Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Leukemia ; 15(12): 1898-905, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11753611

ABSTRACT

There is continued controversy as to the sequential steps and mechanism(s) responsible for the in vivo acquisition of multiple mutations during neoplastic transformation. We investigated the in vivo clonality and mutational spectra of hypoxanthine-guanine phosphoribosyltransferase (HPRT) mutations in T cells from children with acute lymphocytic leukemia (ALL) to gain insight into the mutagenic mechanisms associated with leukemogenesis. We observed several instances of multiple, independent HPRT mutations accumulating in vivo in T cell receptor (TCR) gene defined clones that had undergone extensive pre- and/or post-thymic expansion following chemotherapy. In addition, we also detected the accumulation of multiple unique single mutations within distinct expanding post-thymic T cell clones. This pattern of clonally restricted hypermutability is compatible with extensive cell proliferation and selection alone without postulating genomic instability. These observations provide a paradigm for a continuum of cellular events that eventually results in the clonal accumulation of mutations in selected populations of cells in vivo and may provide insight into the primary genetic events associated with leukemogenesis, as well as the development of second malignancies and drug resistance following chemotherapy.


Subject(s)
Cell Transformation, Neoplastic/genetics , Leukemia/genetics , Mutation , T-Lymphocytes/pathology , Adolescent , Adult , Amino Acid Sequence , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cell Division/physiology , Cell Lineage , Child , Child, Preschool , Clone Cells/enzymology , Clone Cells/metabolism , Clone Cells/pathology , DNA Mutational Analysis , Female , Genes, T-Cell Receptor beta/genetics , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Leukemia/enzymology , Leukemia/etiology , Lymphocyte Activation/genetics , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , T-Lymphocytes/enzymology , T-Lymphocytes/metabolism
2.
Cancer Res ; 61(8): 3432-8, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11309304

ABSTRACT

Recent studies have brought to the forefront the importance of somatic mutations during human fetal development and malignant transformation in children, specifically leukemia. Therefore, a better understanding of the frequency and mutational spectrum of spontaneous in utero mutations is essential for understanding the genetic mechanisms associated with pediatric malignancies. Previously we reported that the frequency of somatic mutations during the late stages of fetal development was dependent on both gestational age and gender. Here we present the hypoxanthine-guanine phosphoribosyltransferase (HPRT) reporter gene mutational spectra analysis for 60 T-cell mutant isolates from the umbilical cord blood of preterm newborns to gain insight into background mutational events during the late stages of fetal development. Logistic regression analyses showed a significant increase in HPRT deletions mediated by V(D)J recombinase in preterm newborns compared with full-term newborns (P = 0.009). A comparative analysis of deletion mutations also revealed that V(D)J recombinase-mediated HPRT deletions increased with decreasing gestational age (P = 0.012) and were significantly higher in females than males of the same developmental status (P = 0.031). Developmental and gender-specific differences in HPRT deletions mediated by V(D)J recombinase provide insight into the gender-specific differences seen in infant leukemia.


Subject(s)
DNA Nucleotidyltransferases/genetics , Gene Deletion , Hypoxanthine Phosphoribosyltransferase/genetics , Infant, Premature/physiology , Base Sequence , Chromosome Breakage , DNA Mutational Analysis , Female , Fetal Blood/cytology , Fetal Blood/enzymology , Fetal Blood/physiology , Gestational Age , Humans , Infant, Newborn , Infant, Premature/blood , Logistic Models , Male , Molecular Sequence Data , Mutation , Sex Factors , T-Lymphocytes/enzymology , T-Lymphocytes/physiology , VDJ Recombinases
3.
Radiats Biol Radioecol ; 40(5): 529-34, 2000.
Article in English | MEDLINE | ID: mdl-11130945

ABSTRACT

We utilize T-cell HPRT mutations to monitor exposure to environmental mutagens in siblings of children who have developed cancer at a persistently high rate in Toms River, New Jersey, U.S.A. A preliminary epidemiological study has found a statistically-significant association between drinking public water (by pregnant mother or infant) and subsequent risk for childhood cancer. Three potential sources of mutagenic exposures in Toms River may have increased the rate of carcinogenic initiation significantly in children: 1. Benzidine-based, other azo dye and anthraquinone dye wastes released by Ciba-Geigy, 2. Styrene-acrylonitrile (SAN) trimer and other plastic wastes of Union Carbide, and 3. Radium-224, present in unusually high concentrations in the Cohansey aquifer. Specific patterns of HPRT mutations are utilized to distinguish these various potential sources of carcinogenic exposures in the drinking water of families with childhood cancer and to differentiate chemically or radiologically induced cancers from those which occur spontaneously.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/genetics , Neoplasms/chemically induced , Water Pollutants, Chemical/toxicity , Water Supply/standards , Child , Environmental Monitoring , Epidemiological Monitoring , Female , Genetic Markers , Humans , Neoplasms/epidemiology , Neoplasms/genetics , Neoplasms, Radiation-Induced/genetics , New Jersey/epidemiology , Pregnancy , Radium/toxicity , Water Pollutants, Radioactive/toxicity
4.
Science ; 288(5465): 514-7, 2000 Apr 21.
Article in English | MEDLINE | ID: mdl-10775110

ABSTRACT

T cells from patients who had received chemotherapy for B-lineage acute lymphocytic leukemia were studied to determine whether genetic instability, a principal characteristic of cancer cells, can also occur in nonmalignant cells. Consistent with expectations for a genetic instability phenotype, multiple mutations were detected in the hypoxanthine-guanine phosphoribosyltransferase (HPRT) reporter gene in independently isolated mutant T cells expressing identical rearranged T cell receptor beta (TCRbeta) gene hypervariable regions. These results indicate that cancer treatment can lead to genetic instability in nonmalignant cells in some individuals. They also suggest a mechanistic paradigm for the induction of second malignancies and drug resistance.


Subject(s)
Antineoplastic Agents/therapeutic use , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/genetics , Genes, T-Cell Receptor , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , T-Lymphocytes , Adolescent , Adult , Alleles , Burkitt Lymphoma/blood , Cell Lineage , Cell Transformation, Neoplastic , Child , Child, Preschool , Clone Cells , Drug Resistance, Neoplasm , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Genes, Reporter , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Infant , Neoplasms, Second Primary/etiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Recurrence
5.
Mutat Res ; 429(2): 249-59, 1999 Oct 19.
Article in English | MEDLINE | ID: mdl-10526209

ABSTRACT

Perinatal treatment with 3'-azido-3'-deoxythymidine (AZT) has been found to reduce the rate of maternal-infant transmission of HIV; however, AZT is genotoxic in mammalian cells in vitro and induces tumors in the offspring of mice treated in utero. The purpose of the present study was to investigate the relationships between incorporation of AZT into DNA, and the frequency and spectrum of mutations at the HPRT locus of the human lymphoblastoid cell line, TK6, following in vitro exposures to AZT. Cells were cultured in medium containing 0 or 300 microM AZT for 1, 3, or 6 day(s) (n = 5/group). The effects of exposure duration on incorporation of AZT into DNA and HPRT mutant frequency were determined using an AZT radioimmunoassay and a cell cloning assay, respectively. AZT accumulated in DNA in a supralinear manner, approaching a plateau at 6 days of treatment (101.9 +/- 14.7 molecules AZT/10(6) nucleotides). After 3 days of AZT exposure, HPRT mutant frequency was significantly increased (1.8-fold, p = 0.016) compared to background (mutant frequency = 3.78 x 10(-6)). Multiplex PCR amplification of genomic DNA was used to determine the frequency of exon deletions in HPRT mutant clones from untreated cells versus AZT-treated cells. Molecular analyses of AZT-induced mutations revealed a significant difference in the frequency of total gene deletions (44/120 vs. 18/114 in controls, p = 0.004 by the Mann-Whitney U-statistic). In fact, the Chi-square test of homogeneity demonstrate that the differences between the control and AZT-treatment groups is attributed mainly to this increase in total gene deletion mutations (p = 0.00001). These data indicate that the primary mechanism of AZT mutagenicity in human TK6 cells is through the production of large deletions which occur as a result of AZT incorporation into DNA and subsequent chain termination. The data imply that perinatal chemoprophylaxis with AZT may put children of HIV-infected women at potential risk for genetic damage.


Subject(s)
Anti-HIV Agents/toxicity , Hypoxanthine Phosphoribosyltransferase/genetics , Zidovudine/toxicity , Cell Line , Cell Survival/drug effects , Dideoxynucleosides/toxicity , Gene Deletion , Humans , Mutagenicity Tests , Point Mutation , Polymerase Chain Reaction , Sequence Deletion , Time Factors
6.
Clin Immunol ; 91(1): 61-7, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10219255

ABSTRACT

Somatic mutant frequencies (Mf) were determined using the HPRT T-cell cloning assay of peripheral blood T-lymphocytes from 14 children with juvenile onset dermatomyositis (JDM). Serologic parameters, specifically muscle enzyme determinations in JDM subjects, were correlated with residual lnMf (delta) in these patients to compare T-cell activation with clinical parameters associated with JDM. In addition TCR analysis was performed to determine T-cell proliferation and clonality on 12 HPRT mutant isolates from two individuals with JDM. Statistically significant correlations were found between residual lnMf and the following serologic parameters: aldolase (r = 0.771, P = 0.015); CPK (r = 0.602, P = 0.023); and SGOT (r = 0.656, P = 0.011) in children with JDM. In addition, identical TCR gene rearrangements were identified in 86 and 40% of the HPRT mutant isolates from the two patient samples analyzed, which is a significantly higher level of clonality than the 10-15% expected in normal individuals. These data suggest that determining HPRT Mf can be a useful antigen-independent method of selecting clonally expanding T-lymphocytes in autoimmune disease where relevant antigens are unknown. Future analysis of HPRT mutant isolates from children with active myositis may increase our understand of the activated T-cells involved in this disease.


Subject(s)
Dermatomyositis/genetics , Dermatomyositis/immunology , Hypoxanthine Phosphoribosyltransferase/genetics , Mutation , T-Lymphocytes/immunology , Adolescent , Age of Onset , Aspartate Aminotransferases/blood , Child , Child, Preschool , Colony-Forming Units Assay , Creatine Kinase/blood , Dermatomyositis/enzymology , Female , Fructose-Bisphosphate Aldolase/blood , Gene Frequency , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Humans , Lymphocyte Activation , Male
7.
Proc Natl Acad Sci U S A ; 96(2): 586-91, 1999 Jan 19.
Article in English | MEDLINE | ID: mdl-9892677

ABSTRACT

Limited information is available regarding the frequency, spectrum, and clinical relevance of somatic mutations in the developing fetus. The goal of this study was to determine somatic mutant frequencies (Mfs) at the hypoxanthine phosphoribosyltransferase (HPRT) reporter gene in cord blood T lymphocytes from preterm infants to gain insight into in utero mutational events. Mf determinations were made by using the HPRT T cell cloning assay on cord blood samples from 52 preterm infants. Natural logarithm Mfs (lnMfs) from preterm infants were compared with results from our database for full-term infants. Our analysis revealed higher lnMfs in cord blood T lymphocytes from preterm compared with full-term infants (P = 0.008). In addition, preterm females had significantly higher lnMfs compared with full-term females (P < 0.001), whereas preterm males were found to have significantly lower lnMfs than preterm females (P = 0.005). Regression analyses also demonstrate a significant relationship between lnMf and gestational age for preterm females that does not exist for preterm males. These results demonstrate the gender-specific association between Mf and age in humans.


Subject(s)
Fetal Blood/enzymology , Hypoxanthine Phosphoribosyltransferase/genetics , Mutation/genetics , Obstetric Labor, Premature/genetics , Sex , Female , Fetus/metabolism , Gestational Age , Humans , Infant, Newborn , Male , Plants, Toxic , Pregnancy , Regression Analysis , Smoking/genetics , T-Lymphocytes/enzymology , Nicotiana/genetics
8.
Nat Med ; 4(10): 1144-51, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9771747

ABSTRACT

We have investigated the molecular effects of passive maternal cigarette exposure in a newborn population and consider the possible implications of the observed genetic changes in the development of neoplastic diseases in children. We present a distribution analysis of somatic mutational events in a reporter gene, HPRT, in cord blood T lymphocytes from newborns after transplacental exposure to cigarette smoke. Analysis of 30 HPRT mutant isolates from 12 newborn infants born to mothers with no evidence of environmental exposure to cigarette smoke and 37 HPRT mutant isolates from 12 infants born to mothers exposed to passive cigarette smoke showed a significant difference in the HPRT mutational spectrum in those exposed in utero to cigarette smoke. The most notable change was an increase in 'illegitimate' genomic deletions mediated by V(D)J recombinase, a recombination event associated with hematopoietic malignancies in early childhood. Recent epidemiological studies of maternal and paternal cigarette smoke exposure and childhood cancers may need to be re-interpreted, given these results.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/genetics , Maternal Exposure , Mutagenesis , T-Lymphocytes/pathology , Tobacco Smoke Pollution/adverse effects , Base Sequence , Cloning, Molecular , Cotinine/blood , DNA Nucleotidyltransferases , Female , Fetal Blood/cytology , Humans , Infant, Newborn , Male , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Deletion , VDJ Recombinases
10.
Mutat Res ; 403(1-2): 35-43, 1998 Jul 17.
Article in English | MEDLINE | ID: mdl-9726004

ABSTRACT

People with Down syndrome are 10-30 fold more likely to develop leukemia than the normal population. To date, little is known regarding the molecular mechanisms underlying this phenomenon. We have previously demonstrated that the spontaneous somatic mutant frequency (Mf) at a reporter gene, hypoxanthine-guanine phosphoribosyl transferase (HPRT), from a normal population showed a strict age dependency with an exponential increase in Mf from birth to late adolescents with a subsequent linear 2-5% increase per year in adults. In this study, we compared HPRT Mf in children and adults with Down syndrome using the HPRT T-cell cloning assay. We determined the Mf at the HPRT locus in 27 subjects with Down syndrome from ages 6 months to 53.4 years. Results demonstrated that background somatic Mf at the HPRT locus in children and adults with Down syndrome are not dependent on age as seen in a normal control population. Results also show that adults with Down syndrome have a significantly lower Mf than normal adults, and that children with Down syndrome have a significantly higher Mf than normal children, although the latter appears to be due to a decreased cloning efficiency (CE). These observations demonstrate that the frequency of spontaneous somatic mutations in children and adults with Down syndrome are atypical compared to normal controls, and suggest that the genetic mechanisms associated with background somatic mutational events in children and adults with Down syndrome may be different.


Subject(s)
Down Syndrome/enzymology , Down Syndrome/genetics , Hypoxanthine Phosphoribosyltransferase/genetics , Mutation , Adolescent , Adult , Age Factors , Case-Control Studies , Child , Child, Preschool , DNA Damage , DNA Repair , Down Syndrome/complications , Female , Genes, Reporter , Humans , Infant , Leukemia/etiology , Leukemia/genetics , Male , Middle Aged , T-Lymphocytes/enzymology
11.
Mutat Res ; 377(1): 115-23, 1997 Jun 09.
Article in English | MEDLINE | ID: mdl-9219586

ABSTRACT

We utilized the hprt T-cell cloning assay to prospectively determined the somatic mutant frequency at the hprt locus of fetal T-lymphocytes exposed in utero to maternal active and passive cigarette smoke. In addition, a maternal questionnaire was administered to evaluate a number of social and medical parameters that may effect hprt mutant frequency. Newborn cord blood plasma cotinine levels were determined on all subjects to compare in utero tobacco metabolite levels with maternal smoking histories. A total of 63 newborns were enrolled and placed into four groups: Group I (n = 21), newborns whose mothers had no history of active or passive cigarette exposure during the pregnancy; Group II (n = 12), newborns whose mothers actively smoked cigarettes throughout the pregnancy; Group III (n = 8), newborns whose mothers actively smoked cigarettes during first trimester only; and Group IV (n = 22), newborns whose mothers were exposed only to passive cigarette smoke. Our analysis showed no statistically significant difference in hprt mutation frequency between any of the four groups. A significant increase in plasma cord blood cotinine was detected in Group II, newborns whose mothers were active cigarette smokers throughout the pregnancy. Our data indicate that exposure to active and passive maternal cigarette smoke in utero does not result in a significant increase in somatic mutant frequency as determined by the hprt T-cell cloning assay.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/genetics , Mutation , Prenatal Exposure Delayed Effects , Smoking/adverse effects , Tobacco Smoke Pollution/adverse effects , Clone Cells , Cotinine/blood , Female , Fetal Blood/chemistry , Humans , Infant, Newborn , Male , Mothers , Pregnancy , Prospective Studies , Surveys and Questionnaires , T-Lymphocytes
12.
Cancer Res ; 56(6): 1405-12, 1996 Mar 15.
Article in English | MEDLINE | ID: mdl-8640832

ABSTRACT

Somatic mutations in the hypoxanthine-guanine phosphoribosyltransferase (hprt) gene are rare occurrences in T-lymphocytes of normal individuals. Lacking pathogenic significance, these events can serve as reporters for assessing environmental genotoxicity. The present molecular analyses of hprt mutations arising spontaneously in normal children show that 30-35% of the genomic hprt changes in children under 5 years of age have approximately 20 Kb deletions encompassing exons 2 and 3. The frequency of these specific changes are dramatically decreased in older children. Sequence analysis of these deletion breakpoint and joining regions reveal the molecular hallmarks of V(D)J recombinase-mediated recombination events. This early childhood hprt mutational spectrum is quite distinct from the adult background spectrum but similar to that reported previously for newborns, as determined in lymphocytes from placental cord blood. The present study also demonstrates that definition of sequences in the hprt deletion joining regions that are analogous to the N-nucleotide insertion hypervariable regions of rearranged T-cell receptor genes allows the same identification of in vivo clonality of mutants as does analysis of the T-cell receptor gene rearrangements themselves. These methods reveal an in vivo clonal amplification of a V(D)J recombinase-mediated hprt mutant clone in one child in the present study. This newly found age-frequency distribution of V(D)J recombinase-mediated hprt mutations correlates with the age-frequency distribution of childhood acute lymphocytic leukemia. A significant number of these malignancies, including acute T-cell leukemia, are also characterized by V(D)J recombinase-mediated recombinations but in critical regions of the genome. hprt, therefore, captures a pathogenic mutagenic mechanism as a harmless mistake which, when it occurs in other genetic regions, may result in malignancy.


Subject(s)
DNA Nucleotidyltransferases/physiology , Exons/genetics , Hypoxanthine Phosphoribosyltransferase/genetics , Mutation/genetics , T-Lymphocytes , Adolescent , Age Factors , Base Sequence , Child , Child, Preschool , Gene Deletion , Humans , Infant , Infant, Newborn , Introns/genetics , Male , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Analysis, DNA , Translocation, Genetic , VDJ Recombinases
13.
Mutat Res ; 313(2-3): 227-47, 1994.
Article in English | MEDLINE | ID: mdl-7523908

