1
|
Li H, Kelley J, Ye Y, Ye ZW, Townsend DM, Zhang J, Wu Y. REDOX Imbalance and Oxidative Stress in the Intervertebral Disc: The Effect of Mechanical Stress and Cigarette Smoking on ER Stress and Mitochondrial Dysfunction. Cells 2025; 14:613. [PMID: 40277939 PMCID: PMC12025608 DOI: 10.3390/cells14080613] [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: 03/11/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/26/2025] Open
Abstract
Low back pain is a widespread condition that significantly impacts quality of life, with intervertebral disc degeneration (IDD) being a major contributing factor. However, the underlying mechanisms of IDD remain poorly understood, necessitating further investigation. Environmental risk factors, such as mechanical stress and cigarette smoke, elevate reactive oxygen species levels from both endogenous and exogenous sources, leading to redox imbalance and oxidative stress. The endoplasmic reticulum (ER) and mitochondria, two key organelles responsible for protein folding and energy production, respectively, are particularly vulnerable to oxidative stress. Under oxidative stress conditions, ER stress and mitochondrial dysfunction occur, resulting in unfolded protein response activation, impaired biosynthetic processes, and disruptions in the tricarboxylic acid cycle and electron transport chain, ultimately compromising energy metabolism. Prolonged and excessive ER stress can further trigger apoptosis through ER-mitochondrial crosstalk. Given the unique microenvironment of the intervertebral disc (IVD)-characterized by hypoxia, glucose starvation, and region-specific cellular heterogeneity-the differential effects of environmental stressors on distinct IVD cell populations require further investigation. This review explores the potential mechanisms through which environmental risk factors alter IVD cell activities, contributing to IDD progression, and discusses future therapeutic strategies aimed at mitigating disc degeneration.
Collapse
Affiliation(s)
- Hui Li
- Department of Bioengineering, Clemson University, Charleston, SC 29425, USA; (H.L.); (J.K.)
| | - Joshua Kelley
- Department of Bioengineering, Clemson University, Charleston, SC 29425, USA; (H.L.); (J.K.)
| | - Yiqing Ye
- Department of Orthopaedics and Physical Medicine & Rehabilitation, Medical University of South Carolina, Charleston, SC 29425, USA
- Academic Magnet High School, North Charleston, SC 29405, USA
| | - Zhi-Wei Ye
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Danyelle M. Townsend
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jie Zhang
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Yongren Wu
- Department of Bioengineering, Clemson University, Charleston, SC 29425, USA; (H.L.); (J.K.)
- Department of Orthopaedics and Physical Medicine & Rehabilitation, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
2
|
Siwakoti RC, Rosario-Pabon Z, Vélez Vega CM, Hao W, Alshawabkeh A, Cordero JF, Watkins DJ, Meeker JD. Assessment of per- and polyfluoroalkyl substances (PFAS) exposure and associations with oxidative stress biomarkers among pregnant women from the PROTECT cohort. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 973:179130. [PMID: 40112543 PMCID: PMC11956163 DOI: 10.1016/j.scitotenv.2025.179130] [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: 12/26/2024] [Revised: 02/22/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
INTRODUCTION Per- and polyfluoroalkyl substances (PFAS) are synthetic chemicals linked with adverse pregnancy outcomes, yet they remain understudied in Puerto Rico. Moreover, underlying biological mechanisms of PFAS are not fully understood, although oxidative stress and inflammation are suspected pathways. OBJECTIVES We aimed to characterize PFAS exposure among pregnant women in Puerto Rico and examine associations between early to mid-pregnancy PFAS concentrations and repeated measures of oxidative stress biomarkers. METHODS This study included 434 participants enrolled in the PROTECT birth cohort. We analyzed serum samples (∼18 weeks of gestation) for nine PFAS, while urinary oxidative stress biomarkers [8-isoprostaglandin F2α (8-IsoP), its metabolite (IsoP-M), and prostaglandin F₂α (PGF2α)] were measured up to three times during pregnancy. We examined associations between each PFAS and oxidative stress biomarker using linear mixed effects regression models and multivariable regression analyses, adjusting for maternal demographic, socioeconomic, and study-related factors. RESULTS PFOS, PFNA, PFHxS, and PFOA were detected in over 50 % of participants, with PFOS being the most dominant. The majority of participants had total PFAS levels above 2 ng/mL. In repeated measures analyses, an interquartile range increase in PFHxS was associated with a 5.35 % (95 % CI: 0.12, 10.86) rise in IsoP-M levels. In categorical analyses, moderate levels of PFOS and PFNA were positively associated with PGF2α, while higher PFDeA was suggestively linked to 8-IsoP and IsoP-M. CONCLUSIONS Despite relatively modest levels compared to the U.S. NHANES, certain PFAS were positively linked with oxidative stress or inflammation, highlighting the need for broader investigations to examine PFAS-related alteration of inflammatory processes during pregnancy.
Collapse
Affiliation(s)
| | - Zaira Rosario-Pabon
- University of Puerto Rico Medical Sciences Campus, San Juan 00921, Puerto Rico
| | - Carmen M Vélez Vega
- University of Puerto Rico Medical Sciences Campus, San Juan 00921, Puerto Rico
| | - Wei Hao
- University of Michigan, Ann Arbor, MI 48105, USA
| | | | | | | | | |
Collapse
|
3
|
Chen Q, Chi Y, Zhu Q, Ma N, Min L, Ji S. Effects of Perfluorooctane Sulfonic Acid Exposure on Intestinal Microbial Community, Lipid Metabolism, and Liver Lesions in Mice. Int J Mol Sci 2025; 26:2648. [PMID: 40141291 PMCID: PMC11942384 DOI: 10.3390/ijms26062648] [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: 02/07/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Perfluorooctane sulfonic acid (PFOS) is a persistent organic pollutant that has attracted much attention due to its wide environmental distribution and potential toxicity. Intestinal microbiota is an important regulator of host health, and its composition and metabolic function are easily interfered with by environmental pollutants. In this study, the effects of PFOS exposure on gut microbiota, lipid metabolism, and host health were investigated in mice. The results showed that PFOS exposure did not significantly change α diversity, but significantly affected the β diversity and community structure of intestinal microflora in mice. At the taxonomic level, the ratio of Firmicutes to Bacteroidetes decreased, and the changes in the abundance of specific bacteria were closely related to liver diseases and lipid metabolism disorders. PFOS exposure also interfered with the gut-liver axis mechanism, increased blood lipids and liver function related indicators in mice, and induced intestinal and liver histological lesions. This study revealed the toxic mechanism of PFOS mediated by intestinal microbiota, providing a new research perspective for health problems caused by environmental pollutants and theoretical support for the formulation of relevant public health policies.
Collapse
Affiliation(s)
- Qianfeng Chen
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou 362000, China
| | - Yulang Chi
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou 362000, China
| | - Qingyu Zhu
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou 362000, China
| | - Nana Ma
- College of Life Science, Hebei University, Baoding 071002, China
| | - Lingli Min
- College of Resources and Environmental Science, Quanzhou Normal University, Quanzhou 362000, China
| | - Shouping Ji
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou 362000, China
| |
Collapse
|
4
|
Chuang YT, Yen CY, Liu W, Chien TM, Chang FR, Tsai YH, Tang JY, Chang HW. The protection of bisphenol A-modulated miRNAs and targets by natural products. ENVIRONMENT INTERNATIONAL 2025; 196:109299. [PMID: 39884249 DOI: 10.1016/j.envint.2025.109299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/04/2024] [Accepted: 01/20/2025] [Indexed: 02/01/2025]
Abstract
Bisphenol A (BPA) is a ubiquitous environmental pollutant with endocrine-disrupting functions. Identifying protective drugs and exploring the mechanisms against BPA are crucial in healthcare. Natural products exhibiting antioxidant properties are considered to be able to protect against BPA toxicity. Although BPA-modulated targets and miRNAs have been individually reported, their connections to natural products were rarely organized. With the help of a protein-protein interaction database (STRING), the relationship between individual BPA-modulated targets was interconnected to provide a systemic view. In this review, BPA-downregulated and -upregulated targets are classified, and their interactive network was innovatively analyzed using the bioinformatic database (STRING). BPA-modulated miRNAs were also retrieved and ingeniously connected to BPA-modulated targets. Moreover, a novel connection between BPA-countering natural products was integrated into BPA-modulated miRNAs and targets. All these targets-associated natural products and/or miRNAs were incorporated into the STRING network, providing systemic relationships. Overall, the BPA-modulated target-miRNA-protecting natural product axis was innovatively constructed, providing a straightforward direction for exploring the integrated BPA-countering effects and mechanisms of natural products.
Collapse
Affiliation(s)
- Ya-Ting Chuang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Ching-Yu Yen
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan; Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan.
| | - Wangta Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Tsu-Ming Chien
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Urology, Kaohsiung Gangshan Hospital, Kaohsiung Medical University, Kaohsiung 820111, Taiwan.
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Yi-Hong Tsai
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung 907101, Taiwan.
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
| |
Collapse
|
5
|
Phelps DW, Connors AM, Ferrero G, DeWitt JC, Yoder JA. Per- and polyfluoroalkyl substances alter innate immune function: evidence and data gaps. J Immunotoxicol 2024; 21:2343362. [PMID: 38712868 PMCID: PMC11249028 DOI: 10.1080/1547691x.2024.2343362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/10/2024] [Indexed: 05/08/2024] Open
Abstract
Per- and polyfluoroalkyl substances (PFASs) are a large class of compounds used in a variety of processes and consumer products. Their unique chemical properties make them ubiquitous and persistent environmental contaminants while also making them economically viable and socially convenient. To date, several reviews have been published to synthesize information regarding the immunotoxic effects of PFASs on the adaptive immune system. However, these reviews often do not include data on the impact of these compounds on innate immunity. Here, current literature is reviewed to identify and incorporate data regarding the effects of PFASs on innate immunity in humans, experimental models, and wildlife. Known mechanisms by which PFASs modulate innate immune function are also reviewed, including disruption of cell signaling, metabolism, and tissue-level effects. For PFASs where innate immune data are available, results are equivocal, raising additional questions about common mechanisms or pathways of toxicity, but highlighting that the innate immune system within several species can be perturbed by exposure to PFASs. Recommendations are provided for future research to inform hazard identification, risk assessment, and risk management practices for PFASs to protect the immune systems of exposed organisms as well as environmental health.
Collapse
Affiliation(s)
- Drake W. Phelps
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC
- Center for Environmental and Health Effects of PFAS, North Carolina State University, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
| | - Ashley M. Connors
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC
- Center for Environmental and Health Effects of PFAS, North Carolina State University, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
- Toxicology Program, North Carolina State University, Raleigh, NC
- Genetics and Genomics Academy, North Carolina State University, Raleigh, NC
| | - Giuliano Ferrero
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC
- Center for Environmental and Health Effects of PFAS, North Carolina State University, Raleigh, NC
| | - Jamie C. DeWitt
- Center for Environmental and Health Effects of PFAS, North Carolina State University, Raleigh, NC
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR
| | - Jeffrey A. Yoder
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC
- Center for Environmental and Health Effects of PFAS, North Carolina State University, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
- Toxicology Program, North Carolina State University, Raleigh, NC
- Genetics and Genomics Academy, North Carolina State University, Raleigh, NC
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC
| |
Collapse
|
6
|
Li L, Ren J, Guo M, An Z, Duan W, Lv J, Tan Z, Yang J, Zhu Y, Yang H, Liu Y, Ma Y, Guo H. SAP130 mediates crosstalk between hepatocyte ferroptosis and M1 macrophage polarization in PFOS-induced hepatotoxicity. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 951:175612. [PMID: 39163934 DOI: 10.1016/j.scitotenv.2024.175612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/30/2024] [Accepted: 08/16/2024] [Indexed: 08/22/2024]
Abstract
Perfluorooctane sulfonate (PFOS) is a persistent organic pollutant widely utilized in industrial manufacturing and daily life, leading to significant environmental accumulation and various public health issues. This study aims to characterize spliceosome-associated protein 130 (SAP130) as a key mediator of crosstalk between hepatocytes and macrophages, elucidating its role in PFOS-induced liver inflammation. The data demonstrate that PFOS exposure induces ferroptosis in mouse liver and AML12 cells. During ferroptosis, SAP130 is released from injured hepatocytes into the microenvironment, binding to macrophage-inducible C-type lectin (Mincle) and activating the Mincle/Syk signaling pathway in macrophages, ultimately promoting M1 polarization and exacerbating liver injury. Treatment with the ferroptosis inhibitor Ferrostatin-1 reduces SAP130 release, inhibits Mincle/Syk signaling activation, and mitigates inflammatory response. Furthermore, siSAP130 suppresses the activation of the Mincle signaling pathway and M1 polarization in BMDM cells. Conversely, treatment with the ferroptosis agonist Erastin enhances paracrine secretion of SAP130 and exacerbates inflammation. These findings emphasize the significance of hepatocyte-macrophage crosstalk as a critical pathway for PFOS-induced liver injury in mice while highlighting SAP130 as a pivotal regulator of ferroptosis and inflammation, thereby elucidating the potential mechanism of PFOS-induced liver injury.
Collapse
Affiliation(s)
- Longfei Li
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Jingyi Ren
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Mingmei Guo
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Ziwen An
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Wenjing Duan
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Junli Lv
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Zhenzhen Tan
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Jing Yang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Yiming Zhu
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Huiling Yang
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, PR China
| | - Yi Liu
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Yuxia Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, PR China.
| | - Huicai Guo
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, PR China; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, PR China.
| |
Collapse
|
7
|
Sethi N, Khokhar M, Mathur M, Batra Y, Mohandas A, Tomo S, Rao M, Banerjee M. Therapeutic Potential of Nutraceuticals against Drug-Induced Liver Injury. Semin Liver Dis 2024; 44:430-456. [PMID: 39393795 DOI: 10.1055/s-0044-1791559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
Drug-induced liver injury (DILI) continues to be a major concern in clinical practice, thus necessitating a need for novel therapeutic approaches to alleviate its impact on hepatic function. This review investigates the therapeutic potential of nutraceuticals against DILI, focusing on examining the underlying molecular mechanisms and cellular pathways. In preclinical and clinical studies, nutraceuticals, such as silymarin, curcumin, and N-acetylcysteine, have demonstrated remarkable efficacy in attenuating liver injury induced by diverse pharmaceutical agents. The molecular mechanisms underlying these hepatoprotective effects involve modulation of oxidative stress, inflammation, and apoptotic pathways. Furthermore, this review examines cellular routes affected by these nutritional components focusing on their influence on hepatocytes, Kupffer cells, and stellate cells. Key evidence highlights that autophagy modulation as well as unfolded protein response are essential cellular processes through which nutraceuticals exert their cytoprotective functions. In conclusion, nutraceuticals are emerging as promising therapeutic agents for mitigating DILI, by targeting different molecular pathways along with cell processes involved in it concurrently.
Collapse
Affiliation(s)
- Namya Sethi
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Manoj Khokhar
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Mitali Mathur
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Yashi Batra
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Amal Mohandas
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Sojit Tomo
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Mahadev Rao
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Karnataka, India
| | - Mithu Banerjee
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| |
Collapse
|
8
|
Ren J, Zuo J, Yin B, Huang D, Wen R, Pei H, Liu J, Zhang Y, Zhu S, Zhen S, Ma Y. Flaxseed Oil Alleviates PFOS-Induced Liver Injury by Regulating Hepatic Cholesterol Metabolism. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23465-23477. [PMID: 39392608 DOI: 10.1021/acs.jafc.4c04438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Perfluorooctanesulfonate (PFOS) is a widespread, persistent environmental pollutant that exerts apparent liver toxicity. Flaxseed oil (FO), a dietary oil rich in α-linolenic acid, has been demonstrated to possess a diverse array of health benefits. However, whether FO protects against PFOS-induced liver injury and its underlying mechanisms remain unclear. C57/BL6 mice were orally treated with different concentrations of FO alone or in combination with 10 mg/kg of PFOS for 28 consecutive days. Blood and liver tissues were collected for proteomic, histopathological, biochemical, immunohistochemical, and molecular examinations. Results demonstrated that FO supplementation reduced PFOS-induced liver injury, as evidenced by a decrease in histopathological changes, serum transaminase (ALT and AST) levels, levels of oxidative stress, and inflammatory cytokine (TNF-α, IL-1β, and IL-6) levels. Proteomic analyses showed that differentially expressed proteins were enriched in cholesterol metabolic pathways when comparing the PFOS group to the FO supplementation groups. The expression of cholesterol metabolism-related proteins was also subsequently measured, revealing that FO supplementation decreased the protein expressions of SREBP2, HMGCR, and LDLR while increasing the expression of CYP7A1. This study demonstrates that FO can alleviate PFOS-induced hepatotoxicity by regulating hepatic cholesterol metabolism, indicating that FO may serve as an effective dietary intervention for preventing liver injury caused by PFOS.
Collapse
Affiliation(s)
- Jingyi Ren
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Jinshi Zuo
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Bowen Yin
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Dan Huang
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang 050017, China
| | - Rui Wen
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Huanting Pei
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Jiarui Liu
- Undergraduate of College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Yadong Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Siqi Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Shuman Zhen
- Department of Radiotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang 050017, China
| | - Yuxia Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| |
Collapse
|
9
|
Zhang X, Gao H, Chen X, Liu Z, Wang H, Cui M, Li Y, Yu Y, Chen S, Xing X, Chen L, Li D, Zeng X, Wang Q. Identification of sanguinarine as a novel antagonist for perfluorooctanoate/perfluorooctane sulfonate-induced senescence of hepatocytes: An integrated computational and experimental analysis. JOURNAL OF HAZARDOUS MATERIALS 2024; 478:135583. [PMID: 39180998 DOI: 10.1016/j.jhazmat.2024.135583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 08/09/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
Perfluorooctanoate (PFOA) and perfluorooctane sulfonate (PFOS), two prominent per- and polyfluoroalkyl substances (PFASs), are potentially harmful to many human organs. However, there only exist limited methods to mitigate their health hazards. The aim of this study is to combine a bioinformatics analysis with in vitro experiments to discover small molecules that can alleviate liver damage caused by PFOA/PFOS. We identified 192 and 82 key genes related to hepatocytes exposed to PFOA and PFOS, respectively. The functional enrichment analysis of key genes suggested cellular senescence may be important in PFOA/PFOS-induced hepatotoxicity. The in vitro models revealed that PFOA/PFOS led to hepatocyte senescence by increasing the activity of SA-β-gal, inducing mitochondrial dysfunction, impacting cell cycle arrest, and elevating the expressions of p21, p53, IL-1β, and SASP-related cytokines. The drug-target gene set enrichment analysis method was employed to compare the transcriptome data from the Gene Expression Omnibus database (GEO), Comparative Toxicogenomics Database (CTD), and the high-throughput experiment- and reference-guided database (HERB), and 21 traditional Chinese medicines (TCMs) were identified that may alleviate PFOA/PFOS-induced liver aging. The experimental results of co-exposure to PFOA/PFOS and TCMs showed that sanguinarine has particular promise in alleviating cellular senescence caused by PFOA/PFOS. Further investigations revealed that the mTOR-p53 signaling pathway was involved in PFOA/PFOS-mediated hepatic senescence and can be blocked using sanguinarine.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Huan Gao
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaoyu Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Ziqi Liu
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Han Wang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Mengxing Cui
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yajie Li
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; Infinitus (China) Company Ltd, Guangzhou 510623, China
| | - Yongjiang Yu
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Shen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiumei Xing
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Liping Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Daochuan Li
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaowen Zeng
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Qing Wang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
10
|
Ren J, Liu J, Zuo J, Zhang Z, Huang D, Liu X, Lu M, Zhang Y, Su Y, Ma Y. Flaxseed oil attenuates PFOS-induced testicular damage by regulating RNA alternative splicing. Food Funct 2024; 15:10007-10019. [PMID: 39282919 DOI: 10.1039/d4fo03486d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Background: Perfluorooctane sulfonate (PFOS) is a persistent, widely present environmental pollutant, and its toxicity to male reproduction has gradually attracted attention. Flaxseed oil (FO) is a dietary oil abundant in α-linolenic acid and has been demonstrated to possess multiple health benefits. However, whether FO protects against PFOS-induced testicular injury and its mechanism remain unclear. Methods: C57/BL6 mice were gavaged with different concentrations of FO or PFOS (10 mg kg-1) for 28 days. Blood and testicular tissues were collected for histopathology, proteomics, and biochemical and molecular analyses. Results: Our results showed that FO supplementation significantly attenuated PFOS-induced testicular injury, as indicated by histopathological changes, decreased oxidative stress level, increased sperm count, decreased rate of sperm malformation, and improved functional markers of spermatogenesis. Proteomic analysis showed that differentially expressed proteins were notably enriched in spliceosome pathways. Machine learning algorithms were used to screen the hub gene, and PRPF3 and PUF60 proteins were found to be important for FO to exert protective benefits to testicular injury. Western blot results confirmed that FO supplementation could increase the protein expression of PRPF3 and decrease the protein expression of PUF60 in PFOS-exposed mice. Conclusions: This study revealed that FO can alleviate PFOS-induced testicular dysfunction by regulating RNA alternative splicing. The spliceosome-related proteins PRPF3 and PUF60 may be the potential targets for FO to alleviate PFOS-induced testicular injury. FO supplementation may be an effective dietary intervention to prevent adverse effects of PFOS on testes.
Collapse
Affiliation(s)
- Jingyi Ren
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China.
| | - Jiarui Liu
- Undergraduate of College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jinshi Zuo
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China.
| | - Zhenao Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China.
| | - Dan Huang
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xuanyi Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China.
| | - Miaomiao Lu
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China.
| | - Yadong Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China.
| | - Yang Su
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yuxia Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China.
| |
Collapse
|
11
|
Orisakwe OE, Ikpeama EU, Orish CN, Ezejiofor AN, Ozoani HA, Okolo KO, Cirovic A, Cirovic A, Nwaogazie IL. African mesquite elicits neuroprotective activity against quaternary metal mixture -induced olfactory bulb-hippocampal oxido-inflammatory stress via nrf2-hmox-1pathway. Nat Prod Res 2024:1-5. [PMID: 39086197 DOI: 10.1080/14786419.2024.2381018] [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: 04/14/2024] [Revised: 06/28/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024]
Abstract
African mesquite AM is widely used as an anti-inflammatory agent in sub-Sahara Africa especially Nigeria. Given its strong anti-inflammatory potency, this study has evaluated the neuroprotective properties of AM in the hippocampus HIP and olfactory bulb OB of rats exposed to Cd, As, Hg, and Pb. Twenty-five albino Sprague Dawley rats were randomly divided into five groups in this experiment. Group 1, the control received only water. Group 2 received heavy metal mixture HMM (PbCl2 (20 mg/kg), CdCl2 (1.61 mg/kg), HgCl2 (0.40 mg/kg), and NaAsO3 (10 mg/kg), for 60 days. Groups 3, 4, and 5 were treated with HMM along with AM at doses of 500, 1000, and 1500 mg/kg, respectively. AM decreased the Cd, As, Hg, and Pb levels in OB and HIP, restored the activities of antioxidants, Hmox-1, reduced the activities of AChE, NRF2 and NFkB and improved histopathology.
Collapse
Affiliation(s)
- Orish E Orisakwe
- African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, Port Harcourt, Choba, Nigeria
- Advanced Research Centre, European University of Lefke, Lefke, Northern Cyprus, Turkey
| | - Evelyn Utomoibor Ikpeama
- World Bank Africa Centre of Excellence in Oilfield Chemicals Research (ACE-CEFOR), University of Port Harcourt, PMB, Port Harcourt, Choba, Nigeria
| | - Chinna N Orish
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Port Harcourt, PMB, Port Harcourt, Choba, Nigeria
| | - Anthonet N Ezejiofor
- African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, Port Harcourt, Choba, Nigeria
| | - Harrison Anezi Ozoani
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Science & Technology, Agbani, Enugu State, Nigeria
| | - Kenneth O Okolo
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Science & Technology, Agbani, Enugu State, Nigeria
| | - Aleksandar Cirovic
- Faculty of Medicine, Institute of Anatomy, University of Belgrade, Belgrade, Serbia
| | - Ana Cirovic
- Faculty of Medicine, Institute of Anatomy, University of Belgrade, Belgrade, Serbia
| | - Ify L Nwaogazie
- World Bank Africa Centre of Excellence in Oilfield Chemicals Research (ACE-CEFOR), University of Port Harcourt, PMB, Port Harcourt, Choba, Nigeria
| |
Collapse
|
12
|
Chen H, Zou Y, Kang X, Yang G, Yang X, Yao Y, Magnuson JT, Cao X, Qiu W, Xu EG, Zheng C. Perfluorooctane Sulfonamide Induced Autotoxic Effects on the Zebrafish Immune System. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024. [PMID: 38976350 DOI: 10.1021/acs.est.4c01153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Perfluorooctane sulfonamide (PFOSA) is an immediate perfluorooctanesulfonate (PFOS) precursor (PreFOS). Previous studies have shown PFOSA to induce stronger toxic responses compared to other perfluorinated compounds (PFCs). However, the specific nature of PFOSA-induced toxicity, whether autonomous or mediated by its metabolite PFOS, has not been fully elucidated. This study systematically investigates the immunomodulatory effects of PFOSA and PFOS in zebrafish (Danio rerio). Exposure to PFOSA compromised the zebrafish's ability to defend against pathogenic infections, as evidenced by increased bacterial adhesion to their skin and reduced levels of the biocidal protein lysozyme (LYSO). Moreover, PFOSA exposure was associated with disruptions in inflammatory markers and immune indicators, along with a decrease in immune cell counts. The findings from this study suggest that the immunotoxicity effects of PFOSA are primarily due to its own toxicity rather than its metabolite PFOS. This conclusion was supported by dose-dependent responses, the severity of observed effects, and multivariate analysis. In addition, our experiments using NF-κB-morpholino knock-down techniques further confirmed the role of the Nuclear factor-κappa B pathway in mediating PFOSA-induced immunotoxicity. In conclusion, this study reveals that PFOSA impairs the immune system in zebrafish through an autotoxic mechanism, providing valuable insights for assessing the ecological risks of PFOSA.
Collapse
Affiliation(s)
- Honghong Chen
- Eastern Institute for Advanced Study, Eastern Institute of Technology, Ningbo 315200, China
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yao Zou
- School of Environment and Energy, South China University of Technology, Guangzhou 510006, China
- Guangdong Society of Environmental Sciences, Guangzhou 510045, China
| | - Xinyuan Kang
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ge Yang
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xin Yang
- Shenzhen Key Laboratory of Precision Measurement and Early Warning Technology for Urban Environmental Health Risks, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yingying Yao
- Institute of Global Environmental Change, Xi'an Jiaotong University, Xi'an 710049, China
| | - Jason T Magnuson
- U.S. Geological Survey, Columbia Environmental Research Center, Columbia, Missouri 65201, United States
| | - Xinde Cao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wenhui Qiu
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Elvis Genbo Xu
- Department of Biology, University of Southern Denmark, Odense 5230, Denmark
| | - Chunmiao Zheng
- Eastern Institute for Advanced Study, Eastern Institute of Technology, Ningbo 315200, China
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
13
|
Kankılıç NA, Küçükler S, Gür C, Akarsu SA, Akaras N, Şimşek H, İleritürk M, Kandemir FM. Naringin protects against paclitaxel-induced toxicity in rat testicular tissues by regulating genes in pro-inflammatory cytokines, oxidative stress, apoptosis, and JNK/MAPK signaling pathways. J Biochem Mol Toxicol 2024; 38:e23751. [PMID: 38879801 DOI: 10.1002/jbt.23751] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 05/18/2024] [Accepted: 05/31/2024] [Indexed: 10/11/2024]
Abstract
Paclitaxel (PTX), which is actively used in the treatment of many types of cancer, has a toxic effect by causing increased oxidative stress in testicular tissues. Naringin (NRG) is a natural flavonoid found in plants, and its antioxidant properties are at the forefront. This study aims to investigate the protective feature of NRG in PTX-induced testicular toxicity. Thirty-five male Sprague rats were divided into five groups: control, NRG, PTX, PTX + NRG50, and PTX + NRG100. Rats were administered PTX (2 mg/kg, BW) intraperitoneally once daily for the first 5 days. Then, between the 6th and 14th days, NRG (50 and 100 mg/kg) was administered orally once a day. NRG reduced PTX-induced lipid peroxidation and increased testicular tissue antioxidant capacity (superoxide dismutase, catalase, glutathione peroxidase, and glutathione). While NRG reduces the mRNA expression levels of nuclear factor kappa B, tumor necrosis factor-alpha, interleukin-1 beta, cyclooxygenase-2, interleukin-6, inducible-nitric oxide synthase, mitogen-activated protein kinase 14 (MAPK)14, MAPK15, c-Jun N-terminal kinase, P53, Apaf1, Caspase3, Caspase6, Caspase9, and Bax in testicular tissues; it caused an increase in Nrf2, HO-1, NQO1 and Bcl-2 levels. NRG also improved the structural and functional integrity of testicular tissue disrupted by PTX. PTX-induced sperm damage was alleviated by NRG. NRG showed a protective effect by alleviating the PTX-induced testicular toxicity by increasing oxidative stress, inflammation, apoptosis, and autophagy.
Collapse
Affiliation(s)
| | - Sefa Küçükler
- Department of Veterinary Biochemistry, Faculty of Veterinary, Atatürk University, Erzurum, Turkey
| | - Cihan Gür
- Department of Medical Laboratory Techniques, Vocational School of Health Services, Atatürk University, Erzurum, Turkey
| | - Serkan Ali Akarsu
- Department of Reproduction and Artificial Insemination, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Nurhan Akaras
- Department of Histology and Embryology, Faculty of Medicine, Aksaray University, Aksaray, Turkey
| | - Hasan Şimşek
- Department of Physiology, Faculty of Medicine, Aksaray University, Aksaray, Turkey
| | - Mustafa İleritürk
- Department of Animal Science, Horasan Vocational College, Atatürk University, Erzurum, Turkey
| | - Fatih Mehmet Kandemir
- Department of Medical Biochemistry, Faculty of Medicine, Aksaray University, Aksaray, Turkey
| |
Collapse
|
14
|
Wen Y, Deng S, Wang B, Zhang F, Luo T, Kuang H, Kuang X, Yuan Y, Huang J, Zhang D. Exposure to polystyrene nanoplastics induces hepatotoxicity involving NRF2-NLRP3 signaling pathway in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 278:116439. [PMID: 38728945 DOI: 10.1016/j.ecoenv.2024.116439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/09/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024]
Abstract
Nanoplastic contamination has been of intense concern by virtue of the potential threat to human and ecosystem health. Animal experiments have indicated that exposure to nanoplastics (NPs) can deposit in the liver and contribute to hepatic injury. To explore the mechanisms of hepatotoxicity induced by polystyrene-NPs (PS-NPs), mice and AML-12 hepatocytes were exposed to different dosages of 20 nm PS-NPs in this study. The results illustrated that in vitro and in vivo exposure to PS-NPs triggered excessive production of reactive oxygen species and repressed nuclear factor erythroid-derived 2-like 2 (NRF2) antioxidant pathway and its downstream antioxidase expression, thus leading to hepatic oxidative stress. Moreover, PS-NPs elevated the levels of NLRP3, IL-1β and caspase-1 expression, along with an activation of NF-κB, suggesting that PS-NPs induced hepatocellular inflammatory injury. Nevertheless, the activaton of NRF2 signaling by tert-butylhydroquinone mitigated PS-NPs-caused oxidative stress and inflammation, and inbihited NLRP3 and caspase-1 expression. Conversely, the rescuing effect of NRF2 signal activation was dramatically supressed by treatment with NRF2 inhibitor brusatol. In summary, our results demonstrated that NRF2-NLRP3 pathway is involved in PS-NPs-aroused hepatotoxicity, and the activation of NRF2 signaling can protect against PS-NPs-evoked liver injury. These results provide novel insights into the hepatotoxicity elicited by NPs exposure.
Collapse
Affiliation(s)
- Yiqian Wen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Shiyi Deng
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Binhui Wang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Fan Zhang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Tao Luo
- Institute of Life Science and School of Life Science, Nanchang University, Nanchang 330031, China
| | - Haibin Kuang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Xiaodong Kuang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Yangyang Yuan
- Clinical Medical Experimental Center of Nanchang University, Nanchang 330031, China
| | - Jian Huang
- Clinical Medical Experimental Center of Nanchang University, Nanchang 330031, China
| | - Dalei Zhang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang 330006, China.
| |
Collapse
|
15
|
Zhang S, Liu Z, Zhang H, Zhou X, Wang X, Chen Y, Miao X, Zhu Y, Jiang W. Effect and mechanism of Qing Gan Zi Shen decoction on heart damage induced by obesity and hypertension. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117163. [PMID: 37741474 DOI: 10.1016/j.jep.2023.117163] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/15/2023] [Accepted: 09/07/2023] [Indexed: 09/25/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qing Gan Zi Shen Decoction (QGZS) is a traditional Chinese formula. It has been extensively used for decades in the treatment of hypertension combined with metabolic diseases, but its cardioprotective effects and underlying mechanisms are poorly understood. AIM OF THE STUDY To explore the cardioprotective effects and potential mechanisms of QGZS in an animal model of obese hypertension. MATERIALS AND METHODS In this study, spontaneously hypertensive rats (SHRs) were utilized as an animal model to examine the effects of a high-fat diet and two concentrations of QGZS. Echocardiography, hematoxylin eosin (H&E) staining, and wheat germ agglutinin (WGA) staining were employed to assess the cardiac structure and function of the SHRs throughout a 16-week therapy period. Furthermore, Western blotting (WB) and immunofluorescence (IF) were employed to identify the levels of Nrf2 expression in the mitochondria, cytoplasm, and nucleus of the myocardium. Additionally, transmission electron microscopy and enzyme-linked immunosorbent assay (ELISA) were utilized to measure mitochondrial morphology and pro-inflammatory cytokine levels, respectively. Furthermore, Western blotting (WB), immunohistochemistry (IHC), and immunofluorescence (IF) techniques were employed to quantify the levels of marker proteins associated with myocardial fibrosis, cardiac inflammation, oxidative stress, and mitochondrial dysfunction. RESULTS QGZS inhibited weight gain and depressed systolic and mean arterial pressures in high-fat-fed SHRs. Echocardiographic results demonstrated that QGZS prevented the increase in left ventricular mass, restricted the growth of left ventricular diameter, and improved ejection fraction (EF), fractional shortening (FS), and the ratio of early diastolic peak velocity of transmitral flow (E) to late diastolic peak velocity (A) in high-fat-fed SHRs. This suggested that QGZS prevented ventricular remodeling and protected cardiac systolic and diastolic functions. H&E and WGA staining showed that QGZS improved cardiomyocyte disorders and restricted cardiomyocyte hypertrophy. The underlying mechanisms, QGZS attenuated the oxidative stress state, including reducing the generation of reactive oxygen species (ROS) in the myocardium, revitalizing the antioxidant enzyme system, and protecting mitochondrial function. Moreover, QGZS alleviated the pro-inflammatory state in high-fat-fed SHRs. What's more, QGZS significantly increased the expression level of Nrf2 in nuclei and mitochondria in rat heart tissues, exerting a proximate Nrf2 agonist effect. CONCLUSIONS QGZS exerted cardioprotective effects, in part due to its increasing expression of Nrf2 protein in the heart, which promoted Nrf2 nuclear expression.
Collapse
Affiliation(s)
- Shujie Zhang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Zitian Liu
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Han Zhang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Xiaonian Zhou
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Xiuming Wang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Yan Chen
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Xiaofan Miao
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Yao Zhu
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China.
| | - Weimin Jiang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China.
| |
Collapse
|
16
|
Wang Z, Zhou Y, Xiao X, Liu A, Wang S, Preston RJS, Zaytseva YY, He G, Xiao W, Hennig B, Deng P. Inflammation and cardiometabolic diseases induced by persistent organic pollutants and nutritional interventions: Effects of multi-organ interactions. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 339:122756. [PMID: 37844865 PMCID: PMC10842216 DOI: 10.1016/j.envpol.2023.122756] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
The development and outcome of inflammatory diseases are associated with genetic and lifestyle factors, which include chemical and nonchemical stressors. Persistent organic pollutants (POPs) are major groups of chemical stressors. For example, dioxin-like polychlorinated biphenyls (PCBs), per- and polyfluoroalkyl substances (PFASs), and polybrominated diphenyl ethers (PBDEs) are closely associated with the incidence of inflammatory diseases. The pathology of environmental chemical-mediated inflammatory diseases is complex and may involve disturbances in multiple organs, including the gut, liver, brain, vascular tissues, and immune systems. Recent studies suggested that diet-derived nutrients (e.g., phytochemicals, vitamins, unsaturated fatty acids, dietary fibers) could modulate environmental insults and affect disease development, progression, and outcome. In this article, mechanisms of environmental pollutant-induced inflammation and cardiometabolic diseases are reviewed, focusing on multi-organ interplays and highlighting recent advances in nutritional strategies to improve the outcome of cardiometabolic diseases associated with environmental exposures. In addition, advanced system biology approaches are discussed, which present unique opportunities to unveil the complex interactions among multiple organs and to fuel the development of precision intervention strategies in exposed individuals.
Collapse
Affiliation(s)
- Zhongmin Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China; Irish Centre for Vascular Biology, School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, Ireland
| | - Yixuan Zhou
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Xia Xiao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Aowen Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Shengnan Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Roger J S Preston
- Irish Centre for Vascular Biology, School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, Ireland
| | - Yekaterina Y Zaytseva
- Superfund Research Center, University of Kentucky, Lexington, KY, USA; Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Guangzhao He
- Department of Pharmacy, Changzhou Cancer Hospital, Soochow University, Changzhou, Jiangsu, China
| | - Wenjin Xiao
- Department of Endocrinology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Bernhard Hennig
- Superfund Research Center, University of Kentucky, Lexington, KY, USA; Department of Animal and Food Sciences, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY, USA
| | - Pan Deng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
17
|
Mohamed EE, Ahmed OM, Zoheir KMA, El-Shahawy AAG, Tamur S, Shams A, Burcher JT, Bishayee A, Abdel-Moneim A. Naringin-Dextrin Nanocomposite Abates Diethylnitrosamine/Acetylaminofluorene-Induced Lung Carcinogenesis by Modulating Oxidative Stress, Inflammation, Apoptosis, and Cell Proliferation. Cancers (Basel) 2023; 15:5102. [PMID: 37894468 PMCID: PMC10605195 DOI: 10.3390/cancers15205102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/29/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Nanotechnology has proven advantageous in numerous scientific applications, one being to enhance the delivery of chemotherapeutic agents. This present study aims to evaluate the mechanisms underlying the chemopreventive action of naringin-dextrin nanocomposites (Nar-Dx-NCs) against diethylnitrosamine (DEN)/2-acetylaminofluorene (2AAF)-induced lung carcinogenesis in male Wistar rats. DEN was administered intraperitoneally (i.p.) (150 mg/kg/week) for two weeks, followed by the oral administration of 2AAF (20 mg/kg) four times a week for three weeks. Rats receiving DEN/2AAF were concurrently treated with naringin or Nar-Dx-NCs orally at a dose of 10 mg/kg every other day for 24 weeks. Naringin and Nar-Dx-NCs treatments prevented the formation of tumorigenic cells within the alveoli of rats exposed to DEN/2AAF. These findings were associated with a significant decrease in lipid peroxidation, upregulation of antioxidant enzyme (glutathione peroxidase and superoxide dismutase) activity, and enhanced glutathione and nuclear factor erythroid 2-related factor 2 expression in the lungs. Naringin and Nar-Dx-NCs exerted anti-inflammatory actions manifested by a decrease in lung protein expression of tumor necrosis factor-α and interleukin-1β and mRNA expression of interleukin-6, interferon-γ, nuclear factor-κB, and inducible nitric oxide synthase, with a concurrent increase in interleukin-10 expression. The anti-inflammatory effect of Nar-Dx-NCs was more potent than naringin. Regarding the effect on apoptosis, both naringin and Nar-Dx-NCs significantly reduced Bcl-2 and increased Bax and P53 expressions. Moreover, naringin or Nar-Dx-NCs induced a significant decrease in the expression of the proliferator marker, Ki-67, and the effect of Nar-Dx-NCs was more marked. In conclusion, Nar-Dx-NCs improved naringin's preventive action against DEN/2AAF-induced lung cancer and exerted anticarcinogenic effects by suppressing oxidative stress and inflammation and improving apoptotic signal induction and propagation.
Collapse
Affiliation(s)
- Eman E. Mohamed
- Physiology Division, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt; (E.E.M.)
| | - Osama M. Ahmed
- Physiology Division, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt; (E.E.M.)
| | - Khairy M. A. Zoheir
- Cell Biology Department, Biotechnology Research Institute, National Research Centre, Cairo 12622, Egypt;
| | - Ahmed A. G. El-Shahawy
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef 62521, Egypt;
| | - Shadi Tamur
- Departement of Pediatrics, College of Medicine, Taif University, Taif 21944, Saudi Arabia;
| | - Anwar Shams
- Departement of Pharmacology, College of Medicine, Taif University, Taif 21944, Saudi Arabia
- Centre of Biomedical Sciences Research, Deanship of Scientific Research, Taif University, Taif 21974, Saudi Arabia
- High Altitude Research Center, Taif University, Taif 21944, Saudi Arabia
| | - Jack T. Burcher
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA; (J.T.B.); or (A.B.)
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA; (J.T.B.); or (A.B.)
| | - Adel Abdel-Moneim
- Physiology Division, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt; (E.E.M.)
| |
Collapse
|
18
|
Gao S, Chen X, Yu Z, Du R, Chen B, Wang Y, Cai X, Xu J, Chen J, Duan H, Cai Y, Zheng G. Progress of research on the role of active ingredients of Citri Reticulatae Pericarpium in liver injury. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 115:154836. [PMID: 37119760 DOI: 10.1016/j.phymed.2023.154836] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/01/2023] [Accepted: 04/18/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND Liver is a vital organ responsible for metabolizing and detoxifying both endogenous and exogenous substances in the body. However, it is susceptible to damage from chemical and natural toxins. The high incidence and mortality rates of liver disease and its associated complications impose a significant economic burden and survival pressure on patients and their families. Various liver diseases exist, including cholestasis, viral and non-viral hepatitis, fatty liver disease, drug-induced liver injury, alcoholic liver injury, and severe end-stage liver diseases such as cirrhosis, hepatocellular carcinoma (HCC), and cholangiocellular carcinoma (CCA). Recent research has shown that flavonoids found in Citri Reticulatae Pericarpium (CRP) have the potential to normalize blood glucose, cholesterol levels, and liver lipid levels. Additionally, these flavonoids exhibit anti-inflammatory properties, prevent oxidation and lipid peroxidation, and reduce liver toxicity, thereby preventing liver injury. Given these promising findings, it is essential to explore the potential of active components in CRP for developing new drugs to treat liver diseases. OBJECTIVE Recent studies have revealed that flavonoids, including hesperidin (HD), hesperetin (HT), naringenin (NIN), nobiletin (NOB), naringin (NRG), tangerine (TN), and erodcyol (ED), are the primary bioactive components in CRP. These flavonoids exhibit various therapeutic effects on liver injury, including anti-oxidative stress, anti-cytotoxicity, anti-inflammatory, anti-fibrosis, and anti-tumor mechanisms. In this review, we have summarized the research progress on the hepatoprotective effects of HD, HT, NIN, NOB, NRG, TN, ED and limonene (LIM), highlighting their underlying molecular mechanisms. Despite their promising effects, the current clinical application of these active ingredients in CRP has some limitations. Therefore, further studies are needed to explore the full potential of these flavonoids and develop new therapeutic strategies for liver diseases. METHODS For this review, we conducted a systematic search of three databases (ScienceNet, PubMed, and Science Direct) up to July 2022, using the search terms "CRP active ingredient," "liver injury," and "flavonoids." The search data followed the PRISMA standard. RESULTS Our findings indicate that flavonoids found in CRP can effectively reduce drug-induced liver injury, alcoholic liver injury, and non-alcoholic liver injury. These therapeutic effects are mainly attributed to the ability of flavonoids to improve liver resistance to oxidative stress and inflammation while normalizing cholesterol and liver lipid levels by exhibiting anti-free radical and anti-lipid peroxidation properties. CONCLUSION Our review provides new insights into the potential of active components in CRP for preventing and treating liver injury by regulating various molecular targets within different cell signaling pathways. This information can aid in the development of novel therapeutic strategies for liver disease.
Collapse
Affiliation(s)
- Shuhan Gao
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xiaojing Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhiqian Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Rong Du
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Baizhong Chen
- Guangdong Xinbaotang Biological Technology Co., Ltd, Guangdong Jiangmen, 529000, China
| | - Yuxin Wang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xiaoting Cai
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jiepei Xu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jiamin Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Huiying Duan
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yi Cai
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Guodong Zheng
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
19
|
Zhang L, Louie A, Rigutto G, Guo H, Zhao Y, Ahn S, Dahlberg S, Sholinbeck M, Smith MT. A systematic evidence map of chronic inflammation and immunosuppression related to per- and polyfluoroalkyl substance (PFAS) exposure. ENVIRONMENTAL RESEARCH 2023; 220:115188. [PMID: 36592815 PMCID: PMC10044447 DOI: 10.1016/j.envres.2022.115188] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 06/01/2023]
Abstract
BACKGROUND The ability to induce chronic inflammation and immunosuppression are two key characteristics of carcinogens and important forms of immunotoxicity. The National Toxicology Program (NTP) evaluated the immunotoxicity of two per- and polyfluoroalkyl substances (PFASs), PFOA (perfluorooctanoic acid) and PFOS (perfluorooctane sulfonate), in 2016. However, the potential pro-inflammatory and immunosuppressive effects of other PFASs remain largely uncharacterized. METHODS We developed an expanded set of search terms pertaining to the chronic inflammatory and immunosuppressive effects of PFASs based on those of the International Agency for Research on Cancer (IARC) and NTP. To confirm searching effectiveness and scope, we compared our search term results with those of IARC and NTP for both PFASs and two other known carcinogens, chromium (VI) and benzene. Systematic evidence maps (SEMs) were also produced using Tableau to visualize the distribution of study numbers and types reporting immunotoxic effects and specific biomarkers elicited by PFAS exposures. RESULTS In total, 1155 PFAS studies were retrieved, of which 321 qualified for inclusion in our dataset. Using our search terms, we identified a greater number of relevant studies than those obtained using IARC and NTP's search terms. From the SEM findings, increased cytokine production strengthened an association between PFAS exposure and chronic inflammation, and decreased B-cell activation and altered levels of T-cell subtypes and immunoglobulins confirmed PFAS-induced immunosuppression. CONCLUSION Our SEM findings confirm that several PFASs commonly found in both in the environment, including those that are lesser-known, may induce immunosuppression and chronic inflammation, two key characteristics of carcinogens. This approach, including development of search terms, study screening process, data coding, and evidence mapping visualizations, can be applied to other key characteristics of chemical carcinogens.
Collapse
Affiliation(s)
- Luoping Zhang
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, 94720, USA.
| | - Allen Louie
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, 94720, USA; Molecular Toxicology Interdepartmental Program, Fielding School of Public Health, University of California, Los Angeles, CA, 90095, USA
| | - Gabrielle Rigutto
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, 94720, USA
| | - Helen Guo
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, 94720, USA
| | - Yun Zhao
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, 94720, USA
| | - Stacy Ahn
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, 94720, USA
| | - Sarah Dahlberg
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, 94720, USA
| | - Michael Sholinbeck
- Bioscience, Natural Resources & Public Health Library, University of California, Berkeley, CA, 94720, USA
| | - Martyn T Smith
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, 94720, USA
| |
Collapse
|
20
|
Ehrlich V, Bil W, Vandebriel R, Granum B, Luijten M, Lindeman B, Grandjean P, Kaiser AM, Hauzenberger I, Hartmann C, Gundacker C, Uhl M. Consideration of pathways for immunotoxicity of per- and polyfluoroalkyl substances (PFAS). Environ Health 2023; 22:19. [PMID: 36814257 PMCID: PMC9944481 DOI: 10.1186/s12940-022-00958-5] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/30/2022] [Indexed: 05/02/2023]
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS) are of public health concern, because of their ubiquitous and extremely persistent occurrence, and depending on their structure, their bio-accumulative, mobile and toxic properties. Human health effects associated with exposure to PFAS include adverse effects on the immune system. In 2020, EFSA (the European Food Safety Authority) defined adverse effects on the immune system as the most critical effect for human health risk assessment, based on reduced antibody responses to childhood vaccines and similar effects observed in experimental animal studies. Likewise, the U.S. EPA (Environmental Protection Agency) considers PFAS-induced immunotoxicity, especially in children, as the critical effect for risk assessment. However, the mechanisms by which antibody concentrations are impacted are not completely understood. Furthermore, other targets of the immune system functions have been reported in the literature. OBJECTIVE The aim of this review is to explore PFAS-associated immune-related effects. This includes, relevant mechanisms that may underlie the observed effects on the immune system, immunosuppression as well as immunoenhancement, such as i) modulation of cell signalling and nuclear receptors, such as NF-κB and PPARs; ii) alteration of calcium signalling and homoeostasis in immune cells; iii) modulation of immune cell populations; iv) oxidative stress and v) impact on fatty acid metabolism & secondary effects on the immune system. METHODS A literature research was conducted using three databases (Web of Science, PubMed, and Scopus), which were searched in July 2021 for relevant studies published in the time frame from 2018 to 2021. In total, 487 publications were identified as potentially eligible and following expert-based judgement, articles relevant for mechanisms of PFAS induced immunotoxicity are discussed. CONCLUSIONS Taken together, we show that there is substantial evidence from both in vitro and in vivo experimental as well as epidemiological studies, supporting that various PFAS, not only PFOA and PFOS, affect multiple aspects of the immune system. Timing of exposure is critical, because the developing immune system is especially vulnerable to toxic insults, resulting in a higher risk of particularly adverse immune effects but also other organs later in life.
Collapse
Affiliation(s)
- Veronika Ehrlich
- Environment Agency Austria (Umweltbundesamt GmbH), Spittelauer Lände 5, 1090, Vienna, Austria
| | - Wieneke Bil
- Centre for Safety of Substances and Products, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Rob Vandebriel
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Berit Granum
- Division of Climate and Environment Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Mirjam Luijten
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Birgitte Lindeman
- Division of Climate and Environment Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Philippe Grandjean
- Department of Public Health, University of Southern Denmark, Odense, Denmark
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - Andreas-Marius Kaiser
- Environment Agency Austria (Umweltbundesamt GmbH), Spittelauer Lände 5, 1090, Vienna, Austria
| | - Ingrid Hauzenberger
- Environment Agency Austria (Umweltbundesamt GmbH), Spittelauer Lände 5, 1090, Vienna, Austria
| | - Christina Hartmann
- Environment Agency Austria (Umweltbundesamt GmbH), Spittelauer Lände 5, 1090, Vienna, Austria
| | - Claudia Gundacker
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Maria Uhl
- Environment Agency Austria (Umweltbundesamt GmbH), Spittelauer Lände 5, 1090, Vienna, Austria.
| |
Collapse
|
21
|
Liu Z, Lin H, Zheng Y, Feng Y, Shi C, Zhu R, Shen X, Han Y, Zhang H, Zhong Y. Perfluorooctanoic acid and perfluorooctanesulfonic acid induce immunotoxicity through the NF-κB pathway in black-spotted frog (Rana nigromaculata). CHEMOSPHERE 2023; 313:137622. [PMID: 36565765 DOI: 10.1016/j.chemosphere.2022.137622] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/18/2022] [Accepted: 12/20/2022] [Indexed: 06/17/2023]
Abstract
Perfluorooctanoic acid (PFOA) and perfluorooctanesulfonic acid (PFOS) are widely detected in the environment and wild animals, thus posing a threat to wildlife and public health; however, knowledge about their immunotoxicity and the underlying mechanism remains limited. In the present study, male black-spotted frogs (Rana nigromaculata) were exposed to environmentally relevant concentrations (0, 1, and 10 μg/L) of PFOA or PFOS for 21 days; subsequently, biochemical analysis, molecular docking, and gene expression determination were conducted. The results indicated that exposure to 10 μg/L PFOA decreased the serum levels of immunoglobulin A. PFOS exposure significantly increased the hepatic levels of interleukin-1β, interleukin-6, tumor necrosis factor-α, interferon-γ, and nitric oxide; but PFOA significantly increased the levels of only tumor necrosis factor-α. Furthermore, PFOA and PFOS exposure significantly decreased the activity of inducible nitric oxide synthase and total nitric oxide synthase. IBRv2 analysis indicated that PFOA and PFOS had a similar effect on these immune indicators, but PFOS was more toxic than PFOA. Molecular docking revealed that PFOA and PFOS can bind to nuclear factor-κB (NF-κB) by forming stable hydrogen bonds. PFOA and PFOS exposure upregulated the gene expression of NF-κB and its downstream genes. Significant correlations between the expression of genes involved in the NF-κB pathway and immune-related indicators suggests that PFOA- and PFOS-induced immunotoxicity was associated with the activation of NF-κB. Our findings provide novel insights into the potential role of NF-κB in immunotoxicity induced by PFOA and PFOS in frogs.
Collapse
Affiliation(s)
- Zhiquan Liu
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, China; School of Engineering, Hangzhou Normal University, Hangzhou, 310018, China
| | - Huikang Lin
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, China
| | - Yueyue Zheng
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, China
| | - Yixuan Feng
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, China
| | - Chaoli Shi
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, China
| | - Ruoxin Zhu
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, China
| | - Xingyao Shen
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, China
| | - Yu Han
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, China; School of Engineering, Hangzhou Normal University, Hangzhou, 310018, China
| | - Hangjun Zhang
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, China; School of Engineering, Hangzhou Normal University, Hangzhou, 310018, China
| | - Yuchi Zhong
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, China; School of Engineering, Hangzhou Normal University, Hangzhou, 310018, China.
| |
Collapse
|
22
|
Chen Y, Wang Y, Cui Z, Liu W, Liu B, Zeng Q, Zhao X, Dou J, Cao J. Endocrine disrupting chemicals: A promoter of non-alcoholic fatty liver disease. Front Public Health 2023; 11:1154837. [PMID: 37033031 PMCID: PMC10075363 DOI: 10.3389/fpubh.2023.1154837] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent liver disorder. With the improvement in human living standards, the prevalence of NAFLD has been increasing in recent years. Endocrine-disrupting chemicals (EDCs) are a class of exogenous chemicals that simulate the effects of hormones in the body. There has been growing evidence regarding the potential effects of EDCs on liver health, especially in NAFLD. This paper aims to summarize the major EDCs that contribute to the growing burden of NAFLD and to raise public awareness regarding the hazards posed by EDCs with the objective of reducing the incidence of NAFLD.
Collapse
|
23
|
Pengnet S, Sumarithum P, Phongnu N, Prommaouan S, Kantip N, Phoungpetchara I, Malakul W. Naringin attenuates fructose-induced NAFLD progression in rats through reducing endogenous triglyceride synthesis and activating the Nrf2/HO-1 pathway. Front Pharmacol 2022; 13:1049818. [PMID: 36588703 PMCID: PMC9797507 DOI: 10.3389/fphar.2022.1049818] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Background: Excessive fructose consumption causes hepatic lipid accumulation via increased triglyceride (TG) synthesis, leading to the development and progression of non-alcoholic fatty liver disease (NALFD). Naringin, a flavanone glycoside found in citrus fruit, has antioxidant and hypolipidemic properties. Therefore, the aim of this study was to investigate the effect of naringin on fructose-induced NAFLD in rats and the possible underlying mechanism. Methods: Male Sprague Dawley rats were given 10% (w/v) fructose in drinking water for 12 weeks. Naringin (100 mg/kg/day) was administered orally to rats for the last 4 weeks of fructose overload. After 12 weeks of treatment, the hepatic lipid content was determined. In addition, the expression of proteins involved in de novo lipogenesis (DNL) and TG synthesis as well as antioxidant and inflammatory mediators in the liver were examined by western blot analysis. Results: Treatment of fructose-fed rats with naringin significantly decreased the hepatic TG and cholesterol content as well as serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) activities. Naringin treatment also decreased the hepatic expression of carbohydrate response element binding protein (ChREBP), sterol regulatory element-binding protein-1c (SREBP-1c) and nuclear SREBP-1c (nSREBP-1c) as well as enzymes involved in DNL (acetyl CoA carboxylase [ACC] and fatty acid synthase [FAS]) and an enzyme involved in TG synthesis (glycerol-3-phosphate acyltransferase 1 [GPAT-1] and diacylglycerol acyltransferase2 [DGAT2]) in fructose-fed rats. In addition, naringin induced a significant decrease in the hepatic expression of nuclear factor kappa B (NF-κB) and tumor necrosis factor α (TNF-α). Furthermore, naringin administration restored the expression of the antioxidant mediators nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and heme oxygenase-1 (HO-1) in the liver of fructose-fed rats. Conclusion: These results demonstrate that oral administration of naringin protects against fructose-induced hepatic steatosis by decreasing DNL and TG synthesis. In addition, naringin could prevent NAFLD progression via targeting the Nrf2/HO-1 and the NF-κB/TNF-α pathways.
Collapse
Affiliation(s)
- Sirinat Pengnet
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao, Thailand
| | - Phinsuda Sumarithum
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Nuttaphong Phongnu
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Sakdina Prommaouan
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Napapas Kantip
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Ittipon Phoungpetchara
- Department of Anatomy, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Wachirawadee Malakul
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand,Centre of Excellence in Medical Biotechnology, Naresuan University, Phitsanulok, Thailand,*Correspondence: Wachirawadee Malakul,
| |
Collapse
|
24
|
Mohamed EE, Ahmed OM, Abdel-Moneim A, Zoheir KMA, Elesawy BH, Al Askary A, Hassaballa A, El-Shahawy AAG. Protective Effects of Naringin-Dextrin Nanoformula against Chemically Induced Hepatocellular Carcinoma in Wistar Rats: Roles of Oxidative Stress, Inflammation, Cell Apoptosis, and Proliferation. Pharmaceuticals (Basel) 2022; 15:1558. [PMID: 36559011 PMCID: PMC9786090 DOI: 10.3390/ph15121558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Nanotechnology holds great promise for the development of treatments for deadly human diseases, such as hepatocellular carcinoma (HCC). In the current study, we compared the hepatoprotective effects of naringin-dextrin nanoparticles (NDNPs) against HCC in male Wistar rats with those of pure naringin and investigated the underlying cellular and molecular mechanisms. HCC was induced by intraperitoneal injection of diethylnitrosamine (DEN, 150 mg/kg body weight (b.w.) per week) for two weeks, followed by oral administration of 2-acetylaminofluorene (2AAF, 20 mg/kg b.w.) four times per week for three weeks. DEN/2AAF-administered rats were divided into three groups that respectively received 1% carboxymethyl cellulose (as vehicle), 10 mg/kg b.w. naringin, or 10 mg/kg b.w. NDNP every other day by oral gavage for 24 weeks. Both naringin and NDNP significantly attenuated the harmful effects of DEN on liver function. Both compounds also suppressed tumorigenesis as indicated by the reduced serum concentrations of liver tumor markers, and this antitumor effect was confirmed by histopathological evaluation. Additionally, naringin and NDNP prevented DEN-induced changes in hepatic oxidative stress and antioxidant activities. In addition, naringin and NDNP suppressed inflammation induced by DEN. Moreover, naringin and NDNP significantly reduced the hepatic expression of Bcl-2 and increased Bax, p53, and PDCD5 expressions. Naringin and NDNP also reduced expression of IQGAP1, IQGAP3, Ras signaling, and Ki-67 while increasing expression of IQGAP2. Notably, NDNP more effectively mitigated oxidative stress and inflammatory signaling than free naringin and demonstrated improved antitumor efficacy, suggesting that this nanoformulation improves bioavailability within nascent tumor sites.
Collapse
Affiliation(s)
- Eman E. Mohamed
- Physiology Division, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef 2722165, Egypt
| | - Osama M. Ahmed
- Physiology Division, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef 2722165, Egypt
| | - Adel Abdel-Moneim
- Physiology Division, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef 2722165, Egypt
| | - Khairy M. A. Zoheir
- Cell Biology Department, Biotechnology Research Institute, National Research Centre, Cairo 12622, Egypt
| | - Basem H. Elesawy
- Department of Pathology, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ahmad Al Askary
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ahmed Hassaballa
- Nutrition and Food Science, College of Liberal Arts and Sciences, Wayne State University, Detroit, MI 48202, USA
- ZeroHarm L.C., Farmington Hills, Farmington, MI 48333, USA
| | - Ahmed A. G. El-Shahawy
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 2722165, Egypt
| |
Collapse
|
25
|
Wu Q, Yu P, Bi Y, Li Z, Guo W, Chen Y, Duan Z. Naringin regulates mitochondrial dynamics to protect against acetaminophen-induced hepatotoxicity by activating the AMPK/Nrf2 signaling pathway in vitro. Braz J Med Biol Res 2022; 55:e12040. [PMID: 36259797 PMCID: PMC9578698 DOI: 10.1590/1414-431x2022e12040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 09/09/2022] [Indexed: 11/22/2022] Open
Abstract
Naringin (Nar) has been reported to exert potential hepatoprotective effects against acetaminophen (APAP)-induced injury. Mitochondrial dysfunction plays an important role in APAP-induced liver injury. However, the protective mechanism of Nar against mitochondrial damage has not been elucidated. Therefore, the aim of this study was to investigate the hepatoprotective effects of Nar against APAP and the possible mechanisms of actions. Primary rat hepatocytes and HepG2 cells were utilized to establish an in vitro model of APAP-induced hepatotoxicity. The effect of APAP and Nar on cell viability was evaluated by a CCK8 assay and detection of the concentrations of alanine aminotransferase, aspartate aminotransferase, and lactate dehydrogenase. The cellular concentrations of biomarkers of oxidative stress were measured by ELISA. The mRNA expression levels of APAP-related phase II enzymes were determined by real-time PCR. The protein levels of Nrf2, phospho (p)-AMPK/AMPK, and biomarkers of mitochondrial dynamics were determined by western blot analysis. The mitochondrial membrane potential (MMP) was measured by high-content analysis and confocal microscopy. JC-1 staining was performed to evaluate mitochondrial depolarization. Nar pretreatment notably prevented the marked APAP-induced hepatocyte injury, increases in oxidative stress marker expression, reductions in the expression of phase II enzymes, significant loss of MMP, mitochondrial depolarization, and mitochondrial fission in vitro. In conclusion, Nar alleviated APAP-induced hepatocyte and mitochondrial injury by activating the AMPK/Nrf2 pathway to reduce oxidative stress in vitro. Applying Nar for the treatment of APAP-induced liver injury might be promising.
Collapse
Affiliation(s)
- Qiao Wu
- Infection Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Pengfei Yu
- Fourth Department of Liver Disease (Difficult & Complicated Liver Diseases and Artificial Liver Center), Beijing You'an Hospital Affiliated to Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing, China
| | - Yanzhen Bi
- Fourth Department of Liver Disease (Difficult & Complicated Liver Diseases and Artificial Liver Center), Beijing You'an Hospital Affiliated to Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing, China
| | - Zhijie Li
- Hepatobiliary Surgery Center, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Wei Guo
- Infection Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yu Chen
- Fourth Department of Liver Disease (Difficult & Complicated Liver Diseases and Artificial Liver Center), Beijing You'an Hospital Affiliated to Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing, China
| | - Zhongping Duan
- Fourth Department of Liver Disease (Difficult & Complicated Liver Diseases and Artificial Liver Center), Beijing You'an Hospital Affiliated to Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing, China
| |
Collapse
|
26
|
Exploration of the Protective Mechanism of Naringin in the Acetaminophen-Induced Hepatic Injury by Metabolomics. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7138194. [PMID: 36160708 PMCID: PMC9507767 DOI: 10.1155/2022/7138194] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/23/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022]
Abstract
Naringin is a dihydroflavone which was found in citrus fruits. Previous studies have indicated the antiapoptotic, antioxidative stress, and anti-inflammatory effects of naringin. It can improve many common diseases, including fibrosis or hepatotoxicity, cardiovascular disease, and diabetes. Acetaminophen (APAP) is a frequently used painkiller, and hepatotoxic side effects limit its use. The purpose of the current examination is to find the impact of naringin on APAP-induced hepatic injury. Firstly, we pretreated mice model groups with naringin. Then, the liver injury model was established by injecting intraperitoneally into mice with APAP. After the mice were euthanized, we obtained serum and liver tissue samples from the mice. Finally, these samples were analyzed using a metabolomics approach to find the underlying mechanism of the effects of naringin on APAP-induced liver injury and provide a new treatment strategy for APAP-induced liver injury. Our data indicate that naringin significantly improves APAP-induced liver injury in mice and reduces the expression levels of liver injury markers in a dose-dependent manner. Furthermore, analysis of differential metabolites in mice with liver injury showed that naringin reduced APAP-induced hepatotoxicity due to reversing multiple metabolite expression levels and the rescue of energy, amino acid, and purine metabolism.
