Network


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

Hotspot


Dive into the research topics where Diane Vannais is active.

Publication


Featured researches published by Diane Vannais.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Radiation risk to low fluences of particles may be greater than we thought

Hongning Zhou; Masao Suzuki; Gerhard Randers-Pehrson; Diane Vannais; Gang Chen; James E. Trosko; Charles A. Waldren; Tom K. Hei

Based principally on the cancer incidence found in survivors of the atomic bombs dropped in Hiroshima and Nagasaki, the International Commission on Radiation Protection (ICRP) and the United States National Council on Radiation Protection and Measurements (NCRP) have recommended that estimates of cancer risk for low dose exposure be extrapolated from higher doses by using a linear, no-threshold model. This recommendation is based on the dogma that the DNA of the nucleus is the main target for radiation-induced genotoxicity and, as fewer cells are directly damaged, the deleterious effects of radiation proportionally decline. In this paper, we used a precision microbeam to target an exact fraction (either 100% or ≤20%) of the cells in a confluent population and irradiated their nuclei with exactly one α particle each. We found that the frequencies of induced mutations and chromosomal changes in populations where some known fractions of nuclei were hit are consistent with non-hit cells contributing significantly to the response. In fact, irradiation of 10% of a confluent mammalian cell population with a single α particle per cell results in a mutant yield similar to that observed when all of the cells in the population are irradiated. This effect was significantly eliminated in cells pretreated with a 1 mM dose of octanol, which inhibits gap junction-mediated intercellular communication, or in cells carrying a dominant negative connexin 43 vector. The data imply that the relevant target for radiation mutagenesis is larger than an individual cell and suggest a need to reconsider the validity of the linear extrapolation in making risk estimates for low dose, high linear-energy-transfer (LET) radiation exposure.


Mutation Research | 1996

A low, adaptive dose of gamma-rays reduced the number and altered the spectrum of S1− mutants in human-hamster hybrid AL cells

Akiko M. Ueno; Diane Vannais; Daniel L. Gustafson; Jenise C. Wong; Charles A. Waldren

We examined the effects of a low, adaptive dose of 137Cs-gamma-irradiation (0.04 Gy) on the number and kinds of mutants induced in AL human-hamster hybrid cells by a later challenge dose of 4 Gy. The yield of S1- mutants was significantly less (by 53%) after exposure to both the adaptive and challenge doses compared to the challenge dose alone. The yield of hprt- mutants was similarly decreased. Incubation with cycloheximide (CX) or 3-aminobenzamide largely negated the decrease in mutant yield. The adaptive dose did not perturb the cell cycle, was not cytotoxic, and did not of itself increase the mutant yield above background. The adaptive dose did, however, alter the spectrum of S1- mutants from populations exposed only to the adaptive dose, as well as affecting the spectrum of S1- mutants generated by the challenge dose. The major change in both cases was a significant increase in the proportion of complex mutations compared to small mutations and simple deletions.


Radiation Research | 1996

Cellular and molecular analysis of mutagenesis induced by charged particles of defined linear energy transfer

Li X. Zhu; Charles A. Waldren; Diane Vannais; Tom K. Hei

Mutation induction by charged particles of defined linear energy transfer (LET) and gamma rays was scored using human-hamster hybrid AL cells. The LET values for charged particles accelerated at the Radiological Research Accelerator Facility ranged from 10 keV/microm protons to 150 keV/microm 4He ions. The induced mutant fractions at both the S1 and HGPRT loci were dependent on the dose and LET. In addition, for each dose examined, the mutant yield at the S1 locus was 30-60 fold higher than at the corresponding HGPRT locus. To determine whether the mutation spectrum was comparably dependent on dose and LET, independent S1- and HGPRT- mutants induced by 150 keV/microm 4He ions and gamma rays were isolated, and their DNA was analyzed by both Southern blotting and multiplex PCR methods. While the majority of radiation-induced mutants showed deletions of varying sizes, the relative percentage of large deletions was found to be related to both the dose and LET of the radiation examined. Using a mutation system that can detect multilocus changes, results of the present study show that radiation-induced chromosomal loss can be in the millions of base pairs.


Radiation and Environmental Biophysics | 1995

Heavy ion mutagenesis: linear energy transfer effects and genetic linkage.

Amy Kronenberg; S. Gauny; K. Criddle; Diane Vannais; Akiko M. Ueno; S. Kraemer; Charles A. Waldren

We have characterized a series of 69 independent mutants at the endogenoushprt locus of human TK6 lymphoblasts and over 200 independent S 1-deficient mutants of the humanxhamster hybrid cell line AL arising spontaneously or following low-fluence exposures to densely ionizing Fe ions (600 MeV/amu, linear energy transfer = 190 keV/µm). We find that large deletions are common. The entirehprt gene (>44 kb) was missing in 19/39 Fe-induced mutants, while only 2/30 spontaneous mutants lost the entirehprt coding sequence. When the gene of interest (S1 locus = M1C1 gene) is located on a nonessential human chromosome 11, multilocus deletions of several million base pairs are observed frequently. The S1 mutation frequency is more than 50-fold greater than the frequency ofhprt mutants in the same cells. Taken together, these results suggest that low-fluence exposures to Fe ions are often cytotoxic due to their ability to create multilocus deletions that may often include the loss of essential genes. In addition, the tumorigenic potential of these HZE heavy ions may be due to the high potential for loss of tumor suppressor genes. The relative insensitivity of thehprt locus to mutation is likely due to tight linkage to a gene that is required for viability.


Advances in Space Research | 1998

Analysis of mutant quantity and quality in human-hamster hybrid AL and AL-179 cells exposed to 137Cs-gamma or HZE-Fe ions.

Charles A. Waldren; Diane Vannais; R Drabek; Daniel L. Gustafson; S. Kraemer; M Lenarczyk; Amy Kronenberg; Tom K. Hei; A Ueno

We measured the number of mutants and the kinds of mutations induced by 137Cs-gamma and by HZE-Fe (56Fe [600 MeV/amu, LET = 190 KeV/micrometer) in standard AL human hamster hybrid cells and in a new variant hybrid, AL-179. We found that HZE-Fe was more mutagenic than 137Cs-gamma per unit dose (about 1.6 fold), but was slightly less mutagenic per mean lethal dose, DO, at both the S1 and hprt- loci of AL cells. On the other hand, HZE-Fe induced about nine fold more complex S1- mutants than 137Cs-gamma rays, 28% vs 3%. 137Cs-gamma rays induced about twice as many S1- mutants and hprt-mutants in AL-179 as in AL cells, and about nine times more of the former were complex, and potentially unstable kinds of mutations.


Radiation Research | 2001

Gamma-ray mutagenesis studies in a new human-hamster hybrid, A(L)CD59(+/-), which has two human chromosomes 11 but is hemizygous for the CD59 gene.

S. Kraemer; Diane Vannais; Amy Kronenberg; Akiko M. Ueno; Charles A. Waldren

Abstract Kraemer, S. M., Vannais, D. B., Kronenberg, A., Ueno, A. and Waldren, C. A. Gamma-Ray Mutagenesis Studies in a New Human–Hamster Hybrid, ALCD59+/–, which has Two Human Chromosomes 11 but is Hemizygous for the CD59 Gene. Radiat. Res. 156, 10–19 (2001). We have developed a human–CHO hybrid cell line, named ALCD59+/–, which has two copies of human chromosome 11 but is hemizygous for the CD59 gene and the CD59 cell surface antigen that it encodes. Our previous studies used the AL and ALC hybrids that respectively contain one or two sets of CHO chromosomes plus a single copy of human chromosome 11. The CD59 gene at 11p13.5 and the CD59 antigen encoded by it are the principal markers used in our mutagenesis studies. The hybrid ALCD59+/– contains two copies of human chromosome 11, only one of which carries the CD59 gene. The incidence of CD59 – mutants (formerly called S1–) induced by 137Cs γ rays is about fivefold greater in ALCD59+/– cells than in AL cells. Evidence is presented that this increase in mutant yield is due to the increased induction of certain classes of large chromosomal mutations that are lethal to AL cells but are tolerated in the ALCD59+/– hybrid. In addition, significantly more of the CD59 – mutants induced by 137Cs γ rays in ALCD59+/– cells display chromosomal instability than in AL cells. On the other hand, the yield of γ-ray-induced CD59 – mutants in ALCD59+/– cells is half that of the ALC hybrid, which also tolerates very large mutations but has only one copy of human chromosome 11. We interpret the difference in mutability as evidence that repair processes involving the homologous chromosomes 11 play a role in determining mutant yields. The ALCD59+/– hybrid provides a useful new tool for quantifying mutagenesis and shedding light on mechanisms of genetic instability and mutagenesis.


Somatic Cell and Molecular Genetics | 1999

Antigen S1, encoded by the MIC1 gene, is characterized as an epitope of human CD59, enabling measurement of mutagen-induced intragenic deletions in the AL cell system

Anne B. Wilson; David J. Seilly; Christine Willers; Diane Vannais; Megan McGraw; Charles A. Waldren; Tom K. Hei; Alexandra Davies

S1 cell membrane antigen is encoded by the MIC1 gene on human chromosome 11. This antigen has been widely used as a marker for studies in gene mapping or in analysis of mutagen-induced gene deletions/mutations, which utilized the human-hamster hybrid cell-line, AL-J1, carrying human chromosome 11. Evidence is presented here which identifies S1 as an epitope of CD59, a cell membrane complement inhibiting protein. E7.1 monoclonal antibody, specific for the S1 determinant, was found to react strongly with membrane CD59 in Western blotting, and to bind to purified, urinary form of CD59 in ELISAs. Cell membrane expression of S1 on various cell lines always correlated with that of CD59 when examined by immunofluorescent staining. In addition, E7.1 antibody inhibited the complement regulatory function of CD59. Identification of S1 protein as CD59 has increased the scope of the AL cell system by enabling analysis of intragenic mutations, and multiplex PCR analysis of mutated cells is described, showing variable loss of CD59 exons.


Advances in Space Research | 1994

Molecular analysis of mutagenesis by high let radiation

Tom K. Hei; Li X. Zhu; Diane Vannais; Charles A. Waldren

Mutation induction by high linear energy transfer [LET] alpha particles and gamma-rays was scored in the human hamster hybrid [AL] cells. Southern blotting technique was used to analyse the molecular changes in the DNA from both the HGPRT- and S1- mutants. Dose dependent mutagenesis in the AL cells irradiated with the charged particles was higher by almost 20 fold at the S1 than the corresponding HGPRT locus. Southern analysis of the mutants induced by the high LET particles showed mostly multilocus deletion at both the HGPRT and S1 genes.


Radiation Research | 2003

The “Pro-drug” RibCys Decreases the Mutagenicity of High-LET Radiation in Cultured Mammalian Cells

M. Lenarczyk; Akiko M. Ueno; Diane Vannais; S. Kraemer; Amy Kronenberg; J. C. Roberts; K. Tatsumi; Tom K. Hei; Charles A. Waldren

Abstract Lenarczyk, M., Ueno, A., Vannais, D. B., Kraemer, S., Kronenberg, A., Roberts, J. C., Tatsumi, K., Hei, T. K. and Waldren, C. A. The “Pro-drug” RibCys Decreases the Mutagenicity of High-LET Radiation in Cultured Mammalian Cells. Radiat. Res. 160, 579–583 (2003). We are carrying out studies aimed at reducing the mutagenic effects of high-LET 56Fe ions and 12C ions (56Fe ions, 143 keV/μm; 12C ions, 100 keV/μm) with certain drugs, including RibCys [2-(R,S)-d-ribo-(1′,2′,3′,4′-tetrahydroxybutyl)-thiazolidine-4(R)-carboxylic acid]. RibCys, formed by condensation of l-cysteine with d-ribose, is designed so that the sulfhydryl amino acid l-cysteine is released intracellularly through nonenzymatic ring opening and hydrolysis leading to increased levels of glutathione (GSH). RibCys (4 or 10 mM), which was present during irradiation and for a few hours after, significantly decreased the yield of CD59− mutants induced by radiation in AL human–hamster hybrid cells. RibCys did not affect the clonogenic survival of irradiated cells, nor was it mutagenic itself. These results, together with the minimal side effects reported in mice and pigs, indicate that RibCys may be useful, perhaps even when used prophylactically, in reducing the mutation load created by high-LET radiation in astronauts or other exposed individuals.


Somatic Cell and Molecular Genetics | 1992

The use of human repetitive DNA to target selectable markers into only the human chromosome of a human-hamster hybrid cell line (AL)

Charles A. Waldren; Mike Braaton; Diane Vannais; Bijan Fouladi; Rodger D. Parker

We used the plasmid BLUR-8 that contains an 800-base pair (bp) sequence of human repetitiveAlu DNA in a cotransfection protocol to target the plasmids pSV2neo or EBO-pcD-leu-2 (hygro) into a single site of the sole human chromosome, number 11, of a Chinese hamster-human hybrid cell line (AL). The neo and hygro plasmids confer resistance to antibiotics G418 and hygromycin, respectively. Of the 33 cotransfected clones with single-site insertions, 1/13 without BLUR-8 and 6/20 with BLUR-8 were only in human chromosome 11. A frequency of insertion of 1/13 is not different than expected by chance (ϱ=0.3512). On the other hand, the probability that 6/20 insertions, as seen with BLUR-8, occurred by chance is low (ϱ=0.0003). We suggest that the human DNA sequences contained in BLUR-8 targeted insertions into only the human chromosome.

Collaboration


Dive into the Diane Vannais's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Akiko M. Ueno

Colorado State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amy Kronenberg

Lawrence Berkeley National Laboratory

View shared research outputs
Top Co-Authors

Avatar

S. Kraemer

Colorado State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark L. Shibuya

Colorado State University

View shared research outputs
Top Co-Authors

Avatar

Megan McGraw

Colorado State University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge