1
|
Liu M, Wang L, Liu Z, Liu D, Li T, Ding L, Zeng S, Wang Z, Wang J, Zhang F, Zhang J, Zhang L, Li M, Liu G, Wang X, Zheng M. MiR-222-3p loaded stem cell nanovesicles repair myocardial ischemia damage via inhibiting mitochondrial oxidative stress. Life Sci 2025; 365:123447. [PMID: 39922425 DOI: 10.1016/j.lfs.2025.123447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/01/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
AIMS Mitochondrial oxidative stress (MOS) is a key contributor to poor cardiac function and a major driver of myocardial ischemia-reperfusion injury (MIRI). Our previous research demonstrated that stem cell-derived nanovesicles (NVs) enhanced cardiac function following ischemia-reperfusion (I/R) injury, although the underlying mechanisms remain unclear. We constructed and characterized miR-222-3p-loaded NVs. MATERIALS AND METHODS An in vitro hypoxia-reoxygenation (H/R) model was established using H9C2 cardiomyocytes. Mitochondrial oxidative respiratory function was assessed using Seahorse XF technology, while mitochondrial reactive oxygen species (mtROS) levels were quantified via flow cytometry. Additional assessments included mitochondrial permeability transition pore (mPTP) status, mitochondrial membrane potential, and mitochondrial DNA (mtDNA) integrity. An in vivo H/R model was developed using C57BL/6 mice. The therapeutic effects of NVs on MOS reduction and cardiac function improvement were evaluated through Masson's staining, immunofluorescence, echocardiography, transmission electron microscopy (TEM), and positron emission tomography/computed tomography (PET/CT). KEY FINDINGS RNA immunoprecipitation (RIP) confirmed that miR-222-3p directly targets cyp1a1. Overexpression of miR-222-3p or knockdown of cyp1a1 significantly improved mitochondrial activity in cardiomyocytes and conferred protection against I/R injury. Conversely, overexpression of cyp1a1 abrogated the protective effects of miR-222-3p. In vivo, NV treatment enhanced cardiac function, reduced MOS, and improved mitochondrial respiratory capacity in MIRI model mice. NV treatment, via miR-222-3p-mediated suppression of cyp1a1, mitigates MOS, enhances mitochondrial respiratory function, and improves cardiac outcomes in MIRI models. SIGNIFICANCE These findings provide a foundational basis for the clinical translation of NV-based therapies.
Collapse
Affiliation(s)
- Mei Liu
- Department of Cardiovascular Medicine, the First Hospital of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China; Hebei Provincial Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang 050031, Hebei Province, China
| | - Le Wang
- Department of Cardiovascular Medicine, the First Hospital of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China; Hebei Provincial Key Laboratory of Heart and Metabolism, Shijiazhuang 050031, Hebei Province, China
| | - Zhao Liu
- Traditional Chinese Medicine Processing Technology Innovation Center of Hebei Province, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang 050091, China; The First Affilfated Hospital of Hebei University of Chinese Medicine, Shijiazhuang 050011, China
| | - Dongyue Liu
- Department of Cardiovascular Medicine, the First Hospital of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China; Hebei Provincial Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang 050031, Hebei Province, China
| | - Tianshuo Li
- Department of Cardiovascular Medicine, the First Hospital of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China; Hebei Provincial Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang 050031, Hebei Province, China
| | - Lini Ding
- Department of Cardiovascular Medicine, the First Hospital of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China; Hebei Provincial Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang 050031, Hebei Province, China
| | - Shasha Zeng
- Department of Cardiovascular Medicine, the First Hospital of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China; Hebei Provincial Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang 050031, Hebei Province, China
| | - Zi Wang
- Department of Cardiovascular Medicine, the First Hospital of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China; Hebei Provincial Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang 050031, Hebei Province, China
| | - Jiaqiu Wang
- Department of Cardiovascular Medicine, the First Hospital of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China; Hebei Provincial Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang 050031, Hebei Province, China
| | - Fan Zhang
- Stem Cell Regenerative Medicine Clinical Research Center, the First Hospital of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China
| | - Jun Zhang
- Stem Cell Regenerative Medicine Clinical Research Center, the First Hospital of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China
| | - Limin Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Meng Li
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
| | - Gang Liu
- Department of Cardiovascular Medicine, the First Hospital of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China; Hebei Provincial Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang 050031, Hebei Province, China; Hebei International Joint Research Center for Structural Heart Disease, Shijiazhuang 050031, Hebei Province, China.
| | - Xianyun Wang
- Department of Cardiovascular Medicine, the First Hospital of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China; Hebei Provincial Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang 050031, Hebei Province, China.
| | - Mingqi Zheng
- Department of Cardiovascular Medicine, the First Hospital of HeBei Medical University, Shijiazhuang 050031, Hebei Province, China; Hebei Provincial Key Laboratory of Heart and Metabolism, Shijiazhuang 050031, Hebei Province, China.
| |
Collapse
|
2
|
Crispim D, Ramos C, Esteves F, Kranendonk M. The Adaptation of MCF-7 Breast Cancer Spheroids to the Chemotherapeutic Doxorubicin: The Dynamic Role of Phase I Drug Metabolizing Enzymes. Metabolites 2025; 15:136. [PMID: 39997761 PMCID: PMC11857127 DOI: 10.3390/metabo15020136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/05/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025] Open
Abstract
Background/Objectives: Drug resistance (DR) is a major challenge in cancer therapy, contributing to approximately 90% of cancer-related deaths. While alterations in drug metabolism are known to be key drivers of DR, their role-particularly in the early stages of acquired chemoresistance-remains understudied. Phase I drug-metabolizing enzymes (DMEs), especially cytochrome P450s (CYPs), significantly influence the metabolic fate of chemotherapeutic agents, directly affecting drug response. This study aimed to investigate the role of Phase I DMEs in the early metabolic adaptation of breast cancer (BC) MCF-7 cells to doxorubicin (DOX). Methods: Four types of spheroids were generated from MCF-7 cells that were either DOX-sensitive (DOXS) or adapted to low concentrations of the chemotherapeutic agent (DOXA 25, 35, and 45 nM). The expression levels of 92 Phase I DMEs and the activities of specific CYP isoforms were assessed in both DOXS and DOXA spheroids. Results: A total of twenty-four DMEs, including fifteen CYPs and nine oxidoreductases, were found to be differentially expressed in DOXA spheroids. Pathway analysis identified key roles for the differentially expressed DMEs in physiologically relevant pathways, including the metabolism of drugs, arachidonic acid, retinoic acid, and vitamin D. Conclusions: The deconvolution of these pathways highlights a highly dynamic process driving early-stage DOX resistance, with a prominent role of CYP3A-dependent metabolism in DOX adaptation. Our findings provide valuable insights into the underlying molecular mechanisms driving the early adaptation of MCF-7 cells to DOX exposure.
Collapse
Affiliation(s)
- Daniel Crispim
- Comprehensive Health Research Centre (CHRC) NOVA Medical School | Faculty of Medical Sciences, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (D.C.); (C.R.); (F.E.)
| | - Carolina Ramos
- Comprehensive Health Research Centre (CHRC) NOVA Medical School | Faculty of Medical Sciences, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (D.C.); (C.R.); (F.E.)
| | - Francisco Esteves
- Comprehensive Health Research Centre (CHRC) NOVA Medical School | Faculty of Medical Sciences, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (D.C.); (C.R.); (F.E.)
- Instituto Politécnico de Setúbal (IPS), Escola Superior de Saúde (ESS), Departamento de Ciências Biomédicas, Estefanilha, 2910-761 Setúbal, Portugal
| | - Michel Kranendonk
- Comprehensive Health Research Centre (CHRC) NOVA Medical School | Faculty of Medical Sciences, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (D.C.); (C.R.); (F.E.)
| |
Collapse
|
3
|
Jiang T, Hou L, Rahman SM, Gong Z, Bai X, Vulpe C, Fasullo M, Gu AZ. Amplified and distinctive genotoxicity of titanium dioxide nanoparticles in transformed yeast reporters with human cytochrome P450 (CYP) genes. JOURNAL OF HAZARDOUS MATERIALS 2024; 474:134850. [PMID: 38850947 PMCID: PMC11948300 DOI: 10.1016/j.jhazmat.2024.134850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/10/2024]
Abstract
Titanium dioxide nanoparticles (nTiO2) have been considered a possible carcinogen to humans, but most existing studies have overlooked the role of human enzymes in assessing the genotoxicity of nTiO2. Here, a toxicogenomics-based in vitro genotoxicity assay using a GFP-fused yeast reporter library was employed to elucidate the genotoxic potential and mechanisms of nTiO2. Moreover, two new GFP-fused yeast reporter libraries containing either human CYP1A1 or CYP1A2 genes were constructed by transformation to investigate the potential modulation of nTiO2 genotoxicity in the presence of human CYP enzymes. This study found a lack of appreciable nTiO2 genotoxicity as indicated by the yeast reporter library in the absence of CYP expression but a significantly elevated indication of genotoxicity in either CYP1A1- or CYP1A2-expressing yeast. The intracellular reactive oxygen species (ROS) measurement indicated significantly higher ROS in yeast expressing either enzyme. The detected mitochondrial DNA damage suggested mitochondria as one of the target sites for oxidative damage by nTiO2 in the presence of either one of the CYP enzymes. The results thus indicated that the genotoxicity of nTiO2 was enhanced by human CYP1A1 or CYP1A2 enzyme and was associated with elevated oxidative stress, which suggested that the similar mechanisms could occur in human cells.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Civil and Environmental Engineering, Northeastern University, Boston, MA 02115, USA; Department of Environmental and Sustainable Engineering, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Liyuan Hou
- Department of Civil and Environmental Engineering, Utah State University, Logan, UT 84322, USA; Utah Water Research Laboratory, Utah State University, Logan, UT 84322, USA
| | - Sheikh Mokhlesur Rahman
- Department of Civil and Environmental Engineering, Northeastern University, Boston, MA 02115, USA; Department of Civil Engineering, Bangladesh University of Engineering and Technology, BUET Central Road, Dhaka 1000, Bangladesh
| | - Zixuan Gong
- Department of Materials, Imperial College London, London LND SW7 2AZ, UK
| | - Xueke Bai
- Department of Chemistry, The University of Manchester, Manchester M13 9PL, UK
| | - Christopher Vulpe
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, USA
| | - Michael Fasullo
- Department of Nanoscale Science and Engineering, University at Albany, State University of New York, Albany, NY 12222, USA
| | - April Z Gu
- School of Civil and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
4
|
Xiao Y, Shen Y, Song H, Gao F, Mao Z, Lv Q, Qin C, Yuan L, Wu D, Chu H, Wang M, Du M, Zheng R, Zhang Z. AKR1C2 genetic variants mediate tobacco carcinogens metabolism involving bladder cancer susceptibility. Arch Toxicol 2024; 98:2269-2279. [PMID: 38662237 DOI: 10.1007/s00204-024-03737-y] [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: 11/07/2023] [Accepted: 03/18/2024] [Indexed: 04/26/2024]
Abstract
Tobacco carcinogens metabolism-related genes (TCMGs) could generate reactive metabolites of tobacco carcinogens, which subsequently contributed to multiple diseases. However, the association between genetic variants in TCMGs and bladder cancer susceptibility remains unclear. In this study, we derived TCMGs from metabolic pathways of polycyclic aromatic hydrocarbons and tobacco-specific nitrosamines, and then explored genetic associations between TCMGs and bladder cancer risk in two populations: a Chinese population of 580 cases and 1101 controls, and a European population of 5930 cases and 5468 controls, along with interaction and joint analyses. Expression patterns of TCMGs were sourced from Nanjing Bladder Cancer (NJBC) study and publicly available datasets. Among 43 TCMGs, we observed that rs7087341 T > A in AKR1C2 was associated with a reduced risk of bladder cancer in the Chinese population [odds ratio (OR) = 0.84, 95% confidence interval (CI) = 0.72-0.97, P = 1.86 × 10-2]. Notably, AKR1C2 rs7087341 showed an interaction effect with cigarette smoking on bladder cancer risk (Pinteraction = 5.04 × 10-3), with smokers carrying the T allele increasing the risk up to an OR of 3.96 (Ptrend < 0.001). Genetically, rs7087341 showed an allele-specific transcriptional regulation as located at DNA-sensitive regions of AKR1C2 highlighted by histone markers. Mechanistically, rs7087341 A allele decreased AKR1C2 expression, which was highly expressed in bladder tumors that enhanced metabolism of tobacco carcinogens, and thereby increased DNA adducts and reactive oxygen species formation during bladder tumorigenesis. These findings provided new insights into the genetic mechanisms underlying bladder cancer.
Collapse
Affiliation(s)
- Yanping Xiao
- Departments of Environmental Genomics and Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health; Institute of Clinical Research, The Affiliated Taizhou People's Hospital of Nanjing Medical University; Department of Urology, The Yancheng School of Clinical Medicine of Nanjing Medical University (The Third People's Hospital of Yancheng), Nanjing Medical University, Nanjing, 211166, China
| | - Yang Shen
- Department of Urology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Second Chinese Medicine Hospital, Nanjing, 210017, China
| | - Hui Song
- Departments of Environmental Genomics and Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Fang Gao
- Departments of Environmental Genomics and Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Zhenguang Mao
- Departments of Environmental Genomics and Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Qiang Lv
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210036, China
| | - Chao Qin
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210036, China
| | - Lin Yuan
- Department of Urology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, 210029, China
| | - Dongmei Wu
- Departments of Environmental Genomics and Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Haiyan Chu
- Departments of Environmental Genomics and Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Meilin Wang
- Departments of Environmental Genomics and Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Mulong Du
- Departments of Environmental Genomics and Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Rui Zheng
- Departments of Environmental Genomics and Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Zhengdong Zhang
- Departments of Environmental Genomics and Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health; Institute of Clinical Research, The Affiliated Taizhou People's Hospital of Nanjing Medical University; Department of Urology, The Yancheng School of Clinical Medicine of Nanjing Medical University (The Third People's Hospital of Yancheng), Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
5
|
Huang D, Su Y, Li M, Xie C, Hu W, Wang S, Zheng N, Chen J, Lin Y, Cai W, Xiao J, Chen B, Hu N, Zhou F. (-)-Epicatechin gallate ameliorates cyprodinil-induced cardiac developmental defects through inhibiting aryl hydrocarbon receptor in zebrafish. Birth Defects Res 2024; 116:e2350. [PMID: 38761027 DOI: 10.1002/bdr2.2350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/31/2024] [Accepted: 04/30/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND Cyprodinil is a widely used fungicide with broad-spectrum activity, but it has been associated with cardiac abnormalities. (-)-Epicatechin gallate (ECG), a natural polyphenolic compound, has been shown to possess protective properties in cardiac development. METHODS In this study, we investigated whether ECG could mitigate cyprodinil-induced heart defects using zebrafish embryos as a model. Zebrafish embryos were exposed to cyprodinil with or without ECG. RESULTS Our results demonstrated that ECG significantly improved the survival rate, embryo movement, and hatching delay induced by cyprodinil. Furthermore, ECG effectively ameliorated cyprodinil-induced cardiac developmental toxicity, including pericardial anomaly and impairment of cardiac function. Mechanistically, ECG attenuated the cyprodinil-induced alterations in mRNA expression related to cardiac development, such as amhc, vmhc, tbx5, and gata4, as well as calcium ion channels, such as ncx1h, atp2a2a, and cdh2. Additionally, ECG was found to inhibit the activity of the aryl hydrocarbon receptor (AhR) signaling pathways induced by cyprodinil. CONCLUSIONS In conclusion, our findings provide evidence for the protective effects of ECG against cyprodinil-induced cardiac developmental toxicity, mediated through the inhibition of AhR activity. These findings contribute to a better understanding of the regulatory mechanisms and safe utilization of pesticide, such as cyprodinil.
Collapse
Affiliation(s)
- Dongqin Huang
- Neonatology, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Yuchao Su
- Neonatology, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Mingmei Li
- Scientific Research Center, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Chengwei Xie
- Scientific Research Center, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Weibin Hu
- Neonatology, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Shuxiang Wang
- Scientific Research Center, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Nanmei Zheng
- Scientific Research Center, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Jianhui Chen
- Neonatology, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Yueyun Lin
- Neonatology, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Weize Cai
- Neonatology, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Jianjia Xiao
- Neonatology, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Baojia Chen
- Scientific Research Center, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Nanping Hu
- Scientific Research Center, Anxi County Hospital, Quanzhou, People's Republic of China
| | - Fushan Zhou
- Scientific Research Center, Anxi County Hospital, Quanzhou, People's Republic of China
| |
Collapse
|
6
|
Chen Q, Wang T, Wu X, Yuan H, Wei Y, Xiao Y. The role of the cytochrome P450 superfamily in the skin. Expert Rev Mol Med 2024; 26:e15. [PMID: 38621674 PMCID: PMC11140544 DOI: 10.1017/erm.2024.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/10/2023] [Accepted: 02/22/2024] [Indexed: 04/17/2024]
Abstract
In mammals, the skin acts as a barrier to prevent harmful environmental stimuli from entering the circulation. CYP450s are involved in drug biotransformation, exogenous and endogenous substrate metabolism, and maintaining the normal physiological function of the skin, as well as facilitating homeostasis of the internal environment. The expression pattern of CYP450s in the skin is tissue-specific and thus differs from the liver and other organs. The development of skin topical medications, and knowledge of the toxicity and side effects of these medications require a detailed understanding of the expression and function of skin-specific CYP450s. Thus, we summarized the expression of CYP450s in the skin, their function in endogenous metabolic physiology, aberrant CYP450 expression in skin diseases and the influence of environmental variables and medications. This information will serve as a crucial foundation for future studies on the skin, as well as for the design and development of new drugs for skin diseases including topical medications.
Collapse
Affiliation(s)
- Qianqian Chen
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Tuan Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Affiliated Hangzhou First People's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xia Wu
- Dermatology Department, Sir Run Run Shaw Hospital, Hangzhou, China
| | - Huipu Yuan
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuan Wei
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Ying Xiao
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
7
|
Ji Z, Deng W, Chen D, Liu Z, Shen Y, Dai J, Zhou H, Zhang M, Xu H, Dai B. Recent understanding of the mechanisms of the biological activities of hesperidin and hesperetin and their therapeutic effects on diseases. Heliyon 2024; 10:e26862. [PMID: 38486739 PMCID: PMC10937595 DOI: 10.1016/j.heliyon.2024.e26862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 03/17/2024] Open
Abstract
Flavonoids are natural phytochemicals that have therapeutic effects and act in the prevention of several pathologies. These phytochemicals can be found in lemon, sweet orange, bitter orange, clementine. Hesperidin and hesperetin are citrus flavonoids from the flavanones subclass that have anti-inflammatory, antioxidant, antitumor and antibacterial potential. Preclinical studies and clinical trials demonstrated therapeutical effects of hesperidin and its aglycone hesperetin in various diseases, such as bone diseases, cardiovascular diseases, neurological diseases, respiratory diseases, digestive diseases, urinary tract diseases. This review provides a comprehensive overview of the biological activities of hesperidin and hesperetin, their therapeutic potential in various diseases and their associated molecular mechanisms. This article also discusses future considerations for the clinical applications of hesperidin and hesperetin.
Collapse
Affiliation(s)
| | | | - Dong Chen
- Binhai County People's Hospital, No.148, Middle Fudong Road, Dongkan Town, Binhai County, Yancheng City, 224500, China
| | - Zhidong Liu
- Binhai County People's Hospital, No.148, Middle Fudong Road, Dongkan Town, Binhai County, Yancheng City, 224500, China
| | - Yucheng Shen
- Binhai County People's Hospital, No.148, Middle Fudong Road, Dongkan Town, Binhai County, Yancheng City, 224500, China
| | - Jiuming Dai
- Binhai County People's Hospital, No.148, Middle Fudong Road, Dongkan Town, Binhai County, Yancheng City, 224500, China
| | - Hai Zhou
- Binhai County People's Hospital, No.148, Middle Fudong Road, Dongkan Town, Binhai County, Yancheng City, 224500, China
| | - Miao Zhang
- Binhai County People's Hospital, No.148, Middle Fudong Road, Dongkan Town, Binhai County, Yancheng City, 224500, China
| | - Hucheng Xu
- Binhai County People's Hospital, No.148, Middle Fudong Road, Dongkan Town, Binhai County, Yancheng City, 224500, China
| | - Bin Dai
- Binhai County People's Hospital, No.148, Middle Fudong Road, Dongkan Town, Binhai County, Yancheng City, 224500, China
| |
Collapse
|
8
|
Ferguson DT, Taka E, Tilghman SL, Womble T, Redmond BV, Gedeon S, Flores-Rozas H, Reed SL, Soliman KFA, Kanga KJW, Darling-Reed SF. The Anticancer Effects of the Garlic Organosulfide Diallyl Trisulfide through the Attenuation of B[a]P-Induced Oxidative Stress, AhR Expression, and DNA Damage in Human Premalignant Breast Epithelial (MCF-10AT1) Cells. Int J Mol Sci 2024; 25:923. [PMID: 38255999 PMCID: PMC10815401 DOI: 10.3390/ijms25020923] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Benzo[a]pyrene (B[a]P) is the most characterized polycyclic aromatic hydrocarbon associated with breast cancer. Our lab previously reported that the organosulfur compound (OSC), diallyl trisulfide (DATS), chemoprevention mechanism works through the induction of cell cycle arrest and a reduction in oxidative stress and DNA damage in normal breast epithelial cells. We hypothesize that DATS will inhibit B[a]P-induced cancer initiation in premalignant breast epithelial (MCF-10AT1) cells. In this study, we evaluated the ability of DATS to attenuate B[a]P-induced neoplastic transformation in MCF-10AT1 cells by measuring biological endpoints such as proliferation, clonogenicity, reactive oxygen species (ROS) formation, and 8-hydroxy-2-deoxyguanosine (8-OHdG) DNA damage levels, as well as DNA repair and antioxidant proteins. The results indicate that B[a]P induced proliferation, clonogenic formation, ROS formation, and 8-OHdG levels, as well as increasing AhR, ARNT/HIF-1β, and CYP1A1 protein expression compared with the control in MCF-10AT1 cells. B[a]P/DATS's co-treatment (CoTx) inhibited cell proliferation, clonogenic formation, ROS formation, AhR protein expression, and 8-OHdG levels compared with B[a]P alone and attenuated all the above-mentioned B[a]P-induced changes in protein expression, causing a chemopreventive effect. This study demonstrates, for the first time, that DATS prevents premalignant breast cells from undergoing B[a]P-induced neoplastic transformation, thus providing more evidence for its chemopreventive effects in breast cancer.
Collapse
Affiliation(s)
- Dominique T. Ferguson
- Pharmaceutical Sciences Division, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (D.T.F.); (E.T.); (S.L.T.); (T.W.); (S.G.); (H.F.-R.); (S.L.R.); (K.F.A.S.)
| | - Equar Taka
- Pharmaceutical Sciences Division, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (D.T.F.); (E.T.); (S.L.T.); (T.W.); (S.G.); (H.F.-R.); (S.L.R.); (K.F.A.S.)
| | - Syreeta L. Tilghman
- Pharmaceutical Sciences Division, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (D.T.F.); (E.T.); (S.L.T.); (T.W.); (S.G.); (H.F.-R.); (S.L.R.); (K.F.A.S.)
| | - Tracy Womble
- Pharmaceutical Sciences Division, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (D.T.F.); (E.T.); (S.L.T.); (T.W.); (S.G.); (H.F.-R.); (S.L.R.); (K.F.A.S.)
| | - Bryan V. Redmond
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Shasline Gedeon
- Pharmaceutical Sciences Division, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (D.T.F.); (E.T.); (S.L.T.); (T.W.); (S.G.); (H.F.-R.); (S.L.R.); (K.F.A.S.)
| | - Hernan Flores-Rozas
- Pharmaceutical Sciences Division, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (D.T.F.); (E.T.); (S.L.T.); (T.W.); (S.G.); (H.F.-R.); (S.L.R.); (K.F.A.S.)
| | - Sarah L. Reed
- Pharmaceutical Sciences Division, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (D.T.F.); (E.T.); (S.L.T.); (T.W.); (S.G.); (H.F.-R.); (S.L.R.); (K.F.A.S.)
| | - Karam F. A. Soliman
- Pharmaceutical Sciences Division, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (D.T.F.); (E.T.); (S.L.T.); (T.W.); (S.G.); (H.F.-R.); (S.L.R.); (K.F.A.S.)
| | - Konan J. W. Kanga
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA;
| | - Selina F. Darling-Reed
- Pharmaceutical Sciences Division, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (D.T.F.); (E.T.); (S.L.T.); (T.W.); (S.G.); (H.F.-R.); (S.L.R.); (K.F.A.S.)
| |
Collapse
|
9
|
Allam AM, AbuBakr HO, Yassin AM, Abdel-Razek AS, Khattab MS, Gouda EM, Mousa SZ. Potential chemopreventive effects of Broccoli extract supplementation against 7, 12 dimethyl Benz(a)anthracene (DMBA) -induced toxicity in female rats. Sci Rep 2023; 13:17234. [PMID: 37821474 PMCID: PMC10567736 DOI: 10.1038/s41598-023-43629-2] [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: 02/18/2023] [Accepted: 09/26/2023] [Indexed: 10/13/2023] Open
Abstract
Dietary components have recently received rapidly expanding attention for their potential to halt or reverse the development of many oxidative stress-mediated diseases after exposure to environmental toxicants. 7, 12 dimethylbenz(a)anthracene (DMBA) is one of the most common environmental pollutants. The present study aimed to evaluate the chemo-preventive effects of broccoli as a nutritional component against DMBA intoxication in rats. A daily dose of aqueous (1 ml/rat) and methanolic (150 mg/kg) broccoli extracts, respectively, was given to 50-day-old female rats for 26 successive weeks after carcinogen intoxication with a single dose of 20 mg/ml of DMBA. DMBA intoxication resulted in a redox imbalance (a decreased GSH level and an increased MDA level) and increased DNA fragmentation in the liver, kidney, and brain. Besides, it affected the level of expression of the bcl2 gene in the liver, kidney, and brain tissue but didn't affect cfos gene expression accompanied by histopathological changes. The aqueous and methanolic broccoli extract supplements ameliorated the adverse effects by increasing the level of GSH, decreasing the MDA level, and reducing DNA fragmentation. Besides, broccoli extracts decreased the expression of bcl2 in the liver and brain and up-regulated bcl2 expression in the kidney, accompanied by lowering NF-κβ 65 expression in the liver and brain and γ-catenin expression in the liver and kidney. In conclusion, broccoli as a dietary component had a strong chemoprotective effect against oxidative stress, DNA damage, and genotoxicity induced by DMBA intoxication in rats.
Collapse
Affiliation(s)
- Aya M Allam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Huda O AbuBakr
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.
| | - Aya M Yassin
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Ahmed S Abdel-Razek
- Department of Microbial Chemistry, Genetic Engineering and Biotechnology Research Division, National Research Centre, Dokki- Giza, Egypt
| | - Marwa S Khattab
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Eman M Gouda
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Said Z Mousa
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| |
Collapse
|
10
|
Vega JMDH, Hong HJ, Loutherback K, Stybayeva G, Revzin A. A Microfluidic Device for Long-Term Maintenance of Organotypic Liver Cultures. ADVANCED MATERIALS TECHNOLOGIES 2023; 8:2201121. [PMID: 36818276 PMCID: PMC9937715 DOI: 10.1002/admt.202201121] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Indexed: 06/03/2023]
Abstract
Liver cultures may be used for disease modeling, testing therapies and predicting drug-induced injury. The complexity of the liver cultures has evolved from hepatocyte monocultures to co-cultures with non-parenchymal cells and finally to precision-cut liver slices. The latter culture format retains liver's native biomolecular and cellular complexity and therefore holds considerable promise for in vitro testing. However, liver slices remain functional for ~72 h in vitro and display limited utility for some disease modeling and therapy testing applications that require longer culture times. This paper describes a microfluidic device for longer-term maintenance of functional organotypic liver cultures. Our microfluidic culture system was designed to enable direct injection of liver tissue into a culture chamber through a valve-enabled side port. Liver tissue was embedded in collagen and remained functional for up to 31 days, highlighted by continued production of albumin and urea. These organotypic cultures also expressed several enzymes involved in xenobiotic metabolism. Conversely, matched liver tissue embedded in collagen in a 96-well plate lost its phenotype and function within 3-5 days. The microfluidic organotypic liver cultures described here represent a significant advance in liver cultivation and may be used for future modeling of liver diseases or for individualized liver-directed therapies.
Collapse
Affiliation(s)
- José M. de Hoyos Vega
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Hye Jin Hong
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Kevin Loutherback
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
11
|
Clarke N, Irvine W. In Silico Design and SAR Study of Dibenzyl Trisulfide Analogues for Improved CYP1A1 Inhibition. Chemistry 2022; 11:e202200016. [PMID: 35610057 PMCID: PMC9130049 DOI: 10.1002/open.202200016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/17/2022] [Indexed: 11/23/2022]
Abstract
Dibenzyl trisulfide (DTS) is a natural compound with potential cancer‐preventive properties occurring in Petiveria alliacea L., an ethnomedicinal plant native to the Americas. Previous studies revealed its inhibitory activity toward cytochrome P450 (CYP)1 enzymes, key in the activation of environmental pollutants. Accordingly, the aim of this study was to design novel DTS analogues, aimed at improving not only inhibitory activity, but also specificity toward CYP1A1. This was achieved by targeting interactions with CYP1A1 residues of identified importance. Three‐dimensional structures for the novel analogues were subjected to molecular docking with several CYP isoforms, before being ranked in terms of binding affinity to CYP1A1. With three hydrogen bond donors, two hydrogen bond acceptors, a molecular mass of 361 Da, and a log P of 3.72, the most promising DTS analogue obeys Lipinski's rule of five. Following synthesis and in vitro validation of its CYP1A1‐inhibitory properties, this compound may be useful in future cancer‐preventive approaches.
Collapse
Affiliation(s)
- Nishani Clarke
- Phillips Academy, 180 Main Street, Andover, MA-01810, USA
| | - William Irvine
- Natural Products Institute, University of the West Indies Mona, Kingston 7, Jamaica
| |
Collapse
|
12
|
Chen Y, Yang J, Wang Y, Shen W, Liu J, Yuan M, Hao X, Zhong L, Guo R. Identification and Analysis of Hub Genes in Diabetic Cardiomyopathy: Potential Role of Cytochrome P450 1A1 in Mitochondrial Metabolism and STZ-Induced Myocardial Dysfunction. Front Cardiovasc Med 2022; 9:835244. [PMID: 35387435 PMCID: PMC8977650 DOI: 10.3389/fcvm.2022.835244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/25/2022] [Indexed: 11/23/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is a primary cause of death in diabetic patients; however, its molecular mechanism is not yet clear, and there is no uniform standard for diagnosis. The aim of this study is to discover the pathogenesis and potential therapeutic targets of DCM through screening and analysis of differentially expressed genes (DEGs) in heart ventricles of DCM, and to testify the role of key hub genes in DCM-induced myocardial dysfunction. Datasets GSE4745 and GSE6880 were downloaded from the GEO database. The difference analysis, visual analysis, cluster analysis and enrichment analysis were performed by using R language, python scripts and bioinformatics software followed by the construction of protein-protein interaction (PPI) network to obtain hub genes. The DCM models were established by streptozocin (STZ) injection to the male mice. The cardiac function and the expressions of hub genes were examined by using echocardiography and real-time quantitative poly-merase chain reaction (RT-qPCR), followed by multiple statistical analyses. Bioinformatic results indicate that mitochondrial dysfunction, disturbed lipid metabolism and decreased collagen synthesis are the main causes of the DCM development. In particular, the hub gene Cyp1a1 that encodes Cytochrome P450 1A1 (CYP4501A1) enzyme has the highest connectivity in the interaction network, and is associated with mitochondrial homeostasis and energy metabolism. It plays a critical role in the oxidation of endogenous or exogenous substrates. Our RT-qPCR results confirmed that ventricular Cyp1a1 mRNA level was nearly 12-fold upregulated in DCM model compared to normal control, which was correlated with abnormal cardiac function in diabetic individuals. CYP4501A1 protein expression in mitochondria was also increased in diabetic hearts. However, we found no significant changes in collagen expressions in cardiac ventricles of mice with DCM. This study provided compact data support for understanding the pathogenesis of DCM. CYP4501A1 might be considered as a potential candidate targeting for DCM therapy. Follow-up animal and clinical verifications need to be further explored.
Collapse
Affiliation(s)
- Yinliang Chen
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Jinbao Yang
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Ying Wang
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Weike Shen
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Jinlin Liu
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Meng Yuan
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Xiaoyu Hao
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Li Zhong
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, China
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Rui Guo
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, China
- The Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, China
- *Correspondence: Rui Guo
| |
Collapse
|
13
|
Ghelli F, Cocchi E, Bellisario V, Buglisi M, Squillacioti G, Santovito A, Bono R. The formation of SCEs as an effect of occupational exposure to formaldehyde. Arch Toxicol 2022; 96:1101-1108. [PMID: 35149893 PMCID: PMC8921006 DOI: 10.1007/s00204-022-03238-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/27/2022] [Indexed: 11/25/2022]
Abstract
Formaldehyde (FA) is a ubiquitous toxic chemical employed worldwide due to its disinfectant and preservative properties. Despite being classified as a human carcinogen, FA is still employed as formalin in pathology wards as standard fixative. We evaluated its relationship with the formation of sister-chromatid exchanges (SCEs) in cultured peripheral blood lymphocytes on 57 pathologists and 48 controls and the risk/protective role played by several genetic polymorphisms. All subjects were assessed for SCEs and genotyped for the most common cancer-associated gene polymorphisms: CYP1A1 exon 7 (A > G), CYP1A1*2A (T > C), CYP2C19*2 (G > A), GSTT1 (presence/absence), GSTM1 (presence/absence), GSTP1 (A > G), XRCC1 (G399A), XRCC1 (C194T), XRCC1 (A280G), XPC exon 15 (A939C), XPC exon 9 (C499T), TNFα − 308 G > A), IL10 − 1082 (G > A), and IL6 − 174 (G > C). Air-FA concentration was assessed through passive personal samplers. Pathologists, exposed to 55.2 μg/m3 of air-FA, showed a significantly higher SCEs frequency than controls, exposed, respectively, to 18.4 μg/m3. Air-FA was directly correlated with SCEs frequency and inversely with the replication index (RI). Regression models showed FA exposure as a significant predictor in developing SCEs, while did not highlight any role of the selected polymorphisms. Our study confirms the role of low air-FA levels as genotoxicity inductor, highlighting the importance to define exposure limits that could be safer for exposed workers.
Collapse
Affiliation(s)
- Federica Ghelli
- Department of Public Health and Pediatrics, University of Turin, Via Santena 5 bis, 10126, Turin, Italy
| | - Enrico Cocchi
- Department of Public Health and Pediatrics, University of Turin, Via Santena 5 bis, 10126, Turin, Italy
| | - Valeria Bellisario
- Department of Public Health and Pediatrics, University of Turin, Via Santena 5 bis, 10126, Turin, Italy
| | - Martina Buglisi
- Department of Public Health and Pediatrics, University of Turin, Via Santena 5 bis, 10126, Turin, Italy
| | - Giulia Squillacioti
- Department of Public Health and Pediatrics, University of Turin, Via Santena 5 bis, 10126, Turin, Italy
| | - Alfredo Santovito
- Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123, Turin, Italy
| | - Roberto Bono
- Department of Public Health and Pediatrics, University of Turin, Via Santena 5 bis, 10126, Turin, Italy.
| |
Collapse
|
14
|
Wei KL, Gao GL, Chou YT, Lin CY, Chen SC, Chen YL, Choi HQ, Cheng CC, Su JGJ. Sorafenib is an antagonist of the aryl hydrocarbon receptor. Toxicology 2022; 470:153118. [PMID: 35124147 DOI: 10.1016/j.tox.2022.153118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 01/29/2022] [Accepted: 02/02/2022] [Indexed: 02/07/2023]
|
15
|
Guo Q, Li S, Wang X, Han HS, Yin XJ, Li JC. Paeoniflorin improves the in vitro maturation of benzo(a)pyrene treated porcine oocytes via effects on the sonic hedgehog pathway. Theriogenology 2021; 180:72-81. [PMID: 34953351 DOI: 10.1016/j.theriogenology.2021.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/20/2021] [Accepted: 12/15/2021] [Indexed: 10/19/2022]
Abstract
Benzo(a)pyrene (BaP) is a toxic substance that people are often exposed to. It has serious harmful effects on the body, and has a destructive effect on oocytes and cumulus cells. Here, we found that paeoniflorin (Pae), a traditional Chinese medicine monomer with antioxidant effects, decreased BaP-induced meiotic failure by increasing the activity of the Sonic hedgehog (SHH) signaling pathway and reducing the level of reactive oxygen species (ROS). We found that the in vitro maturation (IVM) rate was significantly increased (P < 0.05) in the 0.1 μM Pae and BaP (co-treatment) group compared with BaP group due to reduced ROS levels and increased mitochondrial membrane potential (ΔΨ) and ATP content. The mRNA expression levels of oocyte maturation and cumulus cell expansion-related genes were also significantly higher in the co-treatment group. To demonstrate the quality of oocytes, the development capacity of parthenogenetically activated (PA) and in vitro fertilization (IVF) embryos from different treatment groups oocytes were determined.The blastocyst formation rate was significantly higher in PA and IVF embryos derived from oocytes in the co-treatment group than in those derived from oocytes in the BaP group. To further confirm that the SHH signaling pathway was involved in causing these effects of Pae, we treated oocytes with Pae and BaP in the presence or absence of cyclopamine (Cy), an inhibitor of this pathway. Cy abolished the effects of Pae in BaP treated porcine oocytes. In conclusion, Pae improves the IVM capacity of BaP-treated porcine oocytes by activating the SHH signaling pathway, inhibiting ROS production, and increasing ΔΨ.
Collapse
Affiliation(s)
- Qing Guo
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China; Heilongjiang Key Laboratory of Efficient Utilization of Feed Resources and Nutrition Manipulation in Cold Region, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Suo Li
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Xue Wang
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China; Heilongjiang Key Laboratory of Efficient Utilization of Feed Resources and Nutrition Manipulation in Cold Region, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Huan-Sheng Han
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China; Heilongjiang Key Laboratory of Efficient Utilization of Feed Resources and Nutrition Manipulation in Cold Region, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Xi-Jun Yin
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Yanbian University, Yanji, Jilin, 133002, China.
| | - Jing-Chun Li
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China; Heilongjiang Key Laboratory of Efficient Utilization of Feed Resources and Nutrition Manipulation in Cold Region, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China.
| |
Collapse
|
16
|
Coelho NR, Pimpão AB, Correia MJ, Rodrigues TC, Monteiro EC, Morello J, Pereira SA. Pharmacological blockage of the AHR-CYP1A1 axis: a call for in vivo evidence. J Mol Med (Berl) 2021; 100:215-243. [PMID: 34800164 PMCID: PMC8605459 DOI: 10.1007/s00109-021-02163-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 01/21/2023]
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that can be activated by structurally diverse compounds arising from the environment and the microbiota and host metabolism. Expanding evidence has been shown that the modulation of the canonical pathway of AHR occurs during several chronic diseases and that its abrogation might be of clinical interest for metabolic and inflammatory pathological processes. However, most of the evidence on the pharmacological abrogation of the AHR-CYP1A1 axis has been reported in vitro, and therefore, guidance for in vivo studies is needed. In this review, we cover the state-of-the-art of the pharmacodynamic and pharmacokinetic properties of AHR antagonists and CYP1A1 inhibitors in different in vivo rodent (mouse or rat) models of disease. This review will serve as a road map for those researchers embracing this emerging therapeutic area targeting the AHR. Moreover, it is a timely opportunity as the first AHR antagonists have recently entered the clinical stage of drug development.
Collapse
Affiliation(s)
- N R Coelho
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | - A B Pimpão
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | - M J Correia
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | - T C Rodrigues
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | - E C Monteiro
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | - J Morello
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | - S A Pereira
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal.
| |
Collapse
|
17
|
Tooker BC, Quinn K, Armstrong M, Bauer AK, Reisdorph N. Comparing the effects of an exposure to a polycyclic aromatic hydrocarbon mixture versus individual polycyclic aromatic hydrocarbons during monocyte to macrophage differentiation: Mixture exposure results in altered immune metrics. J Appl Toxicol 2021; 41:1568-1583. [PMID: 33559210 PMCID: PMC8349383 DOI: 10.1002/jat.4147] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/13/2021] [Accepted: 01/24/2021] [Indexed: 11/08/2022]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are generated by the incomplete combustion of carbon. Exposures correlate with systemic immune dysfunction and overall immune suppression. Real-world exposures to PAHs are almost always encountered as mixtures; however, research overwhelmingly centers on isolated exposures to a single PAH, benzo[a]pyrene (B[a]P). Here, a human monocyte line (U937) was exposed to B[a]P, benz[a]anthracene (B[a]A), or a mixture of six PAHs (6-MIX) to assess the differential toxicity on monocytes. Further, monocytes were exposed to PAHs with and without CYP1A1 inhibitors during macrophage differentiation to delineate PAH exposure and PAH metabolism-driven alterations to the immune response. U937 monocytes exposed to B[a]P, B[a]A, or 6-MIX had higher levels of cellular health and growth not observed following equimolar exposures to other individual PAHs. PAH exposures during differentiation did not alter monocyte-derived macrophage (MDM) numbers; however, B[a]A and 6-MIX exposures significantly altered M1/M2 polarization in a CYP1A1-dependent manner. U937-MDM adherence was differentially suppressed by all three PAH treatments with 6-MIX exposed U937-MDM having significantly more adhesion than U937-MDM exposed to either individual PAH. Finally, 6-MIX exposures during differentiation reduced U937-MDM endocytic function significantly less than B[a]A exposed cells. Exposure to a unique PAH mixture during U937-MDM differentiation resulted in mixture-specific alterations of pro-inflammatory markers compared to individual PAH exposures. While subtle, these differences highlight the probability that using a model PAH, B[a]P, may not accurately reflect the effects of PAH mixture exposures. Therefore, future studies should include various PAH mixtures that encompass probable real-world PAH exposures for the endpoints under investigation.
Collapse
Affiliation(s)
- Brian C. Tooker
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kevin Quinn
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Michael Armstrong
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Alison K. Bauer
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Nichole Reisdorph
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
18
|
Aichinger G, Del Favero G, Warth B, Marko D. Alternaria toxins-Still emerging? Compr Rev Food Sci Food Saf 2021; 20:4390-4406. [PMID: 34323368 DOI: 10.1111/1541-4337.12803] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 06/18/2021] [Accepted: 06/19/2021] [Indexed: 12/13/2022]
Abstract
Alternaria molds are known to cause the contamination of food with their secondary metabolites, a chemically very heterogeneous group of compounds. Yet, after decades of research on the occurrence and the toxicity of Alternaria toxins in academia, no regulation has been implemented yet, thus leaving these potential food contaminants in the status of so-called "emerging mycotoxins". However, research on this topic has been far from static, leading to the European Food Safety Authority repeatedly calling for more data on the occurrence and toxicity of genotoxic metabolites such as alternariol (AOH) and its monomethyl ether (AME). To give an overview on recent developments in the field, this comprehensive review summarizes published data and addresses current challenges arising from the chemical complexity of Alternaria's metabolome, mixture effects and the emergence of novel biological targets like cell membranes or the interaction with different receptors. Besides toxicodynamics, we review recent research on toxicokinetics, including the first in vivo studies which incorporated the rarely investigated-but highly genotoxic-perylene quinones. Furthermore, a particular focus lies on the advances of liquid chromatography/tandem mass spectrometry (LC-MS/MS)-based analytical tools for determining a broader spectrum of Alternaria toxins including modified/masked forms and assessing exposure via human biomonitoring (HBM).
Collapse
Affiliation(s)
- Georg Aichinger
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Wien, Austria
| | - Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Wien, Austria
| | - Benedikt Warth
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Wien, Austria
| | - Doris Marko
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Wien, Austria
| |
Collapse
|
19
|
El-Ghiaty MA, El-Kadi AO. Arsenic: Various species with different effects on cytochrome P450 regulation in humans. EXCLI JOURNAL 2021; 20:1184-1242. [PMID: 34512225 PMCID: PMC8419240 DOI: 10.17179/excli2021-3890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/02/2021] [Indexed: 11/22/2022]
Abstract
Arsenic is well-recognized as one of the most hazardous elements which is characterized by its omnipresence throughout the environment in various chemical forms. From the simple inorganic arsenite (iAsIII) and arsenate (iAsV) molecules, a multitude of more complex organic species are biologically produced through a process of metabolic transformation with biomethylation being the core of this process. Because of their differential toxicity, speciation of arsenic-based compounds is necessary for assessing health risks posed by exposure to individual species or co-exposure to several species. In this regard, exposure assessment is another pivotal factor that includes identification of the potential sources as well as routes of exposure. Identification of arsenic impact on different physiological organ systems, through understanding its behavior in the human body that leads to homeostatic derangements, is the key for developing strategies to mitigate its toxicity. Metabolic machinery is one of the sophisticated body systems targeted by arsenic. The prominent role of cytochrome P450 enzymes (CYPs) in the metabolism of both endobiotics and xenobiotics necessitates paying a great deal of attention to the possible effects of arsenic compounds on this superfamily of enzymes. Here we highlight the toxicologically relevant arsenic species with a detailed description of the different environmental sources as well as the possible routes of human exposure to these species. We also summarize the reported findings of experimental investigations evaluating the influence of various arsenicals on different members of CYP superfamily using human-based models.
Collapse
Affiliation(s)
- Mahmoud A. El-Ghiaty
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ayman O.S. El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
20
|
Novel Synthetic Analogues of 19(S/R)-Hydroxyeicosatetraenoic Acid Exhibit Noncompetitive Inhibitory Effect on the Activity of Cytochrome P450 1A1 and 1B1. Eur J Drug Metab Pharmacokinet 2021; 46:613-624. [PMID: 34235626 DOI: 10.1007/s13318-021-00699-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND OBJECTIVES Cytochrome P450 (CYP) 1A1 and CYP1B1 enzymes play a significant role in the pathogenesis of cancer and cardiovascular diseases (CVD) such as cardiac hypertrophy and heart failure. Previously, we have demonstrated that R- and S-enantiomers of 19-hydroxyeicosatetraenoic acid (19-HETE), an arachidonic acid endogenous metabolite, enantioselectively inhibit CYP1B1. The current study was conducted to test the possible inhibitory effect of novel synthetic analogues of R- and S-enantiomers of 19-HETE on the activity of CYP1A1, CYP1A2, and CYP1B1. METHODS The O-dealkylation rate of 7-ethoxyresorufin (EROD) by recombinant human CYP1A1 and CYP1B1, in addition to the O-dealkylation rate of 7-methoxyresorufin (MROD) by recombinant human CYP1A2, were measured in the absence and presence of varying concentrations (0-40 nM) of the synthetic analogues of 19(R)- and 19(S)-HETE. Also, the possible inhibitory effect of both analogues on the catalytic activity of EROD and MROD, using RL-14 cells and human liver microsomes, was assessed. RESULTS The results showed that both synthetic analogues of 19(R)- and 19(S)-HETE exhibited direct inhibitory effects on the activity of CYP1A1 and CYP1B1, while they had no significant effect on CYP1A2 activity. Nonlinear regression analysis and comparisons showed that the mode of inhibition for both analogues is noncompetitive inhibition of CYP1A1 and CYP1B1 enzymes. Also, nonlinear regression analysis and Dixon plots showed that the R- and S-analogues have KI values of 15.7 ± 4.4 and 6.1 ± 1.5 nM for CYP1A1 and 26.1 ± 2.9 and 9.1 ± 1.8 nM for CYP1B1, respectively. Moreover, both analogues were able to inhibit EROD and MROD activities in a cell-based assay and human liver microsomes. CONCLUSIONS Therefore, the synthetic analogues of 19-HETE could be considered as a novel therapeutic approach in the treatment of cancer and CVD.
Collapse
|
21
|
Chen CS, Gao GL, Ho DR, Lin CY, Chou YT, Chen SC, Huang MC, Kao WY, Su JGJ. Cyproterone acetate acts as a disruptor of the aryl hydrocarbon receptor. Sci Rep 2021; 11:5457. [PMID: 33750846 PMCID: PMC7943802 DOI: 10.1038/s41598-021-84769-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 02/08/2021] [Indexed: 11/10/2022] Open
Abstract
Prostate cancer is a major cause of death in males. Cyproterone acetate (CPA), the steroidal anti-androgen for part of androgen deprivation therapy, may block the androgen-receptor interaction and then reduce serum testosterone through its weak anti-gonadotropic action. In addition to CPA inducing hepatitis, CPA is known to cause liver tumors in rats also. Aryl hydrocarbon receptor (AhR) is a cytoplasmic receptor and regulates multiple physiological functions. CYP1A1 is an AhR-targeted gene. We found that CPA induced CYP1A1 expression, transcriptional activity of the aryl hydrocarbon response element (AHRE), and the nuclear localization of AhR in mouse Hepa-1c1c7 cells. However, CPA suppressed CYP1A1 mRNA expression and the transcriptional activity of AHRE in human HepG2 and MCF7 cells, and also decreased AhR ligand-induced CYP1A1 protein expression and transcriptional activity of AHRE in HepG2 cells. In summary, CPA is an AhR agonist in mouse cells, but an AhR antagonist in human cells. Accordingly, CPA potentially plays a role as an endocrine disruptor of the AhR. This study helps us to understand why CPA induces acute hepatitis, gene mutation, and many other side effects. In addition, it may trigger further studies investigating the relationships between CPA, glucocorticoid receptor and castration-resistant prostate cancer in the future.
Collapse
Affiliation(s)
- Chih-Shou Chen
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan, ROC
| | - Guan-Lun Gao
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, 60004, Taiwan, ROC.,Department of Biological Resources, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Dong-Ru Ho
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan, ROC
| | - Chih-Yi Lin
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Yu-Ting Chou
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Shan-Chun Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Min-Cong Huang
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan, ROC
| | - Wen-Ya Kao
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Jyan-Gwo Joseph Su
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, 60004, Taiwan, ROC.
| |
Collapse
|
22
|
Ferreira de Oliveira JMP, Santos C, Fernandes E. Therapeutic potential of hesperidin and its aglycone hesperetin: Cell cycle regulation and apoptosis induction in cancer models. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 73:152887. [PMID: 30975541 DOI: 10.1016/j.phymed.2019.152887] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/20/2019] [Accepted: 03/09/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND The ability of cancer cells to divide without restriction and to escape programmed cell death is a feature of the proliferative state. Citrus flavanones are flavonoids with potential multiple anticancer actions, from antioxidant and chemopreventive, to anti-inflammatory, anti-angiogenic, cytostatic and cytotoxic in different cancer models. PURPOSE This review aims to summarize the current knowledge on the antiproliferative actions of the citrus flavanones hesperidin (HSD) and hesperetin (HST), with emphasis on cell cycle arrest and apoptosis. METHODS Cochrane Library, Scopus, Pubmed and Web of Science collection databases were queried for publications reporting antiproliferative effects of HSD and HST in cancer models. RESULTS HSD and HST have been proven to delay cell proliferation in several cancer models. Depending on the compound, dose and cell line studied, different effects have been reported. Cell cycle arrest associated with cytostatic effects has been reported in cells with increased levels of p53 and also cyclin-dependent kinase inhibitors, as well as decreased levels of specific cyclins and cyclin-dependent kinases. Moreover, apoptotic effects have been found to be associated with altered ratios of pro-/antiapoptotic proteins, caspase activation, c-Jun N-terminal kinase (JNK) pathway activation and caspase-independent pathways. CONCLUSION Available scientific literature data indicate complex effects, dependent on cell lines and exposure conditions, suggesting that HSD and HST doses need to be optimized according to the cellular and organismal context. The establishment of the main antiproliferative mechanisms is of utmost importance for a possible therapeutic benefit of citrus flavanones in the context of cancer.
Collapse
Affiliation(s)
- José Miguel P Ferreira de Oliveira
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, 4050-313 Porto, Portugal.
| | - Conceição Santos
- Integrated Biology and Biotechnology Laboratory, Department of Biology, Faculty of Sciences, University of Porto, Rua Campo Alegre, 4169-007 Porto, Portugal; LAQV, REQUIMTE, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal.
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
23
|
Kaur S, Dogra S, Sadwal S, Aniqa A. Chemopreventive activity of hydroethanolic Murraya koenigii leaves extract (HEMKLE) against chemically induced skin carcinogenesis in mice. INT J VITAM NUTR RES 2020; 91:396-410. [PMID: 32580686 DOI: 10.1024/0300-9831/a000660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The present study aimed to examine the chemoprotective effect of Hydroethanolic Murraya koenigii leaves extract (HEMKLE) on murine skin carcinogenesis model. For the study, male LACA mice divided into four groups (n = 15 per group). Group I (Control), Group II (DMBA/TPA), Group III (HEMKLE), and Group IV (HEMKLE + DMBA/TPA). Skin tumors were induced in Group II (DMBA/TPA) and Group IV (HEMKLE + DMBA/TPA) by topical application of 7, 12 dimethylbenz[a]anthracene (DMBA) [500 nmol/100 μL of acetone, twice a week for two weeks] and 12-O-tetradecanoyl phorbol-13-acetate (TPA) [1.7 nmol/100 μL of acetone, twice a week for eighteen weeks] and HEMKLE (200 mg/kg b. w.) was administered orally (instilled by oral gavage). The chemoprotective response of HEMKLE was evident by inhibition in tumor incidence, mean tumor volume, mean tumor burden, total number of tumors, and tumor size in Group IV (HEMKLE + DMBA/TPA) when compared to Group II (DMBA/TPA). HEMKLE administration also decreased the reactive oxygen species (ROS) and lipid peroxidation (LPO) levels and increased the antioxidants enzyme activities in Group IV (HEMKLE + DMBA/TPA) when compared to Group II (DMBA/TPA) that suggests its antioxidant potential. HEMKLE administration also increased the mRNA and protein expression of caspase-9 and caspase-3 and decreased the mRNA and protein expression of Bcl-2 in Group IV (HEMKLE + DMBA/TPA) when compared to Group II (DMBA/TPA) that suggest its apoptosis-inducing effect on DMBA/TPA induced skin carcinogenesis.
Collapse
Affiliation(s)
- Sarvnarinder Kaur
- Department of Biophysics, Basic Medical Sciences, Panjab University, Chandigarh, India
| | - Shilpa Dogra
- Department of Biophysics, Basic Medical Sciences, Panjab University, Chandigarh, India
| | - Shilpa Sadwal
- Department of Biophysics, Basic Medical Sciences, Panjab University, Chandigarh, India
| | - Aniqa Aniqa
- Department of Biophysics, Basic Medical Sciences, Panjab University, Chandigarh, India
| |
Collapse
|
24
|
Kitakaze T, Makiyama A, Nakai R, Kimura Y, Ashida H. Kaempferol modulates TCDD- and t-BHQ-induced drug-metabolizing enzymes and luteolin enhances this effect. Food Funct 2020; 11:3668-3680. [PMID: 32301455 DOI: 10.1039/c9fo02951f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The expression of drug-metabolizing enzymes is deeply involved in chemical-induced cancer progression and prevention. The aryl hydrocarbon receptor (AhR) induces phase I, and certain phase II drug-metabolizing enzymes after the binding of ligands, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). We have previously demonstrated that luteolin inhibited TCDD-induced AhR transformation, and modulated the expression of drug-metabolizing enzymes through not only the AhR, but also the nuclear factor-erythroid-2-related factor 2 (Nrf2). We have examined the effect of kaempferol on the expression of drug-metabolizing enzymes through modulation of the AhR- and Nrf2-pathways, and the effect of co-treatment with kaempferol and luteolin. Kaempferol dose-dependently inhibited not only the TCDD-induced expression of phase I and phase II drug-metabolizing enzymes, but also the tertiary butylhydroquinone (t-BHQ)-induced expression of phase II drug-metabolizing enzymes, by modulating the AhR- and Nrf2-pathways. Co-treatment with kaempferol and luteolin enhanced the inhibitory effect on the expression of drug-metabolizing enzymes, compared with either kaempferol or luteolin alone. Moreover, co-treatment with kaempferol and luteolin increased the cellular levels of kaempferol without affecting the levels of luteolin. An in vivo study was also performed and the results demonstrated that co-treatment with kaempferol and luteolin enhanced the inhibition of benzo[a]pyrene-induced drug-metabolizing enzymes compared with either kaempferol or luteolin alone, in the liver of ICR mice. These results suggest that luteolin promoted the incorporation of kaempferol into hepatocytes and enhanced the inhibitory effect of kaempferol on chemical-induced drug-metabolizing enzymes. Thus, luteolin enhances the kaempferol-inhibited expression of drug-metabolizing enzymes.
Collapse
Affiliation(s)
- Tomoya Kitakaze
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, Japan.
| | - Atsushi Makiyama
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, Japan.
| | - Rika Nakai
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, Japan.
| | - Yuki Kimura
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, Japan.
| | - Hitoshi Ashida
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, Japan.
| |
Collapse
|
25
|
Cubillos-Angulo JM, Fukutani ER, Cruz LAB, Arriaga MB, Lima JV, Andrade BB, Queiroz ATL, Fukutani KF. Systems biology analysis of publicly available transcriptomic data reveals a critical link between AKR1B10 gene expression, smoking and occurrence of lung cancer. PLoS One 2020; 15:e0222552. [PMID: 32097409 PMCID: PMC7041805 DOI: 10.1371/journal.pone.0222552] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 02/11/2020] [Indexed: 12/19/2022] Open
Abstract
Background Cigarette smoking is associated with an increased risk of developing respiratory diseases and various types of cancer. Early identification of such unfavorable outcomes in patients who smoke is critical for optimizing personalized medical care. Methods Here, we perform a comprehensive analysis using Systems Biology tools of publicly available data from a total of 6 transcriptomic studies, which examined different specimens of lung tissue and/or cells of smokers and nonsmokers to identify potential markers associated with lung cancer. Results Expression level of 22 genes was capable of classifying smokers from non-smokers. A machine learning algorithm revealed that AKR1B10 was the most informative gene among the 22 differentially expressed genes (DEGs) accounting for the classification of the clinical groups. AKR1B10 expression was higher in smokers compared to non-smokers in datasets examining small and large airway epithelia, but not in the data from a study of sorted alveolar macrophages. Moreover, AKR1B10 expression was relatively higher in lung cancer specimens compared to matched healthy tissue obtained from nonsmoking individuals. Although the overall accuracy of AKR1B10 expression level in distinction between cancer and healthy lung tissue was 76%, with a specificity of 98%, our results indicated that such marker exhibited low sensitivity, hampering its use for cancer screening such specific setting. Conclusion The systematic analysis of transcriptomic studies performed here revealed a potential critical link between AKR1B10 expression, smoking and occurrence of lung cancer.
Collapse
Affiliation(s)
- Juan M. Cubillos-Angulo
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Bahia, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, Brazil
| | | | - Luís A. B. Cruz
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, Brazil
- Curso de Medicina, Faculdade de Tecnologia e Ciências, Salvador, Bahia, Brazil
| | - María B. Arriaga
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Bahia, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, Brazil
| | - João Victor Lima
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
| | - Bruno B. Andrade
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Bahia, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, Brazil
- Curso de Medicina, Faculdade de Tecnologia e Ciências, Salvador, Bahia, Brazil
- Universidade Salvador (UNIFACS), Laureate Universities, Salvador, Bahia, Brazil
- Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador, Bahia, Brazil
- * E-mail: (BBA); (ATLQ); (KFF)
| | - Artur T. L. Queiroz
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
- * E-mail: (BBA); (ATLQ); (KFF)
| | - Kiyoshi F. Fukutani
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, Brazil
- Curso de Medicina, Faculdade de Tecnologia e Ciências, Salvador, Bahia, Brazil
- * E-mail: (BBA); (ATLQ); (KFF)
| |
Collapse
|
26
|
Rafieian-kopaei M, Hamedi A, Soleiman Dehkordi E, Pasdaran A, Pasdaran A. Phytochemical Investigation on Volatile Compositions and Methoxylated Flavonoids of Agrostis gigantea Roth. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2020; 19:360-370. [PMID: 33224243 PMCID: PMC7667570 DOI: 10.22037/ijpr.2019.15209.12935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In this study, methoxylated flavonoids and volatile constitutions of Agrostis gigantea Roth (Poaceae) were investigated for the first time. The flavonoids were identified by spectroscopic methods (1H-NMR, 13C-NMR, COSY, NOSEY, TCOSY, and HMBC). The volatile constitutions of aerial parts and seeds were analyzed by gas chromatography-mass spectrometry (GC-MS). Two methoxylated flavonoids, luteolin 5-methyl ether (1), and cirsilineol (2) were isolated from the aerial parts of this plant. According to the GC-MS data the main constitutions of these volatile oils belong to the simple phenolic category which include coniferyl alcohol (18.80%) and eugenol (12.19%) in aerial parts and seeds, respectively. By using the computer- aided molecular modeling approaches, the binding affinity of these compounds was predicted in the catalytic domains of aryl hydrocarbon receptor (AhR). These two isolated flavonoids were investigated in-vitro for their inhibitory activity on 4T1 breast carcinoma cells. It was predicted that these compounds could be well-matched in aryl hydrocarbon receptor (3H82) active site, but based on the in-vitro assay, the IC50 values on cytotoxicity were 428.24 ±3.21 and 412.7±3.02 μg/mL for luteolin 5-methyl ether and cirsilineol, respectively. Thus, it can be concluded that these flavonoids exhibit low cytotoxicity against 4T1 breast carcinoma cell line.
Collapse
Affiliation(s)
- Mahmoud Rafieian-kopaei
- Medicinal Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Azadeh Hamedi
- Department of Pharmacognosy, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ebrahim Soleiman Dehkordi
- Medicinal Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Arsalan Pasdaran
- Senior Researcher, Kara Daru & Revive Chemistry Co. Shiraz, Iran.
| | - Aradalan Pasdaran
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
27
|
Le Goff M, Delbrut A, Quinton M, Pradelles R, Bescher M, Burel A, Schoefs B, Sergent O, Lagadic-Gossmann D, Le Ferrec E, Ulmann L. Protective Action of Ostreococcus tauri and Phaeodactylum tricornutum Extracts towards Benzo[a]Pyrene-Induced Cytotoxicity in Endothelial Cells. Mar Drugs 2019; 18:E3. [PMID: 31861403 PMCID: PMC7024323 DOI: 10.3390/md18010003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/13/2019] [Accepted: 12/14/2019] [Indexed: 12/31/2022] Open
Abstract
Marine microalgae are known to be a source of bioactive molecules of interest to human health, such as n-3 polyunsaturated fatty acids (n-3 PUFAs) and carotenoids. The fact that some of these natural compounds are known to exhibit anti-inflammatory, antioxidant, anti-proliferative, and apoptosis-inducing effects, demonstrates their potential use in preventing cancers and cardiovascular diseases (CVDs). Benzo[a]pyrene (B[a]P), a polycyclic aromatic hydrocarbon (PAH), is an ubiquitous environmental pollutant known to contribute to the development or aggravation of human diseases, such as cancer, CVDs, and immune dysfunction. Most of these deleterious effects are related to the activation of the polycyclic aromatic hydrocarbon receptor (AhR). In this context, two ethanolic microalgal extracts with concentrations of 0.1 to 5 µg/mL are tested, Ostreoccoccus tauri (OT) and Phaeodactylum tricornutum (PT), in order to evaluate and compare their potential effects towards B[a]P-induced toxicity in endothelial HMEC-1 cells. Our results indicate that the OT extract can influence the toxicity of B[a]P. Indeed, apoptosis and the production of extracellular vesicles were decreased, likely through the reduction of the expression of CYP1A1, a B[a]P bioactivation enzyme. Furthermore, the B[a]P-induced expression of the inflammatory cytokines IL-8 and IL1-β was reduced. The PT extract only inhibited the expression of the B[a]P-induced cytokine IL-8 expression. The OT extract therefore seems to be a good candidate for counteracting the B[a]P toxicity.
Collapse
Affiliation(s)
- Manon Le Goff
- EA 2160 Mer Molécules Santé—MIMMA, IUML FR-3473 CNRS, Le Mans Université, F-53020 Laval, France; (M.L.G.)
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, F-35000 Rennes, France; (M.B.); (O.S.); (D.L.-G.)
| | - Antoine Delbrut
- Microphyt, 713 Route de Mudaison, 34630 Baillargues, France; (A.D.); (M.Q.); (R.P.)
| | - Marie Quinton
- Microphyt, 713 Route de Mudaison, 34630 Baillargues, France; (A.D.); (M.Q.); (R.P.)
| | - Rémi Pradelles
- Microphyt, 713 Route de Mudaison, 34630 Baillargues, France; (A.D.); (M.Q.); (R.P.)
| | - Maelle Bescher
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, F-35000 Rennes, France; (M.B.); (O.S.); (D.L.-G.)
| | - Agnès Burel
- Univ Rennes, Biosit–UMS 3480, US_S 018, F-35000 Rennes, France; (A.B.)
| | - Benoît Schoefs
- EA 2160 Mer Molécules Santé—MIMMA, IUML FR-3473 CNRS, Le Mans Université, F-72000 Le Mans, France; (B.S.)
| | - Odile Sergent
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, F-35000 Rennes, France; (M.B.); (O.S.); (D.L.-G.)
| | - Dominique Lagadic-Gossmann
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, F-35000 Rennes, France; (M.B.); (O.S.); (D.L.-G.)
| | - Eric Le Ferrec
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, F-35000 Rennes, France; (M.B.); (O.S.); (D.L.-G.)
| | - Lionel Ulmann
- EA 2160 Mer Molécules Santé—MIMMA, IUML FR-3473 CNRS, Le Mans Université, F-53020 Laval, France; (M.L.G.)
| |
Collapse
|
28
|
Shadboorestan A, Tarfiei GA, Montazeri H, Sepand MR, Zangooei M, Khedri A, Ostad SN, Ghahremani MH. Invasion and migration of MDA-MB-231 cells are inhibited by block of AhR and NFAT: role of AhR/NFAT1/β4 integrin signaling. J Appl Toxicol 2018; 39:375-384. [DOI: 10.1002/jat.3728] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 08/04/2018] [Accepted: 08/15/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Amir Shadboorestan
- Department of Toxicology and Pharmacology, Faculty of Pharmacy; Tehran University of Medical Sciences; Tehran Iran
| | - Ghorban Ali Tarfiei
- Department of Molecular Medicine, School of Advanced Technologies in Medicine; Tehran University of Medical Sciences; Tehran Iran
| | - Hamed Montazeri
- School of Pharmacy-International Campus; Iran University of Medical Sciences; Tehran Iran
| | - Mohammad Reza Sepand
- Department of Toxicology and Pharmacology, Faculty of Pharmacy; Tehran University of Medical Sciences; Tehran Iran
| | - Mohammad Zangooei
- Department of Biochemistry, Faculty of Medicine; Tehran University of Medical Sciences; Tehran Iran
| | - Azam Khedri
- Department of Biochemistry, Faculty of Medicine; Tehran University of Medical Sciences; Tehran Iran
| | - Seyed Nasser Ostad
- Department of Toxicology and Pharmacology, Faculty of Pharmacy; Tehran University of Medical Sciences; Tehran Iran
- Toxicology and Poisoning Research Center, Faculty of Pharmacy; Tehran University of Medical Sciences; Tehran Iran
| | - Mohammad Hossein Ghahremani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy; Tehran University of Medical Sciences; Tehran Iran
- Toxicology and Poisoning Research Center, Faculty of Pharmacy; Tehran University of Medical Sciences; Tehran Iran
| |
Collapse
|
29
|
Blachier F, Beaumont M, Portune KJ, Steuer N, Lan A, Audebert M, Khodorova N, Andriamihaja M, Airinei G, Benamouzig R, Davila AM, Armand L, Rampelli S, Brigidi P, Tomé D, Claus SP, Sanz Y. High-protein diets for weight management: Interactions with the intestinal microbiota and consequences for gut health. A position paper by the my new gut study group. Clin Nutr 2018; 38:1012-1022. [PMID: 30274898 DOI: 10.1016/j.clnu.2018.09.016] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/10/2018] [Accepted: 09/13/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS This review examines to what extent high-protein diets (HPD), which may favor body weight loss and improve metabolic outcomes in overweight and obese individuals, may also impact the gut environment, shaping the microbiota and the host-microbe (co)metabolic pathways and products, possibly affecting large intestine mucosa homeostasis. METHODS PubMed-referenced publications were analyzed with an emphasis on dietary intervention studies involving human volunteers in order to clarify the beneficial vs. deleterious effects of HPD in terms of both metabolic and gut-related health parameters; taking into account the interactions with the gut microbiota. RESULTS HPD generally decrease body weight and improve blood metabolic parameters, but also modify the fecal and urinary contents in various bacterial metabolites and co-metabolites. The effects of HPD on the intestinal microbiota composition appear rather heterogeneous depending on the type of dietary intervention. Recently, HPD consumption was shown to modify the expression of genes playing key roles in homeostatic processes in the rectal mucosa, without evidence of intestinal inflammation. Importantly, the effects of HPD on the gut were dependent on the protein source (i.e. from plant or animal sources), a result which should be considered for further investigations. CONCLUSION Although HPD appear to be efficient for weight loss, the effects of HPD on microbiota-derived metabolites and gene expression in the gut raise new questions on the impact of HPD on the large intestine mucosa homeostasis leading the authors to recommend some caution regarding the utilization of HPD, notably in a recurrent and/or long-term ways.
Collapse
Affiliation(s)
- François Blachier
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris, France.
| | - Martin Beaumont
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris, France
| | - Kevin Joseph Portune
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agronomy and Food Technology, Spanish National Research Council, Valencia, Spain
| | - Nils Steuer
- Department of Gastroenterology, Avicenne Hospital, Assistance Publique-Hôpitaux de Paris, Bobigny, France
| | - Annaïg Lan
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris, France
| | - Marc Audebert
- Toxalim, Research Centre in Food Toxicology, INRA, Toulouse, France
| | - Nadezda Khodorova
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris, France
| | | | - Gheorghe Airinei
- Department of Gastroenterology, Avicenne Hospital, Assistance Publique-Hôpitaux de Paris, Bobigny, France
| | - Robert Benamouzig
- Department of Gastroenterology, Avicenne Hospital, Assistance Publique-Hôpitaux de Paris, Bobigny, France
| | - Anne-Marie Davila
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris, France
| | - Lucie Armand
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris, France
| | - Simone Rampelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Patrizia Brigidi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Daniel Tomé
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris, France
| | - Sandrine Paule Claus
- Department of Food Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Yolanda Sanz
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agronomy and Food Technology, Spanish National Research Council, Valencia, Spain
| |
Collapse
|
30
|
Roxithromycin regulates intestinal microbiota and alters colonic epithelial gene expression. Appl Microbiol Biotechnol 2018; 102:9303-9316. [DOI: 10.1007/s00253-018-9257-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 07/09/2018] [Accepted: 07/13/2018] [Indexed: 12/22/2022]
|
31
|
Structure-Based Drug Design for Cytochrome P450 Family 1 Inhibitors. Bioinorg Chem Appl 2018; 2018:3924608. [PMID: 30147715 PMCID: PMC6083639 DOI: 10.1155/2018/3924608] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/17/2018] [Accepted: 06/20/2018] [Indexed: 12/13/2022] Open
Abstract
Cytochromes P450 are a class of metalloproteins which are responsible for electron transfer in a wide spectrum of reactions including metabolic biotransformation of endogenous and exogenous substrates. The superfamily of cytochromes P450 consists of families and subfamilies which are characterized by a specific structure and substrate specificity. Cytochromes P450 family 1 (CYP1s) play a distinctive role in the metabolism of drugs and chemical procarcinogens. In recent decades, these hemoproteins have been intensively studied with the use of computational methods which have been recently developed remarkably to be used in the process of drug design by the virtual screening of compounds in order to find agents with desired properties. Moreover, the molecular modeling of proteins and ligand docking to their active sites provide an insight into the mechanism of enzyme action and enable us to predict the sites of drug metabolism. The review presents the current status of knowledge about the use of the computational approach in studies of ligand-enzyme interactions for CYP1s. Research on the metabolism of substrates and inhibitors of CYP1s and on the selectivity of their action is particularly valuable from the viewpoint of cancer chemoprevention, chemotherapy, and drug-drug interactions.
Collapse
|
32
|
Schiering C, Vonk A, Das S, Stockinger B, Wincent E. Cytochrome P4501-inhibiting chemicals amplify aryl hydrocarbon receptor activation and IL-22 production in T helper 17 cells. Biochem Pharmacol 2018; 151:47-58. [PMID: 29501585 DOI: 10.1016/j.bcp.2018.02.031] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 02/23/2018] [Indexed: 02/07/2023]
Abstract
The aryl hydrocarbon receptor (AHR) controls interleukin 22 production by T helper 17 cells (Th17). IL-22 contributes to intestinal homeostasis but has also been implicated in chronic inflammatory disorders and colorectal cancer, highlighting the need for appropriate regulation of IL-22 production. Upon activation, the AHR induces expression of cytochrome P4501 (CYP1) enzymes which in turn play an important feedback role that curtails the duration of AHR signaling by metabolizing AHR ligands. Recently we described how agents that inhibit CYP1 function potentiate AHR signaling by disrupting metabolic clearance of the endogenous ligand 6-formylindolo[3,2-b]carbazole (FICZ). In the present study, we investigated the immune-modulating effects of environmental pollutants such as polycyclic aromatic hydrocarbons on Th17 differentiation and IL-22 production. Using Th17 cells deficient in CYP1 enzymes (Cyp1a1/1a2/1b1-/-) we show that these chemicals potentiate AHR activation through inhibition of CYP1 enzymes which leads to increases in intracellular AHR agonists. Our findings demonstrate that IL-22 production by Th17 cells is profoundly enhanced by impaired CYP1-function and strongly suggest that chemicals able to modify CYP1 function or expression may disrupt AHR-mediated immune regulation by altering the levels of endogenous AHR agonist(s).
Collapse
Affiliation(s)
- Chris Schiering
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| | - Anne Vonk
- Swetox, Karolinska Institutet, Unit of Toxicology Sciences, Forskargatan 20, 151 36 Södertälje, Sweden.
| | - Srustidhar Das
- Karolinska Institutet, Department of Medicine, Solna (MedS), K2, L2:04 171 76 Stockholm, Sweden.
| | | | - Emma Wincent
- Swetox, Karolinska Institutet, Unit of Toxicology Sciences, Forskargatan 20, 151 36 Södertälje, Sweden; Karolinska Institutet, Institute of Environmental Medicine, Box 210, 171 77 Stockholm, Sweden.
| |
Collapse
|
33
|
Horley NJ, Beresford KJ, Kaduskar S, Joshi P, McCann GJ, Ruparelia KC, Williams IS, Gatchie L, Sonawane VR, Bharate SB, Chaudhuri B. ( E )-3-(3,4,5-Trimethoxyphenyl)-1-(pyridin-4-yl)prop-2-en-1-one, a heterocyclic chalcone is a potent and selective CYP1A1 inhibitor and cancer chemopreventive agent. Bioorg Med Chem Lett 2017; 27:5409-5414. [DOI: 10.1016/j.bmcl.2017.11.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/03/2017] [Accepted: 11/05/2017] [Indexed: 12/13/2022]
|
34
|
Freedland J, Cera C, Fasullo M. CYP1A1 I462V polymorphism is associated with reduced genotoxicity in yeast despite positive association with increased cancer risk. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2017; 815:35-43. [PMID: 28283091 DOI: 10.1016/j.mrgentox.2017.02.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 01/18/2017] [Accepted: 02/13/2017] [Indexed: 12/20/2022]
Abstract
CYP1A1 functions in detoxifying xenobiotics but occasionally converts compounds into potent genotoxins. CYP1A1 activates polyaromatic hydrocarbons, such as benzo[a]pyrene 7,8 dihydrodiol (BaP-DHD), rendering them genotoxic. Particular alleles of CYP1A1, such as CYP1A1 I462V have been correlated with a higher incidence of breast and lung cancer, but it is unknown whether these variants express enzymes in vivo that are more potent in generating genotoxins. We individually expressed CYP1A1 (CYP1A1.1), CYP1A1 T461N (CYP1A1.4) and I462V (CYP1A1.2) alleles in wild-type and DNA repair deficient mutant strains of Saccharomyces cerevisiae (budding yeast) and asked which yeast strains exhibited the highest levels of carcinogen-associated genotoxicity after exposure to BaP-DHD, aflatoxin B1 (AFB1), and heterocyclic aromatic amines (HAAs). We measured carcinogen-associated recombination, Rad51 foci, and carcinogen-associated toxicity in a DNA repair mutant deficient in both nucleotide excision repair and recombinational repair. CYP1A1 activity was confirmed by measuring ethoxyresorufin-O-deethylation (EROD) activities. Our data indicate that CYP1A1 I462V allele confers the least carcinogen-associated genotoxicity, compared to CYP1A1; however, results vary depending on the chemical carcinogen and the genotoxic endpoint. We speculate that the cancer-associated risk of CYP1A1 I462V may be caused by exposure to other xenobiotics.
Collapse
Affiliation(s)
- Julian Freedland
- College of Nanoscale Science and Engineering, State University of New York Polytechnic Institute, Albany, NY 12205, United States
| | - Cinzia Cera
- Center for Medical Sciences,150 New Scotland Road, Albany, NY 12208, United States
| | - Michael Fasullo
- College of Nanoscale Science and Engineering, State University of New York Polytechnic Institute, Albany, NY 12205, United States; Center for Medical Sciences,150 New Scotland Road, Albany, NY 12208, United States.
| |
Collapse
|
35
|
Badal SAM, Asuncion Valenzuela MM, Zylstra D, Huang G, Vendantam P, Francis S, Quitugua A, Amis LH, Davis W, Tzeng TRJ, Jacobs H, Gangemi DJ, Raner G, Rowland L, Wooten J, Campbell P, Brantley E, Delgoda R. Glaucarubulone glucoside from Castela macrophylla suppresses MCF-7 breast cancer cell growth and attenuates benzo[a]pyrene-mediated CYP1A gene induction. J Appl Toxicol 2017; 37:873-883. [PMID: 28138972 DOI: 10.1002/jat.3436] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 11/24/2016] [Accepted: 12/12/2016] [Indexed: 01/04/2023]
Abstract
Quassinoids often exhibit antioxidant and antiproliferative activity. Emerging evidence suggests that these natural metabolites also display chemopreventive actions. In this study, we investigated the potential for the quassinoid glaucarubulone glucoside (Gg), isolated from the endemic Jamaican plant Castela macrophylla (Simaroubaceae), to display potent cytotoxicity and inhibit human cytochrome P450s (CYPs), particularly CYP1A enzymes, known to convert polyaromatic hydrocarbons into carcinogenic metabolites. Gg reduced the viability of MCF-7 breast adenocarcinoma cells (IC50 = 121 nm) to a greater extent than standard of care anticancer agents 5-fluorouracil, tamoxifen (IC50 >10 μm) and the tamoxifen metabolite 4-hydroxytamoxifen (IC50 = 2.6 μm), yet was not cytotoxic to non-tumorigenic MCF-10A breast epithelial cells. Additionally, Gg induced MCF-7 breast cancer cell death. Gg blocked increases in reactive oxygen species in MCF-10A cells mediated by the polyaromatic hydrocarbon benzo[a]pyrene (B[a]P) metabolite B[a]P 1,6-quinone, yet downregulated the expression of genes that promote antioxidant activity in MCF-7 cells. This implies that Gg exhibits antioxidant and cytoprotective actions in non-tumorigenic breast epithelial cells and pro-oxidant, cytotoxic actions in breast cancer cells. Furthermore, Gg inhibited the activities of human CYP1A according to non-competitive kinetics and attenuated the ability of B[a]P to induce CYP1A gene expression in MCF-7 cells. These data indicate that Gg selectively suppresses MCF-7 breast cancer cell growth without impacting non-tumorigenic breast epithelial cells and blocks B[a]P-mediated CYP1A induction. Taken together, our data provide a rationale for further investigations of Gg and similar plant isolates as potential agents to treat and prevent breast cancer. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Simone A M Badal
- Natural Products Institute, Faculty of Science and Technology, University of the West Indies, Mona, Jamaica, West Indies.,Department of Basic Medical Sciences, Faculty of Medical Sciences, University of the West Indies, Mona, Jamaica, West Indies
| | - Malyn M Asuncion Valenzuela
- Department of Basic Sciences, Center for Health Disparities and Molecular Medicine, Loma Linda University Health School of Medicine, Loma Linda, CA, 92350, USA
| | - Dain Zylstra
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University Health School of Pharmacy, Loma Linda, CA, 92350, USA
| | - George Huang
- Department of Biological Sciences, Clemson University, Clemson, SC, 29634, USA
| | - Pallavi Vendantam
- Department of Biological Sciences, Clemson University, Clemson, SC, 29634, USA
| | - Sheena Francis
- Natural Products Institute, Faculty of Science and Technology, University of the West Indies, Mona, Jamaica, West Indies
| | - Ashley Quitugua
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University Health School of Pharmacy, Loma Linda, CA, 92350, USA
| | - Louisa H Amis
- Department of Basic Sciences, Center for Health Disparities and Molecular Medicine, Loma Linda University Health School of Medicine, Loma Linda, CA, 92350, USA
| | - Willie Davis
- Department of Basic Sciences, Center for Health Disparities and Molecular Medicine, Loma Linda University Health School of Medicine, Loma Linda, CA, 92350, USA.,Department of Pharmaceutical and Administrative Sciences, Loma Linda University Health School of Pharmacy, Loma Linda, CA, 92350, USA
| | - Tzuen-Rong J Tzeng
- Department of Biological Sciences, Clemson University, Clemson, SC, 29634, USA
| | - Helen Jacobs
- Department of Chemistry, Faculty of Science and Technology, University of the West Indies, Mona, Jamaica, West Indies
| | - David J Gangemi
- Department of Biological Sciences, Clemson University, Clemson, SC, 29634, USA
| | - Greg Raner
- Department of Chemistry and Biochemistry, The University of North Carolina at Greensboro, Greensboro, NC, 27402, USA.,Department of Biology and Chemistry, Liberty University, Lynchburg, VA, 24515, USA
| | - Leah Rowland
- Department of Basic Sciences, Center for Health Disparities and Molecular Medicine, Loma Linda University Health School of Medicine, Loma Linda, CA, 92350, USA
| | - Jonathan Wooten
- Department of Basic Sciences, Center for Health Disparities and Molecular Medicine, Loma Linda University Health School of Medicine, Loma Linda, CA, 92350, USA
| | - Petreena Campbell
- Department of Basic Sciences, Center for Health Disparities and Molecular Medicine, Loma Linda University Health School of Medicine, Loma Linda, CA, 92350, USA
| | - Eileen Brantley
- Department of Basic Sciences, Center for Health Disparities and Molecular Medicine, Loma Linda University Health School of Medicine, Loma Linda, CA, 92350, USA.,Department of Pharmaceutical and Administrative Sciences, Loma Linda University Health School of Pharmacy, Loma Linda, CA, 92350, USA.,Department of Chemistry, University of California, Riverside, CA, 92521, USA
| | - Rupika Delgoda
- Natural Products Institute, Faculty of Science and Technology, University of the West Indies, Mona, Jamaica, West Indies
| |
Collapse
|
36
|
Ronco AL, Stefani ED, Mendoza B, Deneo-Pellegrini H, Vazquez A, Abbona E. Mate Intake and Risk of Breast Cancer in Uruguay: a Case- Control Study. Asian Pac J Cancer Prev 2017; 17:1453-61. [PMID: 27039789 DOI: 10.7314/apjcp.2016.17.3.1453] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Regarding 'mate' intake (infusion of Ilex paraguariensis herb, a staple beverage in temperate South American regions), most epidemiologic studies showed positive associations with risk of some cancers, (e.g. upper aerodigestive tract), but evidence on breast cancer (BC) risk is limited to a previous multi-site study, which reported a non significant odds ratio [OR]=0.85, 95% confidence interval [95% CI] 0.67-1.09, p for trend=0.31) for the highest quartile of intake. The present study was conducted in order to further assess associations of 'mate' intake with BC risk. We combined two databases of women belonging to public and private healthcare hospitals. The sample included 572 BC incident cases and 889 controls interviewed with a specific questionnaire featured by socio-demographic, reproductive and lifestyle variables, and a food frequency questionnaire of 64 items, also analyzing 'mate' intake (consumer status, daily intake, age at start, age at quit, duration of habit, intensity of intake). ORs and their 95%CI were calculated through unconditional logistic regression, adjusting for relevant potential confounders. The highest quartile of 'mate' intake was inversely associated with BC risk (OR=0.40, 95%CI 0.26-0.57, p for trend <0.001). Stratified analyses also displayed strong significant inverse associations for 'mate' in frequent tea drinkers (OR=0.22), high energy intake (OR=0.23), high body mass index (OR=0.29) and in postmenopausal women (OR=0.36), among other results. As conclusions, we found evidence of a significant inverse association for 'mate' intake and BC risk.
Collapse
Affiliation(s)
- Alvaro L Ronco
- Unit of Oncology and Radiotherapy, Pereira Rossell Women's Hospital, Montevideo, Uruguay E-mail :
| | | | | | | | | | | |
Collapse
|
37
|
Xue Z, Li D, Yu W, Zhang Q, Hou X, He Y, Kou X. Mechanisms and therapeutic prospects of polyphenols as modulators of the aryl hydrocarbon receptor. Food Funct 2017; 8:1414-1437. [DOI: 10.1039/c6fo01810f] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Polyphenolic AhR modulators displayed concentration-, XRE-, gene-, species- and cell-specific agonistic/antagonistic activity.
Collapse
Affiliation(s)
- Zhaohui Xue
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
| | - Dan Li
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
| | - Wancong Yu
- Medical Plant Laboratory
- Tianjin Research Center of Agricultural Biotechnology
- Tianjin 3000381
- China
| | - Qian Zhang
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
| | - Xiaonan Hou
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
| | - Yulong He
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
| | - Xiaohong Kou
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
| |
Collapse
|
38
|
Wei KL, Chen FY, Lin CY, Gao GL, Kao WY, Yeh CH, Chen CR, Huang HC, Tsai WR, Jong KJ, Li WJ, Su JGJ. Activation of aryl hydrocarbon receptor reduces carbendazim-induced cell death. Toxicol Appl Pharmacol 2016; 306:86-97. [DOI: 10.1016/j.taap.2016.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 05/14/2016] [Accepted: 06/06/2016] [Indexed: 01/03/2023]
|
39
|
Li R, Song M, Li Z, Li Y, Watanabe G, Nagaoka K, Taya K, Li C. 4-Nitrophenol exposure alters the AhR signaling pathway and related gene expression in the rat liver. J Appl Toxicol 2016; 37:150-158. [DOI: 10.1002/jat.3332] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 03/09/2016] [Accepted: 03/11/2016] [Indexed: 11/06/2022]
Affiliation(s)
- Ruonan Li
- Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing China
| | - Meiyan Song
- Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing China
| | - Zhi Li
- Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing China
| | - Yansen Li
- Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing China
| | - Gen Watanabe
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Faculty of Agriculture; Tokyo University of Agriculture and Technology; Tokyo Japan
- Department of Basic Veterinary Science, The United Graduate School of Veterinary Sciences; Gifu University; Gifu Japan
| | - Kentaro Nagaoka
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Faculty of Agriculture; Tokyo University of Agriculture and Technology; Tokyo Japan
- Department of Basic Veterinary Science, The United Graduate School of Veterinary Sciences; Gifu University; Gifu Japan
| | - Kazuyoshi Taya
- Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing China
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Faculty of Agriculture; Tokyo University of Agriculture and Technology; Tokyo Japan
| | - Chunmei Li
- Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing China
| |
Collapse
|
40
|
Günther J, Czabanska A, Bauer I, Leigh JA, Holst O, Seyfert HM. Streptococcus uberis strains isolated from the bovine mammary gland evade immune recognition by mammary epithelial cells, but not of macrophages. Vet Res 2016; 47:13. [PMID: 26738804 PMCID: PMC4704416 DOI: 10.1186/s13567-015-0287-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/27/2015] [Indexed: 01/19/2023] Open
Abstract
Streptococcus uberis is frequently isolated from the mammary gland of dairy cattle. Infection with some strains can induce mild subclinical inflammation whilst others induce severe inflammation and clinical mastitis. We compared here the inflammatory response of primary cultures of bovine mammary epithelial cells (pbMEC) towards S. uberis strains collected from clinical or subclinical cases (seven strains each) of mastitis with the strong response elicited by Escherichia coli. Neither heat inactivated nor live S. uberis induced the expression of 10 key immune genes (including TNF, IL1B, IL6). The widely used virulent strain 0140J and the avirulent strain, EF20 elicited similar responses; as did mutants defective in capsule (hasA) or biofilm formation (sub0538 and sub0539). Streptococcus uberis failed to activate NF-κB in pbMEC or TLR2 in HEK293 cells, indicating that S. uberis particles did not induce any TLR-signaling in MEC. However, preparations of lipoteichoic acid (LTA) from two strains strongly induced immune gene expression and activated NF-κB in pbMEC, without the involvement of TLR2. The immune-stimulatory LTA must be arranged in the intact S. uberis such that it is unrecognizable by the relevant pathogen receptors of the MEC. The absence of immune recognition is specific for MEC, since the same S. uberis preparations strongly induced immune gene expression and NF-κB activity in the murine macrophage model cell RAW264.7. Hence, the sluggish immune response of MEC and not of professional immune cells to this pathogen may aid establishment of the often encountered belated and subclinical phenotype of S. uberis mastitis.
Collapse
Affiliation(s)
- Juliane Günther
- Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| | - Anna Czabanska
- Division of Structural Biochemistry, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Parkallee 1-40, 23845, Borstel, Germany.
| | - Isabel Bauer
- Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| | - James A Leigh
- Department Animal Health and Welfare, School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, Leicestershire, LE12 5RD, UK.
| | - Otto Holst
- Division of Structural Biochemistry, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Parkallee 1-40, 23845, Borstel, Germany.
| | - Hans-Martin Seyfert
- Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| |
Collapse
|
41
|
Séïde M, Marion M, Mateescu MA, Averill-Bates DA. The fungicide thiabendazole causes apoptosis in rat hepatocytes. Toxicol In Vitro 2015; 32:232-9. [PMID: 26748015 DOI: 10.1016/j.tiv.2015.12.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 12/02/2015] [Accepted: 12/28/2015] [Indexed: 02/04/2023]
Abstract
Many pharmaceutical drugs cause hepatotoxicity in humans leading to severe liver diseases, representing a serious public health issue. This study investigates the ability of the anthelmintic and antifungal drug thiabendazole to cause cell death by apoptosis and metabolic changes in primary cultures of rat hepatocytes. Thiabendazole (200-500 μM) induced apoptosis in hepatocytes after 1 to 24h, causing loss of mitochondrial membrane potential, cytochrome c release from mitochondria, Fas-associated death domain (FADD) translocation from the cytosol to membranes, and activation of caspases-3, -8 and -9. Thus, thiabendazole activated both the mitochondrial and death receptor pathways of apoptosis. Under these conditions, cell death by necrosis was not detected following exposure to thiabendazole (100-500 μM) for 24-48 h, measured by lactate dehydrogenase release and propidium iodide uptake. Furthermore, thiabendazole increased activities of cytochrome P450 (CYP) isoenzymes CYP1A and CYP2B after 24 and 48 h, determined by 7-ethoxyresorufin-O-deethylase (EROD) and 7-pentoxyresorufin-O-dealkylase (PROD) activities, respectively. An important finding is that thiabendazole can eliminate hepatocytes by apoptosis, which could be a sensitive marker for hepatic damage and cell death. This study improves understanding of the mode of cell death induced by thiabendazole, which is important given that humans and animals are exposed to this compound as a pharmaceutical agent and in an environmental context.
Collapse
Affiliation(s)
- Marilyne Séïde
- Department of Chemistry, Université du Québec à Montréal (University of Quebec at Montreal), Canada; Department of Biological sciences, Université du Québec à Montréal (University of Quebec at Montreal), Canada
| | - Michel Marion
- Department of Chemistry, Université du Québec à Montréal (University of Quebec at Montreal), Canada
| | - Mircea Alexandru Mateescu
- Department of Chemistry, Université du Québec à Montréal (University of Quebec at Montreal), Canada; BioMedical Research Centre, Université du Québec à Montréal (University of Quebec at Montreal), Canada
| | - Diana A Averill-Bates
- Department of Chemistry, Université du Québec à Montréal (University of Quebec at Montreal), Canada; Department of Biological sciences, Université du Québec à Montréal (University of Quebec at Montreal), Canada; Research Centre for Environmental Toxicology (TOXEN), Université du Québec à Montréal (University of Quebec at Montreal), Canada; BioMedical Research Centre, Université du Québec à Montréal (University of Quebec at Montreal), Canada.
| |
Collapse
|
42
|
Korobkova EA. Effect of Natural Polyphenols on CYP Metabolism: Implications for Diseases. Chem Res Toxicol 2015; 28:1359-90. [PMID: 26042469 DOI: 10.1021/acs.chemrestox.5b00121] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cytochromes P450 (CYPs) are a large group of hemeproteins located on mitochondrial membranes or the endoplasmic reticulum. They play a crucial role in the metabolism of endogenous and exogenous molecules. The activity of CYP is associated with a number of factors including redox potential, protein conformation, the accessibility of the active site by substrates, and others. This activity may be potentially modulated by a variety of small molecules. Extensive experimental data collected over the past decade point at the active role of natural polyphenols in modulating the catalytic activity of CYP. Polyphenols are widespread micronutrients present in human diets of plant origin and in medicinal herbs. These compounds may alter the activity of CYP either via direct interactions with the enzymes or by affecting CYP gene expression. The polyphenol-CYP interactions may significantly alter the pharmacokinetics of drugs and thus influence the effectiveness of chemical therapies used in the treatment of different types of cancers, diabetes, obesity, and cardiovascular diseases (CVD). CYPs are involved in the oxidation and activation of external carcinogenic agents, in which case the inhibition of the CYP activity is beneficial for health. CYPs also support detoxification processes. In this case, it is the upregulation of CYP genes that would be favorable for the organism. A CYP enzyme aromatase catalyzes the formation of estrone and estradiol from their precursors. CYPs also catalyze multiple reactions leading to the oxidation of estrogen. Estrogen signaling and oxidative metabolism of estrogen are associated with the development of cancer. Thus, polyphenol-mediated modulation of the CYP's activity also plays a vital role in estrogen carcinogenesis. The aim of the present review is to summarize the data collected over the last five to six years on the following topics: (1) the mechanisms of the interactions of CYP with food constituents that occur via the direct binding of polyphenols to the enzymes and (2) the mechanisms of the regulation of CYP gene expression mediated by polyphenols. The structure-activity relationship relevant to the ability of polyphenols to affect the activity of CYP is analyzed. The application of polyphenol-CYP interactions to diseases is discussed.
Collapse
Affiliation(s)
- Ekaterina A Korobkova
- John Jay College of Criminal Justice, The Department of Sciences, City University of New York, 524 W 59th Street, New York, New York 10019, United States
| |
Collapse
|
43
|
Wang L, Lingappan K, Jiang W, Couroucli XI, Welty SE, Shivanna B, Barrios R, Wang G, Firoze Khan M, Gonzalez FJ, Jackson Roberts L, Moorthy B. Disruption of cytochrome P4501A2 in mice leads to increased susceptibility to hyperoxic lung injury. Free Radic Biol Med 2015; 82:147-59. [PMID: 25680282 PMCID: PMC4418801 DOI: 10.1016/j.freeradbiomed.2015.01.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 01/09/2015] [Accepted: 01/19/2015] [Indexed: 12/16/2022]
Abstract
Hyperoxia contributes to acute lung injury in diseases such as acute respiratory distress syndrome. Cytochrome P450 (CYP) 1A enzymes have been implicated in hyperoxic lung injury, but the mechanistic role of CYP1A2 in pulmonary injury is not known. We hypothesized that mice lacking the gene Cyp1a2 (which is predominantly expressed in the liver) will be more sensitive to lung injury and inflammation mediated by hyperoxia and that CYP1A2 will play a protective role by attenuating lipid peroxidation and oxidative stress in the lung. Eight- to ten-week-old WT (C57BL/6) or Cyp1a2(-/-) mice were exposed to hyperoxia (>95% O2) or maintained in room air for 24-72 h. Lung injury was assessed by determining the ratio of lung weight/body weight (LW/BW) and by histology. Extent of inflammation was determined by measuring the number of neutrophils in the lung as well as cytokine expression. The Cyp1a2(-/-) mice under hyperoxic conditions showed increased LW/BW ratios, lung injury, neutrophil infiltration, and IL-6 and TNF-α levels and augmented lipid peroxidation, as evidenced by increased formation of malondialdehyde- and 4-hydroxynonenal-protein adducts and pulmonary isofurans compared to WT mice. In vitro experiments showed that the F2-isoprostane PGF2-α is metabolized by CYP1A2 to a dinor metabolite, providing evidence for a catalytic role for CYP1A2 in the metabolism of F2-isoprostanes. In summary, our results support the hypothesis that hepatic CYP1A2 plays a critical role in the attenuation of hyperoxic lung injury by decreasing lipid peroxidation and oxidative stress in vivo.
Collapse
Affiliation(s)
- Lihua Wang
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX 77030, USA
| | - Krithika Lingappan
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX 77030, USA
| | - Weiwu Jiang
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xanthi I Couroucli
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stephen E Welty
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX 77030, USA
| | - Binoy Shivanna
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX 77030, USA
| | - Roberto Barrios
- Department of Pathology, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Gangduo Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - M Firoze Khan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Frank J Gonzalez
- Laboratory of Molecular Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - L Jackson Roberts
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Bhagavatula Moorthy
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
44
|
Ashino T, Hakukawa K, Itoh Y, Numazawa S. Inhibitory effect of synthetic cannabinoids on CYP1A activity in mouse liver microsomes. J Toxicol Sci 2015; 39:815-20. [PMID: 25374372 DOI: 10.2131/jts.39.815] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Synthetic cannabinoids developed by chemical modification are believed to bind to cannabinoid receptors and cause neurological effects similar to cannabis; however, their effects on drug metabolizing enzymes are unknown. This study aimed to elucidate the effect of synthetic cannabinoids on cytochrome P450 1A activity. Naphthoylindole, a basic structure of the major synthetic cannabinoids, strongly inhibited CYP1A activity in a competitive manner; the apparent Ki value was 0.40 μM. The N-Alkylated derivatives of naphthoylindole, MAM-2201 and JWH-019, also inhibited CYP1A activity in a concentration-dependent manner; however, their inhibitory effects were weaker than naphthoylindole. An adamantylamidoindole derivative, STS-135, showed inhibition of CYP1A activity in a concentrationdependent manner, but the adamantoylindole derivatives, AB-001 and AM-1248, did not. A tetramethylcyclopropanoylindole derivative, UR-144, showed a weak inhibition of CYP1A activity at high concentrations. These results suggest that synthetic cannabinoids and their basic molecules are capable of inhibiting CYP1A enzymatic activity.
Collapse
Affiliation(s)
- Takashi Ashino
- Division of Toxicology, Department of Pharmacology, Toxicology and Therapeutics, Showa University School of Pharmacy
| | | | | | | |
Collapse
|
45
|
Cho SH, Park SY, Lee EJ, Cho YH, Park HS, Hong SH, Kim WJ. Regulation of CYP1A1 and Inflammatory Cytokine by NCOA7 Isoform 4 in Response to Dioxin Induced Airway Inflammation. Tuberc Respir Dis (Seoul) 2015; 78:99-105. [PMID: 25861343 PMCID: PMC4388907 DOI: 10.4046/trd.2015.78.2.99] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 11/10/2014] [Accepted: 12/10/2014] [Indexed: 11/24/2022] Open
Abstract
Background Aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor, binds to a wide variety of synthetic and naturally occurring compounds. AhR is involved in the regulation of inflammatory response during acute and chronic respiratory diseases. We investigated whether nuclear receptor coactivator 7 (NCOA7) could regulate transcriptional levels of AhR target genes and inflammatory cytokines in 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-treated human bronchial epithelial cells. This study was based on our previous study that NCOA7 was differentially expressed between normal and chronic obstructive pulmonary disease lung tissues. Methods BEAS-2B and A549 cells grown under serum-free conditions were treated with or without TCDD (0.15 nM and 6.5 nM) for 24 hours after transfection of pCMV-NCOA7 isoform 4. Expression levels of cytochrome P4501A1 (CYP1A1), IL-6, and IL-8 were measured by quantitative real-time polymerase chain reaction. Results The transcriptional activities of CYP1A1 and inflammatory cytokines were strongly induced by TCDD treatment in both BEAS-2B and A549 cell lines. The NCOA7 isoform 4 oppositely regulated the transcriptional activities of CYP1A1 and inflammatory cytokines between BEAS-2B and A549 cell lines. Conclusion Our results suggest that NCOA7 could act as a regulator in the TCDD-AhR signaling pathway with dual roles in normal and abnormal physiological conditions.
Collapse
Affiliation(s)
- Sung-Hwan Cho
- Regional Center for Respiratory Diseases, Kangwon National University Hospital, Chuncheon, Korea
| | - Shin Young Park
- Regional Center for Respiratory Diseases, Kangwon National University Hospital, Chuncheon, Korea
| | - Eun Jeong Lee
- Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Yo Han Cho
- Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Hyun Sun Park
- Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Woo Jin Kim
- Regional Center for Respiratory Diseases, Kangwon National University Hospital, Chuncheon, Korea. ; Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| |
Collapse
|
46
|
Kim BR, Seo JY, Sung MK, Park JHY, Suh HJ, Liu KH, Kim JS. Suppression of 7,12-dimethylbenz(a)anthracene-induced mammary tumorigenesis by glyceollins. Mol Nutr Food Res 2015; 59:907-17. [DOI: 10.1002/mnfr.201400726] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 01/23/2015] [Accepted: 01/26/2015] [Indexed: 01/03/2023]
Affiliation(s)
- Bo Ram Kim
- School of Food Science and Biotechnology; Kyungpook National University; Daegu Republic of Korea
| | - Ji-Yeon Seo
- School of Food Science and Biotechnology; Kyungpook National University; Daegu Republic of Korea
| | - Mi-Kyung Sung
- Department of Food and Nutrition; Sookmyung Women's University, Seoul; Republic of Korea
| | - Jung Han Yoon Park
- Department of Food Science and Nutrition; Hallym University; Chuncheon Republic of Korea
| | - Hwa-Jin Suh
- Gyeongbuk Natural Color Industry Institute; Gyeongbuk Republic of Korea
| | - Kwang Hyeon Liu
- College of Pharmacy; Kyungpook National University; Daegu Republic of Korea
| | - Jong-Sang Kim
- School of Food Science and Biotechnology; Kyungpook National University; Daegu Republic of Korea
| |
Collapse
|
47
|
|
48
|
Curran CS, Carrillo ER, Ponik SM, Keely PJ. Collagen density regulates xenobiotic and hypoxic response of mammary epithelial cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 39:114-124. [PMID: 25481308 PMCID: PMC4323890 DOI: 10.1016/j.etap.2014.10.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 10/27/2014] [Indexed: 06/04/2023]
Abstract
Breast density, where collagen I is the dominant component, is a significant breast cancer risk factor. Cell surface integrins interact with collagen, activate focal adhesion kinase (FAK), and downstream cell signals associated with xenobiotics (AhR, ARNT) and hypoxia (HIF-1α, ARNT). We examined if mammary cells cultured in high density (HD) or low density (LD) collagen gels affected xenobiotic or hypoxic responses. ARNT production was significantly reduced by HD culture and in response to a FAK inhibitor. Consistent with a decrease in ARNT, AhR and HIF-1α reporter activation and VEGF production was lower in HD compared to LD. However, P450 production was enhanced in HD and induced by AhR and HIF-1α agonists, possibly in response to increased NF-κB activaton. Thus, collagen density differentially regulates downstream cell signals of AhR and HIF-1α by modulating the activity of FAK, the release of NF-κB transcriptional factors, and the levels of ARNT.
Collapse
Affiliation(s)
- Colleen S Curran
- Department of Cell and Regenerative Biology, University of Wisconsin at Madison, Madison, WI 53706, USA.
| | - Esteban R Carrillo
- Department of Cell and Regenerative Biology, University of Wisconsin at Madison, Madison, WI 53706, USA.
| | - Suzanne M Ponik
- Department of Cell and Regenerative Biology, University of Wisconsin at Madison, Madison, WI 53706, USA.
| | - Patricia J Keely
- Department of Cell and Regenerative Biology, University of Wisconsin at Madison, Madison, WI 53706, USA.
| |
Collapse
|
49
|
Sgrignani J, Bon M, Colombo G, Magistrato A. Computational approaches elucidate the allosteric mechanism of human aromatase inhibition: a novel possible route to Small-molecule regulation of CYP450s activities? J Chem Inf Model 2014; 54:2856-68. [PMID: 25178092 DOI: 10.1021/ci500425y] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Human aromatase (HA) is a P450 cytochrome (CYP) with an essential role in estrogen biosynthesis. Since more than 70% of breast cancers are positive for estrogenic receptor (ER), the reduction of estrogen physiological concentrations through HA inhibition is one of most important therapeutic strategies against this cancer type. Recently, experimental evidence showed that selected taxmoxifen metabolites, which are typically used as estrogen receptor modulators (SERMs), inhibit HA through an allosteric mechanism. In this work, we present a computational protocol to (i) characterize the structural framework and (ii) define the atomistic details of the determinants for the noncompetitive inhibition mechanism. Our calculations identify two putative binding sites able to efficiently bind all tamoxifen metabolites. Analysis of long-scale molecular dynamics simulations reveal that endoxifen, the most effective noncompetitive inhibitor, induces significant enzyme rigidity by binding in one of the possible peripheral sites. The consequence of this binding event is the suppression of one of the functional enzymatic collective motions associated with breathing of the substrate access channel. Moreover, an internal dynamics-based alignment of HA with six other human cytochromes shows that this collective motion is common to other members of the CYP450 protein family. On this basis, our findings may thus be of help for the development of new (pan)inhibitors for the therapeutic treatment of cancer, targeting and modulating the activity of HA and of estrogen receptor, and may also stimulate the development of new drug design strategies for chemoprevention and chemoprotection via allosteric inhibition of CYP450 proteins.
Collapse
Affiliation(s)
- Jacopo Sgrignani
- Istituto di Chimica del Riconoscimento Molecolare, CNR, Via Mario Bianco 9, 20131 Milano, Milano, Italy
| | | | | | | |
Collapse
|