Collapse
|
27
|
Chen Q, Sun S, Mei C, Zhao J, Zhang H, Wang G, Chen W. Capabilities of bio-binding, antioxidant and intestinal environmental repair jointly determine the ability of lactic acid bacteria to mitigate perfluorooctane sulfonate toxicity. ENVIRONMENT INTERNATIONAL 2022; 166:107388. [PMID: 35809485 DOI: 10.1016/j.envint.2022.107388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/26/2022] [Accepted: 06/29/2022] [Indexed: 06/15/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a novel environmental contaminant that can be enriched in humans through the food chain, causing liver diseases, neurotoxicity and metabolic disorders. Lactic acid bacteria (LAB) are safe food-grade microorganisms that exhibit high antioxidant activity and bio-binding capacity towards toxins. Here, strains of LAB with different PFOS binding capacities and antioxidant activities were selected and analyzed for their ability in mitigating the toxic effects of PFOS. The results showed that the PFOS binding capacity and antioxidant activity of LAB largely influenced their ability in alleviating the toxic effects of PFOS. Notably, the individual LAB strains with low PFOS binding capacities and antioxidant activities also attenuated the toxic effects of PFOS, which was shown to up-regulate the contents of short-chain fatty acids (SCFAs) in the cecum and of tight junction proteins in the intestines of mice. Therefore, the mitigation pathway of PFOS-induced toxic damage by LAB is not limited to bio-binding and antioxidant. Repairing the gut environment damaged by PFOS is also essential for LAB to alleviate the toxic damage due to PFOS and may be partly independent of the bio-binding and antioxidant. Therefore, LAB as an alternative pathway for alleviating PFOS toxicity is suggested.
Collapse
Affiliation(s)
- Qian Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China
| | - Shanshan Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China
| | - Chunxia Mei
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, PR China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, PR China; Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi 214122, PR China
| | - Gang Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, PR China.
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China
| |
Collapse
|
28
|
Progress in the treatment of drug-induced liver injury with natural products. Pharmacol Res 2022; 183:106361. [PMID: 35882295 DOI: 10.1016/j.phrs.2022.106361] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 12/13/2022]
Abstract
There are numerous prescription drugs and non-prescription drugs that cause drug-induced liver injury (DILI), which is the main cause of liver disease in humans around the globe. Its mechanism becomes clearer as the disease is studied further. For an instance, when acetaminophen (APAP) is taken in excess, it produces N-acetyl-p-benzoquinone imine (NAPQI) that binds to biomacromolecules in the liver causing liver injury. Treatment of DILI with traditional Chinese medicine (TCM) has shown to be effective. For example, activation of the Nrf2 signaling pathway as well as regulation of glutathione (GSH) synthesis, coupling, and excretion are the mechanisms by which ginsenoside Rg1 (Rg1) treats APAP-induced acute liver injury. Nevertheless, reducing the toxicity of TCM in treating DILI is still a problem to be overcome at present and in the future. Accumulated evidences show that hydrogel-based nanocomposite may be an excellent carrier for TCM. Therefore, we reviewed TCM with potential anti-DILI, focusing on the signaling pathway of these drugs' anti-DILI effect, as well as the possibility and prospect of treating DILI by TCM based on hydrogel materials in the future. In conclusion, this review provides new insights to further explore TCM in the treatment of DILI.
Collapse
|
29
|
Yousuf S, Shabir S, Singh MP. Protection Against Drug-Induced Liver Injuries Through Nutraceuticals via Amelioration of Nrf-2 Signaling. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2022; 42:495-515. [PMID: 35771985 DOI: 10.1080/27697061.2022.2089403] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hepatotoxicity caused by the overdose of various medications is a leading cause of drug-induced liver injury. Overdose of drugs causes hepatocellular necrosis. Nutraceuticals are reported to prevent drug-induced liver failure. The present article aims to review the protection provided by various medicinal plants against hepatotoxic drugs. Ayurveda is considered a conventional restorative arrangement in India. It is consistently used for ages and is still used today to cure drug-induced hepatotoxicity by focusing on antioxidant stress response pathways such as the nuclear factor erythroid-2 (Nrf-2) antioxidant response element signaling pathway. Nrf-2 is a key transcription factor that entangles Kelch-like ECH-associating protein 1, a protein found in the cell cytoplasm. Some antioxidant enzymes, such as gamma glycine cysteine ligase (γ-GCL) and heme oxygenase-1 (HO-1), are expressed in Nrf-2 targeted genes. Their expression, in turn, decreases the stimulation of hepatic macrophages and induces the messenger RNA (mRNA) articulation of proinflammatory factors including tumor necrosis factor α. This review will cover various medicinal plants from a mechanistic view and how they stimulate and interact with Nrf-2, the master regulator of the antioxidant response to counterbalance oxidative stress. Interestingly, therapeutic plants have become popular in the medical sector due to safer yet effective supplementation for the prevention and treatment of new human diseases. The contemporary study is expected to collect information on a variety of therapeutic traditional herbs that have been studied in the context of drug-induced liver toxicity, as nutraceuticals are the most effective treatments for oxidative stress-induced hepatotoxicity. They are less genotoxic, have a lower cost, and are readily available. Together, nutraceuticals exert protective effects against drug-induced hepatotoxicity through the inhibition of oxidative stress, inflammation, and apoptosis. Its mechanism(s) are considered to be associated with the γ-GCL/HO-1 and Nrf-2 signaling pathways. KEY TEACHING POINTSThe liver is the most significant vital organ that carries out metabolic activities of the body such as the synthesis of glycogen, the formation of triglycerides and cholesterol, as well as the formation of bile.Acute liver failure is caused by the consumption of certain drugs; drug-induced liver injury is the major condition.The chemopreventive activity of nutraceuticals may be related to oxidative stress reduction and attenuation of biosynthetic processes involved in hepatic injury via amelioration of the nuclear factor erythroid-2 (Nrf-2) signaling pathway.Nrf-2 is a key transcription factor that is found in the cell cytoplasm resulting in the expression of various genes such as gamma glycine cysteine ligase and heme oxygenase-1.Nutraceutical-rich phytochemicals possess high antioxidant activity, which helps in the prevention of hepatic injury.
Collapse
Affiliation(s)
- Sumaira Yousuf
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Shabnam Shabir
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Mahendra P Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
30
|
Wang P, Liu D, Yan S, Cui J, Liang Y, Ren S. Adverse Effects of Perfluorooctane Sulfonate on the Liver and Relevant Mechanisms. TOXICS 2022; 10:toxics10050265. [PMID: 35622678 PMCID: PMC9144769 DOI: 10.3390/toxics10050265] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a persistent, widely present organic pollutant. PFOS can enter the human body through drinking water, ingestion of food, contact with utensils containing PFOS, and occupational exposure to PFOS, and can have adverse effects on human health. Increasing research shows that the liver is the major target of PFOS, and that PFOS can damage liver tissue and disrupt its function; however, the exact mechanisms remain unclear. In this study, we reviewed the adverse effects of PFOS on liver tissue and cells, as well as on liver function, to provide a reference for subsequent studies related to the toxicity of PFOS and liver injury caused by PFOS.
Collapse
|
31
|
Wan C, Gu T, Ling J, Qin Y, Luo J, Sun L, Hua L, Zhao J, Jiang S. Perfluorooctane sulfonate aggravates CCl4-induced hepatic fibrosis via HMGB1/TLR4/Smad signaling. ENVIRONMENTAL TOXICOLOGY 2022; 37:983-994. [PMID: 34990082 DOI: 10.1002/tox.23458] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 06/14/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a widespread environmental pollutant and may cause a variety of adverse health effects. The hepatotoxicity of PFOS has attracted particular attention, given the fact that the liver has one of the highest PFOS accumulations among human tissues. In this study, we revealed that subchronic PFOS exposure may exacerbate carbon tetrachloride (CCl4 )-induced liver fibrosis in animal models. Administration with 1 mg/kg PFOS every other day for 56 days dramatically enhanced CCl4 -mediated liver injury and hepatic stellate cell (HSC) activation. Furthermore, PFOS exposure may promote the activation of high-mobility group box 1 (HMGB1)/toll-like receptor 4 (TLR4) signaling pathway through inducing the secretion of HMGB1 from hepatocytes. PFOS exposure induced the translocation of HMGB1 from the nucleus into the cytoplasm of hepatocytes and cultured BRL-3A cells at a starting concentration of 50 μM. This process is accompanied with concurrent flux of calcium, suggesting a link between calcium signaling and HMGB1 release following PFOS exposure. Finally, we showed that PFOS-exposed conditional medium (PFOS-CM) of hepatocytes may induce the translocation of Smad2/3 in HSCs in a TLR4-dependent manner. Taken together, subchronic PFOS exposure might play a pro-fibrotic role via a HMGB1/TLR4-dependent Smad signaling in HSCs. Our findings for the first time uncovered an involvement of PFOS exposure in liver fibrosis via HMGB1/TLR4/Smad signaling.
Collapse
Affiliation(s)
- Chunhua Wan
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, China
| | - Tianye Gu
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, China
| | - Junyi Ling
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Yi Qin
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
- Haimen District Center for Disease Control and Prevention, Haimen, Nantong, People's Republic of China
| | - Jiashan Luo
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Lingli Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, China
| | - Lu Hua
- Department of Oncology, Taizhou People's Hospital, Taizhou, China
| | - Jianya Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, China
| | - Shengyang Jiang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| |
Collapse
|
32
|
Hu Q, Liao W, Zhang Z, Shi S, Hou S, Ji N, Zhang X, Zhang Q, Liao Y, Li L, Zhu Z, Chen Y, Chen J, Yu F, Yang Q, Xiao H, Fu C, Du H, Wang Q, Cao H, Xiao H, Li R. The hepatoprotective effects of plant-based foods based on the "gut-liver axis": a prospective review. Crit Rev Food Sci Nutr 2022; 63:9136-9162. [PMID: 35466839 DOI: 10.1080/10408398.2022.2064423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The importance of the "gut-liver axis" in the pathogenesis of liver diseases has been revealed recently; which promotes the process of developing preventive and therapeutic strategies. However, considering that there are still many challenges in the medical treatment of liver diseases, potential preventive dietary intervention may be a good alternative choice. Plant-based foods have received much attention due to their reported health-promoting effects in targeting multiple pathways involved in the pathogenesis of liver diseases as well as the relative safety for general use. Based on the PubMed and Web of Science databases, this review emphatically summarizes the plant-based foods and their chemical constituents with reported effects to impact the LPS/TLR4 signaling pathway of gut-liver axis of various liver diseases, reflecting their health benefits in preventing/alleviating liver diseases. Moreover, some plant-based foods with potential gut-liver effects are specifically analyzed from the reported studies and conclusions. This review intends to provide readers an overview of the current progress in the field of this research topic. We expect to see more hepatoprotective measures for alleviating the current prevalence of liver diseases.
Collapse
Affiliation(s)
- Qiongdan Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Wan Liao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Zhen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Sanjun Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Shuguang Hou
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Ningping Ji
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Xinjie Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Qian Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Yangyang Liao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Linghui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Zongping Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Yi Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Jiao Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Fangkun Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Qingsong Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Hongtao Xiao
- Department of Clinical Pharmacy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Hengjun Du
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Qi Wang
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Heping Cao
- U.S. Department of Agriculture, Agricultural Research Service, Southern Regional Research Center, New Orleans, LA, USA
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Rui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| |
Collapse
|
33
|
Costello E, Rock S, Stratakis N, Eckel SP, Walker DI, Valvi D, Cserbik D, Jenkins T, Xanthakos SA, Kohli R, Sisley S, Vasiliou V, La Merrill MA, Rosen H, Conti DV, McConnell R, Chatzi L. Exposure to per- and Polyfluoroalkyl Substances and Markers of Liver Injury: A Systematic Review and Meta-Analysis. ENVIRONMENTAL HEALTH PERSPECTIVES 2022; 130:46001. [PMID: 35475652 PMCID: PMC9044977 DOI: 10.1289/ehp10092] [Citation(s) in RCA: 194] [Impact Index Per Article: 64.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
BACKGROUND Experimental evidence indicates that exposure to certain pollutants is associated with liver damage. Per- and polyfluoroalkyl substances (PFAS) are persistent synthetic chemicals widely used in industry and consumer products and bioaccumulate in food webs and human tissues, such as the liver. OBJECTIVE The objective of this study was to conduct a systematic review of the literature and meta-analysis evaluating PFAS exposure and evidence of liver injury from rodent and epidemiological studies. METHODS PubMed and Embase were searched for all studies from earliest available indexing year through 1 December 2021 using keywords corresponding to PFAS exposure and liver injury. For data synthesis, results were limited to studies in humans and rodents assessing the following indicators of liver injury: serum alanine aminotransferase (ALT), nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, or steatosis. For human studies, at least three observational studies per PFAS were used to conduct a weighted z-score meta-analysis to determine the direction and significance of associations. For rodent studies, data were synthesized to qualitatively summarize the direction and significance of effect. RESULTS Our search yielded 85 rodent studies and 24 epidemiological studies, primarily of people from the United States. Studies focused primarily on legacy PFAS: perfluorooctanoic acid (PFOA), perfluorooctanesulfonic acid (PFOS), perfluorononanoic acid (PFNA), and perfluorohexanesulfonic acid. Meta-analyses of human studies revealed that higher ALT levels were associated with exposure to PFOA (z-score= 6.20, p<0.001), PFOS (z-score= 3.55, p<0.001), and PFNA (z-score= 2.27, p=0.023). PFOA exposure was also associated with higher aspartate aminotransferase and gamma-glutamyl transferase levels in humans. In rodents, PFAS exposures consistently resulted in higher ALT levels and steatosis. CONCLUSION There is consistent evidence for PFAS hepatotoxicity from rodent studies, supported by associations of PFAS and markers of liver function in observational human studies. This review identifies a need for additional research evaluating next-generation PFAS, mixtures, and early life exposures. https://doi.org/10.1289/EHP10092.
Collapse
Affiliation(s)
- Elizabeth Costello
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Sarah Rock
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Nikos Stratakis
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Sandrah P. Eckel
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Douglas I. Walker
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Damaskini Valvi
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Dora Cserbik
- Barcelona Institute for Global Health, Barcelona, Spain
| | - Todd Jenkins
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Stavra A. Xanthakos
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Rohit Kohli
- Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Stephanie Sisley
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut, USA
| | - Michele A. La Merrill
- Department of Environmental Toxicology, University of California, Davis, Davis, California, USA
| | - Hugo Rosen
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - David V. Conti
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Rob McConnell
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Leda Chatzi
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
34
|
Yen JH, Chio WT, Chuang CJ, Yang HL, Huang ST. Improved Wound Healing by Naringin Associated with MMP and the VEGF Pathway. Molecules 2022; 27:1695. [PMID: 35268795 PMCID: PMC8911856 DOI: 10.3390/molecules27051695] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 02/04/2023] Open
Abstract
This study aims to investigate the wound-healing effectiveness of the phenolic compound, naringin, both in vitro and in vivo. Male mice were shaved on their dorsal skin under isoflurane, a biopsy punch was made in four symmetrical circular resection windows (6 mm) to induce a wound. These excision wounds were used to study the topical effects of naringin in terms of various biochemical, molecular, and histological parameters. We observed a significant recovery in the wound area. Increased levels of MMP-2, 9, 14, TIMP-2, VEGF-A, and VEGF-R1 were induced by naringin in the HaCaT cells. The time course experiments further revealed that levels of VEGF-A and B increased within 36 h; whereas levels of VEGF-C decreased. In line with this, VEGF-R3 levels, but not VEGF-R1 and 2 levels, increased soon after stimulation; although the increase subsided after 36 h. Additionally, naringin cream upregulated wound healing in vitro. The blockage of VEGF by Bevacizumab abolished the function of naringin cream on cell migration. Histological alterations in the wounded skin were restored by naringin cream, which accelerated wound healing via upregulated expression of growth factors (VEGF-A, B, and C and VEGF-R3), and thus increased MMP-2, 9, 14 expressions.
Collapse
Affiliation(s)
- Jia-Hau Yen
- Cancer Research Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung 40402, Taiwan; (J.-H.Y.); (W.-T.C.); (C.-J.C.)
| | - Wan-Ting Chio
- Cancer Research Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung 40402, Taiwan; (J.-H.Y.); (W.-T.C.); (C.-J.C.)
| | - Chia-Ju Chuang
- Cancer Research Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung 40402, Taiwan; (J.-H.Y.); (W.-T.C.); (C.-J.C.)
| | - Hsin-Ling Yang
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung 40402, Taiwan;
| | - Sheng-Teng Huang
- Cancer Research Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung 40402, Taiwan; (J.-H.Y.); (W.-T.C.); (C.-J.C.)
- Department of Chinese Medicine, China Medical University Hospital, Taichung 40402, Taiwan
- School of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
- An-Nan Hospital, China Medical University, Tainan 40402, Taiwan
| |
Collapse
|
35
|
Bioactive Compounds in Oxidative Stress-Mediated Diseases: Targeting the NRF2/ARE Signaling Pathway and Epigenetic Regulation. Antioxidants (Basel) 2021; 10:antiox10121859. [PMID: 34942962 PMCID: PMC8698417 DOI: 10.3390/antiox10121859] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress is a pathological condition occurring due to an imbalance between the oxidants and antioxidant defense systems in the body. Nuclear factor E2-related factor 2 (NRF2), encoded by the gene NFE2L2, is the master regulator of phase II antioxidant enzymes that protect against oxidative stress and inflammation. NRF2/ARE signaling has been considered as a promising target against oxidative stress-mediated diseases like diabetes, fibrosis, neurotoxicity, and cancer. The consumption of dietary phytochemicals acts as an effective modulator of NRF2/ARE in various acute and chronic diseases. In the present review, we discussed the role of NRF2 in diabetes, Alzheimer's disease (AD), Parkinson's disease (PD), cancer, and atherosclerosis. Additionally, we discussed the phytochemicals like curcumin, quercetin, resveratrol, epigallocatechin gallate, apigenin, sulforaphane, and ursolic acid that have effectively modified NRF2 signaling and prevented various diseases in both in vitro and in vivo models. Based on the literature, it is clear that dietary phytochemicals can prevent diseases by (1) blocking oxidative stress-inhibiting inflammatory mediators through inhibiting Keap1 or activating Nrf2 expression and its downstream targets in the nucleus, including HO-1, SOD, and CAT; (2) regulating NRF2 signaling by various kinases like GSK3beta, PI3/AKT, and MAPK; and (3) modifying epigenetic modulation, such as methylation, at the NRF2 promoter region; however, further investigation into other upstream signaling molecules like NRF2 and the effect of phytochemicals on them still need to be investigated in the near future.
Collapse
|
36
|
Koroglu OF, Gunata M, Vardi N, Yildiz A, Ates B, Colak C, Tanriverdi LH, Parlakpinar H. Protective effects of naringin on valproic acid-induced hepatotoxicity in rats. Tissue Cell 2021; 72:101526. [PMID: 33756270 DOI: 10.1016/j.tice.2021.101526] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/22/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023]
Abstract
Valproic acid (VPA) is mainly prescribed to treat epilepsy. VPA has been reported to be associated with many adverse effects, including hepatotoxicity. Naringin (NRG) is a natural, therapeutically active flavanone glycoside with anti-inflammatory, anti-apoptotic, and antioxidant. The current study was therefore designed to investigate the protective effect of NRG against the VPA-induced experimental hepatotoxicity model. For this purpose, 24 Wistar albino rats were randomly divided into three groups as control (Vehicle), VPA (500 mg/kg), and NRG + VPA (100 mg/kg NRG + 500 mg/kg VPA) groups. The agents were administered via oral gavage for 14 days. Blood and liver tissue samples were taken on the end of the experiment. Biochemical analyzes were performed on the blood and liver samples. Also, malondialdehyde (MDA), superoxide dismutase (SOD) enzyme, glutathione (GSH) content, catalase (CAT) enzyme levels were examined in the liver tissue samples. Histopathological changes (hydropic degeneration and congestion) in the VPA group were increased significantly when compared to the control group (p < 0.05). We also found a decrease in enzymes of serum liver function in the VPA group. However, NRG has been shown not to prevent histopathological changes in the VPA group. According to our results with this experiment protocol, NRG could not exert sufficient protection against VPA-induced hepatotoxicity.
Collapse
Affiliation(s)
- Omer Faruk Koroglu
- Medical Student, Faculty of Medicine, Inonu University, Malatya, 44280, Turkey
| | - Mehmet Gunata
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, 44280, Turkey
| | - Nigar Vardi
- Department of Histology and Embryology, Faculty of Medicine, Inonu University, Malatya, 44280, Turkey
| | - Azibe Yildiz
- Department of Histology and Embryology, Faculty of Medicine, Inonu University, Malatya, 44280, Turkey
| | - Burhan Ates
- Department of Chemistry, Faculty of Science and Arts, İnonu University, Malatya, 44280, Turkey
| | - Cemil Colak
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Inonu University, Malatya, 44280, Turkey
| | - Lokman Hekim Tanriverdi
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, 44280, Turkey
| | - Hakan Parlakpinar
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, 44280, Turkey.
| |
Collapse
|
37
|
Yang C, Liu W, Shan H, Yu X, Zhang X, Zeng B, Qian Y. Naringin inhibits titanium particles-induced up-regulation of TNF-α and IL-6 via the p38 MAPK pathway in fibroblasts from hip periprosthetic membrane. Connect Tissue Res 2021; 62:485-494. [PMID: 32500755 DOI: 10.1080/03008207.2020.1778680] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
AIMS Inflammatory responses to wear debris cause osteolysis that leads to aseptic loosening and hip arthroplasty failure. Wear debris stimulate macrophages and fibroblasts to secret proinflammatory cytokines, including TNF-α and IL-6, which have been specifically implicated in periprosthetic osteolysis and osteoclast differentiation. Naringin has anti-inflammatory effect in macrophages. Moreover, naringin inhibited osteoclastogenesis and wear particles-induced osteolysis. In this study, we examined the potential inhibitory effects of naringin on titanium (Ti) particle-induced proinflammatory cytokines secretion in fibroblasts and the possible underlying molecular mechanisms. MATERIALS AND METHODS Fibroblasts were isolated from periprosthetic membrane at the time of revision surgery performed due to aseptic loosening after hip arthroplasty and were cultured in the presence or absence of Ti particles, naringin and mitogen-activated protein kinase (MAPK) inhibitors, PD98059 (a selective inhibitor of ERK), SP600125 (a selective inhibitor of JNK), and SB203580 (a selective inhibitor of p38). TNF-α and IL-6 assays were performed using enzyme-linked immunosorbent assay kits. The phosphorylation levels of p38 and nuclear factor kappa B p65 (NF-κB p65) were examined by western blot. RESULTS Naringin or SB203580 pretreatment significantly suppressed the secretion of TNF-α and IL-6 induced by titanium particles in fibroblasts, while inhibition of ERK or JNK pathways showed no effect on production of TNF-α and IL-6. Moreover, naringin inhibited Ti particle-induced phosphorylation of p38 and p65. CONCLUSIONS These results indicated that naringin could inhibit Ti particle-induced inflammation in fibroblasts by inhibiting p38 MAPK/NF-κB p65 activity and might be a potential drug for the treatment of inflammatory periprosthetic osteolysis after arthroplasty.
Collapse
Affiliation(s)
- Chao Yang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wei Liu
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Haojie Shan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiaowei Yu
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xianlong Zhang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Bingfang Zeng
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yebin Qian
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Orthopedics, Shanghai Sixth People's Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| |
Collapse
|
38
|
Bell EM, De Guise S, McCutcheon JR, Lei Y, Levin M, Li B, Rusling JF, Lawrence DA, Cavallari JM, O'Connell C, Javidi B, Wang X, Ryu H. Exposure, health effects, sensing, and remediation of the emerging PFAS contaminants - Scientific challenges and potential research directions. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 780:146399. [PMID: 33770593 DOI: 10.1016/j.scitotenv.2021.146399] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/02/2021] [Accepted: 03/06/2021] [Indexed: 06/12/2023]
Abstract
Per- and polyfluoroalkyl substances (PFAS) make up a large group of persistent anthropogenic chemicals which are difficult to degrade and/or destroy. PFAS are an emerging class of contaminants, but little is known about the long-term health effects related to exposure. In addition, technologies to identify levels of contamination in the environment and to remediate contaminated sites are currently inadequate. In this opinion-type discussion paper, a team of researchers from the University of Connecticut and the University at Albany discuss the scientific challenges in their specific but intertwined PFAS research areas, including rapid and low-cost detection, energy-saving remediation, the role of T helper cells in immunotoxicity, and the biochemical and molecular effects of PFAS among community residents with measurable PFAS concentrations. Potential research directions that may be employed to address those challenges and improve the understanding of sensing, remediation, exposure to, and health effects of PFAS are then presented. We hope our account of emerging problems related to PFAS contamination will encourage a broad range of scientific experts to bring these research initiatives addressing PFAS into play on a national scale.
