1
|
Su CH, Chen SP, Chen LY, Yang JJ, Lee YC, Lee SS, Chen HH, Ng YY, Kuan YH. 3-Bromofluoranthene-induced cardiotoxicity of zebrafish and apoptosis in the vascular endothelial cells via intrinsic and extrinsic caspase-dependent pathways. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 228:112962. [PMID: 34775346 DOI: 10.1016/j.ecoenv.2021.112962] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/28/2021] [Accepted: 10/31/2021] [Indexed: 06/13/2023]
Abstract
Fluoranthene, a high-molecular-weight polycyclic aromatic hydrocarbon (PAH), is widely present in air pollutants, including fine inhalable particulate matter. 3-Bromofluoranthene (3-BrFlu), which is a brominated fluoranthene and halogenated PAH, is generated from waste combustion, metallurgical processes, cement production, e-waste dismantling, and photoreaction. Vascular endothelial cells have key functions in the homeostasis and the development of the cardiovascular system. The zebrafish model has been widely employed to study cardiotoxicity and embryotoxicity. However, no evidence has indicated that 3-BrFlu induces cytotoxicity in vascular endothelial cells, or cardiotoxicity and embryotoxicity in zebrafish. In this study, 3-BrFlu induced concentration-dependent changes in embryo- and cardiotoxicity. Cytotoxicity was also induced by 3-BrFlu in a concentration-dependent manner through apoptosis and necrosis in vascular endothelial cells, SVEC4-10 cells. The activities of caspase-3, -8, and -9 were induced by 3-BrFlu via an intrinsic pathway constituting Bcl-2 downregulation, Bad upregulation, and mitochondrial dysfunction; the extrinsic pathway included the expression of death receptors, including tumour necrosis factor α and Fas receptors. These results indicated that 3-BrFlu caused cardio- and embryotoxicity in zebrafish through vascular endothelial cells cytotoxicity resulting from caspase-dependent apoptosis through intrinsic and extrinsic pathways.
Collapse
Affiliation(s)
- Chun-Hung Su
- Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan, ROC; Department of Internal Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC; Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC
| | - Shih-Pin Chen
- Department of Internal Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC; Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan, ROC
| | - Li-You Chen
- Department of Anatomy, School of Medicine, College of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC; Department of Medical Education, Chung Shan Medical University Hospital, Taichung, Taiwan, ROC
| | - Jiann-Jou Yang
- Department of BioMedical Sciences, Chung Shan Medical University, Taichung, Taiwan, ROC; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan, ROC
| | - Yi-Chia Lee
- Department of Pharmacology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC; Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan, ROC
| | - Shiuan-Shinn Lee
- School of Public Health, Chung Shan Medical University, Taichung, Taiwan, ROC
| | - Hsin-Hung Chen
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Asia University Hospital, Taichung, Taiwan, ROC; School of Medicine, Institute of Medicine and public health, Chung Shan Medical University, Taichung, Taiwan, ROC; Chung Sheng Clinic, Nantou, Taiwan, ROC
| | - Yan-Yan Ng
- Department of Pediatric, Chung Kang branch, Cheng Ching Hospital, Taichung City, Taiwan, ROC
| | - Yu-Hsiang Kuan
- Department of Pharmacology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC; Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan, ROC.
| |
Collapse
|
2
|
Li M, Wu J, Hu G, Song Y, Shen J, Xin J, Li Z, Liu W, Dong E, Xu M, Zhang Y, Xiao H. Pathological matrix stiffness promotes cardiac fibroblast differentiation through the POU2F1 signaling pathway. SCIENCE CHINA. LIFE SCIENCES 2021; 64:242-254. [PMID: 32617828 DOI: 10.1007/s11427-019-1747-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 05/21/2020] [Indexed: 12/13/2022]
Abstract
Cardiac fibroblast (CF) differentiation into myofibroblasts is a crucial cause of cardiac fibrosis, which increases in the extracellular matrix (ECM) stiffness. The increased stiffness further promotes CF differentiation and fibrosis. However, the molecular mechanism is still unclear. We used bioinformatics analysis to find new candidates that regulate the genes involved in stiffness-induced CF differentiation, and found that there were binding sites for the POU-domain transcription factor, POU2F1 (also known as Oct-1), in the promoters of 50 differentially expressed genes (DEGs) in CFs on the stiffer substrate. Immunofluorescent staining and Western blotting revealed that pathological stiffness upregulated POU2F1 expression and increased CF differentiation on polyacrylamide hydrogel substrates and in mouse myocardial infarction tissue. A chromatin immunoprecipitation assay showed that POU2F1 bound to the promoters of fibrosis repressors IL1R2, CD69, and TGIF2. The expression of these fibrosis repressors was inhibited on pathological substrate stiffness. Knockdown of POU2F1 upregulated these repressors and attenuated CF differentiation on pathological substrate stiffness (35 kPa). Whereas, overexpression of POU2F1 downregulated these repressors and enhanced CF differentiation. In conclusion, pathological stiffness upregulates the transcription factor POU2F1 to promote CF differentiation by inhibiting fibrosis repressors. Our work elucidated the crosstalk between CF differentiation and the ECM and provided a potential target for cardiac fibrosis treatment.
Collapse
Affiliation(s)
- Mingzhe Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Jimin Wu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Guomin Hu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Yao Song
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Jing Shen
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Junzhou Xin
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Zijian Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Wei Liu
- Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Erdan Dong
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
- Institute of Cardiovascular Sciences, Health Science Center, Peking University, Beijing, 100191, China
| | - Ming Xu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Youyi Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| |
Collapse
|
3
|
Jiang Y, Zhang S, Zhang X, Li N, Zhang Q, Guo X, Chi X, Tong M. Peptidomic analysis of zebrafish embryos exposed to polychlorinated biphenyls and their impact on eye development. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 175:164-172. [PMID: 30897415 DOI: 10.1016/j.ecoenv.2019.03.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/22/2019] [Accepted: 03/04/2019] [Indexed: 06/09/2023]
Abstract
Polychlorinated biphenyls (PCBs), a class of persistent organic pollutant, are closely related to abnormal eye development in children. However, little is known regarding the role of peptides in the development of PCB-induced ocular dysplasia. To characterize the nature of PCB exposure on peptides involved in the development of the ocular system, we used liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS) to detect differential expression of peptides between normal and PCB-exposed zebrafish embryos. A total of 7900 peptides were analyzed, 90 of which were differentially expressed, with 29 being up-regulated and 61 down-regulated. These peptides were investigated using ingenuity pathway analysis (IPA) and gene ontology (GO) analysis to explore their role in eye development. This study identified 18 peptides associated with the development of the optic nerve and ocular system in the PCB-exposure group, as well as 10 peptides that are located in the functional domain of their precursor proteins. These peptides provide potential biomarkers for the treatment of ocular dysplasia caused by PCBs and may help us understand the mechanism of abnormal eye development caused by organic pollutants.
Collapse
Affiliation(s)
- Yue Jiang
- Department of Pediatrics, Nanjing Medical University, Nanjing 210004, China
| | - Shuchun Zhang
- Department of Pediatrics, Nanjing Medical University, Nanjing 210004, China
| | - Xin Zhang
- Department of Pediatrics, Nanjing Medical University, Nanjing 210004, China; Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital, Obstetrics and Gynecology Hospital affiliated to Nanjing Medical University, Nanjing, China
| | - Nan Li
- Ningbo First Hospital | Ningbo Hospital of Zhejiang University, Ningbo 315010, China
| | - Qingyu Zhang
- Department of Pediatrics, Nanjing Medical University, Nanjing 210004, China
| | - Xirong Guo
- Department of Pediatrics, Nanjing Medical University, Nanjing 210004, China; Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital, Obstetrics and Gynecology Hospital affiliated to Nanjing Medical University, Nanjing, China
| | - Xia Chi
- Department of Pediatrics, Nanjing Medical University, Nanjing 210004, China; Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital, Obstetrics and Gynecology Hospital affiliated to Nanjing Medical University, Nanjing, China.
| | - Meiling Tong
- Department of Pediatrics, Nanjing Medical University, Nanjing 210004, China; Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital, Obstetrics and Gynecology Hospital affiliated to Nanjing Medical University, Nanjing, China.
| |
Collapse
|
4
|
Hufgard JR, Sprowles JLN, Pitzer EM, Koch SE, Jiang M, Wang Q, Zhang X, Biesiada J, Rubinstein J, Puga A, Williams MT, Vorhees CV. 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. J Appl Toxicol 2018; 39:603-621. [PMID: 30561030 DOI: 10.1002/jat.3751] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 09/17/2018] [Accepted: 10/14/2018] [Indexed: 01/17/2023]
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.
Collapse
Affiliation(s)
- Jillian R Hufgard
- Department of Pediatrics, University of Cincinnati College of Medicine, and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
| | - Jenna L N Sprowles
- Department of Pediatrics, University of Cincinnati College of Medicine, and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
| | - Emily M Pitzer
- Department of Pediatrics, University of Cincinnati College of Medicine, and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
| | - Sheryl E Koch
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Min Jiang
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Qin Wang
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Xiang Zhang
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Jacek Biesiada
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Jack Rubinstein
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Alvaro Puga
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Michael T Williams
- Department of Pediatrics, University of Cincinnati College of Medicine, and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
| | - Charles V Vorhees
- Department of Pediatrics, University of Cincinnati College of Medicine, and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
| |
Collapse
|
5
|
Rebuzzini P, Zuccolo E, Civello C, Fassina L, Arechaga J, Izquierdo A, Faris P, Zuccotti M, Moccia F, Garagna S. Polychlorinated biphenyls reduce the kinematics contractile properties of embryonic stem cells-derived cardiomyocytes by disrupting their intracellular Ca 2+ dynamics. Sci Rep 2018; 8:17909. [PMID: 30559452 PMCID: PMC6297156 DOI: 10.1038/s41598-018-36333-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/19/2018] [Indexed: 12/11/2022] Open
Abstract
Persistent organic pollutants are a group of chemicals that include polychlorinated biphenyls (PCBs). PCBs exposure during adult life increases incidence and severity of cardiomyopathies, whereas in utero exposure determines congenital heart defects. Being fat-soluble, PCBs are passed to newborns through maternal milk, impairing heart functionality in the adult. It is still unknown how PCBs impair cardiac contraction at cellular/molecular levels. Here, we study the molecular mechanisms by which PCBs cause the observed heart contraction defects, analysing the alterations of Ca2+ toolkit components that regulate contraction. We investigated the effect that Aroclor 1254 (Aroclor), a mixture of PCBs, has on perinatal-like cardiomyocytes derived from mouse embryonic stem cells. Cardiomyocytes, exposed to 1 or 2 µg/ml Aroclor for 24 h, were analyzed for their kinematics contractile properties and intracellular Ca2+ dynamics. We observed that Aroclor impairs cardiomyocytes contractile properties by inhibiting spontaneous Ca2+ oscillations. It disrupts intracellular Ca2+ homeostasis by reducing the sarcoplasmic reticulum Ca2+ content and by inhibiting voltage-gated Ca2+ entry. These findings contribute to the understanding of the molecular underpinnings of PCBs-induced cardiovascular alterations, which are emerging as an additional life-threatening hurdle associated to PCBs pollution. Therefore, PCBs-dependent alteration of intracellular Ca2+ dynamics is the most likely trigger of developmental cardiac functional alteration.
Collapse
Affiliation(s)
- Paola Rebuzzini
- Laboratorio di Biologia dello Sviluppo, Dipartimento di Biologia e Biotecnologie, Università degli Studi di Pavia, Pavia, Italy. .,Centre for Health Technologies (C.H.T.), Università degli Studi di Pavia, Pavia, Italy.
| | - Estella Zuccolo
- Laboratorio di Fisiologia Generale, Dipartimento di Biologia e Biotecnologie, Università degli Studi di Pavia, Pavia, Italy
| | - Cinzia Civello
- Laboratorio di Biologia dello Sviluppo, Dipartimento di Biologia e Biotecnologie, Università degli Studi di Pavia, Pavia, Italy
| | - Lorenzo Fassina
- Centre for Health Technologies (C.H.T.), Università degli Studi di Pavia, Pavia, Italy.,Dipartimento di Ingegneria Industriale e dell'Informazione, Università degli Studi di Pavia, Pavia, Italy
| | - Juan Arechaga
- Laboratory of Stem Cells, Development and Cancer, Department of Cell Biology and Histology, Faculty of Medicine and Nursing, Universidad del País Vasco, Vizcaya, Spain
| | - Amaia Izquierdo
- Laboratory of Stem Cells, Development and Cancer, Department of Cell Biology and Histology, Faculty of Medicine and Nursing, Universidad del País Vasco, Vizcaya, Spain
| | - Pawan Faris
- Laboratorio di Fisiologia Generale, Dipartimento di Biologia e Biotecnologie, Università degli Studi di Pavia, Pavia, Italy.,Department of Biology, College of Science, Salahaddin University, Erbil, Kurdistan-Region of Iraq, Iraq
| | - Maurizio Zuccotti
- Laboratorio di Biologia dello Sviluppo, Dipartimento di Biologia e Biotecnologie, Università degli Studi di Pavia, Pavia, Italy. .,Centre for Health Technologies (C.H.T.), Università degli Studi di Pavia, Pavia, Italy.
| | - Francesco Moccia
- Laboratorio di Fisiologia Generale, Dipartimento di Biologia e Biotecnologie, Università degli Studi di Pavia, Pavia, Italy
| | - Silvia Garagna
- Laboratorio di Biologia dello Sviluppo, Dipartimento di Biologia e Biotecnologie, Università degli Studi di Pavia, Pavia, Italy. .,Centre for Health Technologies (C.H.T.), Università degli Studi di Pavia, Pavia, Italy.
| |
Collapse
|
6
|
Jiang Q, Wang C, Xue C, Xue L, Wang M, Li C, Deng Z, Wang Q. Changes in the levels of l-carnitine, acetyl-l-carnitine and propionyl-l-carnitine are involved in perfluorooctanoic acid induced developmental cardiotoxicity in chicken embryo. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 48:116-124. [PMID: 27771504 DOI: 10.1016/j.etap.2016.10.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 10/16/2016] [Accepted: 10/17/2016] [Indexed: 06/06/2023]
Abstract
Perfluorooctanoic acid (PFOA), a persistent organic pollutant, is associated with developmental toxicity. This study investigated the mechanism of PFOA-induced developmental cardiotoxicity in chicken embryo, focusing on the interactions between developmental exposure to PFOA and the levels of l-carnitine (LC), acetyl-l-carnitine (ALC) and propionyl-l-carnitine (PLC) in the heart. To evaluate the developmental cardiotoxicity, fertile chicken eggs were exposed to 0.1, 0.5, 1, 2 or 5mg/kg PFOA via air cell injection. Furthermore, exposure to 2mg/kg PFOA, with or without 100mg/kg LC were applied to investigate the effects of LC supplement. The results of functional and morphological assessments confirmed PFOA induced developmental cardiotoxicity in chicken embryo, which could be alleviated by co-exposure to LC. LC-MS/MS results also revealed remarkable decrease in LC, ALC and PLC levels in embryonic day six (ED6) chicken embryo hearts as well as LC level in embryonic day fifteen (ED15) chicken embryo hearts following developmental exposure to 2mg/kg PFOA. Meanwhile, co-exposure to 100mg/kg LC significantly elevated the levels of LC, ALC and PLC in chicken embryo hearts. Significantly elevated expression level of carnitine acetyltransferase (CRAT) in PFOA-exposed ED6 chicken embryo hearts was observed via western blotting, while LC co-exposure counteracted such changes. In conclusion, changes in the levels of LC, ALC and PLC in early embryonic stages are associated with PFOA induced developmental cardiotoxicity in chicken embryos.
Collapse
Affiliation(s)
- Qixiao Jiang
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong 266021, China.
| | - Chunbo Wang
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong 266021, China.
| | - Chan Xue
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong 266021, China
| | - Lingfang Xue
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong 266021, China
| | - Meiting Wang
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong 266021, China
| | - Changhao Li
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong 266021, China
| | - Ziwen Deng
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong 266021, China
| | - Qian Wang
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong 266021, China
| |
Collapse
|
7
|
Development of cardiac hypertrophy by sunitinib in vivo and in vitro rat cardiomyocytes is influenced by the aryl hydrocarbon receptor signaling pathway. Arch Toxicol 2013; 88:725-38. [PMID: 24247421 DOI: 10.1007/s00204-013-1159-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 11/05/2013] [Indexed: 12/23/2022]
Abstract
Sunitinib (SUN) is a new tyrosine kinase inhibitor that possesses both anti-angiogenic and anti-tumor activities. Although SUN has improved survival rate in cancer patients, cardiotoxicity has been reported as a significant side effect. Several studies suggested a role for the aryl hydrocarbon receptor (AhR) and its regulated genes such as cytochrome P4501A1 (CYP1A1) in the pathogenesis of heart failure and cardiac hypertrophy. To test the hypothesis that SUN induces cardiac hypertrophy through the modulation of AhR, Wistar albino rats were treated for 15 and 30 days with increasing doses of SUN (25, 50, and 100 mg/kg), whereas at the in vitro level, rat cardiomyocyte H9c2 cells were incubated with SUN (1, 2.5, and 5 μM). Thereafter, cardiac hypertrophy parameters were determined at the biochemical, histopathology, and gene expression levels. SUN treatment causes increase in cardiac enzymes, changes in histopathology, and induction in several hypertrophic markers. This was associated with proportional increase in the CYP1A1 gene in a concentration- and time-dependent manner. The direct involvement of AhR in the SUN-induced cardiac hypertrophy in H9c2 cells was supported by the ability of resveratrol, an AhR antagonist, to block the SUN-induced hypertrophy and the ability of SB203580, a novel AhR agonist, to potentiate SUN-induced hypertrophic genes. This is the first demonstration that SUN induces hypertrophic genes in vivo and in vitro rat cardiomyocyte through AhR/CYP1A1-mediated mechanism.
