1
|
Yilmaz E, Gul M. Effects of essential oils on heat-stressed poultry: A review. J Anim Physiol Anim Nutr (Berl) 2024; 108:1481-1497. [PMID: 38808374 DOI: 10.1111/jpn.13992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/11/2024] [Accepted: 05/16/2024] [Indexed: 05/30/2024]
Abstract
While certain animal species are sensitive to heat stress, poultry particularly modern breeds, are more susceptible to high ambient temperatures. This has major implications for the poultry industry, as heat stress causes large financial losses. These economic losses will probably increase as a consequence of a predicted rise in global temperatures. Heat stress adversely affects various aspects of poultry, including physiological responses, growth and production performance, meat quality, egg quality, and reproductive activities. These effects occur through specific molecular and metabolic pathways. To mitigate the impacts of heat stress, it is crucial to go beyond administrative practices and implement dietary interventions during high ambient temperature. Such interventions aim to optimize the development of stressed bird species in terms of performance, health, and profitability. Essential oils have shown promising in mitigating the negative effects of heat stress and improved antioxidant status, growth and yield performance, as well as meat and egg quality in poultry. They actively participate in certain metabolic and molecular pathways that help to counteract the effects of heat stress. The article discusses the impacts of essential oil supplementation on the relationships between antioxidant enzyme activity, these molecular, and metabolic pathways, as well as various parameters such as growth and yield performance, and product quality heat-stressed poultry.
Collapse
Affiliation(s)
- Emre Yilmaz
- Department of Animal Nutrition and Nutritional Diseases, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Türkiye
| | - Mehmet Gul
- Department of Animal Nutrition and Nutritional Diseases, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Türkiye
| |
Collapse
|
2
|
Park JG, Iwata H, Tue NM, Kunisue T, Kim EY. Effects of 1,3,7-tribromodibenzo-p-dioxin, a natural dioxin on chicken embryos: Comparison with effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 237:113538. [PMID: 35483143 DOI: 10.1016/j.ecoenv.2022.113538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/15/2022] [Accepted: 04/16/2022] [Indexed: 06/14/2023]
Abstract
Several naturally occurring dioxins, including 1,3,7-tribromodibenzo-p-dioxin (1,3,7-TriBDD), synthesized by red algae, have been detected in the marine environment. As 1,3,7-TriBDD is accumulated in mussels and fish, predators, such as marine birds, are exposed to this congener, similar to anthropogenic dioxins (including 2,3,7,8-tetrachlorodibenzo-p-dioxin TCDD). However, little is known about the impact of 1,3,7-TriBDD exposure on the bird health. To understand the effects of 1,3,7-TriBDD on birds, the phenotypic effects and hepatic transcriptome were investigated in chicken (Gallus gallus) embryos treated with 27 μM (2.9 ng/g egg) and 137 μM (14.4 ng/g egg) 1,3,7-TriBDD. The blood glucose levels in the 1,3,7-TriBDD-treated groups were lower than those in the control group. The transcriptome analysis of 6520 sequences in the 27 and 137 μM 1,3,7-TriBDD-treated groups identified 733 and 596 differentially expressed genes (DEGs). Cytochrome P450 1A4 and 1A5 were also identified as DEGs, suggesting that the aryl hydrocarbon receptor is activated by this congener. Pathway and network analyses with DEGs suggested that 1,3,7-TriBDD may induce carcinogenic effects and metabolic alterations. These results were similar to the effects on TCDD-treated embryos. Nevertheless, the overall transcriptome results suggested that compared with TCDD, 1,3,7-TriBDD has a unique impact on insulin- and peroxisome-signaling pathways in chicken embryos. Differences in altered transcriptome profiles between 1,3,7-TriBDD- and TCDD-treated embryos may lead to different phenotypic effects: less severe effects of 1,3,7-TriBDD and more fatal effects of TCDD. Collectively, these findings warrant the further assessment of the hazard and risk of 1,3,7-TriBDD on marine animals, considering increased exposure due to climate change.
Collapse
Affiliation(s)
- Jae Gon Park
- Department of Biology, Kyung Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul 130-701, Republic of Korea
| | - Hisato Iwata
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama 790-8577, Japan
| | - Nguyen Minh Tue
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama 790-8577, Japan
| | - Tatsuya Kunisue
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama 790-8577, Japan
| | - Eun-Young Kim
- Department of Biology, Kyung Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul 130-701, Republic of Korea; Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama 790-8577, Japan.
| |
Collapse
|
3
|
Jiang J, Zhu J, Liu Q, Zhang T, Wen J, Xia J, Deng Y. Role of DNA methylation-related chromatin remodeling in aryl hydrocarbon receptor-dependent regulation of T-2 toxin highly inducible Cytochrome P450 1A4 gene. FASEB J 2021; 35:e21469. [PMID: 33788981 DOI: 10.1096/fj.202002570rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 11/11/2022]
Abstract
Mycotoxins are toxic secondary metabolites produced by food-contaminating fungi, which lead to global epigenetic changes and cause toxicity to both farm animals and humans. However, whether mycotoxins induce gene-specific epigenetic alterations associated with inducible downstream gene expression is unclear as are the underlying regulatory mechanisms. Here, we found that T-2 toxin and its deacetylated metabolites but not deoxynivalenol (DON) or other representative mycotoxins highly induced the expression of cytochrome P450 1A4 (CYP1A4) in both Leghorn male hepatoma (LMH) cells and chicken primary hepatocytes, and this effect was related to the regulation of both aryl hydrocarbon receptor (AhR) and DNA methylation. We used methylation-sensitive restriction enzyme digestion-qPCR (MSRE-qPCR) and chromatin immunoprecipitation (ChIP) assays and found that the binding of DNA methyltransferase 1 (DNMT1) and histone deacetylase 2 (HDAC2) to highly methylated CpG island 3-2 at the enhancer of CYP1A4 was accompanied by the recruitment of the repressive histone modification marker H3K27me3, inducing a silent state. In turn, T-2 toxin stimulation enriched the binding of AhR to demethylated CpG island 3-2, which facilitated p300 and H3K9ac recruitment and ultimately generated an activated chromatin structure at the enhancer by increasing the active histone modification markers, including H3K4me3, H3K27ac, and H3K14ac. Interestingly, T-2 toxin-induced AhR activation also facilitated RNA polymerase II binding to CpG island 2, which may form a transcriptionally active chromatin structure at the promoter and ultimately transactivate CYP1A4. Our findings provide novel insights into the epigenetic regulation of T-2 toxin-induced gene expression.
Collapse
Affiliation(s)
- Jun Jiang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, P.R. China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, P.R. China.,Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, P.R. China
| | - Jiahui Zhu
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, P.R. China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, P.R. China.,Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, P.R. China
| | - Qian Liu
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, P.R. China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, P.R. China.,Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, P.R. China
| | - Tingting Zhang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, P.R. China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, P.R. China.,Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, P.R. China
| | - Jikai Wen
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, P.R. China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, P.R. China.,Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, P.R. China
| | - Jianhong Xia
- Key Laboratory of Regenerative Biology of Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, P.R. China
| | - Yiqun Deng
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, P.R. China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, P.R. China.,Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, P.R. China
| |
Collapse
|
4
|
Developmental and lifelong dioxin exposure induces measurable changes in cardiac structure and function in adulthood. Sci Rep 2021; 11:10378. [PMID: 34001975 PMCID: PMC8129097 DOI: 10.1038/s41598-021-89825-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/27/2021] [Indexed: 11/20/2022] Open
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.
Collapse
|
5
|
Luptakova L, Dvorcakova S, Demcisakova Z, Belbahri L, Holovska K, Petrovova E. Dimethyl Sulfoxide: Morphological, Histological, and Molecular View on Developing Chicken Liver. TOXICS 2021; 9:toxics9030055. [PMID: 33809222 PMCID: PMC8001493 DOI: 10.3390/toxics9030055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/28/2021] [Accepted: 03/09/2021] [Indexed: 01/04/2023]
Abstract
Dimethyl sulfoxide (DMSO) is widely used as a solvent for small hydrophobic drug molecules. However, the safe volume allowing to avoid its embryotoxic effect has been poorly studied. In this study, we documented the effects of dimethyl sulfoxide (DMSO) in the developing chicken embryo at morphological, histological, and molecular levels. We focused on the developing chicken liver as the main organ involved in the process of detoxification. In our study, 100% DMSO was administered subgerminally onto the eggshell membrane (membrana papyracea) at various volumes (5, 10, 15, 20, 25, 30, 35, and 50 µL) on 4th embryonic day (ED). We focused on histopathological alterations of the liver structure, and noticed the overall impact of DMSO on developing chicken embryos (embryotoxicity, malformation). At the molecular level, we studied cytochrome P450 complex (CYP) isoform's activities in relation to changes of CYP1A5, CYP3A37, and CYP3A80 gene expression. Total embryotoxicity after application of different doses of DMSO on ED 4, and the embryo lethality increased with increasing DMSO amounts. Overall mortality after DMSO administration ranged below 33%. Mortality was increased with higher amounts of DMSO, mainly from 20 µL. The highest mortality was observed for the highest dose of DMSO over 35 µL. The results also showed a decrease in body weight with increased application volumes of DMSO. At the histological level, we observed mainly the presence of lipid droplets and dilated bile canaliculi and sinusoids in samples over the administration of 25 µL of DMSO. While these findings were not statistically significant, DMSO treatment caused a significant different up-regulation of mRNA expression in all studied genes. For CYP1A5, CYP3A37, and CYP3A80 DMSO volumes needed were 15 µL, 10 µL, and 20 µL, respectively. A significant down-regulation of all studied CYP isoform was detected after application of a DMSO dose of 5 µL. Regarding the morphological results, we can assume that the highest safe dose of DMSO without affecting chicken embryo development and its liver is up to 10 µL. This conclusion is corroborated with the presence of number of malformations and body weight reduction, which correlates with histological findings. Moreover, the gene expression results showed that even the lowest administered DMSO volume could affect hepatocytes at the molecular level causing down-regulation of cytochrome P450 complex (CYP1A5, CYP3A37, CYP3A80).
Collapse
Affiliation(s)
- Lenka Luptakova
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia;
- Correspondence: ; Tel.: +421-918-919-686
| | - Simona Dvorcakova
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia;
| | - Zuzana Demcisakova
- Department of Morphological Disciplines, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia; (Z.D.); (K.H.); (E.P.)
| | - Lassaad Belbahri
- Laboratory of Soil Biodiversity, Department of Biology, University of Neuchatel, 2000 Neuchatel, Switzerland;
| | - Katarina Holovska
- Department of Morphological Disciplines, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia; (Z.D.); (K.H.); (E.P.)
| | - Eva Petrovova
- Department of Morphological Disciplines, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia; (Z.D.); (K.H.); (E.P.)
| |
Collapse
|
6
|
Aromatic hydrocarbon receptor regulates chicken cytochrome P450 1A5 transcription: A novel insight into T-2 toxin-induced gene expression and cytotoxicity in LMH cells. Biochem Pharmacol 2019; 168:319-329. [DOI: 10.1016/j.bcp.2019.07.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 07/23/2019] [Indexed: 12/17/2022]
|
7
|
Mechanisms of 2,3,7,8-tetrachlorodibenzo-p-dioxin- induced cardiovascular toxicity: An overview. Chem Biol Interact 2018; 282:1-6. [PMID: 29317249 DOI: 10.1016/j.cbi.2018.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 12/05/2017] [Accepted: 01/05/2018] [Indexed: 11/21/2022]
Abstract
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is an environmental contaminant and its toxicity is mediated by the aryl hydrocarbon receptor (AHR). Mechanisms of TCDD cardiovascular toxicity consist of oxidative stress, growth factor modulation, and ionic current alteration. It is indicated that the rodent cardiovascular system is a target for TCDD cardiomyopathy. Here, our understanding of TCDD cardiovascular toxicity is reviewed.
Collapse
|
8
|
Regulation of Human Cytochrome P4501A1 (hCYP1A1): A Plausible Target for Chemoprevention? BIOMED RESEARCH INTERNATIONAL 2016; 2016:5341081. [PMID: 28105425 PMCID: PMC5220472 DOI: 10.1155/2016/5341081] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 11/09/2016] [Accepted: 11/13/2016] [Indexed: 12/13/2022]
Abstract
Human cytochrome P450 1A1 (hCYP1A1) has been an object of study due to its role in precarcinogen metabolism; for this reason it is relevant to know more in depth the mechanisms that rule out its expression and activity, which make this enzyme a target for the development of novel chemiopreventive agents. The aim of this work is to review the origin, regulation, and structural and functional characteristics of CYP1A1 letting us understand its role in the bioactivation of precarcinogen and the consequences of its modulation in other physiological processes, as well as guide us in the study of this important protein.
Collapse
|
9
|
Pérez Sáez JM, Bussmann LE, Barañao JL, Bussmann UA. Improvement of Chicken Primordial Germ Cell Maintenance In Vitro by Blockade of the Aryl Hydrocarbon Receptor Endogenous Activity. Cell Reprogram 2016; 18:154-61. [PMID: 27253627 PMCID: PMC4900192 DOI: 10.1089/cell.2016.0015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Primordial germ cells (PGCs) are the undifferentiated progenitors of gametes. Germline competent PGCs can be developed as a cell-based system for genetic modification in chickens, which provides a valuable tool for transgenic technology with both research and industrial applications. This implies manipulation of PGCs, which, in recent years, encouraged a lot of research focused on the study of PGCs and the way of improving their culture. The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that besides mediating toxic responses to environmental contaminants plays pivotal physiological roles in various biological processes. Since a novel compound that acts as an antagonist of this receptor has been reported to promote expansion of hematopoietic stem cells, we conducted the present study with the aim of determining whether addition of an established AHR antagonist to the standard culture medium used nowadays for in vitro chicken PGCs culture improves ex vivo expansion. We have found that addition of α-naphthoflavone in culture medium promotes the amplification of undifferentiated cells and that this effect is exerted by the blockade of AHR action. Our results constitute the first report of the successful use of a readily available AHR antagonist to improve avian PGCs expansion, and they further extend the knowledge of the effects of AHR modulation in undifferentiated cells.
Collapse
Affiliation(s)
- Juan M. Pérez Sáez
- Instituto de Biología y Medicina Experimental-CONICET, Buenos Aires, Argentina
| | | | - J. Lino Barañao
- Instituto de Biología y Medicina Experimental-CONICET, Buenos Aires, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ursula A. Bussmann
- Instituto de Biología y Medicina Experimental-CONICET, Buenos Aires, Argentina
| |
Collapse
|
10
|
Diani-Moore S, Ma Y, Gross SS, Rifkind AB. Increases in levels of epoxyeicosatrienoic and dihydroxyeicosatrienoic acids (EETs and DHETs) in liver and heart in vivo by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and in hepatic EET:DHET ratios by cotreatment with TCDD and the soluble epoxide hydrolase inhibitor AUDA. Drug Metab Dispos 2014; 42:294-300. [PMID: 24311719 PMCID: PMC3912539 DOI: 10.1124/dmd.113.055368] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 12/05/2013] [Indexed: 12/28/2022] Open
Abstract
The environmental toxin and carcinogen 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, dioxin) binds and activates the transcription factor aryl hydrocarbon receptor (AHR), inducing CYP1 family cytochrome P450 enzymes. CYP1A2 and its avian ortholog CYP1A5 are highly active arachidonic acid epoxygenases. Epoxygenases metabolize arachidonic acid to four regioisomeric epoxyeicosatrienoic acids (EETs) and selected monohydroxyeicosatetraenoic acids (HETEs). EETs can be further metabolized by epoxide hydrolases to dihydroxyeicosatrienoic acids (DHETs). As P450-arachidonic acid metabolites affect vasoregulation, responses to ischemia, inflammation, and metabolic disorders, identification of their production in vivo is needed to understand their contribution to biologic effects of TCDD and other AHR activators. Here we report use of an acetonitrile-based extraction procedure that markedly increased the yield of arachidonic acid products by lipidomic analysis over a standard solid-phase extraction protocol. We show that TCDD increased all four EETs (5,6-, 8,9-, 11,12-, and 14,15-), their corresponding DHETs, and 18- and 20-HETE in liver in vivo and increased 5,6-EET, the four DHETs, and 18-HETE in heart, in a chick embryo model. As the chick embryo heart lacks arachidonic acid-metabolizing activity, the latter findings suggest that arachidonic acid metabolites may travel from their site of production to a distal organ, i.e., heart. To determine if the TCDD-arachidonic acid-metabolite profile could be altered pharmacologically, chick embryos were treated with TCDD and the soluble epoxide hydrolase inhibitor 12-(3-adamantan-1-yl-ureido)-dodecanoic acid (AUDA). Cotreatment with AUDA increased hepatic EET-to-DHET ratios, indicating that the in vivo profile of P450-arachidonic acid metabolites can be modified for potential therapeutic intervention.
Collapse
|
11
|
Low-dose trichloroethylene alters cytochrome P450-2C subfamily expression in the developing chick heart. Cardiovasc Toxicol 2013; 13:77-84. [PMID: 22855351 DOI: 10.1007/s12012-012-9180-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Trichloroethylene (TCE) is an organic solvent and common environmental contaminant. TCE exposure is associated with heart defects in humans and animal models. Primary metabolism of TCE in adult rodent models is by specific hepatic cytochrome P450 enzymes (Lash et al. in Environ Health Perspect 108:177-200, 2000). As association of TCE exposure with cardiac defects is in exposed embryos prior to normal liver development, we investigated metabolism of TCE in the early embryo. Developing chick embryos were dosed in ovo with environmentally relevant doses of TCE (8 and 800 ppb) and RNA was extracted from cardiac and extra-cardiac tissue (whole embryo without heart). Real-time PCR showed upregulation of CYP2H1 transcripts in response to TCE exposure in the heart. No detectable cytochrome expression was found in extra-cardiac tissue. As seen previously, the dose response was non-monotonic and 8 ppb elicited stronger upregulation than 800 ppb. Immunostaining for CYP2C subfamily expression confirmed protein expression and showed localization in both myocardium and endothelium. TCE exposure increased protein expression in both tissues. These data demonstrate that the earliest embryonic expression of phase I detoxification enzymes is in the developing heart. Expression of these CYPs is likely to be relevant to the susceptibility of the developing heart to environmental teratogens.
Collapse
|
12
|
Chicken cytochrome P450 1A5 is the key enzyme for metabolizing T-2 toxin to 3'OH-T-2. Int J Mol Sci 2013; 14:10809-18. [PMID: 23702848 PMCID: PMC3709703 DOI: 10.3390/ijms140610809] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 05/12/2013] [Accepted: 05/17/2013] [Indexed: 11/16/2022] Open
Abstract
The transmission of T-2 toxin and its metabolites into the edible tissues of poultry has potential effects on human health. We report that T-2 toxin significantly induces CYP1A4 and CYP1A5 expression in chicken embryonic hepatocyte cells. The enzyme activity assays of CYP1A4 and CYP1A5 heterologously expressed in HeLa cells indicate that only CYP1A5 metabolizes T-2 to 3'OH-T-2 by the 3'-hydroxylation of isovaleryl groups. In vitro enzyme assays of recombinant CYP1A5 expressed in DH5α further confirm that CYP1A5 can convert T-2 into TC-1 (3'OH-T-2). Therefore, CYP1A5 is critical for the metabolism of trichothecene mycotoxin in chickens.
Collapse
|
13
|
Hu S. Effect of age on hepatic cytochrome P450 of Ross 708 broiler chickens. Poult Sci 2013; 92:1283-92. [DOI: 10.3382/ps.2012-02911] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
14
|
Jones SP, Kennedy SW. Chicken embryo cardiomyocyte cultures--a new approach for studying effects of halogenated aromatic hydrocarbons in the avian heart. Toxicol Sci 2009; 109:66-74. [PMID: 19223662 DOI: 10.1093/toxsci/kfp039] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), polychlorinated biphenyls (PCBs), and other halogenated aromatic hydrocarbons (HAHs) elicit a variety of adverse biological effects on the cardiovascular systems of mammalian, piscine and avian species. Many of the cardiotoxic effects of HAHs are mediated by the aryl hydrocarbon receptor (AHR). Induction of cytochrome P4501A (CYP1A) is a well-known AHR-dependent response to HAHs in the liver, but there are a limited number of studies on CYP1A induction by these compounds in the heart. We used an in vitro approach to examine effects of TCDD and 3,3',4,4'-tetrachlorobiphenyl (PCB 77) on CYP1A in the avian heart. The responses of primary cultures of chicken embryo cardiomyocytes (CEC) and chicken embryo hepatocytes (CEH) to TCDD and PCB 77 were compared using immunofluorescence staining for CYP1A, the ethoxyresorufin-O-deethylase (EROD) assay, and real-time RT-PCR analysis of CYP1A4 mRNA and CYP1A5 mRNA. Immunofluorescent detection of CYP1A indicated that induction of CYP1A by TCDD was localized within the cytoplasm of CEC cells. EROD activity and CYP1A4/5 mRNA levels were strongly induced in CEC and CEH cultures by TCDD and PCB 77, and the shapes of the concentration-response curves in CEC and CEH cultures were similar. The studies provide clear evidence that the AHR signaling pathway is induced by TCDD and PCB 77 in CEC, and establish a new in vitro approach for studying the effects of HAHs in the avian heart. Induction of CYP1A5 by TCDD in avian cardiomyocytes is a novel finding, and might help direct future studies on mechanisms of action of HAHs in the heart.
Collapse
Affiliation(s)
- Stephanie P Jones
- Environment Canada, National Wildlife Research Centre, 1125 Colonel By Drive, Ottawa, Ontario, Canada
| | | |
Collapse
|
15
|
Augspurger TP, Tillitt DE, Bursian SJ, Fitzgerald SD, Hinton DE, Di Giulio RT. Embryo toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin to the wood duck (Aix sponsa). ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2008; 55:659-669. [PMID: 18704254 DOI: 10.1007/s00244-008-9198-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2008] [Accepted: 07/05/2008] [Indexed: 05/26/2023]
Abstract
We examined the sensitivity of the wood duck (Aix sponsa) embryo to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) by injecting the toxicant into their eggs. Six groups of wood duck eggs (n = 35 to 211 per trial) were injected with 0 to 4600 pg TCDD/g egg between 2003 and 2005. Injections were made into yolk prior to incubation, and eggs were subsequently incubated and assessed weekly for mortality. Significant TCDD-induced mortality was not observed through day 25 (90% of incubation). Liver, heart, eye, and brain histology were generally unremarkable. Hepatic ethoxyresorufin-O-deethylase activity, a biomarker of dioxin-like compound exposure, was induced by 12-fold in the 4600 pg/g treatment relative to controls. The median lethal dose for chicken (Gallus domesticus) eggs we dosed identically to wood duck eggs was about 100 pg/g, similar to other assessments of chickens. Among dioxin-like compound embryo lethality data for 15 avian genera, the wood duck 4600 pg/g no-observed-effect level ranks near the middle. Because no higher doses were tested, wood ducks may be like other waterfowl (order Anseriformes), which are comparatively tolerant to embryo mortality from polychlorinated dibenzo-p-dioxins and dibenzofurans when exposed by egg injection.
Collapse
Affiliation(s)
- T P Augspurger
- U.S. Fish and Wildlife Service, 551-F Pylon Drive, Raleigh, NC 27606, USA.
| | | | | | | | | | | |
Collapse
|
16
|
Goldstone HMH, Stegeman JJ. Molecular Mechanisms of 2,3,7,8-Tetrachlorodibenzo-p-Dioxin Cardiovascular Embryotoxicity. Drug Metab Rev 2008; 38:261-89. [PMID: 16684661 DOI: 10.1080/03602530600570099] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
2,3,7,8 Tetrachlorodibenzo-p-dioxin (TCDD) and related planar halogenated aromatic hydrocarbons are widespread environmental contaminants and potent developmental toxicants. Hallmarks of embryonic exposure include edema, hemorrhage, and mortality. Recent studies in zebrafish and chicken have revealed direct impairment of cardiac muscle growth that may underlie these overt symptoms. TCDD toxicity is mediated by the aryl hydrocarbon receptor, but downstream targets remain unclear. Oxidative stress and growth factor modulation have been implicated in TCDD cardiovascular toxicity. Gene expression profiling is elucidating additional pathways by which TCDD might act. We review our understanding of the mechanism of TCDD embryotoxicity at morphological and molecular levels.
Collapse
Affiliation(s)
- Heather M H Goldstone
- The Josephine Bay Paul Center for Comparative Molecular Biology and Evolution, Marine Biological Laboratory, Woods Hole, MA 02543, USA.
| | | |
Collapse
|
17
|
Rifkind AB. CYP1A in TCDD toxicity and in physiology-with particular reference to CYP dependent arachidonic acid metabolism and other endogenous substrates. Drug Metab Rev 2006; 38:291-335. [PMID: 16684662 DOI: 10.1080/03602530600570107] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Toxicologic and physiologic roles of CYP1A enzyme induction, the major biochemical effect of aryl hydrocarbon receptor activation by TCDD and other receptor ligands, are unknown. Evidence is presented that CYP1A exerts biologic effects via metabolism of endogenous substrates (i.e., arachidonic acid, other eicosanoids, estrogens, bilirubin, and melatonin), production of reactive oxygen, and effects on K(+) and Ca(2+) channels. These interrelated pathways may connect CYP1A induction to TCDD toxicities, including cardiotoxicity, vascular dysfunction, and wasting. They may also underlie homeostatic roles for CYP1A, especially when transiently induced by common chemical exposures and environmental conditions (i.e., tryptophan photoproducts, dietary indoles, and changes in oxygen tension).
Collapse
Affiliation(s)
- Arleen B Rifkind
- Department of Pharmacology, Weill Medical College of Cornell University, New York, NY 10021, USA.
| |
Collapse
|
18
|
Bruggeman V, Van den Bergh G, Clerens S, Dumez L, Onagbesan O, Arckens L, Decuypere E. Effect of a singlein ovo injection of 2,3,7,8-tetrachlorodibenzo-p-dioxin on protein expression in liver and ovary of the one-day-old chick analyzed by fluorescent two-dimensional difference gel electrophoresis and mass spectrometry. Proteomics 2006; 6:2576-85. [PMID: 16526093 DOI: 10.1002/pmic.200500237] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The polyhalogenated aromatic hydrocarbon 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is an ubiquitously distributed environmental pollutant which can induce a broad spectrum of toxic responses in animals, including birds. In this study, we investigated the impact of 0 or 20 ng TCDD injections into the yolk of chicken eggs before start of development, on liver and ovarian protein expression in hatchlings using fluorescent two-dimensional difference gel electrophoresis (2-D-DIGE) under a pH range of 4-7, combined with MS. Despite considerable interindividual variability, exposure to TCDD prior to the start of embryonic development resulted in significant changes in expression of a small set of proteins. Expression of fibrinogen gamma chain precursor in the liver and 60 kDa heat shock protein in the ovary were significantly higher as a result of the very early exposure to TCDD. NADH ubiquinone oxidoreductase (42 kDa subunit) and regucalcin expression was decreased by early TCDD treatment in the liver and ovary, respectively. These proteins could not be directly linked with drug metabolism per se but are involved in blood clotting, oxidative stress, electron transport, and calcium regulation. It remains to be elucidated how these changes in the hatchling might be linked to the observed long-term consequences during posthatch life of the chicken.
Collapse
Affiliation(s)
- Veerle Bruggeman
- Laboratory of Physiology of Domestic Animals, Katholieke Universiteit Leuven, Kasteelpark Arenberg, Leuven, Belgium.
| | | | | | | | | | | | | |
Collapse
|
19
|
Handley-Goldstone HM, Grow MW, Stegeman JJ. Cardiovascular gene expression profiles of dioxin exposure in zebrafish embryos. Toxicol Sci 2005; 85:683-93. [PMID: 15716485 DOI: 10.1093/toxsci/kfi116] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a widespread environmental contaminant that causes altered heart morphology, circulatory impairment, edema, hemorrhage, and early life stage mortality in fish. TCDD toxicity is dependent, in large part, on the aryl hydrocarbon receptor (AHR), but understanding of the molecular mechanism of cardiovascular embryotoxicity remains incomplete. To identify genes potentially involved in cardiovascular effects, we constructed custom cDNA microarrays consisting of 4896 zebrafish adult heart cDNA clones and over 200 genes with known developmental, toxicological and housekeeping roles. Gene expression profiles were obtained for 3-day-old zebrafish after early embryonic exposure to either 0.5 or 5.0 nM TCDD. In all, 516 clones were significantly differentially expressed (p < 0.005) under at least one treatment condition; 123 high-priority clones were selected for further investigation. Cytochromes P450 1A and 1B1, and other members of the AHR gene battery, were strongly and dose-dependently induced by TCDD. Importantly, altered expression of cardiac sarcomere components, including cardiac troponin T2 and multiple myosin isoforms, was consistent with the hypothesis that TCDD causes dilated cardiomyopathy. Observed increases in expression levels of mitochondrial energy transfer genes also may be related to cardiomyopathy. Other TCDD-responsive genes included fatty acid and steroid metabolism enzymes, ribosomal and signal-transduction proteins, and 18 expressed sequence tags (ESTs) with no known protein homologs. As the first broad-scale study of TCDD-modulated gene expression in a non-mammalian system, this work provides an important perspective on mechanisms of TCDD toxicity.
Collapse
|
20
|
Garrick RA, Woodin BR, Stegeman JJ. CYTOCHROME P4501A INDUCED DIFFERENTIALLY IN ENDOTHELIAL CELLS CULTURED FROM DIFFERENT ORGANS OF ANGUILLA ROSTRATA. ACTA ACUST UNITED AC 2005; 41:57-63. [PMID: 15926861 DOI: 10.1290/0409063.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Endothelial cells are a structural barrier and an active regulator of many bodily processes. Cytochrome P4501A (CYP1A) activity is induced in the endothelium of teleosts and mammals exposed to lipophilic xenobiotics, such as polycyclic aromatic hydrocarbons, and can have significant consequences for endothelial functions. We exposed cultures of characterized endothelial cells from the heart, kidney, and rete mirabile of the eel, Anguilla rostrata, to aryl hydrocarbon receptor (AhR) agonists. In heart endothelial cells, the maximum response (based on O-deethylation of 7-ethoxyresorufin to resorufin [EROD] activity) to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), 113 pmol/mg/min, was at 1 nM TCDD and the peak response to beta-napthoflavone (betaNF), 135 pmol/mg/min, was at 3 microM betaNF. The maximum response to TCDD in the kidney endothelial cells is 12 pmol/mg/min at 0.3 nM TCDD. The rete mirabile capillary endothelial cells responded minimally or not at all to exposure to TCDD and betaNF. Both the heart and kidney endothelial cells (but not the rete mirabile capillary cells) have a low level of EROD activity (12.7 and 5.2 pmol/mg/min, respectively) in untreated or dimethylsulfoxide-treated cells. The robust response of the heart endothelial cells to induction and the lack of response in the rete mirabile capillary endothelial cells indicate that these cells are a good resource to use to investigate the physiological consequences of AhR agonist exposure and CYP1A induction in different areas of the vasculature.
Collapse
Affiliation(s)
- Rita Anne Garrick
- Department of Natural Sciences, Fordham University, 113 W. 60th Street, New York, New York 10023, USA.
| | | | | |
Collapse
|
21
|
Filbrandt CR, Wu Z, Zlokovic B, Opanashuk L, Gasiewicz TA. Presence and functional activity of the aryl hydrocarbon receptor in isolated murine cerebral vascular endothelial cells and astrocytes. Neurotoxicology 2004; 25:605-16. [PMID: 15183014 DOI: 10.1016/j.neuro.2003.08.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2003] [Accepted: 07/11/2003] [Indexed: 10/27/2022]
Abstract
Numerous functions regulated by the central nervous system (CNS) are targeted by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD); however, the cell specific targets and mechanisms of toxicity are unknown. Outside of the brain, the peripheral vascular endothelium has been identified as a significant cellular target of TCDD toxicity resulting in apoptosis, edema, hemorrhaging and vascular dysfunction. Possible effects of TCDD in the vascular endothelium of the CNS have not been examined. Cellular dysfunction in this endothelium may disrupt function of the blood-brain barrier (BBB), which could severely compromise neuronal homeostasis and potentiate neurotoxicity. TCDD toxicity is mediated primarily by the aryl hydrocarbon receptor (AhR), a ligand activated transcription factor that modulates the expression of a large battery of genes. This study examined the presence and functional activity of the AhR in response to TCDD in endothelial cells and astrocytes, the two primary components of the BBB. Primary mouse cortical endothelial cells and astrocytes express the AhR, as shown by immunocytochemical and western blot analyses. AhR activity was assessed by time- and concentration-dependent analyses of CYP1A1 and CYP1B1 protein expression following TCDD treatment. Both CYP1A1 and CYP1B1 proteins were induced in endothelial cells after 4 and 8h, respectively, while only CYP1B1 protein induction was detected in astrocytes after 16h. The CYP450 protein induction was sustained for greater than 72h in both cell types. These changes in protein expression were dependent on AhR activity as indicated by the inhibition of these responses by a receptor antagonist. Together these data indicate endothelial cells and astrocytes are responsive to TCDD through the AhR-mediated pathway and therefore could be targets of toxicity.
Collapse
Affiliation(s)
- Carissa R Filbrandt
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Box EHSC, 575 Elmwood Avenue, Rochester, NY 14642, USA
| | | | | | | | | |
Collapse
|
22
|
Kanzawa N, Kondo M, Okushima T, Yamaguchi M, Temmei Y, Honda M, Tsuchiya T. Biochemical and molecular biological analysis of different responses to 2,3,7,8-tetrachlorodibenzo-p-dioxin in chick embryo heart and liver. Arch Biochem Biophys 2004; 427:58-67. [PMID: 15178488 DOI: 10.1016/j.abb.2004.04.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2004] [Revised: 04/15/2004] [Indexed: 11/25/2022]
Abstract
We studied the mechanism of toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in the chick embryo, which is an organism highly sensitive to TCDD. TCDD was injected into egg yolks prior to embryogenesis, and eggs were incubated for 12 or 18 days. In TCDD-exposed embryos, we observed increased heart wet weight and change in the color of the liver, with abnormal fatty vesicle formation. To determine whether these effects were mediated by the aryl hydrocarbon receptor (AhR), we examined expression levels of AhR, CYP1A4, and CYP1A5. AhR was expressed continuously in the heart and liver during embryogenesis, whereas induction of CYP1A4 and CYP1A5 by TCDD was detected only in the liver. In situ hybridization study of tissue sections revealed induction of CYP1A4 in the abnormal liver tissue in which color change was not observed. To determine whether these different responses to TCDD depended on the cell type, primary cultures of chick hepatocytes and cardiac myocytes were established and 7-ethoxyresorufin-O-deethylase (EROD) activity was measured. Induction of EROD activity following exposure to TCDD was detected in hepatocytes but not in cardiac myocytes. Although the heart is a principal target organ for TCDD toxicity and AhR is expressed throughout embryogenesis, induction of CYP1A was not observed in the chick heart. Thus, we conclude that defects in the heart induced by exposure to TCDD occur via a different pathway than that occurring in the liver.
Collapse
Affiliation(s)
- Nobuyuki Kanzawa
- Department of Chemistry, Faculty of Science and Technology, Sophia University, 7-1 Kioi, Chiyoda-ku, Tokyo 102-8554, Japan.
| | | | | | | | | | | | | |
Collapse
|
23
|
Hilscherova K, Blankenship AL, Nie M, Coady KK, Upham BL, Trosko JE, Giesy JP. Oxidative stress in liver and brain of the hatchling chicken (Gallus domesticus) following in ovo injection with TCDD. Comp Biochem Physiol C Toxicol Pharmacol 2003; 136:29-45. [PMID: 14522597 DOI: 10.1016/s1532-0456(03)00167-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) was injected into chicken eggs prior to incubation to study possible mechanisms of toxicity and teratogenicity. One of the suggested mechanisms of teratogenicity is oxidative stress. Eggs were injected simultaneously with TCDD and cotreatment compounds in an attempt to prevent oxidative stress or to block cytochrome P450 activity. Indicators of oxidative stress were assessed in livers and brains of hatchling chicks. In ovo, exposure to TCDD caused significant effects on indicators of oxidative stress in liver, but not in the brain of the hatchling chicks. TCDD did not significantly affect superoxide production. In liver, TCDD treatment caused a decrease in glutathione content and glutathione peroxidase activity and an increase in the ratio of oxidized to reduced glutathione. TCDD increased the susceptibility to lipid peroxidation and oxidative DNA damage in liver. Administration of the antioxidants vitamin E and vitamin A provided partial protection against TCDD-induced oxidative stress in liver. The lack of effect of TCDD in chicken brain could be due to the low cytochrome P4501A activity in this tissue and little accumulation of TCDD in brain compared to liver. Phenytoin, a known inducer of oxidative stress, caused a decrease in glutathione content and an increase in susceptibility to lipid peroxidation in both liver and brain and increased oxidative DNA damage in brain. Responsiveness varied among individual animals, but measures of the oxidative stress were correlated.
Collapse
Affiliation(s)
- K Hilscherova
- Department of Environmental Chemistry and Ecotoxicology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
24
|
Wood E, Broekman MJ, Kirley TL, Diani-Moore S, Tickner M, Drosopoulos JHF, Islam N, Park JI, Marcus AJ, Rifkind AB. Cell-type specificity of ectonucleotidase expression and upregulation by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Arch Biochem Biophys 2002; 407:49-62. [PMID: 12392715 DOI: 10.1016/s0003-9861(02)00465-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We report here that induction of ectoATPase by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is cell-type specific and not a generalized response to aryl hydrocarbon (Ah) receptor activation. TCDD increased [14C]-ATP and -ADP metabolism in two mouse hepatoma lines, Hepa1c1c7 and Hepa1-6 cells, but not in human hepatoma HepG2 or HuH-7 cells, human umbilical vein endothelial cells (HUVEC), chick hepatoma (LMH) cells, or chick primary hepatocytes or cardiac myocytes, even though all of those cell types were Ah receptor-responsive, as evidenced by cytochrome P4501A induction. To determine whether the differences in ectonucleotidase responsiveness to TCDD might be related to differences in cell-type ectonucleotidase expression, ATP and ADP metabolite patterns, the products of several classes of ectonucleotidases including ectonucleoside triphosphate diphosphohydrolases (E-NTPDases), ectophosphodiesterase/pyrophosphatases (E-NPP enzymes) and ectoalkaline phosphatase activities were examined. Those patterns, together with results of enzyme assays, Western blotting, or semiquantitative RT-PCR show that NTPDase2 is the main ectonucleotidase for murine and human hepatoma cells, NTPDase3 for chick hepatocytes and LMH cells, and an E-NPP enzyme for chick cardiac myocytes. Evidence for NTPDase2 expression was lacking in all cells except the mouse and human hepatoma cells. TCDD increased expression of the NTPDase2 gene but only in the mouse and not in the human hepatoma cells. TCDD did not increase NTPDase3, NTPDase1, E-NPP, or alkaline phosphatase in any of the cell types examined. The failure of TCDD to increase ATP metabolism in HUVEC, chick LMH cells, hepatocytes, and cardiac myocytes can be attributed to their lack of NTPDase2 expression, while the increase in ATP metabolism by TCDD in the mouse but not the human hepatoma cells can be explained by differences in TCDD effects on mouse and human hepatoma NTPDase2 gene expression. In addition to characterizing effects of TCDD on ectonucleotidases, these studies reveal major differences in the complements of ectonucleotidases present in different cell types. It is likely that such differences are important for cell-specific susceptibility to extracellular nucleotide toxicity and responses to purinergic signaling.
Collapse
Affiliation(s)
- Emily Wood
- Department of Pharmacology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Ivnitski I, Elmaoued R, Walker MK. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) inhibition of coronary development is preceded by a decrease in myocyte proliferation and an increase in cardiac apoptosis. TERATOLOGY 2001; 64:201-12. [PMID: 11598926 DOI: 10.1002/tera.1065] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) causes cardiovascular toxicity, culminating in edema, hemorrhage, and mortality in piscine, avian, and mammalian embryos. To elucidate the mechanism of the cardiovascular teratogenicity of TCDD, we used a chick embryo model to determine whether TCDD alters coronary artery development and whether this alteration was associated with apoptosis and/or changes in myocyte proliferation. METHODS Fertile chicken eggs were injected with corn oil (control), 0.24, or 0.40 pmol TCDD/g in corn oil before incubation. To evaluate effects of TCDD on differentiation of coronary arteries, chick embryo hearts from incubation days 8 (D8), D10, and D12 were stained with anti-alpha-smooth muscle actin. Myocyte proliferation was measured by BrdU incorporation on D6, 8, 10, and 12 after TCDD treatment. In addition, temporal and spatial patterns of apoptosis were detected by TUNEL on D3, D5, D6, D8, and D10, and immunohistochemistry was used to identify the origin of apoptotic cells on D6. RESULTS TCDD increased apoptosis in structures where cell death normally occurs, including the outflow tract, endocardial cushion of the atrioventricular canal, and dorsal mesocardium, peaking in intensity on D6. Immunohistochemistry revealed that cells undergoing TCDD-induced apoptosis in the dorsal mesocardium were not neural or epicardial in origin. On D8 and D10 TCDD reduced myocyte proliferation. On D10, TCDD reduced coronary artery size and on D10 and D12 TCDD induced a dose-dependent decrease in coronary artery number. CONCLUSIONS The reduction of myocyte proliferation by TCDD preceded the reduction in coronary artery number and size, suggesting that changes in coronary development may be a consequence of reduced myocyte proliferation and a thinner ventricle wall. The peak of TCDD-induced increase in apoptosis occurred even earlier in embryo development and thus may contribute to changes in myocyte proliferation, coronary development, and cardiac structural malformations; however, a cause-and-effect relationship between apoptosis and these other events has yet to be established.
Collapse
Affiliation(s)
- I Ivnitski
- College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131-5691, USA
| | | | | |
Collapse
|
26
|
Khalil WF, Saitoh T, Shimoda M, Kokue E. In vitro cytochrome P450-mediated hepatic activities for five substrates in specific pathogen free chickens. J Vet Pharmacol Ther 2001; 24:343-8. [PMID: 11696084 DOI: 10.1046/j.1365-2885.2001.00349.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Total hepatic microsomal cytochrome P450 (CYP) content as well as in vitro CYP mediated activities for five substrates [bufuralol 1-hydroxylation, ethoxyresorufin O-deethylation, S-mephenytoin 4-hydroxylation, testosterone 6beta-hydroxylation, and tolbutamide hydroxylation] were measured in specific pathogen free male Japanese leghorn chickens and male beagle dogs. The Vmax, Km and intrinsic clearance (Vmax/Km) for these substrates were calculated and compared between animal species in order to evaluate the drug catalytic activity in chicken liver. The total CYP content in chicken (0.296 +/- 0.04 nmol/mg microsomal protein) was close to levels reported for other species including humans, cats, pigs and some nonmammalian vertebrates (e.g. snakes, frogs and trout fish), but was lower than levels measured in dogs (1.11 +/- 0.22) or recorded in guinea-pigs, hamsters, monkeys, mice, rabbits, rats, horse and ruminants. Bufuralol 1-hydroxylation, ethoxyresorufin O-deethylation, S-mephenytoin 4-hydroxylation, and testosterone 6beta-hydroxylation were lower in chickens than in dogs based on intrinsic clearance. On the other hand, tolbutamide hydroxylation was markedly higher in chickens than in dogs.
Collapse
Affiliation(s)
- W F Khalil
- Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Fuchu, Tokyo183-8509, Japan
| | | | | | | |
Collapse
|
27
|
Guiney PD, Walker MK, Spitsbergen JM, Peterson RE. Hemodynamic dysfunction and cytochrome P4501A mRNA expression induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin during embryonic stages of lake trout development. Toxicol Appl Pharmacol 2000; 168:1-14. [PMID: 11000095 DOI: 10.1006/taap.2000.8999] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Lake trout embryos exposed to [(3)H]2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) manifest toxicity after hatching by subcutaneous edema of the yolk sac, pericardial edema, meningeal edema, subcutaneous hemorrhages, and a marked congestion of blood flow in various vascular beds culminating in death. Our objective was to determine if this syndrome was associated temporally with morphologic lesions in the vascular endothelium, increased vascular permeability, and cytochrome P4501A (CYP1A) mRNA induction. Lake trout embryos exposed as fertilized eggs to TCDD were found to exhibit marked reductions in perfusion of the peripheral vasculature during the early sac fry stage of development (stage F(1)9), which consistently preceded other gross lesions and mortality observed later in sac fry development (stage F(2)10). This reduction in blood flow was manifested as severe capillary congestion and hemoconcentration in certain vascular beds. Transmission electron microscopic (TEM) examination of endothelial cells in these vascular beds failed to reveal cellular necrosis at hatching (stage E(5)8) and throughout sac fry development (stages F(1)9-F(2)10). Rather, only subtle ultrastructural changes in endothelial cells were found consisting of increased vacuolation, separation of intercellular junctions, and cytoplasmic blebbing, consistent with the TCDD dose and time course for developmental cardiovascular toxicity, which began to manifest itself in some embryos approximately 1 week prior to hatching (E(5)8). To assess permeability of yolk sac vasculature to certain constituents in blood, sac fry (stage F(2)10) were analyzed for the presence of plasma proteins, granulocytes, and serum creatine kinase activity in yolk sac subcutaneous edema fluid from control and TCDD-exposed treatment groups. TCDD dose- and time-related increases in yolk sac edema volume, plasma protein content of edema fluid, granulocyte concentration, and creatine kinase activity in the fluid were observed in midstage and late stage of sac fry development (stage F(2)10). Thus, yolk sac subcutaneous edema fluid is an ultrafiltrate of blood and results from increased vascular permeability. In contrast to the changes in vascular blood flow and permeability induced by TCDD during stages F(1)9 and F(2)10 of sac fry development, respectively, CYP1A mRNA levels were induced by TCDD as early as the 10-somite embryo (stage E(2)5). TCDD also caused a dose-related increase in CYP1A mRNA levels in sac fry at hatching (stage E(5)8) and throughout sac fry development (stages F(1)9-F(2)10). We conclude that subtle, ultrastructural changes in vascular endothelial cells consistently precede increases in vascular permeability and sac fry mortality; however, induction of CYP1A mRNA occurs prior to any observable morphological lesions, changes in vascular permeability, or sac fry mortality.
Collapse
Affiliation(s)
- P D Guiney
- NIEHS Aquatic Biomedical Center, University of Wisconsin-Milwaukee, 600 E. Greenfield Avenue, Milwaukee, Wisconsin 53204, USA
| | | | | | | |
Collapse
|
28
|
Fan L, Ovadia M, Friedman DM, Rifkind AB. Ventricular preexcitation sensitive to flecainide in late stage chick embryo ECGs: 2,3,7,8-tetrachlorodibenzo-p-dioxin impairs inotropic but not chronotropic or dromotropic responses to isoproterenol and confers resistance to flecainide. Toxicol Appl Pharmacol 2000; 166:43-50. [PMID: 10873717 DOI: 10.1006/taap.2000.8948] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
ECGs free of movement artefacts were obtained without anesthesia in 16- to 18-day-old chick embryos close to hatching and used to study the effect of the environmental toxin 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD) on cardiac rhythm and conduction. The ECGs of normal late stage chick embryos exhibited short PR intervals, frequent nonisoelectric PR segments, delta waves, and inverted T waves. Those ECG characteristics are found in patients with the Wolff-Parkinson-White syndrome (WPW) in which they reflect ventricular preexcitation associated with the use of accessory conduction pathways and arrhythmias. Isoproterenol (30 microg/egg) did not alter the ECG preexcitation characteristics. Flecainide, a sodium channel blocker used clinically to suppress WPW accessory pathway activity, at 0.5 to 5 mg per egg diminished the preexcitation and caused atrioventricular (AV) block, supporting the use of accessory pathways together with AV-nodal conduction in normal late stage chick embryos. The findings challenge the dogma that accessory pathways are entirely replaced by AV conduction pathways in late fetal development. TCDD, at 1-2 nmol per egg for 48 h, did not affect heart rate, the increase in heart rate by isoproterenol, or the ECG characteristics, suggesting that short-term TCDD treatment did not affect sinus node function or cardiac conduction. The latter results taken together with prior findings indicate that TCDD differentially impairs the inotropic and lusitropic effects but not the chronotropic or dromotropic effects of isoproterenol. In TCDD-treated embryos, flecainide, tested at 5 mg per egg, caused much less inhibition of preexcitation or production of AV block than in the untreated or solvent-treated controls. The resistance to flecainide represents a new TCDD effect consistent with the reported increase of cardiac myocyte [Ca(2+)](i) by TCDD treatment.
Collapse
Affiliation(s)
- L Fan
- Department of Pediatrics, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | |
Collapse
|