ArticlePDF Available

NAT1 (N-acetyltransferase 1 (arylamine N-acetyltransferase))

Authors:
  • University Institute of Molecular Pathology Biomarkers. Universidad de Extremadura
Gene Section
Review
Atlas Genet Cytogenet Oncol Haematol. 2010; 14(1)
39
Atlas of Genetics and Cytogenetics
in Oncology and Haematology
OPEN ACCESS JOURNAL AT INIST-CNRS
NAT1 (N-acetyltransferase 1 (arylamine N-
acetyltransferase))
Jhon D Ruiz, José AG Agúndez, Carmen Martínez, Elena García-Martín
Department of Pharmacology, Medical School, University of Extremadura, Badajoz, Spain (JDR),
Department of Biochemistry & Molecular biology & Genetics, School of Biological Sciences, Badajoz,
Spain (JAGA), Department of Biochemistry & Molecular biology & Genetics, School of Biological
Sciences, Badajoz, Spain (CM, EGM)
Published in Atlas Database: February 2009
Online updated version: http://AtlasGeneticsOncology.org/Genes/NAT1ID41497ch8p22.html
DOI: 10.4267/2042/44659
This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence.
© 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology
Identity
Other names: AAC1; MNAT
HGNC (Hugo): NAT1
Location: 8p22
Picture adapted from an original prepared by Genetics Home
Reference; February 2009.
DNA/RNA
Note
In humans NAT1 is located in the NAT cluster that
comprises 230 kb and includes two functional genes,
NAT1 and NAT2. In other species the number of NAT
genes range from 0 to 4.
Transcription
The human NAT1 gene has nine exons. The coding
region is located in exon 9 and spans 870 bp. Diverse
NAT1 transcripts have been reported and two
promoters exist. The first promoter, designated as P1 is
located in the 5' flanking region of exon 1 and controls
two major (a1 and a2) transcripts. The second promoter
is located upstream of exon 4 and give rise to at least
five (b1 to b5) different transcripts. The different
transcripts appear to have different translational
efficiencies, although the biological significance of this
is unknown (revised in Butcher et al., 2007).
Pseudogene
In humans the NAT locus has a pseudogene designated
as NATP.
Protein
Note
NAT enzymes have been identified in several
vertebrate and microorganism species, whereas NAT
deficiency in the domestic and wild dog is due to
complete absence of NAT genes.
Description
The amino-terminal domain (residues 1-83) consists of
five helices and one short beta-strand. The second
domain comprises residues 85-192 and consists of nine
beta-strands and two short helices. The third domain
has a final helix which precedes the carboxy terminal
region.
Expression
NAT1 activity is expressed in liver and in many
extrahepatic tissues. The transcripts originated from the
first promoter, NATa, are expressed in kidney, liver,
lung and trachea. However the most common
transcripts are those designated as type b in the
NAT1 (N-acetyltransferase 1 (arylamine N-acetyltransferase)) Ruiz JD, et al.
Atlas Genet Cytogenet Oncol Haematol. 2010; 14(1)
40
Structure of the human NAT1 gene and common NAT1 transcripts.
Figure above and have been detected in all tissues
examinated.
Localisation
Arylamine N-acetyltransferases are cytosolic enzymes.
Function
NAT1 is a phase II enzyme that participates in the
metabolism of numerous primary arylamines and
hydrazine drugs and carcinogens. In addition to their
N-acetylation catalytic activity, NAT enzymes have
also O-acetylation activity towards N-
hydroxyarylamines.
Homology
NAT1 and NAT2 share 87% nucleotide homology in
the coding region, whereas NAT1 and NAT2 proteins
share 81% amino-acid sequence identity.
Mutations
Note
In humans NAT1 is highly polymorphic. Several
polymorphisms, most of which are single nucleotide
polymorphisms and at least 26 different haplotypes
have been described. The Figure below shows the
positions of NAT1 polymorphisms, taking as a
reference the start site in the open
reading frame (ORF) in exon 9. Nonsynonymous
polymorphisms are labeled in red font. The association
of different haplotypes with phenotypes is summarized
in the following link:
http://louisville.edu/medschool/pharmacology/Human.
NAT1.pdf.
Implicated in
Lung cancer
Note
Two independent studies have observed a significant
association of the NAT1 polymorphism with lung
cancer risk (Bouchardy et al., 1998; Wikman et al.,
2001). However these studies should be interpreted
cautiously because these do not agree on the NAT1 risk
genotype. Another study identified an increased risk
among carriers of NAT1 plus NAT2 slow genotypes
(Gemignani et al., 2007). In a meta-analysis carried out
with smokers that suffered from non small-cell lung
cancer a relevant association of the NAT1 rapid
acetylation genotype was identified (Zienolddiny et al.,
2008). Although negative associations have been
reported (Perera et al., 2006), NAT1 is emerging as the
NAT gene most likely related to lung cancer (McKay et
al, 2008).
NAT1 (N-acetyltransferase 1 (arylamine N-acetyltransferase)) Ruiz JD, et al.
Atlas Genet Cytogenet Oncol Haematol. 2010; 14(1)
41
Head and neck cancer
Note
Since chemical compounds present in tobacco are
inactivated by phase II enzymes, it has been proposed
that head and neck cancer risk could be modified by
NAT genotypes. Head and neck cancers are strongly
associated with smoking, and a few studies have
explored the role of NAT1 polymorphisms in the risk
of developing head and neck cancer in smokers.
However overall findings are inconsistent and
associations if present are weak, and indicate either a
decreased risk in carriers of the variant NAT1*10
(McKay et al., 2008), an increased risk (Katoh et al.,
1998) or a lack of association (Fronhoffs et al., 2001;
Henning et al., 1999; Agúndez, 2008).
Breast cancer
Note
The NAT1*10 variant allele was associated to
increased breast cancer risk among women who
consumed well-done meat, although the statistical
significance of this finding is low (Krajinovic et al.,
2001). Several studies, however, indicate that no major
association of NAT polymorphisms and breast cancer
risk exists (Agúndez, 2008).
Colorectal cancer
Note
A biologically plausible mechanistic hypothesis
suggest that rapid NAT1 and/or NAT2 acetylators
should more activate heterocyclic amine carcinogens
within the colon to their ultimate carcinogenic forms,
thereby predisposing them to colorectal cancer.
However sufficient evidence is available to rule out a
relevant association of NAT genotypes alone with
colorectal cancer risk. This evidence is based in nearly
thirty studies failed to detect a statistically significant
association for NAT1 genotypes both with colorectal
cancer or adenomas. In addition meta-analyses (Chen et
al., 2005; Ye et al., 2002; Houlston et al., 2001)
consistently confirm a lack of a relevant association of
NAT1 rapid acetylator genotypes and colorectal cancer
risk (revised in Agúndez, 2008).
Bladder cancer
Note
No significant association of the NAT1 genotype with
bladder cancer risk has been observed in a recent meta-
analysis, although the authors found a joint effect of
NAT1 rapid genotypes, slow NAT2 genotypes and
smoking as factors increasing cancer risk (Sanderson et
al., 2007). Overall findings are negative (Okkels et al.,
1997), although a significant risk has been described in
smokers (Taylor et al., 1998; Hsieh et al., 1999;
Cascorbi et al., 2001) and a nearly significant
association was observed in individuals exposed to
benzidine (Carreon et al., 2006).
References
Blum M, Grant DM, Demierre A, Meyer UA. N-acetylation
pharmacogenetics: a gene deletion causes absence of
arylamine N-acetyltransferase in liver of slow acetylator
rabbits. Proc Natl Acad Sci U S A. 1989 Dec;86(23):9554-7
Blum M, Grant DM, McBride W, Heim M, Meyer UA. Human
arylamine N-acetyltransferase genes: isolation, chromosomal
localization, and functional expression. DNA Cell Biol. 1990
Apr;9(3):193-203
Grant DM, Blum M, Beer M, Meyer UA. Monomorphic and
polymorphic human arylamine N-acetyltransferases: a
comparison of liver isozymes and expressed products of two
cloned genes. Mol Pharmacol. 1991 Feb;39(2):184-91
Minchin RF. Acetylation of p-aminobenzoylglutamate, a folic
acid catabolite, by recombinant human arylamine N-
acetyltransferase and U937 cells. Biochem J. 1995 Apr 1;307 (
Pt 1):1-3
Ward A, Summers MJ, Sim E. Purification of recombinant
human N-acetyltransferase type 1 (NAT1) expressed in E. coli
and characterization of its potential role in folate metabolism.
Biochem Pharmacol. 1995 Jun 16;49(12):1759-67
Okkels H, Sigsgaard T, Wolf H, Autrup H. Arylamine N-
acetyltransferase 1 (NAT1) and 2 (NAT2) polymorphisms in
susceptibility to bladder cancer: the influence of smoking.
Cancer Epidemiol Biomarkers Prev. 1997 Apr;6(4):225-31
Trepanier LA, Ray K, Winand NJ, Spielberg SP, Cribb AE.
Cytosolic arylamine N-acetyltransferase (NAT) deficiency in
the dog and other canids due to an absence of NAT genes.
Biochem Pharmacol. 1997 Jul 1;54(1):73-80
Bouchardy C, Mitrunen K, Wikman H, Husgafvel-Pursiainen K,
Dayer P, Benhamou S, Hirvonen A. N-acetyltransferase NAT1
and NAT2 genotypes and lung cancer risk. Pharmacogenetics.
1998 Aug;8(4):291-8
Hughes NC, Janezic SA, McQueen KL, Jewett MA, Castranio
T, Bell DA, Grant DM. Identification and characterization of
variant alleles of human acetyltransferase NAT1 with defective
function using p-aminosalicylate as an in-vivo and in-vitro
probe. Pharmacogenetics. 1998 Feb;8(1):55-66
Kashuba AD, Bertino JS Jr, Kearns GL, Leeder JS, James
AW, Gotschall R, Nafziger AN. Quantitation of three-month
intraindividual variability and influence of sex and menstrual
cycle phase on CYP1A2, N-acetyltransferase-2, and xanthine
oxidase activity determined with caffeine phenotyping. Clin
Pharmacol Ther. 1998 May;63(5):540-51
Katoh T, Kaneko S, Boissy R, Watson M, Ikemura K, Bell DA.
A pilot study testing the association between N-
acetyltransferases 1 and 2 and risk of oral squamous cell
carcinoma in Japanese people. Carcinogenesis. 1998
Oct;19(10):1803-7
Lin HJ, Probst-Hensch NM, Hughes NC, Sakamoto GT, Louie
AD, Kau IH, Lin BK, Lee DB, Lin J, Frankl HD, Lee ER, Hardy
S, Grant DM, Haile RW. Variants of N-acetyltransferase NAT1
and a case-control study of colorectal adenomas.
Pharmacogenetics. 1998 Jun;8(3):269-81
Taylor JA, Umbach DM, Stephens E, Castranio T, Paulson D,
Robertson C, Mohler JL, Bell DA. The role of N-acetylation
polymorphisms in smoking-associated bladder cancer:
evidence of a gene-gene-exposure three-way interaction.
Cancer Res. 1998 Aug 15;58(16):3603-10
Henning S, Cascorbi I, Münchow B, Jahnke V, Roots I.
Association of arylamine N-acetyltransferases NAT1 and NAT2
NAT1 (N-acetyltransferase 1 (arylamine N-acetyltransferase)) Ruiz JD, et al.
Atlas Genet Cytogenet Oncol Haematol. 2010; 14(1)
42
genotypes to laryngeal cancer risk. Pharmacogenetics. 1999
Feb;9(1):103-11
Hsieh FI, Pu YS, Chern HD, Hsu LI, Chiou HY, Chen CJ.
Genetic polymorphisms of N-acetyltransferase 1 and 2 and risk
of cigarette smoking-related bladder cancer. Br J Cancer. 1999
Oct;81(3):537-41
Ilett KF, Kadlubar FF, Minchin RF. 1998 International Meeting
on the Arylamine N-Acetyltransferases: synopsis of the
workshop on nomenclature, biochemistry, molecular biology,
interspecies comparisons, and role in human disease risk.
Drug Metab Dispos. 1999 Sep;27(9):957-9
Jourenkova-Mironova N, Wikman H, Bouchardy C, Mitrunen K,
Dayer P, Benhamou S, Hirvonen A. Role of arylamine N-
acetyltransferase 1 and 2 (NAT1 and NAT2) genotypes in
susceptibility to oral/pharyngeal and laryngeal cancers.
Pharmacogenetics. 1999 Aug;9(4):533-7
Butcher NJ, Ilett KF, Minchin RF. Substrate-dependent
regulation of human arylamine N-acetyltransferase-1 in
cultured cells. Mol Pharmacol. 2000 Mar;57(3):468-73
Hein DW, Doll MA, Fretland AJ, Leff MA, Webb SJ, Xiao GH,
Devanaboyina US, Nangju NA, Feng Y. Molecular genetics
and epidemiology of the NAT1 and NAT2 acetylation
polymorphisms. Cancer Epidemiol Biomarkers Prev. 2000
Jan;9(1):29-42
Cascorbi I, Roots I, Brockmöller J. Association of NAT1 and
NAT2 polymorphisms to urinary bladder cancer: significantly
reduced risk in subjects with NAT1*10. Cancer Res. 2001 Jul
1;61(13):5051-6
Fronhoffs S, Brüning T, Ortiz-Pallardo E, Bröde P, Koch B,
Harth V, Sachinidis A, Bolt HM, Herberhold C, Vetter H, Ko Y.
