Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Mol Phylogenet Evol ; 53(3): 1010-24, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19766197

ABSTRACT

A phylogenetic analysis of 19 sibling taxa in the Vertigo gouldii group was conducted on 73 individuals sampled across North America using DNA sequence data of the mitochondrial genes cytochrome oxidase subunit 1 (CO1) and 16S ribosomal RNA (16S), and the internal transcribed spacer-2 of the nuclear ribosomal RNA (ITS-2) gene. The results of these analyses were found incongruent with previous taxonomic concepts used to define the V. gouldii group and its composite taxa that were based entirely on conchological features. The mtDNA sequence data suggest that some previous members of the traditional V. gouldii group may be more closely related to V. modesta. They also suggest that V. gouldii may itself consist of seven species-level branches spread across two deeply rooted clades. Revision of geographical distributions on the basis of these analyses suggests that these Vertigo species may commonly possess continental-sized ranges in spite of their minute size and limited active dispersal ability. High levels of sympatry within the group are also confirmed, with up to four species being known to co-occur within single microsites. These data also suggest that rates of diversification have been non-constant. Assuming a 1%/my rate of base pair substitution, a 10-fold diversification pulse is indicated from 6.7-7.0 myBP, which would be co-incident with known mid-late Miocene global climate changes.


Subject(s)
Evolution, Molecular , Genetic Speciation , Phylogeny , Snails/genetics , Animals , Bayes Theorem , DNA, Mitochondrial/genetics , DNA, Ribosomal Spacer/genetics , Geography , Likelihood Functions , North America , RNA, Ribosomal, 16S/genetics , Sequence Alignment , Sequence Analysis, DNA , Snails/classification
2.
Int J Cancer ; 117(1): 8-13, 2005 Oct 20.
Article in English | MEDLINE | ID: mdl-15880531

ABSTRACT

Dietary factors appear to be involved in the high incidence of prostate cancer in "Westernized" countries, implicating dietary carcinogens such as heterocyclic amines (HAs) in the initiation of prostate carcinogenesis. We examined 24 human prostate samples with respect to their potential for activation and detoxification of HAs and the presence of DNA adducts formed in vivo. Cytochromes P450 1B1, 3A4 and 3A5 were expressed at low levels (<0.1-6.2 pmol/mg microsomal protein). N-Acetyltransferase (NAT) activities, using p-aminobenzoic acid (NAT1) and sulfamethazine (NAT2) as substrates, were <5-5,500 and <5-43 pmol/min/mg cytosolic protein, respectively. Glutathione S-transferases (GSTs) P1, M2 and M3 were expressed at 0.038-1.284, 0.005-0.126 and 0.010-0.270 microg/mg cytosolic protein, respectively; GSTM1 was expressed in all GSTM1-positive samples (0.012-0.291 microg/mg cytosolic protein); and GSTA1 was expressed at low levels (<0.01-0.11 microg/mg cytosolic protein). Binding of N-hydroxy-PhIP to DNA in vitro occurred primarily by an AcCoA-dependent process (<1-54 pmol/mg/DNA), PAPS- and ATP-dependent binding being <1-7 pmol/mg DNA. In vivo, putative PhIP- or 4-aminobiphenyl-DNA adducts were found in 4 samples (0.4-0.8 adducts/10(8) bases); putative hydrophobic adducts were found in 6 samples (8-64 adducts/10(8) bases). Thus, the prostate appears to have low potential for N-hydroxylation of HAs but greater potential for activation of N-hydroxy HAs to genotoxic N-acetoxy esters. The prostate has potential for GSTP1-dependent detoxification of ATP-activated N-hydroxy-PhIP but little potential for detoxification of N-acetoxy-PhIP by GSTA1. However, there were no significant correlations between expression/activities and DNA adducts formed in vitro or in vivo, DNA adducts in vivo possibly reflecting carcinogen exposure.


Subject(s)
Acetyl Coenzyme A/metabolism , Adenosine Triphosphate/metabolism , Cytochrome P-450 Enzyme System/metabolism , Glutathione Transferase/metabolism , Phosphoadenosine Phosphosulfate/metabolism , Prostate/enzymology , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Aged , Aged, 80 and over , Arylalkylamine N-Acetyltransferase/metabolism , Carcinogens/metabolism , DNA/metabolism , DNA Adducts , Glutathione Transferase/genetics , Humans , Male , Microsomes/enzymology , Middle Aged , Prostatic Hyperplasia/enzymology , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology
3.
Cancer Res ; 65(3): 1105-11, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15705913

ABSTRACT

The proximate cause of cancer cell death by radiation therapy and a number of therapeutic agents is through generation of reactive oxygen species, resulting in DNA damage as well as mitochondrial membrane disruption, triggering the apoptotic cascade. Because mitochondrial manganese superoxide dismutase catalyzes conversion of superoxide radicals to H(2)O(2), with catalase neutralizing H(2)O(2) and myeloperoxidase converting H(2)O(2) to highly reactive hypochlorous acid, we hypothesized that gene variants could impact the efficacy of treatment for breast cancer and improve survival. Women who were treated with radiation and/or chemotherapy for incident breast cancer at the Arkansas Cancer Research Center from 1985 to 1996 were identified. DNA was extracted from paraffin-embedded normal tissue (n = 279), and MnSOD, CAT, and MPO genotypes were determined using mass spectrometry. Cox proportional hazards models were adjusted for age, race, stage with node status, and estrogen receptor and progesterone receptor status. Women who were homozygous for MPO G alleles, associated with increased transcription, had better survival (hazard ratio, 0.60; 95% confidence interval, 0.38-0.95; P = 0.03) than those with common alleles. Both CAT TT and MnSOD CC genotypes were associated with nonsignificant reduced hazard of death. When we combined genotypes associated with higher levels of reactive oxygen species for MnSOD and MPO, women with MnSOD CC and MPO GG genotypes had a 3-fold decrease in hazard of death (hazard ratio, 0.33; 95% confidence interval, 0.13-0.80; P = 0.01). These data indicate that gene variants that impact oxidative stress modify prognosis after treatment for breast cancer.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/therapy , Catalase/genetics , Peroxidase/genetics , Superoxide Dismutase/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/radiotherapy , Catalase/metabolism , Female , Genetic Predisposition to Disease , Humans , Middle Aged , Oxidative Stress , Peroxidase/metabolism , Polymorphism, Genetic , Superoxide Dismutase/metabolism
4.
Methods Enzymol ; 401: 9-42, 2005.
Article in English | MEDLINE | ID: mdl-16399377

ABSTRACT

The human alpha class glutathione S-transferases (GSTs) consist of 5 genes, hGSTA1-hGSTA5, and 7 pseudogenes on chromosome 6p12.1-6p12.2. hGSTA1-hGSTA4 have been well characterized as proteins, but hGSTA5 has not been detected as a gene product. hGSTA1-1 (and to a lesser extent hGSTA2-2) catalyzes the GSH-dependent detoxification of carcinogenic metabolites of environmental pollutants and tobacco smoke (e.g., polycyclic aromatic hydrocarbon diolepoxides) and several alkylating chemotherapeutic agents and has peroxidase activity toward fatty acid hydroperoxides (FA-OOH) and phosphatidyl FA-OOH. hGSTA3-3 has high activity for the GSH-dependent Delta(5)-Delta(4) isomerization of steroids, and hGSTA4-4 has high activity for the GSH conjugation of 4-hydroxynonenal. hGSTA4 is expressed in many tissues; hGSTA1-1 and hGSTA2-2 are expressed at high levels in liver, intestine, kidney, adrenal gland, and testis; and hGSTA3 is expressed in steroidogenic tissues. Functional, allelic, single nucleotide polymorphisms occur in an SP1-binding element of hGSTA1 and in the coding regions of hGSTA2 and hGSTA3. The main effects of these polymorphisms are the low hepatic expression of hGSTA1 in individuals homozygous for hGSTA1*B and the low specific activity of the hGSTA2E-2E variant toward FA-OOH. These properties suggest that alpha class GSTs will be involved in susceptibility to diseases with an environmental component (such as cancer, asthma, and cardiovascular disease) and in response to chemotherapy. Although hGSTM1, hGSTT1, and hGSTP1 have been associated with such diseases (on the basis of genetic polymorphisms as indicators of expression), alpha class GSTs have been little studied in this respect. Nevertheless, hGSTA1*B has been associated with increased susceptibility to colorectal cancer and with increased efficacy of chemotherapy for breast cancer. Methods for identification and quantitation of human alpha class GST protein, mRNA, and genotype are reviewed, and the potential for GST-alpha in plasma to be used as a marker for hepatic expression and induction is discussed.