ABSTRACT

In this paper, we have compared mutant frequency data at the hprt locus in circulating T-lymphocytes from four large datasets obtained in the UK (Sussex), the USA (Vermont), France (Paris) and The Netherlands (Leiden). In total, data from > 500 non-exposed individuals ranging in age from newborns (cord blood samples) to > 80 years old have been included in the analysis. Based on raw data provided by the four laboratories, a model is presented for the analysis of mutant frequency estimations for population monitoring. For three of the laboratories, a considerable body of data was provided on replicate estimates of mutant frequency from single blood samples, as well as estimates from repeat blood samples obtained over a period of time from many of the individual subjects. This enabled us to analyse the sources of variation in the estimation of mutant frequency. Although some variation was apparent in the results from the four laboratories, overall the data were in general agreement. Thus, in all laboratories, cellular cloning efficiency of T-cells was generally high (> 30%), although in each laboratory considerable variation between experiments and subjects was seen. Mutant frequency per clonable T-cell was in general found to be inversely related to cloning efficiency. With the exception of a few outliers (which are to be expected), mutant frequencies at this locus were in the same range in each dataset; no effect of subject gender was found, but an overall clear age effect was apparent. When log mutant frequency was analysed vs log (age + 0.5) a consistent trend from birth to old age was seen. In contrast, the effect of the smoking habit did differ between the laboratories, there being an association of smoking with a significant increase in mutant frequency in the Sussex and Leiden datasets, but not in those from the Vermont or Paris datasets. Possible reasons for this are discussed. One of the objectives of population monitoring is an ability to detect the effect of accidental or environmental exposure to mutagens and carcinogens among exposed persons. The large body of data from non-exposed subjects we have analysed in this paper has enabled us to estimate the size of an effect that could be detected, and the number of individuals required to detect a significant effect, taking known sources of variation into account.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Environmental Exposure , Hypoxanthine Phosphoribosyltransferase/genetics , Mutation , T-Lymphocytes/immunology , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Breast Neoplasms/genetics , Child , Child, Preschool , Databases, Factual , Environmental Monitoring , France , Humans , Infant , Infant, Newborn , Middle Aged , Netherlands , Nurses , Reference Values , Smoking/genetics , United Kingdom , United States
14.
Mutat Res ; 308(2): 223-31, 1994 Jul 16.
Article in English | MEDLINE | ID: mdl-7518049

ABSTRACT

Somatic cell mutant frequencies at the hprt locus of the X-chromosome were measured with the T-lymphocyte cloning assay in a healthy pediatric population. Assays were performed on 49 subjects (29 males and 20 females) ranging in age from 0.08 to 15.2 years. A statistical analysis of the thioguanine-resistant (TGr) mutant frequency (MF), unselected cloning efficiency (CE) and age was performed using data obtained in this study and those previously obtained in our laboratory on 66 newborn umbilical cord blood samples and 230 adult blood samples. For statistical comparisons pediatric subjects were divided into 4 groups. Group I included cord blood samples (age 0 years); Group II were subjects between 0 and 5 years; Group III were between 6 and 11 years and Group IV were between 12 and 17 years. The ln MF of Groups I and II were significantly lower than Groups III and IV (p < 0.05). The mean ln MF for each of Groups I-IV was significantly lower than the adult value. The cloning efficiency for Group I was significantly lower than that for Groups II-IV and adults. The relationships among the ln MF, unselected CE and age were expressed by the equations: ln (MF) = 0.945 -2.453 CE (p < 0.001) and ln (MF) = 0.114 + 0.063 age (p 0.004). The slope coefficients for unselected CE and age were significantly different from adults (p < 0.05). Regression analysis of combined data from Groups I-IV and adults were performed using both age and unselected CE as well as terms to reflect differences in their relationships with ln MF in adults and children. The results showed that the intercept and the age coefficients differ significantly for children and adults after adjustment for CE and yielded the following equations: ln (MF) = 0.548 -1.676 CE + 0.075 age, (Groups I-IV) and ln (MF) = 2.263 -1.676 CE + 0.014 age (adults). An alternative statistical model using ln (age ), ln (MF) = 0.381 -1.767 CE + 0.673 ln (age + 1), (p < 0.001), describes the rapid increase in MF with age that levels off in late adolescence. These findings demonstrate the changing influence of age on mutant frequency in the pediatric population as compared to the adult populations. These studies also illustrate that the increase in background somatic mutant frequencies at the hprt locus in T-lymphocytes is not linear from birth to adolescence and is significantly different from that seen in the adult population.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Aging/genetics , Hypoxanthine Phosphoribosyltransferase/genetics , Models, Statistical , Mutation , Adolescent , Adult , Cell Division , Cell Survival , Child , Child, Preschool , Clone Cells , Drug Resistance/genetics , Female , Gene Frequency , Humans , Infant , Infant, Newborn , Male , Reference Values , Regression Analysis , T-Lymphocytes/physiology , Thioguanine/pharmacology , X Chromosome
15.
Appl Environ Microbiol ; 54(6): 1498-503, 1988 Jun.
Article in English | MEDLINE | ID: mdl-2843094

ABSTRACT

Pseudomonas putida PpF1 degrades toluene through cis-toluene dihydrodiol to 3-methylcatechol. The latter compound is metabolized through the well-established meta pathway for catechol degradation. The first four steps in the pathway involve the sequential action of toluene dioxygenase (todABC1C2), cis-toluene dihydrodiol dehydrogenase (todD), 3-methylcatechol 2,3-dioxygenase (todE), and 2-hydroxy-6-oxo-2,4-heptadienoate hydrolase (todF). The genes for these enzymes form part of the tod operon which is responsible for the degradation of toluene by this organism. A combination of transposon mutagenesis of the PpF1 chromosome, as well as analysis of cloned chromosomal fragments, was used to determine the physical order of the genes in the tod operon. The genes were determined to be transcribed in the order todF, todC1, todC2, todB, todA, todD, todE.


Subject(s)
Dioxygenases , Operon , Pseudomonas/metabolism , Toluene/metabolism , Biodegradation, Environmental , Chemical Phenomena , Chemistry , Cloning, Molecular , DNA Transposable Elements , Genetic Complementation Test , Hydrolases/genetics , Hydrolases/metabolism , Mutation , Oxidoreductases/genetics , Oxidoreductases/metabolism , Oxygenases/genetics , Oxygenases/metabolism , Plasmids , Pseudomonas/enzymology , Pseudomonas/genetics
16.
J Bacteriol ; 160(3): 1003-9, 1984 Dec.
Article in English | MEDLINE | ID: mdl-6501223

ABSTRACT

Pseudomonas putida PpF1 degraded toluene via a dihydrodiol pathway to tricarboxylic acid cycle intermediates. The initial reaction was catalyzed by a multicomponent enzyme, toluene dioxygenase, which oxidized toluene to (+)-cis-1(S),2(R)-dihydroxy-3-methylcyclohexa-3,5-diene (cis-toluene dihydrodiol). The enzyme consisted of three protein components: NADH-ferredoxintol oxidoreductase (reductasetol), ferredoxintol, and a terminal oxygenase which is an iron-sulfur protein (ISPtol). Mutants blocked in each of these components were isolated after mutagenesis with nitrosoguanidine. Mutants occurred as colony morphology variants when grown in the presence of toluene on indicator plates containing agar, mineral salts, a growth-supporting nutrient (arginine), 2,3,5-triphenyltetrazolium chloride (TTC), and Nitro Blue Tetrazolium (NBT). Under these conditions, wild-type colonies appeared large and red as a result of TTC reduction. Colonies of reductasetol mutants were white or white with a light blue center, ferredoxintol strains were light blue with a dark blue center, and strains that lacked ISPtol gave dark blue colonies. Blue color differences in the mutant colonies were due to variations in the extent of NBT reduction. Strains lacking all three components appeared white. Toluene dioxygenase mutants were characterized by assaying toluene dioxygenase activity in crude cell extracts which were complemented with purified preparations of each protein component. Between 40 and 60% of the putative mutants selected from the NBT-TTC indicator plates were unable to grow with toluene as the sole source of carbon and energy. This method should prove extremely useful in isolating mutants in other multicomponent oxygenase enzyme systems.


Subject(s)
Mutation , Oxygenases/genetics , Pseudomonas/genetics , Genetic Complementation Test , Pseudomonas/enzymology , Pseudomonas/isolation & purification , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...