Network


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

Hotspot


Dive into the research topics where Anne-Lee Gustafson is active.

Publication


Featured researches published by Anne-Lee Gustafson.


Reproductive Toxicology | 2012

Inter-laboratory assessment of a harmonized zebrafish developmental toxicology assay—Progress report on phase I

Anne-Lee Gustafson; Donald B. Stedman; Jonathan S. Ball; Jedd M. Hillegass; Annette Flood; Cindy X. Zhang; Julie Panzica-Kelly; J. Cao; Aleasha Coburn; Brian P. Enright; M.B. Tornesi; Malcolm J. Hetheridge; Karen Augustine-Rauch

This report provides a progress update of a consortium effort to develop a harmonized zebrafish developmental toxicity assay. Twenty non-proprietary compounds (10 animal teratogens and 10 animal non-teratogens) were evaluated blinded in 4 laboratories. Zebrafish embryos from pond-derived and cultivated strain wild types were exposed to the test compounds for 5 days and subsequently evaluated for lethality and morphological changes. Each of the testing laboratories achieved similar overall concordance to the animal data (60-70%). Subsequent optimization procedures to improve the overall concordance focused on compound formulation and test concentration adjustments, chorion permeation and number of replicates. These optimized procedures were integrated into a revised protocol and all compounds were retested in one lab using embryos from pond-derived zebrafish and achieved 85% total concordance. To further assess assay performance, a study of additional compounds is currently in progress at two laboratories using embryos from pond-derived and cultivated-strain wild type zebrafish.


Toxicological Sciences | 2009

Valproic acid-induced deregulation in vitro of genes associated in vivo with neural tube defects.

Måns Jergil; Kim Kultima; Anne-Lee Gustafson; Lennart Dencker; Michael Stigson

The utility of an in vitro system to search for molecular targets and markers of developmental toxicity was explored, using microarrays to detect genes susceptible to deregulation by the teratogen valproic acid (VPA) in the pluripotent mouse embryonal carcinoma cell line P19. Total RNA extracted from P19 cells cultured in the absence or presence of 1, 2.5, or 10mM VPA for 1.5, 6, or 24 h was subjected to replicated microarray analysis, using CodeLink UniSet I Mouse 20K Expression Bioarrays. A moderated F-test revealed a significant VPA response for 2972 (p < 10(-3)) array probes (19.4% of the filtered gene list), 421 of which were significant across all time points. In a core subset of VPA target genes whose expression was downregulated (68 genes) or upregulated (125 genes) with high probability (p < 10(-7)) after both 1.5 and 6 h of VPA exposure, there was a significant enrichment of the biological process Gene Ontology term transcriptional regulation among downregulated genes, and apoptosis among upregulated, and two of the downregulated genes (Folr1 and Gtf2i) have a knockout phenotype comprising exencephaly, the major malformation induced by VPA in mice. The VPA-induced gene expression response in P19 cells indicated that approximately 30% of the approximately 200 genes known from genetic mouse models to be associated with neural tube defects may be potential VPA targets, suggestive of a combined deregulation of multiple genes as a possible mechanism of VPA teratogenicity. Gene expression responses related to other known effects of VPA (histone deacetylase inhibition, G(1)-phase cell cycle arrest, induction of apoptosis) were also identified. This study indicates that toxicogenomic responses to a teratogenic compound in vitro may correlate with known in vitro and in vivo effects, and that short-time (< or =6 h) exposures in such an in vitro system could provide a useful component in mechanistic studies and screening tests in developmental toxicology.


Toxicological Sciences | 2011

Short-time gene expression response to valproic acid and valproic acid analogs in mouse embryonic stem cells.

Måns Jergil; Maud Forsberg; Hugh Salter; Kenneth Stockling; Anne-Lee Gustafson; Lennart Dencker; Michael Stigson

Prediction of developmental toxicity in vitro could be based on short-time toxicogenomic endpoints in embryo-derived cell lines. Microarray studies in P19 mouse embryocarcinoma cells and mouse embryos have indicated that valproic acid (VPA), an inducer of neural tube defects, deregulates the expression of many genes, including those critically involved in neural tube development. In this study, we exposed undifferentiated R1 mouse embryonic stem cells to VPA and VPA analogs for 6 h and used CodeLink whole-genome expression microarrays to define VPA-responsive genes correlating with teratogenicity. Compared with the nonteratogenic analog 2-ethyl-4-methylpentanoic acid, VPA and the teratogenic VPA analog (S)-2-pentyl-4-pentynoic acid deregulated a much larger number of genes. Five genes (of ∼2500 array probes correlating with the separation) were sufficient to effectively separate teratogens from nonteratogens. A large fraction of the target genes correlating with teratogenicity are functionally related to embryonic development and morphogenesis, including neural tube formation and closure. Similar responses in R1 were found for most genes previously identified as VPA responsive in P19 and embryos. A subset of target genes was evaluated as candidate markers predictive of potential teratogenicity against a range of known teratogens using TaqMan expression arrays. These marker genes showed a positive predictive value for the teratogens butyrate and trichostatin A, which like VPA and (S)-2-pentyl-4-pentynoic acid are known histone deacetylase (HDAC) inhibitors but not for compounds that are likely to act by other mechanisms. This indicates that HDAC inhibition may be a major mechanism by which VPA induces gene deregulation and possibly teratogenicity.


Toxicology and Applied Pharmacology | 2013

Comparative effects of sodium channel blockers in short term rat whole embryo culture.

Mats Nilsson; Anna-Carin Sköld; Ann-Christin Ericson; Anita Annas; Rodrigo Palma Villar; Gvido Cebers; Heike Hellmold; Anne-Lee Gustafson; William S. Webster

This study was undertaken to examine the effect on the rat embryonic heart of two experimental drugs (AZA and AZB) which are known to block the sodium channel Nav1.5, the hERG potassium channel and the l-type calcium channel. The sodium channel blockers bupivacaine, lidocaine, and the l-type calcium channel blocker nifedipine were used as reference substances. The experimental model was the gestational day (GD) 13 rat embryo cultured in vitro. In this model the embryonic heart activity can be directly observed, recorded and analyzed using computer assisted image analysis as it responds to the addition of test drugs. The effect on the heart was studied for a range of concentrations and for a duration up to 3h. The results showed that AZA and AZB caused a concentration-dependent bradycardia of the embryonic heart and at high concentrations heart block. These effects were reversible on washout. In terms of potency to cause bradycardia the compounds were ranked AZB>bupivacaine>AZA>lidocaine>nifedipine. Comparison with results from previous studies with more specific ion channel blockers suggests that the primary effect of AZA and AZB was sodium channel blockage. The study shows that the short-term rat whole embryo culture (WEC) is a suitable system to detect substances hazardous to the embryonic heart.


Environmental Health Perspectives | 2004

Valproic acid teratogenicity: a toxicogenomics approach

Kim Kultima; Anna-Maja Nyström; Birger Scholz; Anne-Lee Gustafson; Lennart Dencker; Michael Stigson


Toxicological Sciences | 2003

Cadmium-Induced Changes in Apoptotic Gene Expression Levels and DNA Damage in Mouse Embryos Are Blocked by Zinc

Estíbaliz L. Fernández; Anne-Lee Gustafson; Maria Andersson; Björn Hellman; Lennart Dencker


Toxicological Sciences | 2014

Fishing for Teratogens: A Consortium Effort for a Harmonized Zebrafish Developmental Toxicology Assay

Jonathan S. Ball; Donald B. Stedman; Jedd M. Hillegass; Cindy X. Zhang; Julie Panzica-Kelly; Aleasha Coburn; Brian P. Enright; Belen Tornesi; Hamid R. Amouzadeh; Malcolm J. Hetheridge; Anne-Lee Gustafson; Karen Augustine-Rauch


Reproductive Toxicology | 2010

Early transcriptional responses in mouse embryos as a basis for selection of molecular markers predictive of valproic acid teratogenicity

Kim Kultima; Måns Jergil; Hugh Salter; Anne-Lee Gustafson; Lennart Dencker; Michael Stigson


Reproductive Toxicology | 2006

Cadmium-induced gene expression changes in the mouse embryo, and the influence of pretreatment with zinc

Kim Kultima; Estíbaliz L. Fernández; Birger Scholz; Anne-Lee Gustafson; Lennart Dencker; Michael Stigson


Atla-alternatives To Laboratory Animals | 2007

Molecular targets and early response biomarkers for the prediction of developmental toxicity in vitro

Michael Stigson; Kim Kultima; Måns Jergil; Birger Scholz; Henrik Alm; Anne-Lee Gustafson; Lennart Dencker

Collaboration


Dive into the Anne-Lee Gustafson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge