–270.
  • Shepherd, S. R., Baird, S. J., Hallinan, T., Burchell, B. (1989). An investigation of the transverse topology of bilirubin UDP-glucuronosyltransferase in rat hepatic endoplasmic reticulum. Biochem J 259:617–620.
  • Shimomura, K., Kamata, O., Ueki, S., Ida, S., Oguri, K., Yoshimura, H., et al. (1971). Analgesic effect of morphine glucuronides. Tohoku J Exp Med 105:45–52.
  • Soars, M. G., Barbara, B. J., Ring, J., Wrighton, S. A. (2003). The effect of incubation conditions on the enzyme kinetics of UDP-glucuronosyltransferases. Drug Metab Dispos 31:762–767.
  • Subramanian, M., Low, M., Locuson, C. W., Tracy, T. S. (2009). CYP2D6-CYP2C9 protein-protein interactions and isoform-selective effects on substrate binding and catalysis. Drug Metab Dispos 37: 1682–1689.
  • Takeda, S., Ishii, Y., Iwanaga, M., Mackenzie, P. I., Nagata, K., Yamazoe, Y., et al. (2005a). Modulation of UDP-glucuronosyltransferase function by cytochrome P450: evidence for the alteration of UGT2B7-catalyzed glucuronidation of morphine by CYP3A4. Mol Pharmacol 67:665–672.
  • Takeda, S., Ishii, Y., Mackenzie, P. I., Nagata, K., Yamazoe, Y., Oguri, K., et al. (2005b). Modulation of UDP-glucuronosyltransferase 2B7 function by cytochrome P450s in vitro: differential effects of CYP1A2, CYP2C9, and CYP3A4. Biol Pharm Bull 28:2026–2027.
  • Takeda, S., Ishii, Y., Iwanaga, M., Nurrochmad, A., Ito, Y., Mackenzie, P. I., et al. (2009). Interaction of cytochrome P450 3A4 and UDP-glucuronosyltransferase 2B7: evidence for protein-protein association and possible involvement of CYP3A4 J-helix in the interaction. Mol Pharmacol 75:956–964.
  • Tan, Y., Patten, C. J., Smith, T., Yang, C. S. (1997). Competitive interactions between cytochromes P450 2A6 and 2E1 for NADPH-cytochrome P450 oxidoreductase in the microsomal membranes produced by a baculovirus expression system. Arch Biochem Biophys 342:82–91.
  • Taura, K., Naito, E., Ishii Y., Mori, M. A., Oguri, K., Yamada, H. (2004). Cytochrome P450 1A1 (CYP1A1) inhibitor alpha-naphthoflavone interferes with UDP-glucuronosyltransferase (UGT) activity in intact, but not in permeabilized hepatic microsomes, from 3-methylcholanthrene-treated rats: possible involvement of UGT-P450 interactions. Biol Pharm Bull 27:56–60.
  • Taura, K., Yamada, H., Hagino, Y., Ishii, Y., Mori, M., Oguri, K. (2000). Interaction between cytochrome P450 and other drug-metabolizing enzymes: evidence for an association of CYP1A1 with microsomal epoxide hydrolase and UDP-glucuronosyltransferase. Biochem Biophys Res Commun 273:1048–1052.
  • Vanstapel, F., Blanckaert, N. (1988). Topology and regulation of bilirubin UDP-glucuronyltransferase in sealed native microsomes from rat liver. Arch Biochem Biophys 263:216–225.
  • Vatsis, K. P., Weber, W. W., Bell, D. A., Dupret, J. M., Evans, D.A., Grant, D. M., et al. (1995). Nomenclature for N-acetyltransferases. Pharmacogenetics 5:1–17.
  • Whalen, R., Boyer, T. D. (1998). Human glutathione S-transferases. Semin Liver Dis 18:345–358.
  • Williams, P. A., Cosme, J., Sridhar, V., Johnson, E. F., McRee, D. E. (2000). Mammalian microsomal cytochrome P450 monooxygenase: structural adaptations for membrane binding and functional diversity. Mol Cell 5:121–131.
  • Williams, P. A., Cosme, J., Vinkovic, D. M., Ward, A., Angove, H. C., Day, P. J., et al. (2004). Crystal structures of human cytochrome P450 3A4 bound to metyrapone and progesterone. Science 305:683–686.
  • Yamazaki, H., Gillam, E. M. J., Dong, M.-S., Johnson, W. W., Guengerich, F. P., Shimada, T. (1997). Reconstitution of recombinant cytochrome P450 2C10(2C9) and comparison with cytochrome P450 3A4 and other forms: effects of cytochrome P450–P450 and cytochrome P450-b5 interactions. Arch Biochem Biophys 342:329–337.
  • Yano, J. K., Wester, M. R., Schoch, G. A., Griffin, K. J., Stout, C. D., Johnson, E. F. (2004). The structure of human microsomal cytochrome P450 3A4 determined by X-ray crystallography to 2.05-A resolution. J Biol Chem 279:38091–38094.
  • Yoshinari, K., Petrotchenko, E. V., Pedersen, L. C., Negishi, M. (2001). Crystal structure-based studies of cytosolic sulfotransferase. J Biochem Mol Toxicol 15:67–75.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.