Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where John E. French is active.

Publication


Featured researches published by John E. French.


Mutation Research\/reviews in Genetic Toxicology | 1996

Evaluation of transgenic mouse bioassays for identifying carcinogens and noncarcinogens.

Raymond W. Tennant; Judson W. Spalding; John E. French

Data supporting the use of transgenic lines to identify carcinogens and noncarcinogens are thus far based on a limited number of chemicals for which there are also long-term bioassay results in rats and/or mice. Six chemicals have been tested in the heterozygous p53-deficient mice and 13 in the Tg.AC line. The results show that the p53def responds rapidly to mutagenic carcinogens and the Tg.AC responds rapidly to both mutagenic and nonmutagenic carcinogens. Neither transgenic line responded to the noncarcinogens that were tested. The p53def line failed to respond to two nonmutagenic carcinogens (N-methyloacrylamide and reserpine), the Tg.AC line failed to respond to ethyl acrylate, a nonmutagenic chemical that induced tumors of the forestomach when administered by gavage, and to triethanolamine that caused an increase in hepatocellular tumors in B6C3F1 mice via skin painting. Both of the latter chemicals are examples of highly specific responses related to either route of administration or to strain susceptibility. Further efforts to evaluate the range of chemicals to which these transgenic lines respond are currently in progress.


Toxicologic Pathology | 2001

p53 +=¡ Hemizygous Knockout Mouse: Overview of Available Data

Richard D. Storer; John E. French; Joseph K. Haseman; Gerald Hajian; Edmund K. Legrand; Gerald G. Long; Lori Mixson; Ricardo Ochoa; John E. Sagartz; Keith A. Soper

The performance of the p53 +/- transgenic (knockout) mouse model was evaluated through review of the data from 31 short-term carcinogenicity studies with 21 compounds tested as part of the International Life Sciences Institutes (ILSI) Alternatives to Carcinogenicity Testing (ACT) project, together with data from other studies which used comparable protocols. As expected based on the hypothesis for the model, a significant number (12/16 or 75%) of the genotoxic human and/or rodent carcinogens tested were positive and the positive control, p-cresidine, gave reproducible responses across laboratories (18/19 studies positive in bladder). An immunosuppressive human carcinogen, cyclosporin A, was positive for lymphomas but produced a similar response in wild type mice. Two hormones that are human tumorigens, diethylstilbestrol and 17β-estradiol, gave positive and equivocal results, respectively, in the pituitary with p53-defi cient mice showing a greater incidence of proliferative lesions than wild type. None of the 22 nongenotoxic rodent carcinogens that have been tested produced a positive response but 2 compounds in this category, chloroform and diethylhexylphthalate, were judged equivocal based on effects in liver and kidney respectively. Four genotoxic noncarcinogens and 6 nongenotoxic, noncarcinogens were also negative. In total (excluding compounds with equivocal results), 42 of 48 compounds or 88% gave results that were concordant with expectations. The technical lessons learned from the ILSI ACT-sponsored testing in the p53+/- model are discussed.


Toxicologic Pathology | 1997

Phenolphthalein Induces Thymic Lymphomas Accompanied by Loss of the p53 Wild Type Allele in Heterozygous p53-Deficient (±) Mice

June K. Dunnick; Jerry F. Hardisty; Ronald A. Herbert; John C. Seely; E. Marianna Furedi-Machacek; Julie F. Foley; Gregory D. Lacks; Stanley Stasiewicz; John E. French

Epidemiology studies have indicated that many human cancers are influenced by environmental factors. Genetically altered mouse model systems offer us the opportunity to study the interaction of chemicals with genetic predisposition to cancer. Using the heterozygous p53-deficient (±) mouse, an animal model carrying one wild type p53 gene and one p53 null allele, we studied the effects of phenolphthalein on tumor induction and p53 gene alterations. Earlier studies showed that phenolphthalein caused carcinogenic effects in Fisher 344 rats and B6C3F, mice after a 2-yr dosing period (Dunnick and Hailey, Cancer Res. 56: 4922-4926, 1996). The p53 (±) mice received phenolphthalein in the feed at concentrations of 200, 375, 750, 3,000, or 12,000 ppm (approximately 43, 84, 174, 689, or 2,375 mg/kg body weight/day or 129, 252, 522, 2,867, or 7,128 mg/m2 body surface area/day) for up to 6 mo. A target organ cancer site that accumulated p53 protein in the B6C3F, mouse (i.e., thymic lymphoma) was also a target site for cancer in the p53 (±) mouse. In the p53 (±) mouse, treatment-related atypical hyperplasia and malignant lymphoma of thymic origin were seen in the control and dosed groups at a combined incidence of 0, 5, 5, 25, 100, and 95%, respectively. Twenty-one of the thymic lymphomas were examined for p53 gene changes, and all showed loss of the p53 wild type allele. Chemical-induced ovarian tumors in the B6C3F, mouse showed no evidence for p53 protein accumulation and did not occur in the p53 (±) mouse. The p53-deficient (±) mouse model responded to phenolphthalein treatment with a carcinogenic response in the thymus after only 4 mo of dosing. This carcinogenic response took 2 yr to develop in the conventional B6C3F, mouse bioassay. The p53-deficient (±) mouse is an important model for identifying a carcinogenic response after short-term (<6 mo) exposures. Our studies show that exposure to phenolphthalein combined with a genetic predisposition to cancer can potentiate the carcinogenic process and cause p53 gene alterations, a gene alteration found in many human cancers.


Toxicologic Pathology | 2001

The Nature of the Heterozygous Trp53 Knockout Model for Identifi cation of Mutagenic Carcinogens

John E. French; Richard D. Storer; Lawrence A. Donehower

The heterozygous Trp 53 null allele C57BL/6 (N5) mouse is susceptible to the rapid development of neoplasia by mutagenic carcinogens relative to control strains. This mouse model of chemical carcinogenesis demonstrates 1) dose-related rapid induction of tumors (26 wks), 2) multiple sites of carcinogen-specifi c tissue susceptibility, and 3) carcinogen-induced loss of heterozygosity involving the Trp53 wild-type allele or a p53 haploinsufficiency permitting mutation of other critical protooncogenes and/or inactivation of tumor suppressor genes driving tumorigenesis. Demonstration of mutation or loss ofheterozygosity involving the Trp53 locus is consistent with a common finding in human cancers and supports extrapolation between rodents and humans. Using diverse experimental protocols, almost all mutagenic rodent carcinogens (including all mutagens that are carcinogenic to humans), but not nonmutagenic rodent carcinogens, induce tumors within 26 weeks of continuous exposure. These characteristics and results indicate that the mouse heterozygous for the Trp53 null allele may be of significant use for the prospective identification of mutagenic carcinogens of potential risk to human health.


Journal of Toxicology and Environmental Health | 1988

Evaluation of tissue disposition, myelopoietic, and immunologic responses in mice after long‐term exposure to nickel sulfate in the drinking water

M. P. Dieter; C. W. Jameson; A. N. Tucker; M. I. Luster; John E. French; H. L. Hong; G. A. Boorman

Female B6C3F1 mice were exposed to graded doses of nickel sulfate to determine a threshold response for myelotoxicity and immunotoxicity, and to identify which of the populations of lymphoreticular cells were most sensitive to the toxic effects of nickel. Animals were given free access to the chemical in the drinking water at 0, 1, 5, or 10 g/l for 180 d. Water consumption, blood and tissue nickel concentrations, body and organ weights, histopathology, immune responses, bone marrow cellularity and proliferation, and cellular enzyme activities were evaluated. There was no mortality. Mice in the 5-g/l and 10-g/l dose groups drank less water than controls; the responses measured in the 10-g/l group may have been due to a combination of dehydration and chemical toxicity. Decreases in body and organ weights were confined to mice in the 10-g/l dose group, except for the dose-related reductions in thymus weights. Blood nickel was measured at 4, 8, 16, and 23 wk of exposure. The mean blood nickel values showed increases between 4 and 8 wk that were proportional to time and dose; thereafter there was no substantial increase in blood nickel in any of the dose groups, except for an increase in the mean blood concentration in the 10-g/l group at 23 wk. The kidney was the major organ of nickel accumulation. The primary toxic effects of nickel sulfate were expressed in the myeloid system. There were dose-related decreases in bone marrow cellularity, and in granulocyte-macrophage and pluripotent stem-cell proliferative responses. In unfractionated bone marrow cells glucose-6-phosphate dehydrogenase enzyme activity from the hexose monophosphate shunt was more sensitive to nickel sulfate than were representative glycolytic or Krebs cycle enzymes, with 25-35% maximum inhibition at 5 g/l and 10 g/l. Aliquots of bone marrow cells were separated into enriched bands of lymphocytes, granulocyte-macrophages, and erythrocytes; enzyme inhibition that occurred in unfractionated bone marrow cell aliquots was only expressed after cell separation in the enriched granulocyte-macrophage cell population, suggesting that these committed stem cells were a primary target of nickel sulfate toxicity. There was one example of systemic immunotoxicity, reduction in the lymphoproliferative response to lipopolysaccharide, and it was regarded as secondary to the primary effect of nickel sulfate on the myeloid system, since this was the only significant change among a panel of seven immune parameters that were evaluated.


Environmental Health Perspectives | 2014

Diversity Outbred Mice Identify Population-Based Exposure Thresholds and Genetic Factors that Influence Benzene-Induced Genotoxicity

John E. French; Daniel M. Gatti; Daniel L. Morgan; Grace E. Kissling; Keith R. Shockley; Gabriel A. Knudsen; Kim G. Shepard; Herman C. Price; Deborah King; Kristine L. Witt; Lars C. Pedersen; Steven C. Munger; Karen L. Svenson; Gary A. Churchill

Background Inhalation of benzene at levels below the current exposure limit values leads to hematotoxicity in occupationally exposed workers. Objective We sought to evaluate Diversity Outbred (DO) mice as a tool for exposure threshold assessment and to identify genetic factors that influence benzene-induced genotoxicity. Methods We exposed male DO mice to benzene (0, 1, 10, or 100 ppm; 75 mice/exposure group) via inhalation for 28 days (6 hr/day for 5 days/week). The study was repeated using two independent cohorts of 300 animals each. We measured micronuclei frequency in reticulocytes from peripheral blood and bone marrow and applied benchmark concentration modeling to estimate exposure thresholds. We genotyped the mice and performed linkage analysis. Results We observed a dose-dependent increase in benzene-induced chromosomal damage and estimated a benchmark concentration limit of 0.205 ppm benzene using DO mice. This estimate is an order of magnitude below the value estimated using B6C3F1 mice. We identified a locus on Chr 10 (31.87 Mb) that contained a pair of overexpressed sulfotransferases that were inversely correlated with genotoxicity. Conclusions The genetically diverse DO mice provided a reproducible response to benzene exposure. The DO mice display interindividual variation in toxicity response and, as such, may more accurately reflect the range of response that is observed in human populations. Studies using DO mice can localize genetic associations with high precision. The identification of sulfotransferases as candidate genes suggests that DO mice may provide additional insight into benzene-induced genotoxicity. Citation French JE, Gatti DM, Morgan DL, Kissling GE, Shockley KR, Knudsen GA, Shepard KG, Price HC, King D, Witt KL, Pedersen LC, Munger SC, Svenson KL, Churchill GA. 2015. Diversity Outbred mice identify population-based exposure thresholds and genetic factors that influence benzene-induced genotoxicity. Environ Health Perspect 123:237–245; http://dx.doi.org/10.1289/ehp.1408202


Environmental Health Perspectives | 2007

Acquisition of apoptotic resistance in cadmium-transformed human prostate epithelial cells: Bcl-2 overexpression blocks the activation of JNK signal transduction pathway.

Wei Qu; Hengning Ke; Jingbo Pi; Daniel Broderick; John E. French; Mukta M. Webber; Michael P. Waalkes

Background We have recently shown that cadmium can induce malignant transformation of the human prostate epithelial cell line (RWPE-1) and that these cadmium-transformed prostate epithelial (CTPE) cells acquire apoptotic resistance concurrently with malignant phenotype. Objective The present study was designed to define the mechanism of acquired apoptotic resistance in CTPE cells. Methods Various molecular events associated with apoptosis were assessed in control and CTPE cells that were obtained after 8 weeks of continuous cadmium exposure. Results Compared with control, CTPE cells showed a generalized resistance to apoptosis induced by cadmium, cisplatin, or etoposide. Signal-regulated mitogen-activated protein kinases, extracellular signal-regulated kinases 1 and 2, c-Jun N-terminal kinases (JNK1 and JNK2), and p38 were phosphorylated in a cadmium concentration-dependent fashion in CTPE and control cells. However, phosphorylated JNK1/2 levels and JNK kinase activity were much lower in CTPE cells. The pro-apoptotic gene Bax showed lower transcript and protein levels, whereas the anti-apoptotic gene Bcl-2 showed higher levels in CTPE cells. The ratio of Bcl-2/Bax, a key determinant in apoptotic commitment, increased more than 4-fold in CTPE cells. In Bcl-2–transfected PT-67 cells, phosphorylated JNK1/2 levels were much lower after apoptogenic stimulus, and apoptosis induced by cadmium or etoposide was reduced compared with control. Mutation of tyrosine to serine at the 21st amino acid of the Bcl-2 protein BH4 domain resulted in a loss both of suppression of JNK1/2 phosphorylation and its anti-apoptotic function. Conclusions CTPE cells become resistant to apoptosis during malignant transformation, and disruption of the JNK pathway and Bcl-2 overexpression play important roles in this resistance. Bcl-2 BH4 domain is required for modulating JNK phosphorylation and anti-apoptotic function.


Cell Research | 2010

BCL2 inhibits cell adhesion, spreading, and motility by enhancing actin polymerization.

Hengning Ke; Vandy I. Parron; Jeff Reece; Jennifer Y. Zhang; Steven K. Akiyama; John E. French

BCL2 is best known as a multifunctional anti-apoptotic protein. However, little is known about its role in cell-adhesive and motility events. Here, we show that BCL2 may play a role in the regulation of cell adhesion, spreading, and motility. When BCL2 was overexpressed in cultured murine and human cell lines, cell spreading, adhesion, and motility were impaired. Consistent with these results, the loss of Bcl2 resulted in higher motility observed in Bcl2-null mouse embryonic fibroblast (MEF) cells compared to wild type. The mechanism of BCL2 regulation of cell adhesion and motility may involve formation of a complex containing BCL2, actin, and gelsolin, which appears to functionally decrease the severing activity of gelsolin. We have observed that the lysate from MCF-7 and NIH3T3 cells that overexpressed BCL2 enhanced actin polymerization in cell-free in vitro assays. Confocal immunofluorescent localization of BCL2 and F-actin during spreading consistently showed that increased expression of BCL2 resulted in increased F-actin polymerization. Thus, the formation of BCL2 and gelsolin complexes (which possibly contain other proteins) appears to play a critical role in the regulation of cell adhesion and migration. Given the established correlation of cell motility with cancer metastasis, this result may explain why the expression of BCL2 in some tumor cell types reduces the potential for metastasis and is associated with improved patient prognosis.


Toxicology and Applied Pharmacology | 1979

Saturable metabolism and the acute toxicity of 1,1-dichloroethylene

Melvin E. Andersen; John E. French; Michael L. Gargas; Robert A. Jones; Lawrence J. Jenkins

Abstract The toxic effects of single, oral doses of 1,1-dichloroethylene (1,1-DCE) are caused by its metabolites. When saturable, enzymatic production of a toxic intermediate regulates toxicity, the observed inhalation toxicity, at sufficiently high concentrations, will depend on the duration of exposure and be relatively independent of the ambient concentration. The inhalation toxicity of 1,1-DCE was examined in fasted male rats to ascertain its dependence on both 1,1-DCE concentration and the duration of exposure. Immature rats (100–150 g) were exposed to 200 ppm 1,1-DCE for durations up to 2 hr. Pentobarbital sleep times (PBST) were significantly increased after exposures as brief as 0.5 hr. Intubation of immature rats with 1,1-DCE produced extensive dilation and vacuolization of the endoplasmic reticulum (ER). While young rats were more susceptible to 1,1-DCE than older rats (> 200 g), extensive damage to the ER (the presumed site of 1,1-DCE metabolism) during exposure precluded their use in determining the inhalation dose-effect curves. With older rats, PBST were not increased during exposure. The concentration-mortality curve for 4-hr exposures increased sharply between 100 and 200 ppm, but was virtually flat (i.e., concentration independent) between 200 and 1000 ppm. The LT50 (the time of exposure required to kill half an exposed group at a given concentration) only varied between 4.1 and 2.4 hr as concentration increased from 200 to 1000 ppm. Failure of 1,1-DCE to adhere to a concentration × time relationship is further evidence of a role of saturable, enzymatic activation in the expression of its toxicity.


PLOS Genetics | 2015

A Multi-Megabase Copy Number Gain Causes Maternal Transmission Ratio Distortion on Mouse Chromosome 2

John P. Didion; Andrew P. Morgan; Amelia M.-F. Clayshulte; Rachel C. McMullan; Liran Yadgary; Petko M. Petkov; Timothy A. Bell; Daniel M. Gatti; James J. Crowley; Kunjie Hua; David L. Aylor; Ling Bai; Mark Calaway; Elissa J. Chesler; John E. French; Thomas R. Geiger; Terry J. Gooch; Theodore Garland; Alison H. Harrill; Kent W. Hunter; Leonard McMillan; Matt Holt; Darla R. Miller; Deborah A. O'Brien; Kenneth Paigen; Wenqi Pan; Lucy B. Rowe; Ginger D. Shaw; Petr Simecek; Patrick F. Sullivan

Significant departures from expected Mendelian inheritance ratios (transmission ratio distortion, TRD) are frequently observed in both experimental crosses and natural populations. TRD on mouse Chromosome (Chr) 2 has been reported in multiple experimental crosses, including the Collaborative Cross (CC). Among the eight CC founder inbred strains, we found that Chr 2 TRD was exclusive to females that were heterozygous for the WSB/EiJ allele within a 9.3 Mb region (Chr 2 76.9 – 86.2 Mb). A copy number gain of a 127 kb-long DNA segment (designated as responder to drive, R2d) emerged as the strongest candidate for the causative allele. We mapped R2d sequences to two loci within the candidate interval. R2d1 is located near the proximal boundary, and contains a single copy of R2d in all strains tested. R2d2 maps to a 900 kb interval, and the number of R2d copies varies from zero in classical strains (including the mouse reference genome) to more than 30 in wild-derived strains. Using real-time PCR assays for the copy number, we identified a mutation (R2d2WSBdel1) that eliminates the majority of the R2d2WSB copies without apparent alterations of the surrounding WSB/EiJ haplotype. In a three-generation pedigree segregating for R2d2WSBdel1, the mutation is transmitted to the progeny and Mendelian segregation is restored in females heterozygous for R2d2WSBdel1, thus providing direct evidence that the copy number gain is causal for maternal TRD. We found that transmission ratios in R2d2WSB heterozygous females vary between Mendelian segregation and complete distortion depending on the genetic background, and that TRD is under genetic control of unlinked distorter loci. Although the R2d2WSB transmission ratio was inversely correlated with average litter size, several independent lines of evidence support the contention that female meiotic drive is the cause of the distortion. We discuss the implications and potential applications of this novel meiotic drive system.

Collaboration


Dive into the John E. French's collaboration.

Top Co-Authors

Avatar

Raymond W. Tennant

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Judson W. Spalding

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Daniel M. Gatti

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Leena A. Nylander-French

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Raymond R. Tice

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Andrew P. Morgan

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Elissa J. Chesler

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

James J. Crowley

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

John P. Didion

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Liran Yadgary

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge