Elsevier

Chemico-Biological Interactions

Volume 162, Issue 3, 25 September 2006, Pages 195-211
Chemico-Biological Interactions

Mini review
Structure, function and regulation of carboxylesterases

https://doi.org/10.1016/j.cbi.2006.07.001Get rights and content

Abstract

This review covers current developments in molecular-based studies of the structure and function of carboxylesterases. To allay the confusion of the classic classification of carboxylesterase isozymes, we have proposed a novel nomenclature and classification of mammalian carboxylesterases on the basis of molecular properties. In addition, mechanisms of regulation of gene expression of carboxylesterases by xenobiotics and involvement of carboxylesterase in drug metabolism and enzyme induction are also described.

Introduction

The present review highlights the importance of structure in delineating overall function, substrate specificity, regulation and localization of mammalian carboxylesterases (CarbEs). They are α,β-hydrolase-fold protein and comprise a multigene superfamily [1]. Structural considerations emerge from the genes encoding the family. Diversity in the structure and ultimately function and cellular localization of the gene product is achieved through gene doubling, alternative mRNA processing and post-translational modification.

The expression of CarbEs is ubiquitous with high levels in various tissues. Some of the isozymes are destined for export into the plasma. Others are associated with cell membrane such as endoplasmic reticulum (ER) with its catalytic function directed extracellularly [2], [3], [4], [5], [6], [7], [8], [9], [10], [11], [12].

Expression profiles of gene encoding CarbEs are highly regulated during development by nutritional status, hormonal factors and xenobiotics. Although the consequences of regulation of CarbEs by drugs and chemicals have been intensively studied [11], [13], [16], [17], relatively little is known about the mechanisms by which esterases are regulated by physiological factors. Recent developments have included more detailed biochemical characterization of mammalian CarbE enzymes and genes, leading to a better understanding of the biochemical significance and physiological role of CarbEs.

This review addresses the significant differences of molecular structure and function of recently identified CarbEs, and proposes a novel nomenclature for mammalian CarbE isozymes that is based on the nucleotide sequences of the genes encoding the individual CarbE isozymes.

Section snippets

Novel classification and nomenclature of mammalian CarbEs

According to the classification of esterases by Aldridge [18], the serine super family of esterases, i.e., acetylcholinesterase(AChE), butyrylcholinesterase and CarbE, fall into the B-esterase group. CarbE iszymes were initially classified by their substrate specificity and pI. However, this classification is ambiguous in overlapping substrate specificities. A single esterolytic reaction is frequently mediated by several kinds of enzymes. Recent studies on esterases, as with other enzymes

Structure–activity relationship of mammalian CarbEs

The CarbEs comprise a multigene superfamily, the gene products of which are localized in the endoplasmic reticulum (ER) of many tissues [2], [3], [4], [5], [6], [7], [8], [9], [10], [11], [23], [24]. These enzymes efficiently catalyze the hydrolysis of a variety of drugs or prodrugs containing ester- and amide-bonds to the respective free acids and alcohol. Since ester derivatives of therapeutic agents have been used as prodrugs, CarbEs are major determinants of the pharmacokinetic behavior of

Gene structure and regulation of CarbE isozymes

Both the murine [11] and human [4], [38], [46]CES1 genes span about 30 kb and contain 14 small exons. Recently, sequencing of the mouse and human genomes were completed, enabling detailed sequence comparisons. The previously published sequences of the individual exons, splice junctions, size of the introns and restriction sites within the murine and human CarbE genes are consistent with their respective genes sequenced by the mouse and human genome projects. Therefore, the organization of the

Drug metabolism

Drug-metabolizing enzymes that are present predominantly in the liver are involved in biotransformation of both endogenous and exogenous compounds to polar products to facilitate their elimination. These reactions are categorized into phases I and II reactions. CarbEs show ubiquitous tissue expression profiles with the highest levels of CarbE activity present in liver microsomes in many mammals [4], [5], [11]. CarbEs are categorized as phase-I drug-metabolizing enzymes that can hydrolyze a

