169 results on '"Alvaro Puga"'
Search Results
2. Hexavalent chromium promotes differential binding of CTCF to its cognate sites in Euchromatin
- Author
-
Andrew VonHandorf, Hesbon A. Zablon, Jacek Biesiada, Xiang Zhang, Mario Medvedovic, and Alvaro Puga
- Subjects
Genetics ,QH426-470 - Abstract
Hexavalent chromium compounds are well-established respiratory carcinogens to which humans are commonly exposed in industrial and occupational settings. In addition, natural and anthropogenic sources of these compounds contribute to the exposure of global populations through multiple routes, including dermal, ingestion and inhalation that elevate the risk of cancer by largely unresolved mechanisms. Cr(VI) has genotoxic properties that include ternary adduct formation with DNA, increases in DNA damage, mostly by double-strand break formation, and altered transcriptional responses. Our previous work using ATAC-seq showed that CTCF motifs were enriched in Cr(VI)-dependent differentially accessible chromatin, suggesting that CTCF, a key transcription factor responsible for the regulation of the transcriptome, might be a chromium target. To test this hypothesis, we investigated the effect of Cr(VI) treatment on the binding of CTCF to its cognate sites and ensuing changes in transcription-related histone modifications. Differentially bound CTCF sites were enriched by Cr(VI) treatment within transcription-related regions, specifically transcription start sites and upstream genic regions. Functional annotation of the affected genes highlighted biological processes previously associated with Cr(VI) exposure. Notably, we found that differentially bound CTCF sites proximal to the promoters of this subset of genes were frequently associated with the active histone marks H3K27ac, H3K4me3, and H3K36me3, in agreement with the concept that Cr(VI) targets CTCF in euchromatic regions of the genome. Our results support the conclusion that Cr(VI) treatment promotes the differential binding of CTCF to its cognate sites in genes near transcription-active boundaries, targeting these genes for dysregulation.
- Published
- 2021
- Full Text
- View/download PDF
3. Developmental and lifelong dioxin exposure induces measurable changes in cardiac structure and function in adulthood
- Author
-
Matthew de Gannes, Sheryl E. Koch, Alvaro Puga, and Jack Rubinstein
- Subjects
Medicine ,Science - Abstract
Abstract Congenital heart disease (CHD) is the most common congenital abnormality. A precise etiology for CHD remains elusive, but likely results from interactions between genetic and environmental factors during development, when the heart adapts to physiological and pathophysiological conditions. Further, it has become clearer that early exposure to toxins that do not result in overt CHD may be associated with adverse cardiac outcomes that are not manifested until later life. Previously, interference with endogenous developmental functions of the aryl hydrocarbon receptor (AHR), either by gene ablation or by in utero exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a potent AHR ligand, was shown to cause structural, molecular and functional cardiac abnormalities and altered heart physiology in mouse embryos. Here, we show that continuous exposure to TCDD from fertilization throughout adulthood caused male mice to underperform at exercise tolerance tests compared to their control and female counterparts, confirming previous observations of a sexually dimorphic phenotype. Renin-angiotensin stimulation by angiotensin II (Ang II) caused measurable increases in blood pressure and left ventricle mass, along with decreased end diastolic volume and preserved ejection fraction. Interestingly, TCDD exposure caused measurable reductions in the myocardial hypertrophic effects of Ang II, suggesting that endogenous AHR signaling present in adulthood may play a role in the pathogenesis of hypertrophy. Overall, the findings reported in this pilot study highlight the complex systems underlying TCDD exposure in the development of cardiac dysfunction in later life.
- Published
- 2021
- Full Text
- View/download PDF
4. Chromium disrupts chromatin organization and CTCF access to its cognate sites in promoters of differentially expressed genes
- Author
-
Andrew VonHandorf, Francisco Javier Sánchez-Martín, Jacek Biesiada, Hongxia Zhang, Xiang Zhang, Mario Medvedovic, and Alvaro Puga
- Subjects
chromatin organization ,nucleosome architecture ,atac-seq ,ctcf ,hexavalent chromium ,transcription ,Genetics ,QH426-470 - Abstract
Hexavalent chromium compounds are well-established respiratory carcinogens used in industrial processes. While inhalation exposure constitutes an occupational risk affecting mostly chromium workers, environmental exposure from drinking water is a widespread gastrointestinal cancer risk, affecting millions of people throughout the world. Cr(VI) is genotoxic, forming protein-Cr-DNA adducts and silencing tumor suppressor genes, but its mechanism of action at the molecular level is poorly understood. Our prior work using FAIRE showed that Cr(VI) disrupted the binding of transcription factors CTCF and AP-1 to their cognate chromatin sites. Here, we used two complementary approaches to test the hypothesis that chromium perturbs chromatin organization and dynamics. DANPOS2 analyses of MNase-seq data identified several chromatin alterations induced by Cr(VI) affecting nucleosome architecture, including occupancy changes at specific genome locations; position shifts of 10 nucleotides or more; and changes in position amplitude or fuzziness. ATAC-seq analysis revealed that Cr(VI) disrupted the accessibility of chromatin regions enriched for CTCF and AP-1 binding motifs, with a significant co-occurrence of binding sites for both factors in the same region. Cr(VI)-enriched CTCF sites were confirmed by ChIP-seq and found to correlate with evolutionarily conserved sites occupied by CTCF in vivo, as determined by comparison with ENCODE-validated CTCF datasets from mouse liver. In addition, more than 30% of the Cr(VI)-enriched CTCF sites were located in promoters of genes differentially expressed from chromium treatment. Our results support the conclusion that Cr(VI) exposure promotes broad changes in chromatin accessibility and suggest that the subsequent effects on transcription regulation may result from disruption of CTCF binding and nucleosome spacing, implicating transcription regulatory mechanisms as primary Cr(VI) targets.
- Published
- 2018
- Full Text
- View/download PDF
5. Pluripotency factors and Polycomb Group proteins repress aryl hydrocarbon receptor expression in murine embryonic stem cells
- Author
-
Chia-I Ko, Qin Wang, Yunxia Fan, Ying Xia, and Alvaro Puga
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The aryl hydrocarbon receptor (AHR) is a transcription factor and environmental sensor that regulates expression of genes involved in drug-metabolism and cell cycle regulation. Chromatin immunoprecipitation analyses, Ahr ablation in mice and studies with orthologous genes in invertebrates suggest that AHR may also play a significant role in embryonic development. To address this hypothesis, we studied the regulation of Ahr expression in mouse embryonic stem cells and their differentiated progeny. In ES cells, interactions between OCT3/4, NANOG, SOX2 and Polycomb Group proteins at the Ahr promoter repress AHR expression, which can also be repressed by ectopic expression of reprogramming factors in hepatoma cells. In ES cells, unproductive RNA polymerase II binds at the Ahr transcription start site and drives the synthesis of short abortive transcripts. Activation of Ahr expression during differentiation follows from reversal of repressive marks in Ahr promoter chromatin, release of pluripotency factors and PcG proteins, binding of Sp factors, establishment of histone marks of open chromatin, and engagement of active RNAPII to drive full-length RNA transcript elongation. Our results suggest that reversible Ahr repression in ES cells holds the gene poised for expression and allows for a quick switch to activation during embryonic development.
- Published
- 2014
- Full Text
- View/download PDF
6. Old Receptor, New Tricks—The Ever-Expanding Universe of Aryl Hydrocarbon Receptor Functions. Report from the 4th AHR Meeting, 29–31 August 2018 in Paris, France
- Author
-
Charlotte Esser, B. Paige Lawrence, David H. Sherr, Gary H. Perdew, Alvaro Puga, Robert Barouki, and Xavier Coumoul
- Subjects
transcription factor ,translational science ,environmental health ,barrier organs ,stem cells ,nervous system ,obesity ,cancer ,immunity ,development ,diet ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
In a time where “translational„ science has become a mantra in the biomedical field, it is reassuring when years of research into a biological phenomenon suddenly points towards novel prevention or therapeutic approaches to disease, thereby demonstrating once again that basic science and translational science are intimately linked. The studies on the aryl hydrocarbon receptor (AHR) discussed here provide a perfect example of how years of basic toxicological research on a molecule, whose normal physiological function remained a mystery for so long, has now yielded a treasure trove of actionable information on the development of targeted therapeutics. Examples are autoimmunity, metabolic imbalance, inflammatory skin and gastro-intestinal diseases, cancer, development and perhaps ageing. Indeed, the AHR field no longer asks, “What does this receptor do in the absence of xenobiotics?„ It now asks, “What doesn’t this receptor do?„.
- Published
- 2018
- Full Text
- View/download PDF
7. Disruption of Ah Receptor Signaling during Mouse Development Leads to Abnormal Cardiac Structure and Function in the Adult.
- Author
-
Vinicius S Carreira, Yunxia Fan, Hisaka Kurita, Qin Wang, Chia-I Ko, Mindi Naticchioni, Min Jiang, Sheryl Koch, Xiang Zhang, Jacek Biesiada, Mario Medvedovic, Ying Xia, Jack Rubinstein, and Alvaro Puga
- Subjects
Medicine ,Science - Abstract
The Developmental Origins of Health and Disease (DOHaD) Theory proposes that the environment encountered during fetal life and infancy permanently shapes tissue physiology and homeostasis such that damage resulting from maternal stress, poor nutrition or exposure to environmental agents may be at the heart of adult onset disease. Interference with endogenous developmental functions of the aryl hydrocarbon receptor (AHR), either by gene ablation or by exposure in utero to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a potent AHR ligand, causes structural, molecular and functional cardiac abnormalities and altered heart physiology in mouse embryos. To test if embryonic effects progress into an adult phenotype, we investigated whether Ahr ablation or TCDD exposure in utero resulted in cardiac abnormalities in adult mice long after removal of the agent. Ten-months old adult Ahr-/- and in utero TCDD-exposed Ahr+/+ mice showed sexually dimorphic abnormal cardiovascular phenotypes characterized by echocardiographic findings of hypertrophy, ventricular dilation and increased heart weight, resting heart rate and systolic and mean blood pressure, and decreased exercise tolerance. Underlying these effects, genes in signaling networks related to cardiac hypertrophy and mitochondrial function were differentially expressed. Cardiac dysfunction in mouse embryos resulting from AHR signaling disruption seems to progress into abnormal cardiac structure and function that predispose adults to cardiac disease, but while embryonic dysfunction is equally robust in males and females, the adult abnormalities are more prevalent in females, with the highest severity in Ahr-/- females. The findings reported here underscore the conclusion that AHR signaling in the developing heart is one potential target of environmental factors associated with cardiovascular disease.
- Published
- 2015
- Full Text
- View/download PDF
8. Formaldehyde-Assisted Isolation of Regulatory Elements (FAIRE) analysis uncovers broad changes in chromatin structure resulting from hexavalent chromium exposure.
- Author
-
Jerald L Ovesen, Yunxia Fan, Xiang Zhang, Jing Chen, Mario Medvedovic, Ying Xia, and Alvaro Puga
- Subjects
Medicine ,Science - Abstract
The ability of chromatin to switch back and forth from open euchromatin to closed heterochromatin is vital for transcriptional regulation and genomic stability, but its dynamic structure is subject to disruption by exposure to environmental agents such as hexavalent chromium. Cr(VI) exposure disrupts chromatin remodeling mechanisms and causes chromosomal damage through formation of free radicals, Cr-DNA adducts, and DNA-Cr-protein cross-links. In addition, acute, high-concentration, and chronic, low-concentration exposures to Cr(VI) lead to significantly different transcriptional and genomic stability outcomes. We used mouse hepatoma Hepa-1c1c7 cells to investigate how transcriptional responses to chromium treatment might correlate with structural chromatin changes. We used Formaldehyde-Assisted Isolation of Regulatory Elements (FAIRE) analysis coupled with deep sequencing to identify regions of the genome that may switch between open and closed chromatin in response to exposure to varying Cr(VI) concentrations. At either Cr(VI) concentration, chromatin domains surrounding binding sites for AP-1 transcription factors become significantly open, whereas BACH2 and CTCF binding sites are open solely at the low and high concentrations, respectively. Parallel gene expression profiling using RNA-seq indicates that the structural chromatin changes caused by Cr(VI) affect gene expression levels in the target areas that vary depending on Cr(VI) concentration, but show no correlation between global changes in the overall transcriptional response and Cr(VI) concentration. Our results suggest that FAIRE may be a useful technique to map chromatin elements targeted by DNA damaging agents for which there is no prior knowledge of their specificity, and to identify subsequent transcriptomic changes induced by those agents.
- Published
- 2014
- Full Text
- View/download PDF
9. Lead induces similar gene expression changes in brains of gestationally exposed adult mice and in neurons differentiated from mouse embryonic stem cells.
- Author
-
Francisco Javier Sánchez-Martín, Yunxia Fan, Diana M Lindquist, Ying Xia, and Alvaro Puga
- Subjects
Medicine ,Science - Abstract
Exposure to environmental toxicants during embryonic life causes changes in the expression of developmental genes that may last for a lifetime and adversely affect the exposed individual. Developmental exposure to lead (Pb), an ubiquitous environmental contaminant, causes deficits in cognitive functions and IQ, behavioral effects, and attention deficit hyperactivity disorder (ADHD). Long-term effects observed after early life exposure to Pb include reduction of gray matter, alteration of myelin structure, and increment of criminal behavior in adults. Despite growing research interest, the molecular mechanisms responsible for the effects of lead in the central nervous system are still largely unknown. To study the molecular changes due to Pb exposure during neurodevelopment, we exposed mice to Pb in utero and examined the expression of neural markers, neurotrophins, transcription factors and glutamate-related genes in hippocampus, cortex, and thalamus at postnatal day 60. We found that hippocampus was the area where gene expression changes due to Pb exposure were more pronounced. To recapitulate gestational Pb exposure in vitro, we differentiated mouse embryonic stem cells (ESC) into neurons and treated ESC-derived neurons with Pb for the length of the differentiation process. These neurons expressed the characteristic neuronal markers Tubb3, Syp, Gap43, Hud, Ngn1, Vglut1 (a marker of glutamatergic neurons), and all the glutamate receptor subunits, but not the glial marker Gafp. Importantly, several of the changes observed in Pb-exposed mouse brains in vivo were also observed in Pb-treated ESC-derived neurons, including those affecting expression of Ngn1, Bdnf exon IV, Grin1, Grin2D, Grik5, Gria4, and Grm6. We conclude that our ESC-derived model of toxicant exposure during neural differentiation promises to be a useful model to analyze mechanisms of neurotoxicity induced by Pb and other environmental agents.
- Published
- 2013
- Full Text
- View/download PDF
10. Induction of Oxidative Stress Responses by Dioxin and other Ligands of the Aryl Hydrocarbon Receptor
- Author
-
John F. Reichard, Timothy P. Dalton, Howard G. Shertzer, and Alvaro Puga
- Subjects
Therapeutics. Pharmacology ,RM1-950 - Abstract
TCDD and other polyhalogenated aromatic hydrocarbon ligands of the aryl hydrocarbon receptor (AHR) have been classically considered as non-genotoxic compounds because they fail to be directly mutagenic in either bacteria or most in vitro assay systems. They do so in spite of having repeatedly been linked to oxidative stress and to mutagenic and carcinogenic outcomes. Oxidative stress, on the other hand, has been used as a marker for the toxicity of dioxin and its congeners. We have focused this review on the connection between oxidative stress induction and the toxic effects of fetal and adult dioxin exposure, with emphasis on the large species difference in sensitivity to this agent. We examine the roles that the dioxin-inducible cytochromes P450s play in the cellular and toxicological consequences of dioxin exposure with emphasis on oxidative stress involvement. Many components of the health consequences resulting from dioxin exposure may be attributable to epigenetic mechanisms arising from prolonged reactive oxygen generation.
- Published
- 2005
- Full Text
- View/download PDF
11. The Role of MAP3K1 in the Development of the Female Reproductive Tract
- Author
-
Eiki Kimura, Maureen Mongan, Bo Xiao, Jingjing Wang, Vinicius S Carreira, Brad Bolon, Xiang Zhang, Katherine A. Burns, Jacek Biesiada, Mario Medvedovic, Alvaro Puga, and Ying Xia
- Subjects
Article - Abstract
Mitogen-Activated Protein 3 Kinase 1 (MAP3K1) is a dynamic signaling molecule with a plethora of cell-type specific functions, most of which are yet to be understood. Here we describe a role for MAP3K1 in the development of female reproductive tract (FRT). MAP3K1 kinase domain-deficient (Map3k1ΔKD) females exhibit imperforate vagina, labor failure, and infertility. These defects correspond to a shunted Müllerian duct (MD), the principle precursor of the FRT, in embryos, while they manifest as a contorted caudal vagina with abrogated vaginal-urogenital sinus fusion in neonates. In epithelial cells, MAP3K1 acts through JNK and ERK to activate WNT, yetin vivoMAP3K1 is crucial for WNT activity in mesenchyme associated with the caudal MD. Expression ofWnt7bis high in wild type, but low inMap3k1knockout MD epithelium and MAP3K1-deficient keratinocytes. Correspondingly, conditioned media derived from MAP3K1-competent epithelial cells activate TCF/Lef-luciferase reporter in fibroblasts, suggesting that MAP3K1-induced factors released from epithelial cells trans-activate WNT signaling in fibroblasts. Our results reveal a temporal-spatial and paracrine MAP3K1-WNT crosstalk contributing to MD caudal elongation and FRT development.HighlightsMAP3K1 deficient female mice exhibit imperforate vagina and infertilityLoss of MAP3K1 kinase activity impedes Müllerian duct (MD) caudal elongation and fusion with urogenital sinus (UGS) in embryogenesisThe MAP3K1-MAPK pathway up-regulates WNT signaling in epithelial cellsMAP3K1 deficiency down-regulates Wnt7b expression in the MD epithelium and prevents WNT activity in mesenchyme of the caudal MD
- Published
- 2023
12. Supplementary Table 1 from The Aryl Hydrocarbon Receptor Functions as a Tumor Suppressor of Liver Carcinogenesis
- Author
-
Alvaro Puga, Ying Xia, Daniel W. Nebert, Erik S. Knudsen, Gregory P. Boivin, and Yunxia Fan
- Abstract
Supplementary Table 1 from The Aryl Hydrocarbon Receptor Functions as a Tumor Suppressor of Liver Carcinogenesis
- Published
- 2023
13. Data from The Aryl Hydrocarbon Receptor Functions as a Tumor Suppressor of Liver Carcinogenesis
- Author
-
Alvaro Puga, Ying Xia, Daniel W. Nebert, Erik S. Knudsen, Gregory P. Boivin, and Yunxia Fan
- Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that mediates the biological and toxic effects of its xenobiotic ligands. Previous cell culture studies have shown that, in addition to controlling the xenobiotic detoxification response, AHR activation leads to G0-G1 arrest, diminished capacity for DNA replication, and inhibition of cell proliferation. In fact, recent work from our own and from other laboratories suggests that AHR may function as a tumor suppressor gene that becomes silenced during the process of tumor formation. To test this hypothesis and determine whether the mouse Ahr gene acts as a tumor suppressor gene in vivo, we have examined the role of Ahr ablation in liver tumorigenesis induced by the genotoxic chemical diethylnitrosamine (DEN), a hepatic carcinogen that is not an AHR ligand. In mice given a single i.p. injection of DEN, AHR antagonized liver tumor formation and growth by regulating cell proliferation, inflammatory cytokine expression, and DNA damage, parameters which were significantly elevated in the livers of control and, more so, of DEN-exposed Ahr−/− mice. Ahr−/− hepatocytes also showed significantly higher numbers of 4N cells, increased expression of proliferative markers, and repression of tumor suppressor genes. These data support the concept that in its basal state in the absence of a xenobiotic ligand, the Ahr gene functions as a tumor suppressor gene, and that its silencing may be associated with cancer progression. Cancer Res; 70(1); 212–220
- Published
- 2023
14. Mechanisms of chromate carcinogenesis by chromatin alterations
- Author
-
Hesbon A. Zablon, Andrew VonHandorf, and Alvaro Puga
- Published
- 2023
15. The aryl hydrocarbon receptor directs the differentiation of murine progenitor blastomeres
- Author
-
Chia-I, Ko, Jacek, Biesiada, Hesbon A, Zablon, Xiang, Zhang, Mario, Medvedovic, and Alvaro, Puga
- Subjects
Health, Toxicology and Mutagenesis ,Cell Biology ,Toxicology - Abstract
Key regulatory decisions during cleavage divisions in mammalian embryogenesis determine the fate of preimplantation embryonic cells. Single-cell RNA sequencing of early-stage—2-cell, 4-cell, and 8-cell—blastomeres show that the aryl hydrocarbon receptor (AHR), traditionally considered as an environmental sensor, directs blastomere differentiation. Disruption of AHR functions in Ahr knockout embryos or in embryos from dams exposed to dioxin, the prototypic xenobiotic AHR agonist, significantly impairs blastocyst formation, causing repression and loss of transcriptional heterogeneity of OCT4 and CDX2 and incidence of nonspecific downregulation of pluripotency. Trajectory—the path of differentiation—and gene variability analyses further confirm that deregulation of OCT4 functions and changes of transcriptional heterogeneity resulting from disruption of AHR functions restrict the emergence of differentiating blastomeres in 4-cell embryos. It appears that AHR directs the differentiation of progenitor blastomeres and that disruption of preimplantation AHR functions may significantly perturb embryogenesis leading to long-lasting conditions at the heart of disease in offspring’s adulthood.
- Published
- 2022
16. Hexavalent Chromium Disrupts Chromatin Architecture
- Author
-
Alvaro Puga, Hesbon A Zablon, and Andrew VonHandorf
- Subjects
Chromium ,Cancer Research ,Euchromatin ,Transcription, Genetic ,Chemistry ,Carcinogenesis ,Article ,Chromatin ,Cell biology ,AP-1 transcription factor ,CTCF ,Regulatory sequence ,Gene expression ,Animals ,Humans ,Transcription factor ,Gene - Abstract
Accessibility of DNA elements and the orchestration of spatiotemporal chromatin-chromatin interactions are critical mechanisms in the regulation of gene transcription. Thus, in an ever-changing milieu, cells mount an adaptive response to environmental stimuli by modulating gene expression that is orchestrated by coordinated changes in chromatin architecture. Correspondingly, agents that alter chromatin structure directly impact transcriptional programs in cells. Heavy metals, including hexavalent chromium (Cr(VI)), are of special interest because of their ability to interact directly with cellular protein, DNA and other macromolecules, resulting in general damage or altered function. In this review we highlight the chromium-mediated mechanisms that promote disruption of chromatin architecture and how these processes are integral to its carcinogenic properties. Emerging evidence shows that Cr(VI) targets nucleosomal architecture in euchromatin, particularly in genomic locations flanking binding sites of the essential transcription factors CTCF and AP1. Ultimately, these changes contribute to an altered chromatin state in critical gene regulatory regions, which disrupts gene transcription in functionally relevant biological processes.
- Published
- 2021
17. Chromium exposure disrupts chromatin architecture upsetting the mechanisms that regulate transcription
- Author
-
Alvaro Puga, Hesbon A Zablon, and Andrew VonHandorf
- Subjects
0301 basic medicine ,chemistry.chemical_element ,010501 environmental sciences ,medicine.disease_cause ,01 natural sciences ,Chemical used ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,Chromatin ,03 medical and health sciences ,chemistry.chemical_compound ,Chromium ,030104 developmental biology ,chemistry ,Transcription (biology) ,medicine ,Epigenetics ,Hexavalent chromium ,Genotoxicity ,Carcinogen ,0105 earth and related environmental sciences - Abstract
Hexavalent chromium (Cr(VI)) is a highly mutagenic and carcinogenic chemical used in many industrial processes. Occupational exposure to chromium, occurring mostly by inhalation, constitutes a major lung cancer risk affecting chromium workers. Environmental exposure, on the other hand, mainly by ingestion of contaminated drinking water, is a widespread gastrointestinal cancer risk, affecting millions of people throughout the world. One of the major mechanisms through which Cr(VI) causes carcinogenic transformation is thought to be the disruption of transcriptional regulation. Indeed, Cr(VI)-directed DNA damage and crosslinking occurs preferentially at sites where active DNA replication and transcription processes take place. Accordingly, numerous studies have shown that Cr(VI) causes gene expression changes in a wide range of cell signaling pathways, resulting from Cr(VI)-induced direct macromolecular damage, alteration in transcription factor function, and disruption of epigenetic signatures. This brief review highlights past and current information on the impact of Cr(VI) on the various mechanisms of transcriptional regulation. Impact statement This mini-review highlights current evidence on the mechanisms through which hexavalent chromium (Cr(VI)) disrupts transcriptional regulation, an emerging area of interest and one of the central processes by which chromium induces carcinogenesis. Several studies have shown that Cr(VI) causes widespread DNA damage and disrupts epigenetic signatures, suggesting that chromatin may be a direct Cr(VI) target. The findings discussed here suggest that Cr(VI) disrupts transcriptional regulation by causing genomic architecture changes.
- Published
- 2019
18. Prenatal exposure to PCBs in Cyp1a2 knock‐out mice interferes with F 1 fertility, impairs long‐term potentiation, reduces acoustic startle and impairs conditioned freezing contextual memory with minimal transgenerational effects
- Author
-
Charles V. Vorhees, Sheryl E. Koch, Jenna L.N. Sprowles, Xiang Zhang, Qin Wang, Michael T. Williams, Alvaro Puga, Jacek Biesiada, Min Jiang, Emily M. Pitzer, Jillian R. Hufgard, and Jack Rubinstein
- Subjects
0303 health sciences ,medicine.medical_specialty ,Perinatal Exposure ,Offspring ,Morris water navigation task ,Hippocampus ,Long-term potentiation ,010501 environmental sciences ,Biology ,Toxicology ,Aryl hydrocarbon receptor ,01 natural sciences ,03 medical and health sciences ,Endocrinology ,Internal medicine ,Toxicity ,Knockout mouse ,medicine ,biology.protein ,030304 developmental biology ,0105 earth and related environmental sciences - Abstract
Polychlorinated biphenyls (PCBs) are toxic environmental pollutants. Humans are exposed to PCB mixtures via contaminated food or water. PCB exposure causes adverse effects in adults and after exposure in utero. PCB toxicity depends on the congener mixture and CYP1A2 gene activity. For coplanar PCBs, toxicity depends on ligand affinity for the aryl hydrocarbon receptor (AHR). Previously, we found that perinatal exposure of mice to a three-coplanar/five-noncoplanar PCB mixture induced deficits in novel object recognition and trial failures in the Morris water maze in Cyp1a2-/- ::Ahrb1 C57BL6/J mice compared with wild-type mice (Ahrb1 = high AHR affinity). Here we exposed gravid Cyp1a2-/- ::Ahrb1 mice to a PCB mixture on embryonic day 10.5 by gavage and examined the F1 and F3 offspring (not F2 ). PCB-exposed F1 mice exhibited increased open-field central time, reduced acoustic startle, greater conditioned contextual freezing and reduced CA1 hippocampal long-term potentiation with no change in spatial learning or memory. F1 mice also had inhibited growth, decreased heart rate and cardiac output, and impaired fertility. F3 mice showed few effects. Gene expression changes were primarily in F1 PCB males compared with wild-type males. There were minimal RNA and DNA methylation changes in the hippocampus from F1 to F3 with no clear relevance to the functional effects. F0 PCB exposure during a period of rapid DNA de-/remethylation in a susceptible genotype produced clear F1 effects with little evidence of transgenerational effects in the F3 generation. While PCBs show clear developmental neurotoxicity, their effects do not persist across generations for effects assessed herein.
- Published
- 2018
19. Converging Roles of the Aryl Hydrocarbon Receptor in Early Embryonic Development, Maintenance of Stemness, and Tissue Repair
- Author
-
Chia-I Ko, Alvaro Puga, and Hesbon A Zablon
- Subjects
0301 basic medicine ,Homeobox protein NANOG ,biology ,Embryonic Development ,Context (language use) ,Cell Differentiation ,Toxicology ,Aryl hydrocarbon receptor ,Embryonic stem cell ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Mediator ,Gene Expression Regulation ,Receptors, Aryl Hydrocarbon ,Cancer stem cell ,030220 oncology & carcinogenesis ,Contemporary Review ,biology.protein ,Humans ,Signal transduction ,Transcription factor ,Embryonic Stem Cells - Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor well-known for its adaptive role as a sensor of environmental toxicants and mediator of the metabolic detoxification of xenobiotic ligands. In addition, a growing body of experimental data has provided indisputable evidence that the AHR regulates critical functions of cell physiology and embryonic development. Recent studies have shown that the naïve AHR—that is, unliganded to xenobiotics but activated endogenously—has a crucial role in maintenance of embryonic stem cell pluripotency, tissue repair, and regulation of cancer stem cell stemness. Depending on the cellular context, AHR silences the expression of pluripotency genes Oct4 and Nanog and potentiates differentiation, whereas curtailing cellular plasticity and stemness. In these processes, AHR-mediated contextual responses and outcomes are dictated by changes of interacting partners in signaling pathways, gene networks, and cell-type-specific genomic structures. In this review, we focus on AHR-mediated changes of genomic architecture as an emerging mechanism for the AHR to regulate gene expression at the transcriptional level. Collective evidence places this receptor as a physiological hub connecting multiple biological processes whose disruption impacts on embryonic development, tissue repair, and maintenance or loss of stemness.
- Published
- 2021
20. Dioxin Disrupts Dynamic DNA Methylation Patterns in Genes That Govern Cardiomyocyte Maturation
- Author
-
Matthew de Gannes, Xiang Zhang, Alvaro Puga, Mario Medvedovic, Chia-I Ko, Jack Rubinstein, Liang Niu, Jacek Biesiada, and Sheryl E. Koch
- Subjects
0301 basic medicine ,Polychlorinated Dibenzodioxins ,Biology ,Toxicology ,Dioxins ,Cell Line ,Transcriptome ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Gene expression ,Animals ,Myocytes, Cardiac ,Epigenetics ,Gene ,Selectable marker ,Embryonic Stem Cells ,Heart development ,DNA Methylation ,Genetic and Epigenetic Toxicology ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,chemistry ,Puromycin ,030220 oncology & carcinogenesis ,DNA methylation - Abstract
Congenital heart disease (CHD), the leading birth defect worldwide, has a largely unknown etiology, likely to result from complex interactions between genetic and environmental factors during heart development, at a time when the heart adapts to diverse physiological and pathophysiological conditions. Crucial among these is the regulation of cardiomyocyte development and postnatal maturation, governed by dynamic changes in DNA methylation. Previous work from our laboratory has shown that exposure to the environmental toxicant tetrachlorodibenzo-p-dioxin (TCDD) disrupts several molecular networks responsible for heart development and function. To test the hypothesis that the disruption caused by TCDD in the heart results from changes in DNA methylation and gene expression patterns of cardiomyocytes, we established a stable mouse embryonic stem cell line expressing a puromycin resistance selectable marker under control of the cardiomyocyte-specific Nkx2-5 promoter. Differentiation of these cells in the presence of puromycin induces the expression of a large suite of cardiomyocyte-specific markers. To assess the consequences of TCDD treatment on gene expression and DNA methylation in these cardiomyocytes, we subjected them to transcriptome and methylome analyses in the presence of TCDD. Unlike control cardiomyocytes maintained in vehicle, the TCDD-treated cardiomyocytes showed extensive gene expression changes, with a significant correlation between differential RNA expression and DNA methylation in 111 genes, many of which are key elements of pathways that regulate cardiovascular development and function. Our findings provide an important clue toward the elucidation of the complex interactions between genetic and epigenetic mechanisms after developmental TCDD exposure that may contribute to CHD.
- Published
- 2020
21. Regulation of a long noncoding RNA MALAT1 by aryl hydrocarbon receptor in pancreatic cancer cells and tissues
- Author
-
Sung-Gook Cho, Kyounghyun Kim, Alvaro Puga, Seong-Gyu Ko, Ji-eun Lee, and Syed A. Ahrmad
- Subjects
0301 basic medicine ,Polychlorinated Dibenzodioxins ,Biophysics ,Biochemistry ,Article ,03 medical and health sciences ,0302 clinical medicine ,Pancreatic cancer ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Enhancer of Zeste Homolog 2 Protein ,Molecular Biology ,Transcription factor ,Pancreas ,MALAT1 ,biology ,Chemistry ,EZH2 ,Cancer ,Cell Biology ,Environmental exposure ,Aryl hydrocarbon receptor ,medicine.disease ,Mice, Inbred C57BL ,Pancreatic Neoplasms ,030104 developmental biology ,Receptors, Aryl Hydrocarbon ,030220 oncology & carcinogenesis ,Histone methyltransferase ,biology.protein ,Cancer research ,Environmental Pollutants ,RNA, Long Noncoding ,Signal Transduction - Abstract
Environmental toxicants such as dioxins and polycyclic aromatic carbons are risk factors for pancreatitis and pancreatic cancer. These toxicants activate aryl hydrocarbon receptor (AHR), a ligand-activated transcription factor, of which activation regulates many downstream biological events, including xenobiotic metabolism, inflammation, and cancer cell growth and transformation. Here, we identified that environmental toxicant-activated AHR increased expression of metastasis associated lung adenocarcinoma transcript 1 (MALAT1) in pancreatic cancer cells and pancreatic tissues. The MALAT1 is a long noncoding (lnc) RNA which interacts with Enhancer of Zeste 2 (EZH2), a histone methyltransferase with epigenetic silencer activity, and the MALAT1-EZH2 interaction increased its epigenetic silencing activity. In contrast, AHR antagonist, CH223191 or resveratrol, counteracted the AHR-mediated MALAT1 induction and MALAT1-enahnced EZH2 activity. Collectively, these results revealed a novel pathway of how environmental exposure leads to epigenetic alteration via activation of AHR-MALAT1-EZH2 signaling axis under pancreatic tissue- and cancer cell-context.
- Published
- 2020
22. Hexavalent Chromium Promotes Differential Binding of CTCF to its Cognate Sites in Euchromatin
- Author
-
Hesbon A Zablon, Xiang Zhang, Andrew VonHandorf, Jacek Biesiada, Mario Medvedovic, and Alvaro Puga
- Subjects
0301 basic medicine ,Chromium ,Cancer Research ,Euchromatin ,biology ,Promoter ,DNA Methylation ,Chromatin ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Histone ,CTCF ,030220 oncology & carcinogenesis ,biology.protein ,H3K4me3 ,Humans ,Molecular Biology ,Gene ,Transcription factor ,Research Paper - Abstract
Hexavalent chromium compounds are well-established respiratory carcinogens to which humans are commonly exposed in industrial and occupational settings. In addition, natural and anthropogenic sources of these compounds contribute to the exposure of global populations through multiple routes, including dermal, ingestion and inhalation that elevate the risk of cancer by largely unresolved mechanisms. Cr(VI) has genotoxic properties that include ternary adduct formation with DNA, increases in DNA damage, mostly by double-strand break formation, and altered transcriptional responses. Our previous work using ATAC-seq showed that CTCF motifs were enriched in Cr(VI)-dependent differentially accessible chromatin, suggesting that CTCF, a key transcription factor responsible for the regulation of the transcriptome, might be a chromium target. To test this hypothesis, we investigated the effect of Cr(VI) treatment on the binding of CTCF to its cognate sites and ensuing changes in transcription-related histone modifications. Differentially bound CTCF sites were enriched by Cr(VI) treatment within transcription-related regions, specifically transcription start sites and upstream genic regions. Functional annotation of the affected genes highlighted biological processes previously associated with Cr(VI) exposure. Notably, we found that differentially bound CTCF sites proximal to the promoters of this subset of genes were frequently associated with the active histone marks H3K27ac, H3K4me3, and H3K36me3, in agreement with the concept that Cr(VI) targets CTCF in euchromatic regions of the genome. Our results support the conclusion that Cr(VI) treatment promotes the differential binding of CTCF to its cognate sites in genes near transcription-active boundaries, targeting these genes for dysregulation.
- Published
- 2020
- Full Text
- View/download PDF
23. Repression of the Aryl Hydrocarbon Receptor Is Required to Maintain Mitotic Progression and Prevent Loss of Pluripotency of Embryonic Stem Cells
- Author
-
Qin Wang, Chia-I Ko, Ying Xia, Alvaro Puga, Matthew de Gannes, and Yunxia Fan
- Subjects
Pluripotent Stem Cells ,0301 basic medicine ,Rex1 ,Cellular differentiation ,Mitosis ,Biology ,Cell Line ,S Phase ,Mice ,03 medical and health sciences ,0302 clinical medicine ,SOX2 ,Animals ,Cell Lineage ,Myocytes, Cardiac ,Induced pluripotent stem cell ,Psychological repression ,Cell Differentiation ,Mouse Embryonic Stem Cells ,Cell Biology ,Embryonic stem cell ,Cell biology ,Repressor Proteins ,030104 developmental biology ,Gene Expression Regulation ,Receptors, Aryl Hydrocarbon ,Cancer research ,Molecular Medicine ,Stem cell ,Neuroglia ,030217 neurology & neurosurgery ,Signal Transduction ,Developmental Biology - Abstract
Lack of cell cycle checkpoints and uninterrupted passage through S-phase continuously renew the embryonic stem (ES) cell population and maintain pluripotency. Here, we show that to regulate mitotic progression and pluripotency ES cells must keep the aryl hydrocarbon receptor (AHR), an environmental sensor and transcriptional regulator, in a persistent state of repression. This repression, however, is not always absolute, causing the AHR to fluctuate between reversible states of expression and repression, with a fraction of the cells escaping repression at any one time. Cells that escape AHR repression exhibit reduced levels of the pluripotency factors OCT4 and SOX2 and show an extended mitotic traverse time due to AHR-dependent MID1 repression and the subsequent disruption of the MID1-PP2A-CDC25B-CDK1 signaling pathway that regulates mitosis. Unlike the bulk of the cell population that differentiates into cardiomyocytes upon stimulation, AHR-expressing ES cells restrict cardiogenesis and commit to a neuroglia cell fate. It appears that the untimely expression of the Ahr gene needs to be repressed to maintain ES cell mitotic progression and prevent premature loss of pluripotency.
- Published
- 2016
24. Lead Exposure during Early Human Development and DNA Methylation of Imprinted Gene Regulatory Elements in Adulthood
- Author
-
Yue Li, Alvaro Puga, Cathrine Hoyo, Randy L. Jirtle, David Skaar, Changchun Xie, Monica D. Nye, Kim M. Cecil, Adriana C. Vidal, Kim N. Dietrich, and Susan K. Murphy
- Subjects
Male ,0301 basic medicine ,Health, Toxicology and Mutagenesis ,Cell Cycle Proteins ,Biology ,03 medical and health sciences ,Tumor suppressor proteins ,Child Development ,Insulin-Like Growth Factor II ,Humans ,Transcription factor ,Insulin-like growth factor-II ,Genetics ,Tumor Suppressor Proteins ,Infant, Newborn ,Public Health, Environmental and Occupational Health ,Infant ,Environmental Exposure ,Environmental exposure ,DNA Methylation ,Infant newborn ,3. Good health ,030104 developmental biology ,Lead ,Child, Preschool ,Children's Health ,Lead exposure ,DNA methylation ,Environmental Pollutants ,Female ,Genomic imprinting ,Transcription Factors - Abstract
Background: Lead exposure during early development causes neurodevelopmental disorders by unknown mechanisms. Epidemiologic studies have focused recently on determining associations between lead exposure and global DNA methylation; however, such approaches preclude the identification of loci that may alter human disease risk. Objectives: The objective of this study was to determine whether maternal, postnatal, and early childhood lead exposure can alter the differentially methylated regions (DMRs) that control the monoallelic expression of imprinted genes involved in metabolism, growth, and development. Methods: Questionnaire data and serial blood lead levels were obtained from 105 participants (64 females, 41 males) of the Cincinnati Lead Study from birth to 78 months. When participants were adults, we used Sequenom EpiTYPER assays to test peripheral blood DNA to quantify CpG methylation in peripheral blood leukocytes at DMRs of 22 human imprinted genes. Statistical analyses were conducted using linear regression. Results: Mean blood lead concentration from birth to 78 months was associated with a significant decrease in PEG3 DMR methylation (β = –0.0014; 95% CI: –0.0023, –0.0005, p = 0.002), stronger in males (β = –0.0024; 95% CI: –0.0038, –0.0009, p = 0.003) than in females (β = –0.0009; 95% CI: –0.0020, 0.0003, p = 0.1). Elevated mean childhood blood lead concentration was also associated with a significant decrease in IGF2/H19 (β = –0.0013; 95% CI: –0.0023, –0.0003, p = 0.01) DMR methylation, but primarily in females, (β = –0.0017; 95% CI: –0.0029, –0.0006, p = 0.005) rather than in males, (β = –0.0004; 95% CI: –0.0023, 0.0015, p = 0.7). Elevated blood lead concentration during the neonatal period was associated with higher PLAGL1/HYMAI DMR methylation regardless of sex (β = 0.0075; 95% CI: 0.0018, 0.0132, p = 0.01). The magnitude of associations between cumulative lead exposure and CpG methylation remained unaltered from 30 to 78 months. Conclusions: Our findings provide evidence that early childhood lead exposure results in sex-dependent and gene-specific DNA methylation differences in the DMRs of PEG3, IGF2/H19, and PLAGL1/HYMAI in adulthood. Citation: Li Y, Xie C, Murphy SK, Skaar D, Nye M, Vidal AC, Cecil KM, Dietrich KN, Puga A, Jirtle RL, Hoyo C. 2016. Lead exposure during early human development and DNA methylation of imprinted gene regulatory elements in adulthood. Environ Health Perspect 124:666–673; http://dx.doi.org/10.1289/ehp.1408577
- Published
- 2016
25. Ah receptor expression in cardiomyocytes protects adult female mice from heart dysfunction induced by TCDD exposure
- Author
-
Mindi Naticchioni, Alvaro Puga, Hisaka Kurita, Jack Rubinstein, Min Jiang, Sheryl E. Koch, Yunxia Fan, and Vinicius Carreira
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Polychlorinated Dibenzodioxins ,Time Factors ,Cre recombinase ,Blood Pressure ,Toxicology ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Sex Factors ,0302 clinical medicine ,Fibrosis ,Internal medicine ,medicine ,Animals ,Myocytes, Cardiac ,heterocyclic compounds ,Mice, Knockout ,Regulation of gene expression ,biology ,Body Weight ,Aryl hydrocarbon receptor ,medicine.disease ,stomatognathic diseases ,030104 developmental biology ,Endocrinology ,Gene Expression Regulation ,Receptors, Aryl Hydrocarbon ,chemistry ,Toxicity ,Knockout mouse ,biology.protein ,Calcium ,Environmental Pollutants ,Female ,Signal transduction ,030217 neurology & neurosurgery ,Signal Transduction ,Toxicant - Abstract
Epidemiological studies in humans and experimental work in rodents suggest that exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a persistent environmental toxicant, is associated with incidence of heart disease. Although TCDD toxicity depends by and large on the aryl hydrocarbon receptor (AHR), the role of the cardiac AHR in TCDD induced cardiovascular disease is not well defined. To determine whether the Ahr gene mediates disruption of heart function by TCDD, we generated a cardiomyocyte-specific Ahr knockout mouse by crossing Ahr(fx/fx) mice with βMhc:cre/+ mice, in which expression of Cre recombinase is driven by the promoter of the βMhc (myosin heavy chain-beta) gene. Starting at three months of age, mice with cardiomyocyte-specific Ahr ablation were exposed to 1μg/kg/week of TCDD or control vehicle by oral gavage for an additional three months. Relative to unexposed controls, TCDD-exposure induced cardiomyocyte Ahr-independent changes in males but not females, including a significant increase in body weight, blood pressure, and cardiac hypertrophy and a decrease in cardiac ejection fraction. TCDD exposure also induced cardiomyocyte Ahr-dependent changes in fibrosis and calcium signaling gene expression in both males and females. TCDD exposure appears to cause sexually dimorphic effects on heart function and induce fibrosis and changes in calcium signaling in both males and females through activation of the cardiomyocyte-specific Ahr.
- Published
- 2016
26. Quinone-mediated induction of cytochrome P450 1A1 in HepG2 cells through increased interaction of aryl hydrocarbon receptor with aryl hydrocarbon receptor nuclear translocator
- Author
-
Hsinyu Lee, Yoshito Kumagai, Yumi Abiko, Alvaro Puga, and Fang-Yu Lin
- Subjects
0301 basic medicine ,Aryl hydrocarbon receptor nuclear translocator ,Metabolite ,Active Transport, Cell Nucleus ,Fluorescent Antibody Technique ,Transfection ,Toxicology ,03 medical and health sciences ,chemistry.chemical_compound ,Downregulation and upregulation ,Basic Helix-Loop-Helix Transcription Factors ,Benzoquinones ,Cytochrome P-450 CYP1A1 ,Humans ,Cytochrome P450, family 1, member A1 ,RNA, Messenger ,Naphthalene ,Dose-Response Relationship, Drug ,biology ,Chemistry ,Aryl Hydrocarbon Receptor Nuclear Translocator ,Quinones ,Cytochrome P450 ,Hep G2 Cells ,respiratory system ,Aryl hydrocarbon receptor ,Quinone ,030104 developmental biology ,Receptors, Aryl Hydrocarbon ,Biochemistry ,Enzyme Induction ,Hepatocytes ,biology.protein ,Naphthoquinones - Abstract
While it has long been believed that benzenes and naphthalenes are unable to activate the aryl hydrocarbon receptor (AhR) because they are poor ligands, we recently reported that these quinoid metabolites upregulated cytochrome P450 1A1 (CYP1A1) in Hepa1c1c7 cells (Abiko et al., 2015). In the current study, AhR activation, measured with a bioluminescence-based cell free assay, was induced by 1,2-naphthoquinone (1,2-NQ), a metabolite of naphthalene. Consistent with this, 1,4-benzoquinone (1,4-BQ), tert-butyl-1,4-BQ, and 1,4-NQ, as well as 1,2-NQ, all electrophilic mono- and bi-cyclic quinones, upregulated CYP1A1 mRNA and protein in HepG2 cells, whereas their parent aromatic hydrocarbons had little effect. Furthermore, immunofluorescence analysis confirmed that these quinones enhanced translocation of AhR to the nucleus.
- Published
- 2016
27. Editorial overview: Nrf2 in Toxicology: An update
- Author
-
Alvaro Puga, Kendall B. Wallace, Jeffrey S. Johnson, and Donna D. Zhang
- Subjects
Engineering ,business.industry ,Engineering ethics ,Toxicology ,business - Published
- 2016
28. Old Receptor, New Tricks—The Ever-Expanding Universe of Aryl Hydrocarbon Receptor Functions. Report from the 4th AHR Meeting, 29–31 August 2018 in Paris, France
- Author
-
B. Paige Lawrence, Gary H. Perdew, David H. Sherr, Robert Barouki, Alvaro Puga, Xavier Coumoul, Charlotte Esser, Leibniz Research Institute for Environmental Medicine [Düsseldorf, Germany] ( IUF), Environmental Medicine [Rochester, NY, USA], University of Rochester Medical Center (URMC), Department of Environmental Health [Boston, MA, USA], Boston University [Boston] (BU), Department of Veterinary and Biomedical Sciences [University Park, PA, USA] (Center for Molecular Toxicology and Carcinogenesis), Pennsylvania State University (Penn State), Penn State System-Penn State System, Department of Environmental Health [Cincinnati, OH, USA] (College of Medicine), University of Cincinnati (UC), Toxicité environnementale, cibles thérapeutiques, signalisation cellulaire (T3S - UMR_S 1124), Université Paris Descartes - Paris 5 (UPD5)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris Descartes - Faculté des Sciences Fondamentales et Biomédicales (UPD5 Sciences), Université Paris Descartes - Paris 5 (UPD5), The meeting was financially supported by L’alliance nationale pour les sciences de la vie et de la santé (AVIESAN), the Institut National de la Santé et de la Recherche Médicale (INSERM), the Fondation ARC pour la Recherche sur le Cancer (ARC), and the University of Paris Descartes. C.E. is funded by the Deutsche Forschungsgemeinschaft (ES103/7-1 and ES103/9-1). G.H.P. is funded by the National Institutes of Health (ES028244). D.H.S. is supported by P42 ES07381 and a grant from the Find The Cause Breast Cancer Foundation., Leibniz Research Institute for Environmental Medicine [Düsseldorf, Germany] (IUF), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), and Benoist, David
- Subjects
0301 basic medicine ,obesity ,translational science ,environmental health ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,barrier organs ,Catalysis ,lcsh:Chemistry ,Inorganic Chemistry ,03 medical and health sciences ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,stem cells ,Political science ,[SDV.EE.SANT] Life Sciences [q-bio]/Ecology, environment/Health ,cancer ,Physical and Theoretical Chemistry ,Receptor ,lcsh:QH301-705.5 ,Molecular Biology ,development ,Spectroscopy ,transcription factor ,[SDV.EE.SANT]Life Sciences [q-bio]/Ecology, environment/Health ,Physiological function ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,biology ,Biological phenomenon ,Organic Chemistry ,nervous system ,Conference Report ,General Medicine ,Aryl hydrocarbon receptor ,immunity ,3. Good health ,Computer Science Applications ,[SDV.TOX] Life Sciences [q-bio]/Toxicology ,030104 developmental biology ,lcsh:Biology (General) ,lcsh:QD1-999 ,[SDV.TOX]Life Sciences [q-bio]/Toxicology ,biology.protein ,Translational science ,diet ,Neuroscience ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
In a time where “translational„ science has become a mantra in the biomedical field, it is reassuring when years of research into a biological phenomenon suddenly points towards novel prevention or therapeutic approaches to disease, thereby demonstrating once again that basic science and translational science are intimately linked. The studies on the aryl hydrocarbon receptor (AHR) discussed here provide a perfect example of how years of basic toxicological research on a molecule, whose normal physiological function remained a mystery for so long, has now yielded a treasure trove of actionable information on the development of targeted therapeutics. Examples are autoimmunity, metabolic imbalance, inflammatory skin and gastro-intestinal diseases, cancer, development and perhaps ageing. Indeed, the AHR field no longer asks, “What does this receptor do in the absence of xenobiotics?„ It now asks, “What doesn’t this receptor do?„.
- Published
- 2018
29. Prenatal exposure to PCBs in Cyp1a2 knock-out mice interferes with F
- Author
-
Jillian R, Hufgard, Jenna L N, Sprowles, Emily M, Pitzer, Sheryl E, Koch, Min, Jiang, Qin, Wang, Xiang, Zhang, Jacek, Biesiada, Jack, Rubinstein, Alvaro, Puga, Michael T, Williams, and Charles V, Vorhees
- Subjects
Mice, Knockout ,Reflex, Startle ,Conditioning, Classical ,Long-Term Potentiation ,Motor Activity ,Polychlorinated Biphenyls ,Mice, Inbred C57BL ,Mice ,Fertility ,Cytochrome P-450 CYP1A2 ,Pregnancy ,Prenatal Exposure Delayed Effects ,Animals ,Environmental Pollutants ,Female ,Maze Learning ,CA1 Region, Hippocampal ,Spatial Memory - Abstract
Polychlorinated biphenyls (PCBs) are toxic environmental pollutants. Humans are exposed to PCB mixtures via contaminated food or water. PCB exposure causes adverse effects in adults and after exposure in utero. PCB toxicity depends on the congener mixture and CYP1A2 gene activity. For coplanar PCBs, toxicity depends on ligand affinity for the aryl hydrocarbon receptor (AHR). Previously, we found that perinatal exposure of mice to a three-coplanar/five-noncoplanar PCB mixture induced deficits in novel object recognition and trial failures in the Morris water maze in Cyp1a2
- Published
- 2018
30. Chromium disrupts chromatin organization and CTCF access to its cognate sites in promoters of differentially expressed genes
- Author
-
Hongxia Zhang, Andrew VonHandorf, Xiang Zhang, Mario Medvedovic, Jacek Biesiada, Francisco J. Sánchez-Martín, and Alvaro Puga
- Subjects
0301 basic medicine ,Chromium ,Cancer Research ,CCCTC-Binding Factor ,ATAC-seq ,Biology ,Cell Line ,03 medical and health sciences ,Mice ,Transcription (biology) ,Nucleosome ,Animals ,Promoter Regions, Genetic ,Molecular Biology ,Transcription factor ,Binding Sites ,Drinking Water ,Promoter ,Environmental exposure ,Sequence Analysis, DNA ,Chromatin Assembly and Disassembly ,Chromatin ,Cell biology ,030104 developmental biology ,Gene Expression Regulation ,Liver ,CTCF ,Research Paper ,Protein Binding - Abstract
Hexavalent chromium compounds are well-established respiratory carcinogens used in industrial processes. While inhalation exposure constitutes an occupational risk affecting mostly chromium workers, environmental exposure from drinking water is a widespread gastrointestinal cancer risk, affecting millions of people throughout the world. Cr(VI) is genotoxic, forming protein-Cr-DNA adducts and silencing tumor suppressor genes, but its mechanism of action at the molecular level is poorly understood. Our prior work using FAIRE showed that Cr(VI) disrupted the binding of transcription factors CTCF and AP-1 to their cognate chromatin sites. Here, we used two complementary approaches to test the hypothesis that chromium perturbs chromatin organization and dynamics. DANPOS2 analyses of MNase-seq data identified several chromatin alterations induced by Cr(VI) affecting nucleosome architecture, including occupancy changes at specific genome locations; position shifts of 10 nucleotides or more; and changes in position amplitude or fuzziness. ATAC-seq analysis revealed that Cr(VI) disrupted the accessibility of chromatin regions enriched for CTCF and AP-1 binding motifs, with a significant co-occurrence of binding sites for both factors in the same region. Cr(VI)-enriched CTCF sites were confirmed by ChIP-seq and found to correlate with evolutionarily conserved sites occupied by CTCF in vivo, as determined by comparison with ENCODE-validated CTCF datasets from mouse liver. In addition, more than 30% of the Cr(VI)-enriched CTCF sites were located in promoters of genes differentially expressed from chromium treatment. Our results support the conclusion that Cr(VI) exposure promotes broad changes in chromatin accessibility and suggest that the subsequent effects on transcription regulation may result from disruption of CTCF binding and nucleosome spacing, implicating transcription regulatory mechanisms as primary Cr(VI) targets.
- Published
- 2018
31. Epigenetics as a mechanism linking developmental exposures to long-term toxicity
- Author
-
Rémy Slama, Jia Chen, Toshihiro Kawamoto, Alvaro Puga, Karine Audouze, Zdenko Herceg, K Nohara, Margaret R. Karagas, Erik Melén, W Yan, Robert Barouki, Y Xia, Philippe Grandjean, L Chadwick, J.J. Heindel, and J Beckers
- Subjects
Male ,0301 basic medicine ,Prenatal Programming ,Disease ,010501 environmental sciences ,Biology ,Long term toxicity ,01 natural sciences ,Article ,Mechanistic toxicology ,Epigenesis, Genetic ,Developmental psychology ,03 medical and health sciences ,Pregnancy ,Humans ,Epigenetics ,lcsh:Environmental sciences ,0105 earth and related environmental sciences ,General Environmental Science ,Developmental Vulnerability ,Endocrine Disrupting Compounds ,Mechanistic Toxicology ,Particulate Matter ,Tobacco Smoking ,lcsh:GE1-350 ,Mechanism (biology) ,Stressor ,Environmental Exposure ,Environmental exposure ,DNA Methylation ,Tobacco smoking ,030104 developmental biology ,Developmental vulnerability ,Endocrine disrupting compounds ,Environmental Pollutants ,Female ,Biological plausibility ,Particulate matter - Abstract
A variety of experimental and epidemiological studies lend support to the Developmental Origin of Health and Disease (DOHaD) concept. Yet, the actual mechanisms accounting for mid- and long-term effects of early-life exposures remain unclear. Epigenetic alterations such as changes in DNA methylation, histone modifications and the expression of certain RNAs have been suggested as possible mediators of long-term health effects of environmental stressors.This report captures discussions and conclusions debated during the last Prenatal Programming and Toxicity meeting held in Japan. Its first aim is to propose a number of criteria that are critical to support the primary contribution of epigenetics in DOHaD and intergenerational transmission of environmental stressors effects. The main criteria are the full characterization of the stressors, the actual window of exposure, the target tissue and function, the specificity of the epigenetic changes and the biological plausibility of the linkage between those changes and health outcomes. The second aim is to discuss long-term effects of a number of stressors such as smoking, air pollution and endocrine disruptors in order to identify the arguments supporting the involvement of an epigenetic mechanism. Based on the developed criteria, missing evidence and suggestions for future research will be identified. The third aim is to critically analyze the evidence supporting the involvement of epigenetic mechanisms in intergenerational and transgenerational effects of environmental exposure and to particularly discuss the role of placenta and sperm.While the article is not a systematic review and is not meant to be exhaustive, it critically assesses the contribution of epigenetics in the long-term effects of environmental exposures as well as provides insight for future research. Keywords: Endocrine disrupting compounds, Tobacco smoking, Particulate matter, Developmental vulnerability, Mechanistic toxicology
- Published
- 2018
32. Aryl Hydrocarbon Receptor
- Author
-
Qin Wang, Andrew VonHandorf, and Alvaro Puga
- Published
- 2018
33. Gene-Environment Interactions Target Mitogen-activated Protein 3 Kinase 1 (MAP3K1) Signaling in Eyelid Morphogenesis
- Author
-
Jingjing Wang, Winston W.-Y. Kao, Alvaro Puga, Ying Xia, Maureen Mongan, and Qinghang Meng
- Subjects
medicine.medical_specialty ,Mutant ,Morphogenesis ,MAP Kinase Kinase Kinase 1 ,Gene mutation ,Dioxins ,Biochemistry ,Epithelium ,Mice ,Internal medicine ,medicine ,Animals ,Phosphorylation ,Molecular Biology ,biology ,JNK Mitogen-Activated Protein Kinases ,Wnt signaling pathway ,Wild type ,Eyelids ,Gene Expression Regulation, Developmental ,Cell Biology ,Embryo, Mammalian ,Aryl hydrocarbon receptor ,Cell biology ,Endocrinology ,Receptors, Aryl Hydrocarbon ,biology.protein ,Environmental Pollutants ,Gene-Environment Interaction ,Signal transduction ,Signal Transduction - Abstract
Gene-environment interactions determine the biological outcomes through mechanisms that are poorly understood. Mouse embryonic eyelid closure is a well defined model to study the genetic control of developmental programs. Using this model, we investigated how exposure to dioxin-like environmental pollutants modifies the genetic risk of developmental abnormalities. Our studies reveal that mitogen-activated protein 3 kinase 1 (MAP3K1) signaling is a focal point of gene-environment cross-talk. Dioxin exposure, acting through the aryl hydrocarbon receptor (AHR), blocked eyelid closure in genetic mutants in which MAP3K1 signaling was attenuated but did not disturb this developmental program in either wild type or mutant mice with attenuated epidermal growth factor receptor or WNT signaling. Exposure also markedly inhibited c-Jun phosphorylation in Map3k1(+/-) embryonic eyelid epithelium, suggesting that dioxin-induced AHR pathways can synergize with gene mutations to inhibit MAP3K1 signaling. Our studies uncover a novel mechanism through which the dioxin-AHR axis interacts with the MAP3K1 signaling pathways during fetal development and provide strong empirical evidence that specific gene alterations can increase the risk of developmental abnormalities driven by environmental pollutant exposure.
- Published
- 2015
34. Ah Receptor Signaling Controls the Expression of Cardiac Development and Homeostasis Genes
- Author
-
Qing Wang, Mario Medvedovic, Yunxia Fan, Sheryl E. Koch, Mindi Naticchioni, Chia-I Ko, Min Jiang, Ying Xia, Jack Rubinstein, Xiang Zhang, Vinicius Carreira, Hisaka Kurita, and Alvaro Puga
- Subjects
Male ,Cardiac function curve ,medicine.medical_specialty ,Heart disease ,Mitochondrion ,Biology ,Toxicology ,Mitochondria, Heart ,Mice ,Microscopy, Electron, Transmission ,Internal medicine ,medicine ,Animals ,Homeostasis ,Transcription factor ,Ejection fraction ,Myocardium ,Heart ,Aryl hydrocarbon receptor ,medicine.disease ,Mice, Inbred C57BL ,Endocrinology ,Receptors, Aryl Hydrocarbon ,Echocardiography ,biology.protein ,Aryl Hydrocarbon Receptor Mediation of Heart Development ,Female ,Signal transduction ,Signal Transduction - Abstract
Congenital heart disease (CHD) is the most common congenital abnormality and one of the leading causes of newborn death throughout the world. Despite much emerging scientific information, the precise etiology of this disease remains elusive. Here, we show that the aryl hydrocarbon receptor (AHR) regulates the expression of crucial cardiogenesis genes and that interference with endogenous AHR functions, either by gene ablation or by agonist exposure during early development, causes overlapping structural and functional cardiac abnormalities that lead to altered fetal heart physiology, including higher heart rates, right and left ventricle dilation, higher stroke volume, and reduced ejection fraction. With striking similarity between AHR knockout (Ahr � /� ) and agonist-exposed wild type (Ahr þ/þ ) embryos, in utero disruption of endogenous AHR functions converge into dysregulation of molecular mechanisms needed for attainment and maintenance of cardiac differentiation, including the pivotal signals regulated by the cardiogenic transcription factor NKH2.5, energy balance via oxidative phosphorylation and TCA cycle and global mitochondrial function and homeostasis. Our findings suggest that AHR signaling in the developing mammalian heart is central to the regulation of pathways crucial for cellular metabolism, cardiogenesis, and cardiac function, which are potential targets of environmental factors associated with CHD.
- Published
- 2015
35. Long-term Coexposure to Hexavalent Chromium and B[a]P Causes Tissue-Specific Differential Biological Effects in Liver and Gastrointestinal Tract of Mice
- Author
-
Alvaro Puga, Andrew VonHandorf, Jerald L. Ovesen, Ying Xia, Francisco J. Sánchez-Martín, Yunxia Fan, and Vinicius Carreira
- Subjects
Chromium ,medicine.medical_specialty ,DNA damage ,Enterocyte ,Chromium and Benzo(A)Pyrene Co-Exposure Leads to Variable Liver and GI Tract Effects ,Inflammation ,Biology ,Toxicology ,medicine.disease_cause ,Mice ,chemistry.chemical_compound ,Internal medicine ,Benzo(a)pyrene ,medicine ,Animals ,Gastrointestinal tract ,Gastrointestinal Tract ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Endocrinology ,Liver ,chemistry ,Immunology ,medicine.symptom ,Drug metabolism ,Oxidative stress ,Toxicant - Abstract
Complex mixtures of environmental agents often cause mixture-specific health effects that cannot be accounted for by a single mechanism. To study the biological effects of exposure to a mixture of chromium-VI and benzo[a]pyrene (B[a]P), often found together in the environment, we exposed mice for 60 days to 0, 55, 550, or 5500 ppb Cr(VI) in drinking water followed by 90 days of coexposure to B[a]P at 0, 1.25, 12.5, or 125 mg/kg/day and examined liver and gastrointestinal (GI) tract for exposure effects. In the liver, the mixture caused more significant histopathology than expected from the sum of effects of the individual components, while in the GI tract, Cr(VI) alone caused significant enterocyte hypertrophy and increases in cell proliferation and DNA damage that were also observed in mice coexposed to B[a]P. Expression of genes involved in drug metabolism, tumor suppression, oxidative stress, and inflammation was altered in mixed exposures relative to control and to singly exposed mice. Drug metabolism and oxidative stress genes were upregulated and tumor suppressor and inflammation genes downregulated in the proximal GI tract, whereas most markers were upregulated in the distal GI tract and downregulated in the liver. Oral exposure to Cr(VI) and B[a]P mixtures appears to have tissue-specific differential consequences in liver and GI tract that cannot be predicted from the effects of each individual toxicant. Tissue specificity may be particularly critical in cases of extended exposure to mixtures of these agents, as may happen in the occupational setting or in areas where drinking water contains elevated levels of Cr(VI).
- Published
- 2015
36. Sex- and tissue-specific methylome changes in brains of mice perinatally exposed to lead
- Author
-
Jing Chen, Julio A. Landero-Figueroa, Mario Medvedovic, Francisco J. Sánchez-Martín, Diana M. Lindquist, Kim M. Cecil, Xiang Zhang, and Alvaro Puga
- Subjects
Male ,Hippocampus ,Biology ,Toxicology ,Article ,Andrology ,Mice ,Animals ,RNA, Messenger ,Epigenetics ,Regulation of gene expression ,Sex Characteristics ,General Neuroscience ,Brain ,Chromosome Mapping ,Gene Expression Regulation, Developmental ,Methylation ,DNA Methylation ,Molecular biology ,Mice, Inbred C57BL ,Differentially methylated regions ,Animals, Newborn ,Lead ,CpG site ,Lead acetate ,DNA methylation ,CpG Islands ,Female - Abstract
A B S T R A C T Changes in DNA methylation and subsequent changes in gene expression regulation are the hallmarks of age- and tissue-dependent epigenetic drift and plasticity resulting from the combinatorial integration of genetic determinants and environmental cues. To determine whether perinatal lead exposure caused persistent DNA methylation changes in target tissues, we exposed mouse dams to 0, 3 or 30 ppm of lead acetate in drinking water for a period extending from 2 months prior to mating, through gestation, until weaning of pups at postnatal day-21, and analyzed whole-genome DNA methylation in brain cortex and hippocampus of 2-month old exposed and unexposed progeny. Lead exposure resulted in hypermethylation of three differentially methylated regions in the hippocampus of females, but not males. These regions mapped to Rn4.5s, Sfi1, and Rn45s loci in mouse chromosomes 2, 11 and 17, respectively. At a conservative fdr < 0.001, 1623 additional CpG sites were differentially methylated in female hippocampus, corresponding to 117 unique genes. Sixty of these genes were tested for mRNA expression and showed a trend toward negative correlation between mRNA expression and methylation in exposed females but not males. No statistically significant methylome changes were detected in male hippocampus or in cortex of either sex. We conclude that exposure to lead during embryonic life, a time when the organism is most sensitive to environmental cues, appears to have a sex- and tissue-specific effect on DNA methylation that may produce pathological or physiological deviations from the epigenetic plasticity operative in unexposed mice.
- Published
- 2015
37. Covalent binding of quinones activates the Ah receptor in Hepa1c1c7 cells
- Author
-
Yumi Abiko, Yoshito Kumagai, and Alvaro Puga
- Subjects
Carcinoma, Hepatocellular ,Aryl hydrocarbon receptor nuclear translocator ,NF-E2-Related Factor 2 ,Active Transport, Cell Nucleus ,Cellular homeostasis ,Plasma protein binding ,Toxicology ,Mice ,chemistry.chemical_compound ,Cytochrome P-450 CYP1A1 ,Tumor Cells, Cultured ,Animals ,Luciferase ,Promoter Regions, Genetic ,biology ,Aryl Hydrocarbon Receptor Nuclear Translocator ,Liver Neoplasms ,Quinones ,Cytochrome P450 ,respiratory system ,Aryl hydrocarbon receptor ,Antioxidant Response Elements ,Receptors, Aryl Hydrocarbon ,chemistry ,Biochemistry ,biology.protein ,Signal transduction ,Xenobiotic ,Protein Binding ,Signal Transduction - Abstract
Highly reactive quinone species produced by photooxidation and/or metabolic activation of mono- or bi-aromatic hydrocarbons modulate cellular homeostasis and electrophilic signal transduction pathways through the covalent modification of proteins. Polycyclic aromatic hydrocarbons, but not mono- or bi-aromatic hydrocarbons, are well recognized as ligands for the aryl hydrocarbon receptor (AhR). However, quinone species produced from mono- and bi-aromatic hydrocarbons could potentially cause AhR activation. To clarify the AhR response to mono- and bi-aromatic hydrocarbon quinones, we studied Cyp1a1 (cytochrome P450 1A1) induction and AhR activation by these quinones. We detected Cyp1a1 induction during treatment with quinones in Hepa1c1c7 cells, but not their parent compounds. Nine of the twelve quinones with covalent binding capability for proteins induced Cyp1a1. Cyp1a1 induction mediated by 1,2-naphthoquinone (1,2-NQ), 1,4-NQ, 1,4-benzoquinone (1,4-BQ) and tert-butyl-1,4-BQ was suppressed by a specific AhR inhibitor and was not observed in c35 cells, which do not have a functional AhR. These quinones stimulated AhR nuclear translocation and interaction with the AhR nuclear translocator. Interestingly, 1,2-NQ covalently modified AhR, which was detected by an immunoprecipitation assay using a specific antibody against 1,2-NQ, resulting in enhancement of xenobiotic responsive element (XRE)-derived luciferase activity and binding of AhR to the Cyp1a1 promoter region. While mono- and bi-aromatic hydrocarbons are generally believed to be poor ligands for AhR and hence unable to induce Cyp1a1, our study suggests that the quinones of these molecules are able to modify AhR and activate the AhR/XRE pathway, thereby inducing Cyp1a1. Since we previously reported that 1,2-NQ and tert-butyl-1,4-BQ also activate NF-E2-related factor 2, it seems likely that some of quinones are bi-functional inducers for phase-I and phase-II reaction of xenobiotics.
- Published
- 2015
38. Aryl Hydrocarbon Receptor Ablation in Cardiomyocytes Protects Male Mice From Heart Dysfunction Induced by NKX2.5 Haploinsufficiency
- Author
-
Jack Rubinstein, Min Jiang, Hisaka Kurita, Qin Wang, Sheryl E. Koch, Alvaro Puga, Marepalli B Rao, and Yunxia Fan
- Subjects
0301 basic medicine ,Cardiac function curve ,Cardiomyopathy, Dilated ,Male ,medicine.medical_specialty ,Polychlorinated Dibenzodioxins ,Heart disease ,Cre recombinase ,Aryl Hydrocarbon Receptor in Cardiac Differentiation and Dysfunction ,Cardiomegaly ,Haploinsufficiency ,Toxicology ,03 medical and health sciences ,Sex Factors ,Internal medicine ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Ventricular Dysfunction ,Animals ,Ventricular Function ,Genetic Predisposition to Disease ,Myocytes, Cardiac ,Promoter Regions, Genetic ,Gene knockout ,Mice, Knockout ,Ejection fraction ,biology ,business.industry ,Age Factors ,Dilated cardiomyopathy ,Stroke Volume ,respiratory system ,medicine.disease ,Aryl hydrocarbon receptor ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Phenotype ,Receptors, Aryl Hydrocarbon ,biology.protein ,cardiovascular system ,Homeobox Protein Nkx-2.5 ,Environmental Pollutants ,Female ,Gene-Environment Interaction ,business - Abstract
Epidemiological studies in humans and research in vertebrates indicates that developmental exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a ubiquitous and biopersistent environmental toxicant, is associated with incidence of early congenital heart disease in the embryo and later in the adult. TCDD-mediated toxicity depends on the aryl hydrocarbon receptor (AHR) but the role of the TCDD-activated AHR in cardiac function is not well-defined. To characterize the mechanisms responsible for AHR-mediated disruption of heart function, we generated several mouse strains with cardiomyocyte-specific Ahr gene knockout. Here, we report results on one of these strains in which the Ahr gene was deleted by cre recombinase regulated by the promoter of the cardiomyocyte-specific Nkx2.5 gene. We crossed mice with loxP-targeted Ahrfx/fx alleles with Nkx2.5+/cre mice bearing a "knock-in" cre recombinase gene integrated into one of the Nkx2.5 alleles. In these mice, loss of one Nkx2.5 allele is associated with disrupted cardiac development. In males, Nkx2.5 hemizygosity resulted in cardiac haploinsufficiency characterized by hypertrophy, dilated cardiomyopathy, and impaired ejection fraction. Ahr ablation protected Nkx2.5+/cre haploinsufficient males from cardiac dysfunction while inducing a significant increase in body weight. These effects were absent or largely blunted in females. Starting at 3 months of age, mice were exposed by oral gavage to 1 μg/kg/week of TCDD or control vehicle for an additional 2 months. TCDD exposure restored cardiac physiology in aging males, appearing to compensate for the heart dysfunction caused by Nkx2.5 hemizygosity. Our findings underscore the conclusion that deletion of the Ahr gene in cardiomyocytes protects males from heart dysfunction due to NKX2.5 haploinsufficiency.
- Published
- 2017
39. Does the Aryl Hydrocarbon Receptor Regulate Pluripotency?
- Author
-
Alvaro Puga and Chia-I Ko
- Subjects
0301 basic medicine ,Genetics ,Homeobox protein NANOG ,Cellular differentiation ,Biology ,Cell fate determination ,respiratory system ,Toxicology ,Blastula ,Aryl hydrocarbon receptor ,Embryonic stem cell ,Article ,Chromatin ,Cell biology ,respiratory tract diseases ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,embryonic structures ,biology.protein ,Reprogramming ,030217 neurology & neurosurgery - Abstract
Recent evidence from embryonic stem cells suggests that the aryl hydrocarbon receptor (AHR) plays a central role in the regulation of pluripotency, a short-lived property of cells in the early blastula inner cell mass (ICM). Four key observations support this conclusion. The first is the temporal association between upregulation of AHR expression and the onset of cell differentiation, which argues for the AHR as a determinant of cell fate decisions. The second is the repression of the pluripotency factors OCT4 and NANOG by the AHR, which depresses their function and contributes to the cell's exit from pluripotency. The third is the temporal association between changes in global DNA methylation and stage-dependent AHR expression, which parallel each other during embryonic development, suggesting that AHR helps configure a repressive chromatin structure that controls differentiation. The fourth is the incidence of early developmental aberrations that take place in Ahr-null mice and cause the disruption of their embryonic program, which is likely to be a consequence of the loss of pluripotency of the Ahr−/− ICM cells. In this short review, we will focus on the modulation of pluripotency as a novel function of the AHR, and on the potentially detrimental developmental outcomes that may result from exposure to environmental toxicants. This line of enquiry brings us to the tantalizing conclusion that by activating mechanisms that modulate pluripotency, AHR regulates embryonic development. The likelihood that exposure to environmental AHR ligands might disrupt developmental processes is a reasonable corollary to this conclusion.
- Published
- 2017
40. Pluripotency factors and Polycomb Group proteins repress aryl hydrocarbon receptor expression in murine embryonic stem cells
- Author
-
Ying Xia, Alvaro Puga, Qin Wang, Chia-I Ko, and Yunxia Fan
- Subjects
Homeobox protein NANOG ,Cellular differentiation ,Polycomb-Group Proteins ,Trithorax-group proteins ,Article ,Histones ,Kruppel-Like Factor 4 ,Mice ,SOX2 ,Polycomb-group proteins ,Animals ,Phosphorylation ,Promoter Regions, Genetic ,lcsh:QH301-705.5 ,Cells, Cultured ,Embryonic Stem Cells ,Medicine(all) ,biology ,Cell Differentiation ,Cell Biology ,General Medicine ,respiratory system ,Aryl hydrocarbon receptor ,Molecular biology ,Chromatin ,respiratory tract diseases ,Mice, Inbred C57BL ,Sp3 Transcription Factor ,lcsh:Biology (General) ,Gene Expression Regulation ,Receptors, Aryl Hydrocarbon ,biology.protein ,RNA Polymerase II ,Transcription Initiation Site ,Chromatin immunoprecipitation ,Protein Binding ,Transcription Factors ,Developmental Biology - Abstract
The aryl hydrocarbon receptor (AHR) is a transcription factor and environmental sensor that regulates expression of genes involved in drug-metabolism and cell cycle regulation. Chromatin immunoprecipitation analyses, Ahr ablation in mice and studies with orthologous genes in invertebrates suggest that AHR may also play a significant role in embryonic development. To address this hypothesis, we studied the regulation of Ahr expression in mouse embryonic stem cells and their differentiated progeny. In ES cells, interactions between OCT3/4, NANOG, SOX2 and Polycomb Group proteins at the Ahr promoter repress AHR expression, which can also be repressed by ectopic expression of reprogramming factors in hepatoma cells. In ES cells, unproductive RNA polymerase II binds at the Ahr transcription start site and drives the synthesis of short abortive transcripts. Activation of Ahr expression during differentiation follows from reversal of repressive marks in Ahr promoter chromatin, release of pluripotency factors and PcG proteins, binding of Sp factors, establishment of histone marks of open chromatin, and engagement of active RNAPII to drive full-length RNA transcript elongation. Our results suggest that reversible Ahr repression in ES cells holds the gene poised for expression and allows for a quick switch to activation during embryonic development.
- Published
- 2014
41. Disruption of Aryl Hydrocarbon Receptor Homeostatic Levels during Embryonic Stem Cell Differentiation Alters Expression of Homeobox Transcription Factors that Control Cardiomyogenesis
- Author
-
Ying Xia, Alvaro Puga, Chia-I Ko, Vinicius Carreira, Jing Chen, Yinglei Chen, Mario Medvedovic, Qin Wang, and Yunxia Fan
- Subjects
Polychlorinated Dibenzodioxins ,Health, Toxicology and Mutagenesis ,Green Fluorescent Proteins ,Drug Resistance ,Homeobox A1 ,Fluorescent Antibody Technique ,Biology ,Muscle Development ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Mediator ,Animals ,Homeostasis ,Cell Lineage ,RNA, Small Interfering ,Transcription factor ,Embryonic Stem Cells ,030304 developmental biology ,Homeodomain Proteins ,0303 health sciences ,Reverse Transcriptase Polymerase Chain Reaction ,Sequence Analysis, RNA ,Gene Expression Profiling ,Research ,Public Health, Environmental and Occupational Health ,Cell Differentiation ,Heart ,Flavones ,Aryl hydrocarbon receptor ,Myocardial Contraction ,Molecular biology ,Embryonic stem cell ,Cell biology ,Mice, Inbred C57BL ,Gene Expression Regulation ,Receptors, Aryl Hydrocarbon ,chemistry ,Gene Knockdown Techniques ,biology.protein ,Pyrazoles ,Homeobox ,Puromycin ,Xenobiotic ,Azo Compounds ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Background: The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that regulates the expression of xenobiotic detoxification genes and is a critical mediator of gene–environment interactions. Many AHR target genes identified by genome-wide gene expression profiling have morphogenetic functions, suggesting that AHR may play a role in embryonic development. Objectives: To characterize the developmental functions of the AHR, we studied the consequences of AHR activation by the agonist 2,3,7,8-tetrachlorodibenzo-p-doxin (TCDD), and the result of its repression by the antagonists 6,2,4-trimethoxyflavone and CH 223191 or by short-hairpin RNA (shRNA)-mediated Ahr knockdown during spontaneous differentiation of embryonic stem (ES) cells into cardiomyocytes. Methods: We generated an AHR-positive cardiomyocyte lineage differentiated from mouse ES cells that expresses puromycin resistance and enhanced green fluorescent protein (eGFP) under the control of the Cyp1a1 (cytochrome P450 1a1) promoter. We used RNA sequencing (RNA.Seq) to analyze temporal trajectories of TCDD-dependent global gene expression in these cells during differentiation. Results: Activation, inhibition, and knockdown of Ahr significantly inhibited the formation of contractile cardiomyocyte nodes. Global expression analysis of AHR-positive cells showed that activation of the AHR/TCDD axis disrupted the concerted expression of genes that regulate multiple signaling pathways involved in cardiac and neural morphogenesis and differentiation, including dozens of genes encoding homeobox transcription factors and Polycomb and trithorax group proteins. Conclusions: Disruption of AHR expression levels resulted in gene expression changes that perturbed cardiomyocyte differentiation. The main function of the AHR during development appears to be the coordination of a complex regulatory network responsible for attainment and maintenance of cardiovascular homeostasis. Citation: Wang Q, Chen J, Ko CI, Fan Y, Carreira V, Chen Y, Xia Y, Medvedovic M, Puga A. 2013. Disruption of aryl hydrocarbon receptor homeostatic levels during embryonic stem cell differentiation alters expression of homeobox transcription factors that control cardiomyogenesis. Environ Health Perspect 121:1334–1343; http://dx.doi.org/10.1289/ehp.1307297
- Published
- 2013
42. Loss of NR2E3 represses AHR by LSD1 reprogramming, is associated with poor prognosis in liver cancer
- Author
-
Yuet-Kin Leung, Alvaro Puga, Tilak Khanal, Shuk-Mei Ho, Kyounghyun Kim, Dasom Kim, Vinothini Janakiram, Kwangmin Choi, Jiang Wang, and Sung-Gook Cho
- Subjects
0301 basic medicine ,medicine.medical_specialty ,lcsh:Medicine ,Kaplan-Meier Estimate ,Biology ,Models, Biological ,Article ,03 medical and health sciences ,Internal medicine ,Cell Line, Tumor ,Coactivator ,medicine ,Humans ,Gene Regulatory Networks ,lcsh:Science ,Regulation of gene expression ,Histone Demethylases ,Sp1 transcription factor ,Multidisciplinary ,lcsh:R ,Liver Neoplasms ,Cancer ,Computational Biology ,High-Throughput Nucleotide Sequencing ,respiratory system ,medicine.disease ,Aryl hydrocarbon receptor ,Cellular Reprogramming ,Orphan Nuclear Receptors ,Prognosis ,3. Good health ,respiratory tract diseases ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Endocrinology ,Histone ,Cell Transformation, Neoplastic ,Receptors, Aryl Hydrocarbon ,Cancer research ,biology.protein ,Demethylase ,lcsh:Q ,Liver cancer - Abstract
The aryl hydrocarbon receptor (AHR) plays crucial roles in inflammation, metabolic disorder, and cancer. However, the molecular mechanisms regulating AHR expression remain unknown. Here, we found that an orphan nuclear NR2E3 maintains AHR expression, and forms an active transcriptional complex with transcription factor Sp1 and coactivator GRIP1 in MCF-7 human breast and HepG2 liver cancer cell lines. NR2E3 loss promotes the recruitment of LSD1, a histone demethylase of histone 3 lysine 4 di-methylation (H3K4me2), to the AHR gene promoter region, resulting in repression of AHR expression. AHR expression and responsiveness along with H3K4me2 were significantly reduced in the livers of Nr2e3rd7 (Rd7) mice that express low NR2E3 relative to the livers of wild-type mice. SP2509, an LSD1 inhibitor, fully restored AHR expression and H3K4me2 levels in Rd7 mice. Lastly, we demonstrated that both AHR and NR2E3 are significantly associated with good clinical outcomes in liver cancer. Together, our results reveal a novel link between NR2E3, AHR, and liver cancer via LSD1-mediated H3K4me2 histone modification in liver cancer development.
- Published
- 2016
43. Epigenetic Histone Changes in the Toxicologic Mode of Action of Arsenic
- Author
-
Alvaro Puga and John F. Reichard
- Subjects
Genetics ,Histone ,biology ,chemistry ,biology.protein ,chemistry.chemical_element ,Cancer epigenetics ,Epigenetics ,Mode of action ,Arsenic - Published
- 2012
44. Distinct Signaling Properties of Mitogen-activated Protein Kinase Kinases 4 (MKK4) and 7 (MKK7) in Embryonic Stem Cell (ESC) Differentiation
- Author
-
Lin Zhang, Alvaro Puga, Liang Chen, Jingcai Wang, Chia-I Ko, and Ying Xia
- Subjects
Pluripotent Stem Cells ,Cell signaling ,MAP Kinase Kinase 4 ,MAP Kinase Signaling System ,Cellular differentiation ,p38 mitogen-activated protein kinases ,MAP Kinase Kinase 7 ,Cell fate determination ,Biology ,p38 Mitogen-Activated Protein Kinases ,Biochemistry ,Gene Expression Regulation, Enzymologic ,MAP2K7 ,Mice ,Animals ,Myocytes, Cardiac ,Phosphorylation ,Molecular Biology ,Cells, Cultured ,Embryonic Stem Cells ,Mice, Knockout ,Regulation of gene expression ,Activating Transcription Factor 2 ,MEF2 Transcription Factors ,Kinase ,Cell Differentiation ,Cell Biology ,Molecular biology ,Cell biology ,Enzyme Activation ,Myogenic Regulatory Factors ,Signal transduction ,Developmental Biology - Abstract
Signal transduction pathways are integral components of the developmental regulatory network that guides progressive cell fate determination. MKK4 and MKK7 are upstream kinases of the mitogen-activated protein kinases (MAPKs), responsible for channeling physiological and environmental signals to their cellular responses. Both kinases are essential for survival of mouse embryos, but because of embryonic lethality, their precise developmental roles remain largely unknown. Using gene knock-out mouse ESCs, we studied the roles of MKK4 and MKK7 in differentiation in vitro. While MKK4 and MKK7 were dispensable for ESC self-renewal and pluripotency maintenance, they exhibited unique signaling and functional properties in differentiation. MKK4 and MKK7 complemented each other in activation of the JNK-c-Jun cascades and loss of both led to senescence upon cell differentiation. On the other hand, MKK4 and MKK7 had opposite effects on activation of the p38 cascades during differentiation. Specifically, MKK7 reduced p38 activation, while Mkk7(-/-) ESCs had elevated phosphorylation of MKK4, p38, and ATF2, and increased MEF2C expression. Consequently, Mkk7(-/-) ESCs had higher expression of MHC and MLC and enhanced formation of contractile cardiomyocytes. In contrast, MKK4 was required for p38 activation and Mkk4(-/-) ESCs exhibited diminished p-ATF2 and MEF2C expression, resulting in impaired MHC induction and defective cardiomyocyte differentiation. Exogenous MKK4 expression partially restored the ability of Mkk4(-/-) ESCs to differentiate into cardiomyocytes. Our results uncover complementary and interdependent roles of MKK4 and MKK7 in development, and identify the essential requirement for MKK4 in p38 activation and cardiomyocyte differentiation.
- Published
- 2012
45. Mitogen-activated protein kinase kinase kinase 1 (MAP3K1) integrates developmental signals for eyelid closure
- Author
-
Atsushi Takatori, Esmond Geh, Alvaro Puga, Jingcai Wang, Qinghang Meng, Maureen Mongan, Richard A. Lang, Yi Zheng, and Ying Xia
- Subjects
rho GTP-Binding Proteins ,RHOA ,MAP Kinase Signaling System ,Proto-Oncogene Proteins c-jun ,Morphogenesis ,MAP Kinase Kinase Kinase 1 ,MAP3K1 ,Mitogen-activated protein kinase kinase ,Models, Biological ,Gene Expression Regulation, Enzymologic ,Mice ,Transactivation ,medicine ,Animals ,Promoter Regions, Genetic ,Protein kinase A ,Mice, Knockout ,rho-Associated Kinases ,Multidisciplinary ,biology ,Kinase ,Eyelids ,Gene Expression Regulation, Developmental ,Transforming Growth Factor alpha ,Biological Sciences ,eye diseases ,Cell biology ,ErbB Receptors ,Transcription Factor AP-1 ,medicine.anatomical_structure ,biology.protein ,Eyelid ,rhoA GTP-Binding Protein ,Signal Transduction - Abstract
Developmental eyelid closure is an evolutionarily conserved morphogenetic event requiring proliferation, differentiation, cytoskeleton reorganization, and migration of epithelial cells at the tip of the developing eyelid. Many signaling events take place during eyelid closure, but how the signals converge to regulate the morphogenetic process remains an open and intriguing question. Here we show that mitogen-activated protein kinase kinase kinase 1 (MAP3K1) highly expressed in the developing eyelid epithelium, forms with c-Jun, a regulatory axis that orchestrates morphogenesis by integrating two different networks of eyelid closure signals. A TGF-α/EGFR-RhoA module initiates one of these networks by inducing c-Jun expression which, in a phosphorylation-independent manner, binds to the Map3k1 promoter and causes an increase in MAP3K1 expression. RhoA knockout in the ocular surface epithelium disturbs this network by decreasing MAP3K1 expression, and causes delayed eyelid closure in Map3k1 hemizygotes. The second network is initiated by the enzymatic activity of MAP3K1, which phosphorylates and activates a JNK-c-Jun module, leading to AP-1 transactivation and induction of its downstream genes, such as Pai-1 . MAP3K1 inactivation reduces AP-1 activity and PAI-1 expression both in cells and developing eyelids. MAP3K1 is therefore the nexus of an intracrine regulatory loop connecting the TGF-α/EGFR/RhoA-c-Jun and JNK-c-Jun-AP-1 pathways in developmental eyelid closure.
- Published
- 2011
46. Aryl Hydrocarbon Receptor Ligands of Widely Different Toxic Equivalency Factors Induce Similar Histone Marks in Target Gene Chromatin
- Author
-
Michael Schnekenburger, Alvaro Puga, and Jerald L. Ovesen
- Subjects
Base Sequence ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,HDAC10 ,respiratory system ,SAP30 ,Ligands ,Toxicology ,Aryl hydrocarbon receptor ,Molecular biology ,Chromatin ,Histones ,Mice ,Histone H3 ,Histone ,Receptors, Aryl Hydrocarbon ,Molecular Toxicology ,Cell Line, Tumor ,Histone methyltransferase ,Histone methylation ,Histone H2A ,biology.protein ,Animals ,DNA Primers - Abstract
Posttranslational histone modifications are a critical regulatory mechanism of gene transcription. Previous studies from our laboratory have shown that contingent on binding to its cognate promoter motifs in the Cyp1a1 gene, activation of the aryl hydrocarbon receptor (AHR) by benzo[a]pyrene (BaP) treatment induces histone modifications in the Cyp1a1 promoter that are required for activation of gene transcription. Here, we have studied different AHR ligands, including polychlorinated biphenyls (PCBs) of different toxic equivalency factors (TEF), to determine whether changes in histone modifications are linked to different levels of Cyp1a1 expression or dependent on AHR-ligand affinity. We find that all ligands lead to the same pattern of histone modifications in a relationship that parallels the strength of their AHR-ligand affinity. Thus, whereas PCB126 (TEF 0.1), 3-methylcholanthrene, β-naphthoflavone, and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) initiate a pattern of histone marks similar to those induced by BaP, PCB77 (TEF 0.0001) causes a lower level of change in the same marks and requires a longer activation time than PCB126, BaP, or TCDD. In contrast, the non-dioxin-like PCB153 recruits AHR to the Cyp1a1 enhancer causing a displacement of enhancer-associated histone H3 but does not cause the other observed histone mark changes nor does it induce transcription. These results indicate that AHR recruitment to the promoter is not sufficient to induce the histone modifications needed to activate gene expression and show that there is a good correlation between the regulatory chromatin changes associated with ligand-induced AHR target gene transcription and the resultant toxicity of the ligand.
- Published
- 2011
47. Inhibitor of κB Kinase β Regulates Redox Homeostasis by Controlling the Constitutive Levels of Glutathione
- Author
-
Esmond Geh, Yunxia Fan, Ying Xia, Maureen A. Sartor, Zhimin Peng, Liang Chen, Howard G. Shertzer, Qinghang Meng, Zheng-Gang Liu, and Alvaro Puga
- Subjects
Cell Survival ,Blotting, Western ,Apoptosis ,Biology ,Mice ,chemistry.chemical_compound ,Genes, Reporter ,Animals ,Homeostasis ,Luciferases ,Cells, Cultured ,Tissue homeostasis ,DNA Primers ,Mice, Knockout ,Pharmacology ,chemistry.chemical_classification ,Reactive oxygen species ,Kinase ,GCLM ,NF-kappa B ,I-Kappa-B Kinase ,Articles ,Glutathione ,NFKB1 ,Molecular biology ,I-kappa B Kinase ,GCLC ,chemistry ,Molecular Medicine ,Reactive Oxygen Species ,Oxidation-Reduction ,Plasmids - Abstract
Cytokine-activated inhibitor of κB kinase β (IKKβ) is a key mediator of immune and inflammatory responses, but recent studies suggest that IKKβ is also required for tissue homeostasis in physiopathological processes. Here we report a novel role for IKKβ in maintenance of constitutive levels of the redox scavenger GSH. Inactivation of IKKβ by genetic or pharmacological means results in low cellular GSH content and marked reduction of redox potential. Similar to Ikkβ(−/−) cells, Tnfr1(−/−) and p65(−/−) cells are also GSH-deficient. As a consequence, cells deficient in IKKβ signaling are extremely susceptible to toxicity caused by environmental and pharmacological agents, including oxidants, genotoxic agents, microtubule toxins, and arsenic. GSH biosynthesis depends on the activity of the rate-limiting enzyme glutamate-cysteine ligase (GCL), consisting of a catalytic subunit (GCLC) and a modifier subunit (GCLM). We found that loss of IKKβ signaling significantly reduces basal NF-κB activity and decreases binding of NF-κB to the promoters of Gclc and Gclm, leading to reduction of GCLC and GCLM expression. Conversely, overexpression of GCLC and GCLM in IKKβ-null cells partially restores GSH content and prevents stress-induced cytotoxicity. We suggest that maintenance of GSH is a novel physiological role of the IKKβ-NF-κB signaling cascade to prevent oxidative damage and preserve the functional integrity of the cells.
- Published
- 2010
48. Effects of arsenic exposure on DNA methylation and epigenetic gene regulation
- Author
-
John F. Reichard and Alvaro Puga
- Subjects
Cancer Research ,Transsulfuration ,Biology ,medicine.disease_cause ,Article ,Arsenic ,Epigenesis, Genetic ,Mice ,Pregnancy ,Neoplasms ,Genetics ,medicine ,Animals ,Humans ,Nutritional Physiological Phenomena ,Epigenetics ,skin and connective tissue diseases ,Gene ,Regulation of gene expression ,Oncogene ,Methyltransferases ,DNA Methylation ,Oxidative Stress ,Prenatal Exposure Delayed Effects ,DNA methylation ,Cancer cell ,Arsenates ,Female ,sense organs ,Carcinogenesis ,Oxidation-Reduction - Abstract
Arsenic is a nonmutagenic human carcinogen that induces tumors through unknown mechanisms. A growing body of evidence suggests that its carcinogenicity results from epigenetic changes, particularly in DNA methylation. Changes in gene methylation status, mediated by arsenic, have been proposed to activate oncogene expression or silence tumor suppressor genes, leading to long-term changes in the activity of genes controlling cell transformation. Mostly descriptive, and often contradictory, studies have demonstrated that arsenic exposure is associated with both hypo- and hyper-methylation at various genetic loci in vivo or in vitro. This ambiguity has made it difficult to assess whether the changes induced by arsenic are causally involved in the transformation process or are simply a reflection of the altered physiology of rapidly dividing cancer cells. Here, we discuss the evidence supporting changes in DNA methylation as a cause of arsenic carcinogenesis and highlight the strengths and limitations of these studies, as well as areas where consistencies and inconsistencies exist.
- Published
- 2010
49. The Aryl Hydrocarbon Receptor Functions as a Tumor Suppressor of Liver Carcinogenesis
- Author
-
Alvaro Puga, Erik S. Knudsen, Daniel W. Nebert, Gregory P. Boivin, Ying Xia, and Yunxia Fan
- Subjects
Cancer Research ,medicine.medical_specialty ,Tumor suppressor gene ,Cell growth ,DNA damage ,respiratory system ,Biology ,Aryl hydrocarbon receptor ,medicine.disease_cause ,Endocrinology ,Oncology ,Cell culture ,Internal medicine ,medicine ,biology.protein ,Cancer research ,Gene silencing ,Carcinogenesis ,Transcription factor - Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that mediates the biological and toxic effects of its xenobiotic ligands. Previous cell culture studies have shown that, in addition to controlling the xenobiotic detoxification response, AHR activation leads to G0-G1 arrest, diminished capacity for DNA replication, and inhibition of cell proliferation. In fact, recent work from our own and from other laboratories suggests that AHR may function as a tumor suppressor gene that becomes silenced during the process of tumor formation. To test this hypothesis and determine whether the mouse Ahr gene acts as a tumor suppressor gene in vivo, we have examined the role of Ahr ablation in liver tumorigenesis induced by the genotoxic chemical diethylnitrosamine (DEN), a hepatic carcinogen that is not an AHR ligand. In mice given a single i.p. injection of DEN, AHR antagonized liver tumor formation and growth by regulating cell proliferation, inflammatory cytokine expression, and DNA damage, parameters which were significantly elevated in the livers of control and, more so, of DEN-exposed Ahr−/− mice. Ahr−/− hepatocytes also showed significantly higher numbers of 4N cells, increased expression of proliferative markers, and repression of tumor suppressor genes. These data support the concept that in its basal state in the absence of a xenobiotic ligand, the Ahr gene functions as a tumor suppressor gene, and that its silencing may be associated with cancer progression. Cancer Res; 70(1); 212–220
- Published
- 2010
50. Distinct Contributions of JNK and p38 to Chromium Cytotoxicity and Inhibition of Murine Embryonic Stem Cell Differentiation
- Author
-
Alvaro Puga, Jerald L. Ovesen, Liang Chen, and Ying Xia
- Subjects
MAPK/ERK pathway ,Chromium ,Health, Toxicology and Mutagenesis ,Cellular differentiation ,p38 mitogen-activated protein kinases ,Blotting, Western ,p38 ,MAP Kinase Kinase 7 ,Biology ,p38 Mitogen-Activated Protein Kinases ,MAP2K7 ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,MAPKs ,Animals ,Phosphorylation ,Cytotoxicity ,Cells, Cultured ,Embryonic Stem Cells ,030304 developmental biology ,0303 health sciences ,Kinase ,Reverse Transcriptase Polymerase Chain Reaction ,Research ,Public Health, Environmental and Occupational Health ,JNK Mitogen-Activated Protein Kinases ,ROS ,Cell Differentiation ,Embryonic stem cell ,3. Good health ,Cell biology ,Enzyme Activation ,ERK ,chemistry ,embryonic stem cell differentiation ,030220 oncology & carcinogenesis ,cytotoxicity ,Environmental Pollutants ,JNK ,Mitogen-Activated Protein Kinases ,Toxicant - Abstract
Background Potassium dichromate [Cr(VI)] is a widespread environmental toxicant responsible for increased risk of several human diseases. Cr(VI) exposure leads to activation of mitogen-activated protein kinases (MAPKs), including c-Jun N-terminal kinase (JNK)1/2, p38, and extracellular-signal regulated kinase (ERK)1/2. Objectives We evaluated the contribution of MAPKs to Cr(VI) toxicity. Methods Phosphorylation of MAPKs and their downstream effectors was evaluated by Western immunoblotting; reactive oxygen species were measured by DCFDA (5′,6′-chloromethyl-2′-7′-dichlorofluorescin diacetate) labeling and flow cytometry, and glutathione and glutathione disulfide levels were determined by monochrome graphic spectroflurometer. Cytotoxicity was assessed by the MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] assay and colony formation. Embryoid body (EB) differentiation was evaluated by contracting cardiomyocyte formation, and real-time polymerase chain reaction (RT-PCR) was used for cardiomyocyte-specific and stem-cell-specific gene expression. Results Acute treatment of mouse embryonic stem (ES) cells with 50 μM Cr(VI) induced the rapid phosphorylation of JNK, p38, and ERK and their respective downstream transcription factors, c-JUN, activating transcription factor-2, and ELK1. MAPK activation and cytotoxicity induction were partially blocked by pretreatment with the antioxidant N-acetyl cysteine. Ablation of the upstream MAP kinase kinase (MAP2K7) in ES cells prevented JNK activation, whereas ablation of MAP2K4 prevented both JNK and p38 activation. Using specific MAPK inhibitors and MAP2K4- and MAP2K7-deficient ES cells, we showed that JNK reduced acute Cr(VI) cytotoxicity, p38 potentiated it, and ERK had no effect. At low submicromolar concentrations, Cr(VI) caused MAP2K4/7-dependent JNK activation and MAP2K4-dependent p38 activation and strongly inhibited contracting cardiomyocyte development in wild-type ES cells, but much less so in Map2k7(−/−) cells. Conclusion Each MAPK distinctly contributes to chromium toxicity. Whereas JNK prevents and p38 promotes acute cytotoxicity, JNK contributes to optimal inhibition of ES cell differentiation by chromium.
- Published
- 2009
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.