Real-time PCR analysis of the N-acetyltransferase NAT1 allele
*3, *4, *10, *11, *14 and *17 polymorphism in squamous cell
cancer of head and neck. Carcinogenesis. 2001
Sep;22(9):1405-12
Houlston RS, Tomlinson IP.. Polymorphisms and colorectal
tumor risk. Gastroenterology. 2001 Aug;121(2):282-301.
Krajinovic M, Ghadirian P, Richer C, Sinnett H, Gandini S,
Perret C, Lacroix A, Labuda D, Sinnett D. Genetic susceptibility
to breast cancer in French-Canadians: role of carcinogen-
metabolizing enzymes and gene-environment interactions. Int
J Cancer. 2001 Apr 15;92(2):220-5
Rodrigues-Lima F, Deloménie C, Goodfellow GH, Grant DM,
Dupret JM. Homology modelling and structural analysis of
human arylamine N-acetyltransferase NAT1: evidence for the
conservation of a cysteine protease catalytic domain and an
active-site loop. Biochem J. 2001 Jun 1;356(Pt 2):327-34
Upton A, Johnson N, Sandy J, Sim E. Arylamine N-
acetyltransferases - of mice, men and microorganisms. Trends
Pharmacol Sci. 2001 Mar;22(3):140-6
Wikman H, Thiel S, Jäger B, Schmezer P, Spiegelhalder B,
Edler L, Dienemann H, Kayser K, Schulz V, Drings P, Bartsch
H, Risch A. Relevance of N-acetyltransferase 1 and 2 (NAT1,
NAT2) genetic polymorphisms in non-small cell lung cancer
susceptibility. Pharmacogenetics. 2001 Mar;11(2):157-68
Hein DW. Molecular genetics and function of NAT1 and NAT2:
role in aromatic amine metabolism and carcinogenesis. Mutat
Res. 2002 Sep 30;506-507:65-77
Ye Z, Parry JM. Meta-analysis of 20 case-control studies on
the N-acetyltransferase 2 acetylation status and colorectal
cancer risk. Med Sci Monit. 2002 Aug;8(8):CR558-65
Butcher NJ, Arulpragasam A, Minchin RF. Proteasomal
degradation of N-acetyltransferase 1 is prevented by
acetylation of the active site cysteine: a mechanism for the
slow acetylator phenotype and substrate-dependent down-
regulation. J Biol Chem. 2004 May 21;279(21):22131-7
Husain A, Barker DF, States JC, Doll MA, Hein DW.
Identification of the major promoter and non-coding exons of
the human arylamine N-acetyltransferase 1 gene (NAT1).
Pharmacogenetics. 2004 Jul;14(7):397-406
Butcher NJ, Arulpragasam A, Goh HL, Davey T, Minchin RF.
Genomic organization of human arylamine N-acetyltransferase
Type I reveals alternative promoters that generate different 5'-
UTR splice variants with altered translational activities.
Biochem J. 2005 Apr 1;387(Pt 1):119-27
Chen K, Jiang QT, He HQ. Relationship between metabolic
enzyme polymorphism and colorectal cancer. World J
Gastroenterol. 2005 Jan 21;11(3):331-5
Gu J, Liang D, Wang Y, Lu C, Wu X. Effects of N-acetyl
transferase 1 and 2 polymorphisms on bladder cancer risk in
Caucasians. Mutat Res. 2005 Mar 7;581(1-2):97-104
Barker DF, Husain A, Neale JR, Martini BD, Zhang X, Doll MA,
States JC, Hein DW. Functional properties of an alternative,
tissue-specific promoter for human arylamine N-
acetyltransferase 1. Pharmacogenet Genomics. 2006
Jul;16(7):515-25
Carreón T, Ruder AM, Schulte PA, Hayes RB, Rothman N,
Waters M, Grant DJ, Boissy R, Bell DA, Kadlubar FF,
Hemstreet GP 3rd, Yin S, LeMasters GK. NAT2 slow
acetylation and bladder cancer in workers exposed to
benzidine. Int J Cancer. 2006 Jan 1;118(1):161-8
Dolled-Filhart M, Rydén L, Cregger M, Jirström K, Harigopal M,
Camp RL, Rimm DL. Classification of breast cancer using
genetic algorithms and tissue microarrays. Clin Cancer Res.
2006 Nov 1;12(21):6459-68
Jensen LE, Hoess K, Mitchell LE, Whitehead AS. Loss of
function polymorphisms in NAT1 protect against spina bifida.