Subject(s)
Genetic Predisposition to Disease , Glutathione Transferase , Isoenzymes , Polymorphism, Genetic , Animals , Genotype , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Tissue Distribution
5.
Environ Mol Mutagen ; 44(4): 265-76, 2004.
Article in English | MEDLINE | ID: mdl-15468054

ABSTRACT

Coffee drinking has been associated with reduced incidence of colorectal cancer, possibly via chemoprotection/modification of the metabolism of dietary heterocyclic amine carcinogens such as 2-amino-1-methyl-6-phenylimidazo-[4,5-b]pyridine (PhIP) by kahweol and cafestol palmitates (K/C), two components of unfiltered coffee. Using the PhIP-exposed male Fisher F344 rat as a model, K/C have been shown to reduce colonic PhIP-DNA adducts by > 50%. We have used the male F344 rat to investigate the effects of dietary K/C (0.02-0.2% as a 1:1 mixture) on the metabolism of PhIP by N-acetyltransferase- (NAT), sulfotransferase- (SULT), and glutathione-dependent pathways. K/C decreased hepatic NAT-dependent PhIP activation by up to 80% in a dose-dependent manner. Conversely, hepatic glutathione S-transferase (GST) activity/expression increased, e.g., 3-4 fold toward 1-chloro-2,4-dinitrobenzene (total activity), up to 23-fold toward 4-vinylpyridine (rGSTP1), and approximately 7-fold for rGSTA2 protein. These effects had fully developed after 5 days of the test diet and persisted for at least 5 days after withdrawal of K/C. Hepatic glutathione increased two- to threefold and this increase was more short-lived than other changes. K/C did not modify hepatic SULT activity or colon NAT and GST activities. Benzylisothiocyanate and black tea, which have also been shown to reduce the formation of PhIP-DNA adducts in this model, had little effect on hepatic NAT, SULT, GST, or GSH. In primary culture of rat hepatocytes, both kahweol and cafestol palmitates reduced NAT activity by 80%. In summary, the unique potential of K/C to convert rapid acetylators to a slow acetylator phenotype, accompanied by GST induction, might contribute to chemoprevention against cancers associated with heterocyclic amines.


Subject(s)
Acetyltransferases/metabolism , Carcinogens/metabolism , Diterpenes/pharmacology , Glutathione Transferase/metabolism , Imidazoles/metabolism , Liver/drug effects , Animals , Coffee/chemistry , Colon/drug effects , Colon/enzymology , DNA Adducts/metabolism , Dose-Response Relationship, Drug , Isothiocyanates/pharmacology , Liver/enzymology , Rats , Rats, Inbred F344 , Tea/chemistry , Time Factors
6.
Lung Cancer ; 44(3): 281-6, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15140540

ABSTRACT

The formation of DNA adducts is thought to be a critical step for the induction of chemically induced cancer. O(6)-Methylguanine-DNA methyltransferase (MGMT) is a ubiquitously expressed enzyme that repairs DNA adducts formed by alkylating carcinogens. Thus, genetic polymorphisms of the MGMT that could result in differences in MGMT activity are potential risk factors for cancer. In the present study, we established a convenient and reliable genotyping method for the MGMT codon 178 polymorphism, a Lys (AAG) to Arg (AGG) substitution, using restriction fragment length polymorphism (RFLP), and studied differences in the distribution of this polymorphism in 92 Caucasian lung cancer patients and 85 controls. Frequencies of the "A" and "G" alleles (MGMT codon 178, AAG and AGG, respectively) were 0.91 and 0.09, respectively. The genetic polymorphism of the MGMT codon 178 was linked with that of the MGMT codon 143 (P < 0.05). The distribution of the MGMT codon 178 genetic polymorphism was not significantly different between lung cancer patients and controls. Thus, our study suggests that the MGMT codon 178 (and possibly 143) polymorphisms do not appear to markedly affect lung cancer risk for this population. In addition, we found an apparent 10bp-deletion in the intron before exon 5 by DNA sequencing. Because this "deletion" was observed in all sequenced samples (N = 20), the previously reported human (Caucasian) MGMT gene sequence should be revised to exclude this 10bp segment.


Subject(s)
Lung Neoplasms/genetics , O(6)-Methylguanine-DNA Methyltransferase/genetics , Codon/genetics , DNA Adducts/genetics , Female , Genetic Predisposition to Disease/ethnology , Genetic Predisposition to Disease/genetics , Humans , Lung Neoplasms/ethnology , Male , Middle Aged , Polymorphism, Genetic/genetics , Risk Factors , Sequence Analysis, DNA/methods , White People/genetics
7.
Pharmacogenetics ; 14(1): 35-44, 2004 Jan.
Article in English | MEDLINE | ID: mdl-15128049

ABSTRACT

Variability of expression of the major glutathione S-transferases (GSTs) of liver, GSTA1 and GSTA2, is thought to affect the efficiency of detoxification of xenobiotics, including chemical carcinogens. Polymorphism of the GSTA1 regulatory sequence determines some of the variation of hepatic GSTA1 expression, but the polymorphisms in GSTA2 (exons 5 and 7) were not thought to affect GSTA2 activity. By examining GST protein expression for a set of human liver and pancreas samples (coupled with a cloning/polymerase chain reaction-restriction fragment length polymorphism strategy), we identified a novel substitution Pro110Ser (328C>T) and the corresponding novel variant GSTA2*E (Ser110Ser112Lys196Glu210), and confirmed the presence of variants GSTA2*A (Pro110Ser112Lys196Glu210), GSTA2*B (Pro110Ser112Lys196Ala210) and GSTA2*C (Pro110Thr112Lys196Glu210). GSTA2*C occurred at 30-60% (i.e. approximately 100-fold more frequent than previously reported) and GSTA2*E occurred (heterozygous) at approximately 11%. Hepatic expression of the Ser112 variants (GSTA2*A, GSTA2*B or GSTA2*E) was approximately four-fold higher than that of the Thr112 variant (GSTA2*C). Compared to any other variant, GSTA2E had lower rates of catalysis towards 1-chloro-2,4-dinitrobenzene (CDNB), 4-vinylpyridine, and cumene-, t-butyl- and arachidonic acid hydroperoxides, although kcat/Km for CDNB were similar for all four variants. Using a prostate cancer case-control population, it was found that GSTA1*A/GSTA2 C335 and GSTA1*B/GSTA2 G335 were in linkage disequilibrium in Caucasians but not in African-Americans. However, there were no significant differences in the distribution of these polymorphisms or resultant haplotypes by case status. Nevertheless, the rare genotypes, GSTA2*E/*E and GSTA1*B/*B + GSTA2*C/*C (potential low GSTA2 activity and low hepatic GSTA1 and GSTA2 expression, respectively) could increase the risk of adverse effects of xenobiotics via compromised efficiency of detoxification.


Subject(s)
Glutathione Transferase/genetics , Polymorphism, Genetic , Prostatic Neoplasms/enzymology , Base Sequence , Case-Control Studies , Catalysis , Chromatography, High Pressure Liquid , Glutathione Transferase/metabolism , Humans , Liver/enzymology , Male , Molecular Sequence Data , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Prostatic Neoplasms/genetics , Sequence Homology, Amino Acid
8.
J Biol Chem ; 279(16): 16246-53, 2004 Apr 16.
Article in English | MEDLINE | ID: mdl-14742434

ABSTRACT

Kappa class glutathione S-transferase (GST) cDNA sequences have been identified in rat, mouse, and human. In the present study, we determined the structure and chromosomal location of the human GST Kappa 1 (hGSTK1) gene, characterized the protein, and demonstrated its subcellular localization. The human gene spans approximately 5 kb, has 8 exons, and maps onto chromosome 7q34. The 5'-flanking region lacks TATA or CCAAT boxes, but there is an initiator element overlapping the transcription start site. hGSTK1 amino acid sequence showed homology to bacterial 2-hydroxychromene-2-carboxylate isomerase, an enzyme involved in naphthalene degradation pathway. hGSTK1 mRNA was expressed in all of the organs examined. Subcellular fractionation of HepG2 cells showed that the protein was located in peroxisomes and mitochondria and was not detectable in cytoplasm. The peroxisomal localization was confirmed by transfection of HepG2 cells with a plasmid coding a green fluorescent protein fused inframe to the N terminus of hGSTK1. The C terminus of hGSTK1 was essential for localization of the protein to peroxisomes, and the C-terminal sequence Ala-Arg-Leu represents a peroxisome targeting signal. This is the first time that a human GST has been found in peroxisomes, suggesting a new function for this family of enzymes.


Subject(s)
Chromosomes, Human, Pair 7 , Glutathione Transferase/analysis , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Peroxisomes/metabolism , Amino Acid Sequence , Base Sequence , Chromosome Mapping , Humans , Molecular Sequence Data , Organ Specificity , Sequence Alignment , Sequence Analysis
9.
Biofactors ; 17(1-4): 115-30, 2003.
Article in English | MEDLINE | ID: mdl-12897434

ABSTRACT

The glutathione S-transferases (GSTs) catalyze the GSH-dependent detoxification of reactive electrophiles such as genotoxic chemical carcinogens and cytotoxic chemotherapeutic agents. Allelic polymorphism in the GSTs has been used to investigate the hypothesis that GSTs are involved in susceptibility to human cancers. Such studies have resulted in low penetrance, high prevalence associations between cancer risk and GST polymorphisms. By examination of interindividual variation of GST expression it becomes clear that GST genotype alone is not an accurate predictor of GST expression. GST expression is tissue specific and interindividual variation of expression is at least 7-fold in normal tissues. Thus, populations of the same genotype are actually heterogeneous as regards expression. Similarly, polymorphisms are not effective in all tissues and GST induction is not independent of genotype. Mechanistic models for chemical aspects of colorectal cancer and chemotherapy for breast cancer demonstrate some of the ways by which such interactions can be studied and the potential for future studies.


Subject(s)
Antineoplastic Agents/metabolism , Carcinogens/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Inactivation, Metabolic , Alleles , Antineoplastic Agents, Alkylating , Catalysis , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/genetics , Gene Expression , Genetic Predisposition to Disease , Genetic Variation , Genotype , Humans , Polymorphism, Genetic
10.
Int J Cancer ; 103(6): 810-4, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12516103

ABSTRACT

Glutathione S-transferase (GST) enzymes detoxify chemotherapeutic drugs, and several studies have reported differences in survival for cancer patients who have variant genotypes for GSTP1, GSTM1 or GSTT1 enzymes. A recently described polymorphism alters hepatic expression of GSTA1, a GST with high activity in glutathione conjugation of metabolites of cyclophosphamide (CP). To consider the possible influence of the reduced-expression GSTA1*B allele on cancer patient survival, we have conducted a pilot study of breast cancer patients treated with CP-containing combination chemotherapy. GSTA1 genotype was determined by polymerase chain reaction and restriction fragment length polymorphism. Kaplan-Meier methods and Cox proportional hazards models were used to evaluate survival in relation to genotype. Among 245 subjects, 35% were GSTA1*A/*A, 49% GSTA1*A/*B and 16% GSTA1*B/*B; the genotype distribution did not differ by ethnic group, age or stage at diagnosis. Among patients who had 0 or 1 GSTA1*B allele, the proportion surviving at 5 years was 0.66 (95% CI = 0.59-0.72), whereas for GSTA1*B/*B subjects the proportion was higher, 0.86 (95% CI = 0.67-0.95). Significantly reduced hazard of death was observed for GSTA1*B/*B subjects during the first 5 years after diagnosis, hazard ratio (HR) = 0.3, 95% CI = 0.1-0.8. The association varied with time, with no survival difference observed for subjects who survived beyond 5 years. These results, although based on a small study population, describe an apparent difference in survival after treatment for breast cancer according to GSTA1 genotype. Further studies should consider the possible association between the novel GSTA1*B variant and outcomes of cancer therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/mortality , Glutathione Transferase/genetics , Polymorphism, Genetic , Promoter Regions, Genetic/genetics , Adult , Aged , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/enzymology , Carcinoma, Ductal, Breast/mortality , Carcinoma, Lobular/drug therapy , Carcinoma, Lobular/enzymology , Carcinoma, Lobular/mortality , Case-Control Studies , Cohort Studies , Cyclophosphamide/administration & dosage , Female , Genotype , Humans , Isoenzymes , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Pilot Projects , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Retrospective Studies , Survival Rate
11.
Toxicology ; 181-182: 83-7, 2002 Dec 27.
Article in English | MEDLINE | ID: mdl-12505289

ABSTRACT

Red meats cooked at high temperatures generate mutagenic heterocyclic amines, which undergo metabolic activation by hepatic cytochrome P450 1A2 and N-acetyltransferase-2. A primary detoxification pathway involves glutathione S-transferase A1 (GSTA1), which catalyzes the reduction of the carcinogenic N-acetoxy derivative back to the parent amine. Recently, we described a polymorphism in the GSTA1 proximal promoter; the variant (GSTA1*B) allele significantly lowers enzyme expression. In a case-control study, GSTA1*B/*B genotype was associated with an increased risk of colorectal cancer, particularly among consumers of well-done meat. Dietary nitrosamines, which are bioactivated by CYP2A6, represent another potential etiologic factor for colorectal cancer. CYP2A6 converts the caffeine metabolite 1,7-dimethylxanthine (17X) to 1,7-dimethyluric acid (17U); we investigated CYP2A6 activity using the 17U/17X urinary metabolite ratio from case-control subjects who completed a caffeine phenotype assay. The distribution of CYP2A6 activity was significantly different between CRCa cases and controls, with subjects in the medium and high activity groups having an increased risk (P for trend=0.001). GSTA1 genotype and CYP2A6 phenotype should be evaluated as markers of susceptibility to dietary carcinogens in future studies.


Subject(s)
Bacterial Proteins , Carcinogens/toxicity , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/epidemiology , Diet/adverse effects , Meat/analysis , Aryl Hydrocarbon Hydroxylases/genetics , Biomarkers , Caffeine , Carrier Proteins/genetics , Case-Control Studies , Cytochrome P-450 CYP2A6 , DNA/analysis , DNA/genetics , Genotype , Glutathione Transferase , Humans , Intracellular Signaling Peptides and Proteins , Lymphocytes/chemistry , Meat/adverse effects , Mixed Function Oxygenases/genetics , Nitrosamines/analysis , Phenotype , Phosphodiesterase Inhibitors , Promoter Regions, Genetic/genetics , Risk Assessment
12.
Lung Cancer ; 38(1): 15-21, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12367788

ABSTRACT

Individual differences in lung cancer susceptibility should be considered for effective lung cancer prevention. We investigated the CYP2E1, ADH3, and GSTP1 genetic polymorphisms that biotransform xenobiotic carcinogens, and variations of their enzyme activity in Caucasian lung tissues (N=28), and found a variant distribution in pulmonary ADH and CYP2E1 activity. The ADH3*1/*1 subjects (N=8) showed significantly higher ADH activity than ADH3*2/*2 (N=3) subjects (P<0.01). On the other hand, we found a 5-fold variation in the pulmonary CYP2E1 activity using a sensitive HLPC/EC based technique. A subject with the CYP2E1-c/t allele showed 2-fold higher CYP2E1 activity than subjects with the c/c allele (N=14). GSTP1 expression comprised 83% of the total pulmonary GSTs. However, neither the GSTP1 polymorphism, nor other lifestyle factors, such as age, gender, smoking status, were found to be associated with pulmonary GST expression. In conclusion, subjects with the ADH3*1 allele showed higher ADH activity and acetaldehyde-DNA adducts in lung than other subjects; thus, the ADH3*1 allele could be considered a risk factor for lung cancer.


Subject(s)
Alcohol Dehydrogenase/biosynthesis , Alcohol Dehydrogenase/genetics , Cytochrome P-450 CYP2E1/biosynthesis , Cytochrome P-450 CYP2E1/genetics , DNA Adducts , Gene Expression Regulation, Neoplastic , Glutathione Transferase/biosynthesis , Glutathione Transferase/genetics , Isoenzymes/biosynthesis , Isoenzymes/genetics , Lung Neoplasms/genetics , Lung Neoplasms/physiopathology , Lung/physiology , Polymorphism, Genetic , White People/genetics , Acetaldehyde/chemistry , Adolescent , Adult , Age Factors , Aged , Alcohol Dehydrogenase/pharmacology , Biotransformation , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP2E1/pharmacology , Female , Glutathione S-Transferase pi , Glutathione Transferase/pharmacology , Humans , Isoenzymes/pharmacology , Life Style , Male , Middle Aged , Risk Factors , Sex Factors
13.
Mutat Res ; 506-507: 175-85, 2002 Sep 30.
Article in English | MEDLINE | ID: mdl-12351157

ABSTRACT

A case-control study of colorectal cancer, consisting of 157 cases and 380 controls matched by sex, ethnicity, decade of age and county of residence was performed to explore the associations between environmental exposure, metabolic polymorphisms and cancer risk. Participants were required to provide a blood sample, undergo caffeine phenotyping and complete an in-person interview that evaluated meat consumption, cooking methods and degree of doneness. A color atlas of foods cooked to different degrees of doneness was used to estimate food preparation techniques and food models were used to estimate serving portion sizes. Data was analyzed using a reference database of heterocyclic amine (HCA) exposure based on the food preferences chosen from the atlas. Data regarding individual food items cooked to different levels of doneness, as well as summary variables of foods and of food groups cooked to different degrees of doneness were also evaluated in a univariate analysis for association with colorectal cancer case status. Three measures of metabolic variation, hGSTA1 genotype, SULT1A1 genotype and the phenotype for CYP2A6 were also evaluated for possible association with colon cancer. While higher exposure to HCAs was strongly associated with colorectal cancer risk, increased consumption of five red meats cooked well done or very well done produced comparable odds ratios (OR) for colorectal cancer risk (OR=4.36, 95% CI 2.08-9.60) for the highest quartile of exposure. Similarly, individuals in the most rapid CYP2A6 phenotype quartile showed an odds ratio (OR = 4.18, 95% CI 2.03-8.90). The ORs for the low activity hGSTA1 and low activity SULT1A1 alleles were 2.0, 95% CI 1.0-3.7 and 0.6, 95% CI 0.3-1.1, respectively. Individual measures of specific HCAs provided little improvement in risk assessment over the measure of meat consumption, suggesting that exposure to other environmental or dietary carcinogens such as nitrosamines or undefined HCAs may contribute to colorectal cancer risk.


Subject(s)
Arylsulfotransferase , Carcinogens/toxicity , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/genetics , Imidazoles/metabolism , Meat Products/adverse effects , Quinoxalines/metabolism , Adult , Aged , Aged, 80 and over , Aryl Hydrocarbon Hydroxylases/genetics , Case-Control Studies , Colorectal Neoplasms/etiology , Cooking , Cytochrome P-450 CYP2A6 , Diet , Eating , Environmental Exposure , Female , Genotype , Glutathione Transferase/genetics , Humans , Imidazoles/pharmacology , Male , Middle Aged , Mixed Function Oxygenases/genetics , Polymerase Chain Reaction , Quinoxalines/pharmacology , Sulfotransferases/genetics
14.
Arch Biochem Biophys ; 403(2): 270-6, 2002 Jul 15.
Article in English | MEDLINE | ID: mdl-12139976

ABSTRACT

Glutathione S-transferase (GST) protein in gastrointestinal (GI) tracts of 16 organ donors, from whom all or substantial portions of the GI tract (stomach-colon) were available, was quantitated by HPLC and examined for interindividual variability/consistency of organ-specific patterns of expression. GSTP1, GSTA1, and GSTA2 were major components, and GSTM1 and GSTM3 were minor components. Consistent patterns of organ-specific expression were evident despite a high degree of interindividual variation of expression. GSTP1 was expressed throughout the GI tract and showed a decrease of expression from stomach to colon. GSTA1 and GSTA2 were expressed at high levels in duodenum and small intestine and expression decreased from proximal to distal small intestine. In contrast, GSTA1 and GSTA2 expression in colon and stomach of all subjects was low, particularly for colon where GSTA1 expression was 20- to 800-fold lower than that in corresponding small intestine. These consistent patterns of expression would suggest that compared to duodenum and small intestine, colon and to a lesser extent stomach always have low potential for GST-dependent detoxification of chemical carcinogens and are therefore at greater risk of genotoxic effects, particularly via substrates that are specific for GSTA1. This may be a factor in the greater susceptibility of stomach and colon to cancers compared to duodenum/small intestine.


Subject(s)
Digestive System/enzymology , Glutathione Transferase/metabolism , Adolescent , Adult , Cytosol/enzymology , Female , Gastric Mucosa/enzymology , Glutathione S-Transferase pi , Humans , Isoenzymes/metabolism , Male , Middle Aged , Organ Specificity , Reference Values
15.
Pharmacogenetics ; 12(4): 277-86, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12042665

ABSTRACT

By searching the human genome sequence database with human hGSTA1 and hGSTA4 cDNA sequences, we identified three PAC and one BAC clones covering more than 400 kilobases and containing the entire GST alpha gene cluster. The cluster consists of five genes: hGSTA1, hGSTA2, hGSTA3, hGSTA4 and hGSTA5, and seven pseudogenes that are distinguished as such by single-base and/or complete exon deletions. Using gene-specific probes we demonstrated that hGSTA1, hGSTA2 and hGSTA4 mRNAs are widely expressed in human tissues, whereas hGSTA3 mRNA appears to be a rare message subject to splicing defects. Although examination of the hGSTA5 gene sequence suggests that it is a functional gene, hGSTA5 mRNA could not be detected in human tissues we studied. hGSTA1 expression has been shown to be influenced by a genetic polymorphism, that consists of two alleles hGSTA1*A and hGSTA1*B, containing three linked base substitutions in the proximal promoter, at positions -567, -69 and -52. Constructs consisting of the luciferase gene controlled by variant hGSTA1 promoters showed differential expression when transfected into HepG2, GLC4 and Caco-2 cells: hGSTA1*A > hGSTA1*B. Directed mutagenesis for each base substitution indicated that the base change -52G>A was responsible for the differential promoter activity of hGSTA1*A and hGSTA1*B. The base at position -52 also altered binding of the ubiquitous transcription factor Sp1, as determined by gel shift analysis. Thus it may be postulated that hGSTA1 genotyping will be of importance to determine individual susceptibility to certain cancers or the efficacy of chemotherapeutics via its effect on hGSTA1 expression.


Subject(s)
Glutathione Transferase/genetics , Isoenzymes/genetics , Polymorphism, Genetic , Promoter Regions, Genetic/genetics , Amino Acid Sequence , Cell Line , Chromosomes, Human, Pair 6/genetics , DNA Primers/chemistry , Databases, Factual , Electrophoretic Mobility Shift Assay , Exons , Gene Expression , Genotype , Glutathione Transferase/metabolism , Humans , Introns , Isoenzymes/metabolism , Luciferases/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...