Collapse
|
8
|
The role of aryl hydrocarbon receptor signaling pathway in cardiotoxicity of acute lead intoxication in vivo and in vitro rat model. Toxicology 2013; 306:40-9. [DOI: 10.1016/j.tox.2013.01.024] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 01/26/2013] [Accepted: 01/29/2013] [Indexed: 11/17/2022]
|
9
|
Guida M, Marra M, Zullo F, Guida M, Trifuoggi M, Biffali E, Borra M, De Mieri G, D’Alessandro R, De Felice B. Association between exposure to dioxin-like polychlorinated biphenyls and miR-191 expression in human peripheral blood mononuclear cells. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2013; 753:36-41. [DOI: 10.1016/j.mrgentox.2012.12.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 11/02/2012] [Accepted: 12/22/2012] [Indexed: 10/27/2022]
|
10
|
Al-Arifi MN, Maayah ZH, Alshamrani AA, Korashy HM. Impact of cigarette smoke exposure on the expression of cardiac hypertrophic genes, cytochrome P450 enzymes, and oxidative stress markers in rats. J Toxicol Sci 2013; 37:1083-90. [PMID: 23038017 DOI: 10.2131/jts.37.1083] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Various experimental and clinical studies strongly support a cigarette smoke-heart disease association and suggest possible mechanisms, unfortunately, the involvement of genetic modulations remain unexplored. Thus, the main aim of the current study was to evaluate the effects of sub-chronic cigarette smoke exposure on the mRNA expression of cardiac hypertrophy genes, cytochrome P450 (CYP) enzymes, and the oxidative stress markers in heart rats. For this purpose, Wistar albino rats were exposed to increasing doses of passive cigarette smoke 2, 4, 8, and 24 cigarettes per day for 7 consecutive days. The mRNA expression of fifteen cardiac genes was determined using real-time polymerase chain reaction. Our results showed that the levels of hypertrophic genes; atrial natriuretic peptide, brain natriuretic peptide, and β-myosin heavy chain were significantly induced, whereas the anti-hypertrophic gene α-myosin heavy chain was dramatically inhibited, in heart tissues of passive-smoke-exposed groups compared with normal-control groups. This was accompanied with a significant induction of CYP enzymes; CYP1A1, CYP2C11, CYP2E1, and CYP3A2, and the expression of oxidative stress genes, heme oxygenase 1, catalase, cyclooxygenase, and glutathione S-Transferase. The ability of cigarette smoke to induce cardiac hypertrophic genes, CYPs enzymes, and oxidative stress, collectively explore the molecular mechanism of cigarette smoke-induced cardiac diseases and brings further investigative attention to the public health issue of the injurious effects of chronic passive smoke exposure. In conclusion, sub-chronic environmental tobacco smoke exposure increases the incidence of cardiovascular diseases through modulation of cardiac genes.
Collapse
|
11
|
Inhibition of Soluble Epoxide Hydrolase Confers Cardioprotection and Prevents Cardiac Cytochrome P450 Induction by Benzo(a)pyrene. J Cardiovasc Pharmacol 2011; 57:273-81. [DOI: 10.1097/fjc.0b013e3182055baf] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
12
|
Gillardin V, Silvestre F, Dieu M, Delaive E, Raes M, Thomé JP, Kestemont P. Protein expression profiling in the African clawed frog Xenopus laevis tadpoles exposed to the polychlorinated biphenyl mixture aroclor 1254. Mol Cell Proteomics 2008; 8:596-611. [PMID: 19011258 DOI: 10.1074/mcp.m800323-mcp200] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Exposure to environmental pollutants such as polychlorinated biphenyls (PCBs) is now taken into account to partly explain the worldwide decline of amphibians. PCBs induce deleterious effects on developing amphibians including deformities and delays in metamorphosis. However, the molecular mechanisms by which they express their toxicity during the development of tadpoles are still largely unknown. A proteomics analysis was performed on developing Xenopus laevis tadpoles exposed from 2 to 5 days postfertilization to either 0.1 or 1 ppm Aroclor 1254, a PCB mixture. Two-dimensional DIGE with a minimal labeling method coupled to nanoflow liquid chromatography-tandem mass spectrometry was used to detect and identify proteins differentially expressed under PCBs conditions. Results showed that 59 spots from the 0.1 ppm Aroclor 1254 condition and 57 spots from the 1 ppm Aroclor 1254 condition displayed a significant increase or decrease of abundance compared with the control. In total, 28 proteins were identified. The results suggest that PCBs induce mechanisms against oxidative stress (peroxiredoxins 1 and 2), adaptative changes in the energetic metabolism (enolase 1, glycerol-3-phosphate dehydrogenase, and creatine kinase muscle and brain types), and the implication of the unfolded protein response system (glucose-regulated protein, 58 kDa). They also affect, at least at the highest concentration tested, the synthesis of proteins involved in normal cytogenesis (alpha-tropomyosin, myosin heavy chain, and alpha-actin). For the first time, proteins such as aldehyde dehydrogenase 7A1, CArG binding factor-A, prolyl 4-hydroxylase beta, and nuclear matrix protein 200 were also shown to be up-regulated by PCBs in developing amphibians. These data argue that protein expression reorganization should be taken into account while estimating the toxicological hazard of wild amphibian populations exposed to PCBs.
Collapse
Affiliation(s)
- Virginie Gillardin
- Unité de Recherche en Biologie des Organismes, Facultés Universitaires Notre-Dame de la Paix, Rue de Bruxelles 61, B-5000 Namur, Belgium
| | | | | | | | | | | | | |
Collapse
|
13
|
Blickwede M, Borlak J. Isolation and characterization of metabolically competent pulmonary epithelial cells from pig lung tissue. Xenobiotica 2008; 35:927-41. [PMID: 16393853 DOI: 10.1080/00498250500296264] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Administration of drugs by inhalation opens new possibilities for entry into the systemic circulation and cultures of porcine pulmonary epithelial cells (PECs) may prove to be valuable in the prediction of pulmonary metabolism of drugs in humans. This paper, therefore, reports a method for the routine isolation and cultivation of PECs from slaughterhouse animals. On average 1.5x10(6) cells g-1 tissue were isolated by discontinuous density-gradient centrifugation. Cells were subsequently cultivated on collagen-coated plates and characterized by staining for alkaline phosphatase, by tannic acid staining of lamellar bodies and by surfactant protein (SP) expression at days 0, 3 and 6 in culture. Over 70% of purified cells were positive for SP-C and tannic acid staining and thus defined as epithelial cells of alveolar origin (AECs). The AEC phenotype was also confirmed by specific binding of marker lectins (Maclura pomifera and Helix pomatia) and by studying gene expression and activity of cytochrome P450 monooxygenases. Testosterone, ethoxyresorufin, benzyloxyresorufin and verapamil were used as substrates for cytochrome P450-catalysed oxidations and cultured cells were found to be differentiated as well as metabolically competent during cultivation. Therefore, this culture system enables in depth pulmonary biotransformation and toxicity studies.
Collapse
Affiliation(s)
- M Blickwede
- Department of Drug Research and Medical Biotechnology, Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
| | | |
Collapse
|
14
|
Aboutabl ME, El-Kadi AOS. Constitutive expression and inducibility of CYP1A1 in the H9c2 rat cardiomyoblast cells. Toxicol In Vitro 2007; 21:1686-91. [PMID: 17719738 DOI: 10.1016/j.tiv.2007.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2007] [Revised: 06/13/2007] [Accepted: 07/09/2007] [Indexed: 11/29/2022]
Abstract
Cardiomyocytes are a valuable tool for studying the drug metabolizing enzymes in the heart. However, isolated cardiomyocytes are rather fragile and difficult to isolate. Therefore, there is an urgent need for an in vitro cell line model. The H9c2 cells are commonly used as an in vitro model for studying the cellular mechanisms and signaling pathways involved in drug-induced cardiotoxicity. These cells maintain many molecular markers of cardiomyocytes and show morphological characteristics of immature embryonic cardiomyocytes. Therefore, in the present study we examined the expression and inducibility of CYP1A1 in the H9c2 rat cardiomyoblast cells. Our results showed that treatment of H9c2 cells with the CYP1A1 inducer, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) significantly induced CYP1A1 at mRNA, protein, and activity levels in a concentration-dependent manner. The RNA synthesis inhibitor, actinomycin D, completely blocked the CYP1A1 mRNA induction by TCDD, indicating the requirement of de novo RNA synthesis through transcriptional activation. In conclusion, we demonstrated for the first time the constitutive expression and inducibility of CYP1A1 in H9c2 cells. Therefore, this cell line offers a unique in vitro model to study the role of CYP1A1 in the pathogenesis of various cardiovascular diseases.
Collapse
Affiliation(s)
- Mona E Aboutabl
- Faculty of Pharmacy and Pharmaceutical Sciences, 3126 Dentistry/Pharmacy Centre, University of Alberta, Edmonton, Alberta, Canada T6G 2N8
| | | |
Collapse
|
15
|
Menzel R, Yeo HL, Rienau S, Li S, Steinberg CEW, Stürzenbaum SR. Cytochrome P450s and short-chain dehydrogenases mediate the toxicogenomic response of PCB52 in the nematode Caenorhabditis elegans. J Mol Biol 2007; 370:1-13. [PMID: 17499272 DOI: 10.1016/j.jmb.2007.04.058] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2007] [Revised: 04/13/2007] [Accepted: 04/19/2007] [Indexed: 11/28/2022]
Abstract
Although non-coplanar PCBs are ubiquitous organic chemicals known to induce numerous biological responses and thus are toxic to man and wildlife, little is known about the toxic mode of action. Using PCB52, an ortho-substituted, 2,2',5,5'-tetrachlorobiphenyl, it was possible to pinpoint the relationship between induced gene expression and observed toxicity in the model nematode Caenorhabditis elegans. On the basis of the calculated EC20 for brood size (5 mg/l), whole genome DNA microarray experiments were performed to identify differentially expressed genes. Gene knockdown by RNAi was used to determine the consequences in reproductive fitness in the presence and in the absence of PCB52. On the basis of altered phenotype, several gene classes were identified to have a pivotal role in PCB52 toxicogenesis, most notably cytochrome P450s, short-chain dehydrogenases and lipases. In addition to this, four of six selected cytochrome P450s were shown to be involved in fat storage, with PCB52 exposure increasing the fat content in N2 wild-type as indicated by staining with Nile red. Furthermore, exposure to PCB52 induces a general detoxification response via small heat-shock proteins and caspases. Our data provide strong evidence of the molecular mechanisms that underlie the toxicity of non-coplanar PCBs, and confirms that, despite the ability to metabolize PCB, alterations in lipid metabolism and storage are major factors that drive the toxic effect of PCB52.
Collapse
Affiliation(s)
- Ralph Menzel
- Department of Biology, Freshwater and Stress Ecology, Humboldt University at Berlin, Unter den Linden 6, 10099 Berlin, Germany
| | | | | | | | | | | |
Collapse
|
16
|
Korashy HM, El-Kadi AOS. The role of aryl hydrocarbon receptor in the pathogenesis of cardiovascular diseases. Drug Metab Rev 2006; 38:411-50. [PMID: 16877260 DOI: 10.1080/03602530600632063] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Numerous experimental and epidemiological studies have demonstrated that polycyclic aromatic hydrocarbons (PAHs), which are major constituents of cigarette tobacco tar, are strongly involved in the pathogenesis of the cardiovascular diseases (CVDs). Knowing that PAH-induced toxicities are mediated by the activation of a cytosolic receptor, aryl hydrocarbon receptor (AhR), which regulates the expression of a group of xenobiotic metabolizing enzymes (XMEs) such as CYP1A1, CYP1A2, CYP1B1, NQO1, and GSTA1, suggests a direct link between AhR-regulated XMEs and CVDs. Therefore, identifying the localization and expression of the AhR and its regulated XMEs in the cardiovascular system (CVS) is of major importance in understanding their physiological and pathological roles. Generally, it was believed that the levels of AhR-regulated XMEs are lower in the CVS than in the liver; however, it has been shown that similar or even higher levels of expression are demonstrated in the CVS in a tissue- and species-specific manner. Moreover, most, if not all, AhR-regulated XMEs are differentially expressed in most of the CVS, particularly in the endothelium cells, aorta, coronary arteries, and ventricles. Although the exact mechanisms of PAH-mediated cardiotoxicity are not fully understood, several mechanisms are proposed. Generally, induction of CYP1A1, CYP1A2, and CYP1B1 is considered cardiotoxic through generating reactive oxygen species (ROS), DNA adducts, and endogenous arachidonic acid metabolites. However the cardioprotective properties of NQO1 and GSTA1 are mainly attributed to the antioxidant effect by decreasing ROS and increasing the levels of endogenous antioxidants. This review provides a clear understanding of the role of AhR and its regulated XMEs in the pathogenesis of CVDs, in which imbalance in the expression of cardioprotective and cardiotoxic XMEs is the main determinant of PAH-mediated cardiotoxicity.
Collapse
Affiliation(s)
- Hesham M Korashy
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
17
|
Huang X, Lessner L, Carpenter DO. Exposure to persistent organic pollutants and hypertensive disease. ENVIRONMENTAL RESEARCH 2006; 102:101-6. [PMID: 16458884 DOI: 10.1016/j.envres.2005.12.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2005] [Revised: 12/06/2005] [Accepted: 12/12/2005] [Indexed: 05/06/2023]
Abstract
Previous research suggests that exposure to persistent organic pollutants (POPs) increases the risk of chronic diseases such as hypertension. We identified the zip codes of more than 800 waste sites contaminated with POPs and other pollutants, based on which we classified zip codes of upstate New York into three groups: "POPs sites", zip codes containing hazardous waste sites with POPs; "other waste sites", zip codes containing hazardous waste sites but not with POPs; and "clean sites", zip codes without any known hazardous waste sites. Age, gender, race, and zip code of residence of patients diagnosed with hypertension (ICD-9 codes 401-404) were identified using the New York Statewide Planning and Research Cooperative System (SPARCS) for the years 1993-2000. A generalized linear model, the negative binomial model, was used to assess the effect of living in a zip code with a hazardous waste site on the discharge rate of hypertension. After control for the aforementioned covariates, we found a statistically significant elevation of 19.2% (95% CI = 8.5%, 31%) in hypertension discharge rate for "POPs sites" and a 10% elevation in discharge rates for "other waste sites" as compared to "clean sites". In a subset of "POPs sites" where people have higher income, smoke less, exercise more and have healthier diets, there was still a 13.9% elevation of hypertension discharge rate as compared to "clean sites". The results support the hypothesis that living near hazardous waste sites, particularly sites containing POPs, may constitute a risk of exposure and of developing hypertension.
Collapse
Affiliation(s)
- Xiaoyu Huang
- Department of Biometry and Statistics, School of Public Health, University at Albany, SUNY, One University Place, Rm. 131, Rensselaer, NY 12144, USA
| | | | | |
Collapse
|
18
|
Klein G, Schaefer A, Hilfiker-Kleiner D, Oppermann D, Shukla P, Quint A, Podewski E, Hilfiker A, Schröder F, Leitges M, Drexler H. Increased Collagen Deposition and Diastolic Dysfunction but Preserved Myocardial Hypertrophy After Pressure Overload in Mice Lacking PKCε. Circ Res 2005; 96:748-55. [PMID: 15761199 DOI: 10.1161/01.res.0000161999.86198.1e] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Overexpression and activation of protein kinase C-epsilon (PKCepsilon) results in myocardial hypertrophy. However, these observations do not establish that PKCepsilon is required for the development of myocardial hypertrophy. Thus, we subjected PKCepsilon-knockout (KO) mice to a hypertrophic stimulus by transverse aortic constriction (TAC). KO mice show normal cardiac morphology and function. TAC caused similar cardiac hypertrophy in KO and wild-type (WT) mice. However, KO mice developed more interstitial fibrosis and showed enhanced expression of collagen Ialpha1 and collagen III after TAC associated with diastolic dysfunction, as assessed by tissue Doppler echocardiography (Ea/Aa after TAC: WT 2.1+/-0.3 versus KO 1.0+/-0.2; P<0.05). To explore underlying mechanisms, we analyzed the left ventricular (LV) expression pattern of additional PKC isoforms (ie, PKCalpha, PKCbeta, and PKCdelta). After TAC, expression and activation of PKCdelta protein was increased in KO LVs. Moreover, KO LVs displayed enhanced activation of p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK), whereas p42/p44-MAPK activation was attenuated. Under stretch, cultured KO fibroblasts showed a 2-fold increased collagen Ialpha1 (col Ialpha1) expression, which was prevented by PKCdelta inhibitor rottlerin or by p38 MAPK inhibitor SB 203580. In conclusion, PKCepsilon is not required for the development of a pressure overload-induced myocardial hypertrophy. Lack of PKCepsilon results in upregulation of PKCdelta and promotes activation of p38 MAPK and JNK, which appears to compensate for cardiac hypertrophy, but in turn, is associated with increased collagen deposition and impaired diastolic function.
Collapse
Affiliation(s)
- Gunnar Klein
- Department of Cardiovascular Medicine, Hannover Medical School, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|