Hum Genet. 2006 Aug;120(1):52-7
Liu F, Zhang N, Zhou X, Hanna PE, Wagner CR, Koepp DM,
Walters KJ. Arylamine N-acetyltransferase aggregation and
constitutive ubiquitylation. J Mol Biol. 2006 Aug 18;361(3):482-
92
Perera FP, Tang D, Brandt-Rauf P, Santella RM, Mooney LV,
Tu YH, Bendkowska I, Bell DA. Lack of associations among
cancer and albumin adducts, ras p21 oncoprotein levels, and
CYP1A1, CYP2D6, NAT1, and NAT2 in a nested case-control
study of lung cancer within the physicians' health study.
Cancer Epidemiol Biomarkers Prev. 2006 Jul;15(7):1417-9
Ring BZ, Seitz RS, Beck R, Shasteen WJ, Tarr SM, Cheang
MC, Yoder BJ, Budd GT, Nielsen TO, Hicks DG, Estopinal NC,
Ross DT. Novel prognostic immunohistochemical biomarker
panel for estrogen receptor-positive breast cancer. J Clin
Oncol. 2006 Jul 1;24(19):3039-47
Butcher NJ, Tetlow NL, Cheung C, Broadhurst GM, Minchin
RF. Induction of human arylamine N-acetyltransferase type I
by androgens in human prostate cancer cells. Cancer Res.
2007 Jan 1;67(1):85-92
Gemignani F, Landi S, Szeszenia-Dabrowska N, Zaridze D,
Lissowska J, Rudnai P, Fabianova E, Mates D, Foretova L,
Janout V, Bencko V, Gaborieau V, Gioia-Patricola L, Bellini I,
Barale R, Canzian F, Hall J, Boffetta P, Hung RJ, Brennan P.
Development of lung cancer before the age of 50: the role of
xenobiotic metabolizing genes. Carcinogenesis. 2007
Jun;28(6):1287-93
Husain A, Zhang X, Doll MA, States JC, Barker DF, Hein DW.
Functional analysis of the human N-acetyltransferase 1 major
promoter: quantitation of tissue expression and identification of
NAT1 (N-acetyltransferase 1 (arylamine N-acetyltransferase)) Ruiz JD, et al.
Atlas Genet Cytogenet Oncol Haematol. 2010; 14(1)
43
critical sequence elements. Drug Metab Dispos. 2007
Sep;35(9):1649-56
Minchin RF, Hanna PE, Dupret JM, Wagner CR, Rodrigues-
Lima F, Butcher NJ. Arylamine N-acetyltransferase I. Int J
Biochem Cell Biol. 2007;39(11):1999-2005
Sanderson S, Salanti G, Higgins J. Joint effects of the N-
acetyltransferase 1 and 2 (NAT1 and NAT2) genes and
smoking on bladder carcinogenesis: a literature-based
systematic HuGE review and evidence synthesis. Am J
Epidemiol. 2007 Oct 1;166(7):741-51
Wu H, Dombrovsky L, Tempel W, Martin F, Loppnau P,
Goodfellow GH, Grant DM, Plotnikov AN. Structural basis of
substrate-binding specificity of human arylamine N-
acetyltransferases. J Biol Chem. 2007 Oct 12;282(41):30189-
97
Agúndez JA. Polymorphisms of human N-acetyltransferases
and cancer risk. Curr Drug Metab. 2008 Jul;9(6):520-31
McKay JD, Hashibe M, Hung RJ, Wakefield J, Gaborieau V,
Szeszenia-Dabrowska N, Zaridze D, Lissowska J, Rudnai P,
Fabianova E, Mates D, Foretova L, Janout V, Bencko V,
Chabrier A, Hall J, Boffetta P, Canzian F, Brennan P.
Sequence variants of NAT1 and NAT2 and other
xenometabolic genes and risk of lung and aerodigestive tract
cancers in Central Europe. Cancer Epidemiol Biomarkers Prev.