Enzyme induction of CarbE

Much interest has been shown by both clinicians and reserchers in the induction of expression of drug-metabolizing enzymes by chemicals, including medical agents, since it is one of the main reasons for drug-drug interaction causing adverse effects and for the reduction in pharmacological potencies of drugs. As for CarbEs, it has been shown that rodent CarbE isozyme(s) was induced by phenobarbital [21], aminopyrine [32], peroxisome proliferators (clofibrate, di(2-ethylhexyl)phthalate, and

A novel biomarker of organophosphate insecticide exposure

Egasyn, an isozyme of CarbE, is an accessary protein of β-glucuronidase in the liver microsomes [4], [36], [84]. Egasyn-β-glucuronidase complexes are located at the luminal sites of liver microsomal endoplasmic reticulum membrane [85]. When the organophosphorus insecticides (OP) are incorporated into the liver microsomes, the OP is tightly bound to egasyn, and subsequently, β-glucuronidase is dissociated and released into the blood. Consequently, the increase of plasma BG activity is good

Conclusion

Multiple CarbEs play an important role in the hydrolytic biotransformation of a vast number of structurally diverse drugs. These enzymes are a major determinant of the pharmacokinetic behavior of most therapeutic agents containing an ester or amide bond. There are several factors that influence CarbE activity, either directly or at the level of enzyme regulation. In the clinical field, drug elimination is decreased and the incidence of drug–drug interactions increases when two or more drugs

Acknowledgements

The authors wish to acknowledge Professor Palmer Taylor, Univresity of California at San Diego, La Jolla, and Professor William Bosron, Indiana University, Indianapolis, Indiana for providing us with valuable suggestions. We also acknowledge the colleagues at the HAB Research laboratories for preparation of this manuscript.

References (89)

  • C.A. Fox et al.

    Altered expression of TFF-1 and CES-2 in Barrett's Esophagus and associated adenocarcinomas

    Neoplasia

    (2005)
  • H.R. Pelham

    The retention signal for soluble proteins of the endoplasmic reticulum

    Trends Biochem. Sci.

    (1990)
  • G. Korza et al.

    Complete covalent structure of 60-kDa esterase isolated from 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced rabbit liver

    J. Biol. Chem.

    (1988)
  • J. Ozols

    Isolation, properties, and the complete amino acid sequence of a second form of 60-kDa glycoprotein esterase

    J. Biol. Chem.

    (1989)
  • M. Robbi et al.

    Cloning and sequencing of rat liver carboxylesterase ES-3 (egasyn)

    Biochem. Biophys. Res. Commun.

    (1994)
  • S. Medda et al.

    An accessory protein identical to mouse egasyn is complexed with rat microsomal beta-glucuronidase and is identical to rat esterase-3

    J. Biol. Chem.

    (1987)
  • M. Ovnic et al.

    Characterization and functional expression of a cDNA encoding egasyn (esterase-22): the endoplasmic reticulum-targeting protein of beta-glucuronidase

    Genomics

    (1991)
  • F. Shibata et al.

    Molecular cloning and characterization of a human carboxylesterase gene

    Genomics

    (1993)
  • S. Bencharit et al.

    Crystal structure of human carboxylesterase 1 complexed with the Alzheimer's drug tacrine: from binding promiscuity to selective inhibition

    Chem. Biol.

    (2003)
  • H. Wong et al.

    The lipase gene family

    J. Lipid. Res.

    (2002)
  • T. Langmann et al.

    Structural organization and characterization of the promoter region of a human carboxylesterase gene

    Biochim. Biophys. Acta

    (1997)
  • F. Zschunke et al.

    cDNA cloning and characterization of human monocyte/macrophage serine esterase-1

    Blood

    (1991)
  • T. Furihata et al.

    Hepatocyte nuclear factor-4alpha plays pivotal roles in the regulation of mouse carboxylesterase 2 gene transcription in mouse liver

    Arch. Biochem. Biophys.

    (2006)
  • S.R. Kim et al.

    Twelve novel single nucleotide polymorphisms in the CES2 gene encoding human carboxylesterase 2 (hCE-2)

    Drug Metab. Pharmacokenet.

    (2003)
  • M. Ovnic et al.

    Characterization of a murine cDNA encoding a member of the carboxylesterase multigene family

    Genomics

    (1991)
  • K. Ohtsuka et al.

    Intracellular conversion of irinotecan to its active form, SN-38, by native carboxylesterase in human non-small cell lung cancer

    Lung Cancer

    (2003)
  • S. Marsh et al.

    Pharmacogenomics assessment of carboxylesterases 1 and 2

    Genomics

    (2004)
  • T.Y. Lynch et al.

    Cocaine detoxification by human plasma butyrylcholinesterase

    Toxicol. Appl. Pharmacol.

    (1997)
  • E.V. Pindel et al.

    Purification and cloning of a broad substrate specificity human liver carboxylesterase that catalyzes the hydrolysis of cocaine and heroin

    J. Biol. Chem.

    (1997)
  • M. Inoue et al.

    Species difference and characterization of intestinal esterase on the hydrolizing activity of ester-type drugs

    Jpn. J. Pharmacol.

    (1979)
  • C.D. Fleming et al.

    Structural insights into drug processing by human carboxylesterase 1: tamoxifen, mevastatin, and inhibition by benzyl

    J. Mol. Biol.

    (2005)
  • J.A. Howarth et al.

    Effects on male rats of di-(2-ethylhexyl) phthalate and di-n-hexylphthalate administered alone or in combination

    Toxicol. Lett.

    (2001)
  • K. Aida et al.

    Cloning and nucleotide sequence of a novel, male-predominant carboxylesterase in mouse liver

    Biochim. Biophys. Acta

    (1993)
  • V.W. Dolinsky et al.

    The cloning and expression of a murine triacylglycerol hydrolase cDNA and the structure of its corresponding gene

    Biochim. Biophys. Acta

    (2001)
  • K. Hattori et al.

    Purification and characterization of glucocorticoid-inducible steroid esterase in rat hepatic microsomes: specific induction by glucocorticoids of steroid esterase in rat hepatic microsomes and its release into serum

    Biochem. Pharmacol.

    (1992)
  • T. Satoh et al.

    Toxicological significance in the cleavage of esterase-B-glucuronidase complex in liver microsomes by organophosphorus compounds

    Chem. Biol. Interact.

    (1999)
  • M.K. Ross et al.

    Hydrolyutic metabolism of pyrethroids by human and other mammalian carboxylesterases

    Biochem. Pharmacol.

    (2006)
  • M. Cygler et al.

    Relationship between sequence conservation and three-dimensional structure in a large family of esterases, lipases, and related proteins

    Protein Sci.

    (1993)
  • T. Satoh et al.

    Current progress on esterases: from molecular structure to function

    Drug Metab. Dispos.

    (2002)
  • M. Hosokawa et al.

    Multiplicity and regulation of hepatic microsomal carboxylesterases in rats

    Mol. Pharmacol.

    (1987)
  • M. Hosokawa et al.

    Interindividual variation in carboxylesterase levels in human liver microsomes

    Drug Metab. Dispos.

    (1995)
  • T. Maki et al.

    Changes in carboxylesterase isoenzymes of rat liver microsomes during hepatocarcinogenesis

    Jpn. J. Cancer Res.

    (1991)
  • T. Satoh et al.

    Metabolic activation of CPT-11, 7-ethyl-10-[4-(1-piperidino)-1-piperidinol] carbonyloxycamptothecin, a novel antitumor agent, by carboxylesterase

    Biol. Pharm. Bull.

    (1994)
  • T. Yamada et al.

    Immunohistochemistry with an antibody to human liver carboxylesterase in human brain tissues

    Brain Res.

    (1993)
  • Cited by (0)

    View full text