2008 Jan;17(1):141-7
Zienolddiny S, Campa D, Lind H, Ryberg D, Skaug V,
Stangeland LB, Canzian F, Haugen A. A comprehensive
analysis of phase I and phase II metabolism gene
polymorphisms and risk of non-small cell lung cancer in
smokers. Carcinogenesis. 2008 Jun;29(6):1164-9
This article should be referenced as such:
Ruiz JD, Agúndez JAG, Martínez C, García-Martín E. NAT1
(N-acetyltransferase 1 (arylamine N-acetyltransferase)). Atlas
Genet Cytogenet Oncol Haematol. 2010; 14(1):39-43.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Arylamine N-acetyltransferases (EC 2.3.1.5) (NATs) catalyse the biotransformation of many primary arylamines, hydrazines and their N-hydroxylated metabolites, thereby playing an important role in both the detoxification and metabolic activation of numerous xenobiotics. The recently published crystal structure of the Salmonella typhimurium NAT (StNAT) revealed the existence of a cysteine protease-like (Cys-His-Asp) catalytic triad. In the present study, a three-dimensional homology model of human NAT1, based upon the crystal structure of StNAT [Sinclair, Sandy, Delgoda, Sim and Noble (2000) Nat. Struct. Biol. 7, 560-564], is demonstrated. Alignment of StNAT and NAT1, together with secondary structure predictions, have defined a consensus region (residues 29-131) in which 37% of the residues are conserved. Homology modelling provided a good quality model of the corresponding region in human NAT1. The location of the catalytic triad was found to be identical in StNAT and NAT1. Comparison of active-site structural elements revealed that a similar length loop is conserved in both species (residues 122-131 in NAT1 model and residues 122-133 in StNAT). This observation may explain the involvement of residues 125, 127 and 129 in human NAT substrate selectivity. Our model, and the fact that cysteine protease inhibitors do not affect the activity of NAT1, suggests that human NATs may have adapted a common catalytic mechanism from cysteine proteases to accommodate it for acetyl-transfer reactions.
Article
Breast cancer is the most frequent malignancy among women. Since genetic factors such as BRCA1 and BRCA2 as well as reproductive history constitute only 30% of the cause, environmental exposure may play a significant role in the development of breast cancer. Likewise, the relevant enzymes involved in the biotransformation of xenobiotics (from tobacco smoke, diet or other environmental sources) might play a role in breast carcinogenesis. Since individuals with modified ability to metabolize these carcinogens could have a different risk for breast cancer, we investigated the role of cytochromes P‐450 (CYP1A1, CYP2D6), glutathione‐S‐transferases (GSTM1, GSTT1, GSTP1) and N‐acetyltransferases (NAT1, NAT2) gene variants in breast carcinogenesis. A case‐control study was conducted on 149 women with breast carcinoma and 207 healthy controls, both of French‐Canadian origin. The CYP1A1*4 allele was found to be a significant risk determinant of breast carcinoma (OR = 3.3, 95% CI 1.1–9.7), particularly among post‐menopausal women (OR = 4.0, 95% CI 1.2–13.8). The frequency of NAT2 rapid acetylators was increased among smokers (OR = 2.6, 95% CI 0.8–8.2), while the NAT1*10 allele conferred a 4‐fold increase in risk among women who consumed well‐done meat (OR = 4.4, 95% CI 1.0–18.9). These data suggest that CYP1A1*4, NAT1 and NAT2 variants are involved in the susceptibility to breast carcinoma by modifying the impact of exogenous and/or endogenous exposures. © 2001 Wiley‐Liss, Inc.
Article
Arylamine N-acetyltransferases (NATs) catalyse the transfer of an acetyl group from acetyl CoA to the terminal nitrogen of hydrazine and arylamine drugs and carcinogens. These enzymes are polymorphic and have an important place in the history of pharmacogenetics, being first identified as responsible for the polymorphic inactivation of the anti-tubercular drug isoniazid. NAT has recently been identified within Mycobacterium tuberculosis itself and is an important candidate for modulating the response of mycobacteria to isoniazid. The first three-dimensional structure of the unique NAT family shows the active-site cysteine to be aligned with conserved histidine and aspartate residues to form a catalytic triad, thus providing an activation mechanism for transfer of the acetyl group from acetyl CoA to cysteine. The unique fold could allow different members of the NAT family to play a variety of roles in endogenous and xenobiotic metabolism.
Article
Arylamine N-acetyltransferase I (NAT1) is a phase II enzyme that acetylates a wide range of arylamine and hydrazine substrates. The NAT1 gene is located on chromosome 8 and shares homology to NAT genes found in most mammalian species. Gene expression occurs from at least two promoters and a number of tissue-specific transcripts have been identified. The gene is polymorphic with most mutations identified to date producing an unstable protein that is subject to polyubiquitination. The NAT1 protein contains a catalytic triad similar to a number of cysteine proteases and transglutaminases. NAT1 is widely distributed in the body, but the only endogenous substrate identified to date is the folate catabolite p-aminobenzoylglutamate. Recent links between NAT1 genotypes and susceptibility to spina bifida suggests that the enzyme has an important role in folate homeostasis.
Article
Human arylamine N-acetyltransferases (CoASAc; NAT, EC 2.3.1.5) NAT1 and NAT2 play a key role in the metabolism of drugs and environmental chemicals and in the metabolic activation and detoxification of procarcinogens. Phenotyping analyses have revealed an association between NAT enzyme activities and the risk of developing several forms of cancer. As genotyping procedures have become available for NAT1 and NAT2 gene variations, hundreds of association studies on NAT polymorphisms and cancer risk have been conducted. Here we review the findings obtained from these studies. Evidence for a putative association of NAT1 polymorphism and myeloma, lung and bladder cancer, as well as association of NAT2 polymorphisms with non-Hodgkin lymphoma, liver, colorectal and bladder cancer have been reported. In contrast, no consistent evidence for a relevant association of NAT polymorphisms with brain, head & neck, breast, gastric, pancreatic or prostate cancer have been described. Although preliminary data are available, further well-powered studies are required to fully elucidate the role of NAT1 in most human cancers, and that of NAT2 in astrocytoma, meningioma, esophageal, renal, cervical and testicular cancers, as well as in leukaemia and myeloma. This review discusses controversial findings on cancer risk and putative causes of heterogeneity in the proposed associations, and it identifies topics that require further investigation, particularly mechanisms underlying association of NAT polymorphisms and risk for subsets of cancer patients with specific exposures, putative epistatic contribution of polymorphism for other xenobiotic-metabolising enzymes such as glutathione S-transferases of Cytochrome P450 enzymes, and genetic plus environmental interaction.
Article
The highly polymorphic N-acetyltransferases (NAT1 and NAT2) are involved in both activation and inactivation reactions of numerous carcinogens, such as tobacco derived aromatic amines. The potential effect of the NAT genotypes in individual susceptibility to lung cancer was examined in a hospital based case-control study consisting of 392 Caucasian lung cancer patients [152 adenocarcinomas, 173 squamous cell carcinomas (SCC) and 67 other primary lung tumours] and 351 controls. In addition to the wild-type allele NAT1*4, seven variant NAT1 alleles (NAT1*3, *10, *11, *14, *15, *17 and *22) were analysed. A new method based on the LightCycler (Roche Diagnostics Inc.) technology was applied for the detection of the polymorphic NAT1 sites at nt 1088 and nt 1095. The NAT2 polymorphic sites at nt 481, 590, 803 and 857 were detected by polymerase chain reaction-restriction fragment length polymorphism or LightCycler. Multivariate logistic regression analyses were performed taking into account levels of smoking, age, gender and occupational exposure. An increased risk for adenocarcinoma among the NAT1 putative fast acetylators [odds ratio (OR) 1.92 (1.16-3.16)] was found but could not be detected for SCC or the total case group. NAT2 genotypes alone appeared not to modify individual lung cancer risk, however, individuals with combined NAT1 fast and NAT2 slow genotype had significantly elevated adenocarcinoma risk [OR 2.22 (1.03-4.81)] compared to persons with other genotype combinations. These data clearly show the importance of separating different histological lung tumour subtypes in studies on genetic susceptibility factors and implicate the NAT1*10 allele as a risk factor for adenocarcinoma.
Article
Increasingly, studies of the relationship between common genetic variants and colorectal tumor risk are being proposed. To assess the evidence that any of these confers a risk, a systematic review and meta-analysis of published studies was undertaken. Fifty studies of the effect of common alleles of 13 genes on risk were identified. To clarify the impact of individual polymorphisms on risk, pooled analyses were performed. Of the 50 studies identified, significant associations were seen in 16, but only 3 were reported in more than one study. Pooling studies, significant associations were only seen for 3 of the polymorphisms: adenomatosis polyposis coli (APC)-I1307K (odds ratio [OR] = 1.58, 95% confidence interval [CI]: 1.21-2.07); Harvey ras-1 variable number tandem repeat polymorphism (HRAS1-VNTR; OR = 2.50, 95% CI: 1.54-4.05); and methylenetetrahydrofolate reductase (MTHFR)(Val/Val) (OR = 0.76, 95% CI: 0.62-0.92). For tumor protein 53 (TP53), N-acetyl transferase 1 (NAT1), NAT2, glutathione-S transferase Mu (GSTM1), glutathione-S transferase Theta (GSTT1), and glutathione-S transferase Pi (GSTP1) polymorphisms, the best estimates are sufficient to exclude a 1.7-fold increase in risk of colorectal cancer. APC-I1307K, HRAS1-VNTR, and MTHFR variants represent the strongest candidates for low penetrance susceptibility alleles identified to date. Although their genotypic risks are modest, their high frequency in the population implies that they may well have considerable impact on colorectal cancer incidence. Determining precise risk estimates associated with other variants and gene-gene and gene-environment interactions will be contingent on further studies with sample sizes larger than typically used to date.
Article
Rapid NAT2 acetylation has been considered as a risk factor for developing colon cancer in a number of studies, however the overall results of such studies are inconsistent. To clarify the influence of NAT2 rapid acetylation status on colon cancer risk, we have performed a meta-analysis of 20 published case-control studies (4431 cases, 4547 controls). Odd ratio was employed to evaluate the risk of colon cancer and NAT2 rapid acetylation status. To take into account the possibility of heterogeneity across the studies, a statistical test for heterogeneity across the studies was performed. The summary odds ratios were assessed by calculating a weighted average of odds ratios for all of the studies. The pooling of studies based on phenotyping methods indicated that the overall odds ratio of colon cancer risk associated with rapid acetylator was 1.51 (95%CI: 1.07-2.12). However, the risk of colon cancer associated with rapid acetylator from the studies based on genotyping method was lower with a calculated overall odds ratio of 1.06 (95%CI: 0.97-1.15). Pooling studies were also conducted on specific tumour sites and ethnic groups. The results show that effect of rapid acetylator on colon cancer risk was not obviously different. The results of our meta-analyses do not support the hypothesis that NAT2 alone is an important risk factor for colon cancer and suggests that NAT2 rapid acetylation status has no specific effect on the risk of developing colon cancer.
Article
Aromatic and heterocyclic amines require metabolic activation to electrophilic intermediates that initiate carcinogenesis. N-Acetyltransferase 1 (NAT1) and 2 (NAT2) are important enzymes in the biotransformation of these carcinogens and exhibit genetic polymorphism. Human NAT1 and NAT2 alleles are listed at: http://www.louisville.edu/medschool/pharmacology/NAT.html by an international gene nomenclature committee. The high frequency of the NAT1 and NAT2 acetylation polymorphisms in human populations together with ubiquitous exposure to aromatic and heterocyclic amines suggest that NAT1 and NAT2 acetylator genotypes are important modifiers of human cancer susceptibility. For cancers in which N-acetylation is a detoxification step such as aromatic amine-related urinary bladder cancer, NAT2 slow acetylator phenotype is at higher risk. Multiple studies have shown that the urinary bladder cancer risk is particularly high in the slowest NAT2 acetylator phenotype or genotype (NAT2(*)5). In contrast, for cancers in which N-acetylation is negligible and O-acetylation is an activation step such as for heterocyclic amine-related colon cancer, NAT2 rapid acetylator phenotype is at higher risk. Although studies have found associations between NAT1 genotype and various cancers, the findings are less consistent and are not well understood. Since cancer risk requires exposure to aromatic and/or heterocyclic amine carcinogens modified by NAT1 and/or NAT2 acetylator genotype, the results from human epidemiology studies are dependent upon the quality and accuracy of the exposure assessment and genotype determination. Conclusions require understanding the relationship between genotype and phenotype, as well as the role of genetic variation in carcinogen metabolism, DNA repair, and host susceptibility. Investigations have been carried out in rapid and slow acetylator rodent models in which both exposure and genetic variability are tightly controlled. Human NAT1 and NAT2 alleles have been characterized by recombinant expression to further understand the effects of nucleotide polymorphisms on function and phenotype.
Article
Many drugs and chemicals found in the environment are either detoxified by N-acetyltransferase 1 (NAT1, EC 2.3.1.5) and eliminated from the body or bioactivated to metabolites that have the potential to cause toxicity and/or cancer. NAT1 activity in the body is regulated by genetic polymorphisms as well as environmental factors such as substrate-dependent down-regulation and oxidative stress. Here we report the molecular mechanism for the low protein expression from mutant NAT1 alleles that gives rise to the slow acetylator phenotype and show that a similar process accounts for enzyme down-regulation by NAT1 substrates. NAT1 allozymes NAT1 14, NAT1 15, NAT1 17, and NAT1 22 are devoid of enzyme activity and have short intracellular half-lives ( approximately 4 h) compared with wild-type NAT1 4 and the active allozyme NAT1 24. The inactive allozymes are unable to be acetylated by cofactor, resulting in ubiquitination and rapid degradation by the 26 S proteasome. This was confirmed by site-directed mutagenesis of the active site cysteine 68. The NAT1 substrate p-aminobenzoic acid induced ubiquitination of the usually stable NAT1 4, leading to its rapid degradation. From this study, we conclude that NAT1 exists in the cell in either a stable acetylated state or an unstable non-acetylated state and that mutations in the NAT1 gene that prevent protein acetylation produce a slow acetylator phenotype.