Collapse
Affiliation(s)
- Erin M Bell
- Department of Environmental Health Sciences, University at Albany - State University of New York, Rensselaer, NY 12144, USA
| | - Sylvain De Guise
- Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06269, USA
| | - Jeffrey R McCutcheon
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Yu Lei
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA.
| | - Milton Levin
- Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06269, USA
| | - Baikun Li
- Department of Civil and Environmental Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - James F Rusling
- Department of Chemistry and Institute of Material Science, University of Connecticut, Storrs, CT 06269, USA; Department of Surgery and Neag Cancer Center, UConn Health, Farmington, CT 06032, USA; School of Chemistry, National University of Ireland at Galway, Ireland
| | - David A Lawrence
- Department of Environmental Health Sciences, University at Albany - State University of New York, Rensselaer, NY 12144, USA; Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA
| | - Jennifer M Cavallari
- Department of Public Health Sciences, University of Connecticut, Farmington, CT 06030, USA
| | - Caitlin O'Connell
- Office of the Vice President for Research, University of Connecticut, Storrs, CT 06269, USA
| | - Bethany Javidi
- Office of the Vice President for Research, University of Connecticut, Storrs, CT 06269, USA
| | - Xinyu Wang
- Department of Civil and Environmental Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Heejeong Ryu
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
39
|
Naringin alleviates methotrexate-induced liver injury in male albino rats and enhances its antitumor efficacy in HepG2 cells. Biosci Rep 2021; 40:225020. [PMID: 32458964 PMCID: PMC7286878 DOI: 10.1042/bsr20193686] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 05/23/2020] [Accepted: 05/27/2020] [Indexed: 12/24/2022] Open
Abstract
Methotrexate (MTX) is an efficient chemotherapeutic and immunosuppressant drug, but the hepatotoxicity of MTX limits its clinical use. Naringin (Nar) is a flavonoid derived from Citrus paradise, and has been shown to possess several pharmacological activities, including free-radical scavenging and antioxidant properties. In the present study, we first tested the possible protective effects of multiple doses of Nar against MTX-induced acute hepatotoxicity in rats, and then we investigated the growth inhibition and apoptotic effects of MTX and/or Nar against the HepG2 hepatocarcinoma cell line. Our in vivo results showed that Nar significantly reduced MTX-induced increases in serum alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase and total bilirubin levels. Nar also reduced MTX-induced oxidative stress by significantly reducing liver malondialdehyde (MDA) and nitric oxide (NO) content and increasing superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione reductase (GR), and glutathione (GSH). In addition, Nar significantly counteracted MTX-induced increases in hepatic interleukin-6 and tumor necrosis factor-α (TNF-α). Further, Nar greatly protected hepatocyte ultrastructure against MTX-induced injury. In contrast, in vitro MTX and/or Nar treatment of HepG2 cells for 48 h exhibited a cytotoxic effect and induced apoptosis in a dose-dependent manner mediated by a significant increase in the Bax/Bcl-2 protein expression ratio. Noticeably, Nar potentiated the MTX effect on the Bax/Bcl-2 ratio. In conclusion, Nar decreased MTX-induced functional and ultrastructural liver damage in a tumor-free animal model. Also, our data introduce MTX and Nar as promising antiproliferative agents with a distinctive mode of action, inducing apoptosis in HepG2 tumor cells through activation of Bax and down-regulation of Bcl-2 protein expression.
Collapse
|
40
|
Attenuation of Perfluorooctane Sulfonate-Induced Steatohepatitis by Grape Seed Proanthocyanidin Extract in Mice. BIOMED RESEARCH INTERNATIONAL 2021; 2020:8818160. [PMID: 33457418 PMCID: PMC7787751 DOI: 10.1155/2020/8818160] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/11/2020] [Accepted: 11/30/2020] [Indexed: 01/09/2023]
Abstract
Perfluorooctane sulfonate (PFOS), an environmentally persistent pollutant, has been revealed to elicit hepatic toxicity. In the current study, we investigated the protective role of grape seed proanthocyanidin extract (GSPE) against PFOS-caused steatohepatitis in mice. Animals were exposed intragastrically to PFOS (10 mg/kg/day), GSPE (150 mg/kg/day), or their combination. After 21 days of treatment, mice exposed to PFOS exhibited steatosis, oxidative stress, and inflammation in the liver. Nevertheless, simultaneous administration of GSPE resumed the declined serum hepatic enzyme activities and histological abnormalities in PFOS-exposed mice. Furthermore, GSPE supplementation reduced the contents of triglyceride (TG) and total cholesterol (TC) and expression of lipid metabolism-associated genes CD36 and fatty acid-binding protein 4 (FABP4) in the liver of mice treated with PFOS. Moreover, GSPE suppressed the generation of lipid peroxidative product malondialdehyde and restored the activity of superoxide dismutase in the liver of PFOS-exposed mice. In addition, GSPE repressed the PFOS-induced hepatic overproduction of proinflammatory cytokines interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). Our results demonstrate that GSPE attenuates PFOS-caused steatohepatitis in mice by regulating lipid metabolism, oxidative stress, and inflammatory response.
Collapse
|
41
|
Flavonoids from Aurantii Fructus Immaturus and Aurantii Fructus: promising phytomedicines for the treatment of liver diseases. Chin Med 2020; 15:89. [PMID: 32863858 PMCID: PMC7449045 DOI: 10.1186/s13020-020-00371-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/19/2020] [Indexed: 12/13/2022] Open
Abstract
Background Liver diseases and related complications are major sources of morbidity and mortality, which places a huge financial burden on patients and lead to nonnegligible social problems. Therefore, the discovery of novel therapeutic drugs for the treatment of liver diseases is urgently required. Aurantii Fructus Immaturus (AFI) and Aurantii Fructus (AF) are frequently used herbal medicines in traditional Chinese medicine (TCM) formulas for the treatment of diverse ailments. A variety of bioactive ingredients have been isolated and identified from AFI and AF, including alkaloids, flavonoids, coumarins and volatile oils. Main body Emerging evidence suggests that flavonoids, especially hesperidin (HD), naringenin (NIN), nobiletin (NOB), naringin (NRG), tangeretin (TN), hesperetin (HT) and eriodictyol (ED) are major representative bioactive ingredients that alleviate diseases through multi-targeting mechanisms, including anti-oxidative stress, anti-cytotoxicity, anti-inflammation, anti-fibrosis and anti-tumor mechanisms. In the current review, we summarize the recent progress in the research of hepatoprotective effects of HD, NIN, NOB, NRG, TN, HT and ED and highlight the potential underlying molecular mechanisms. We also point out the limitations of the current studies and shed light on further in-depth pharmacological and pharmacokinetic studies of these bioactive flavonoids. Conclusion This review outlines the recent advances in the literature and highlights the potential of these flavonoids isolated from AFI and AF as therapeutic agents for the treatment of liver diseases. Further pharmacological studies will accelerate the development of natural products in AFI and AF and their derivatives as medicines with tantalizing prospects in the clinical application.
Collapse
|
42
|
Implications for glycosylated compounds and their anti-cancer effects. Int J Biol Macromol 2020; 163:1323-1332. [PMID: 32622770 DOI: 10.1016/j.ijbiomac.2020.06.281] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 06/20/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023]
Abstract
Glycosylated compounds are major secondary metabolites of plants, which have various therapeutic effects on human diseases, by acting as anti-cancer, antioxidant, and anti-inflammatory agents. Glycosylation increases stability, bioactivity, and solubility of compounds and improves their pharmacological properties. Two well-known examples of glycosylated compounds include cardiac and flavonoid, the anti-tumor activities of which have been emphasized by several studies. However, little is known about their role in the treatment or prevention of cancer. In this review, recent studies on anti-tumor properties of cardiac and flavonoid glycosides, and their mechanisms of action, have been investigated. More specifically, this review is aimed at focusing on the multifactorial properties of cardiac and flavonoid compounds as well as their correlation with signaling pathways in the treatment of cancer.
Collapse
|
43
|
Jiang X, Li A, Wang Y, Iqbal M, Waqas M, Yang H, Li Z, Mehmood K, Qamar H, Li J. Ameliorative effect of naringin against thiram-induced tibial dyschondroplasia in broiler chicken. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:11337-11348. [PMID: 31960246 DOI: 10.1007/s11356-020-07732-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 01/12/2020] [Indexed: 06/10/2023]
Abstract
Tetramethyl thiuram disulfide (thiram) is widely used in agricultural production as an insecticide and fungicide, which can also lead to tibial dyschondroplasia (TD) in poultry. TD is characterized by leg disorders and growth performance retardation, and no targeted drugs have been found to treat TD until now. Therefore, the objective of the present study was to explore the ameliorative effect of traditional Chinese medicine naringin on thiram-induced TD chickens. A total of 180 one-day-old Arbor Acres (AA) broiler chickens were randomly divided into three equal groups (n = 60): control group (standard diet), thiram-induced group (thiram 50 mg/kg from day 3 to day 7), and naringin-treated group (naringin 30 mg/kg from day 8 to day 18). During the 18-day experiment, the growth performance, tibial bone parameters, antioxidant property of liver, serum biochemical changes and clinical symptoms were recorded to evaluate the protective effect of naringin in thiram-induced TD broiler chickens. Additionally, mRNA expressions and protein levels of Ihh and PTHrP genes were determined via quantitative real-time polymerase chain reaction and western blot. Administration of naringin showed significant results by alleviating lameness, increased growth performance, recuperated growth plate (GP) width, and improved functions and antioxidant enzyme level of liver in broilers affected by TD. Moreover, naringin treatment restored the development of damaged tibia bone via downregulating Ihh and upregulating PTHrP mRNA and protein expressions. In conclusion, our study determines naringin could be used as an effective medicine to treat TD.
Collapse
Affiliation(s)
- Xiong Jiang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
- Hubei Three Gorges Polytechnic, Yichang, 443000, Hubei province, People's Republic of China
| | - Aoyun Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yaping Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Mudassar Iqbal
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
- University College of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Muhammad Waqas
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
- Faculty of Veterinary & Animal Sciences, University of the Poonch, District Poonch, Rawalakot, Azad Jammu & Kashmir, 12350, Pakistan
| | - Hao Yang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Zhixing Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Khalid Mehmood
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
- University College of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Hammad Qamar
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Jiakui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
- College of Animals Husbandry and Veterinary Medicine, Tibet Agricultural and Animal Husbandry University, Linzhi, 860000, Tibet, People's Republic of China.
| |
Collapse
|
44
|
Shirani K, Yousefsani BS, Shirani M, Karimi G. Protective effects of naringin against drugs and chemical toxins induced hepatotoxicity: A review. Phytother Res 2020; 34:1734-1744. [DOI: 10.1002/ptr.6641] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/16/2020] [Accepted: 01/31/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Kobra Shirani
- Department of Toxicology, Faculty of Medical SciencesTarbiat Modares University Tehran Iran
| | - Bahare Sadat Yousefsani
- Research Institute for Islamic and Complementary MedicineIran University of Medical Sciences Tehran Iran
- School of Persian MedicineIran University of Medical Sciences
| | - Maryam Shirani
- Department of Toxicology, Faculty of PharmacyAhvaz Jundishapur University of Medical Sciences Ahvaz Iran
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of PharmacyMashhad University of Medical Sciences Mashhad Iran
- Pharmaceutical Research Center, Pharmaceutical Technology InstituteMashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
45
|
Sun LJ, Qiao W, Xiao YJ, Cui L, Wang X, Ren WD. Naringin mitigates myocardial strain and the inflammatory response in sepsis-induced myocardial dysfunction through regulation of PI3K/AKT/NF-κB pathway. Int Immunopharmacol 2019; 75:105782. [PMID: 31376623 DOI: 10.1016/j.intimp.2019.105782] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 07/18/2019] [Accepted: 07/24/2019] [Indexed: 12/27/2022]
Abstract
Sepsis-induced myocardial dysfunction (SIMD) is a manifestation of severe sepsis and is the main cause of increased mortality in sepsis patients. Naringin (Nar) has been reported to possess various biological activities and pharmacological properties. Therefore, the present study was undertaken to evaluate whether Nar can protect rats from the effects of LPS-induced SIMD. SD Rats were pre-treated with Nar (50 and 100 mg/kg) for 7 days before administration of a single dose of LPS (10 mg/kg, i.p.) on the seventh day. We found that Nar treatment markedly improved the global strain and strain rate of longitudinal, circumference, and radial direction (GLS/GLSr, GCS/GCSr, GRS/GRSr) compared to the LPS group. The layer-specific strain decreased gradually from the endocardial layer to epicardial layer, and the most serious damage occurred in the endocardial layer. Moreover, Nar significantly decreased the levels of pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6) and myocardial enzymes (CK, LDH, and AST) induced by LPS and attenuated the inflammation response. Finally, Nar also inhibited NF-κB nuclear translocation and the activity of iNOS in H9c2 cardiomyocytes by activating PI3K/AKT signaling pathway. These results suggest that naringin may possess novel therapeutic potential for protection against LPS-induced myocardial dysfunction.
Collapse
Affiliation(s)
- Li-Juan Sun
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, PR China; Department of Ultrasound, The First Hospital of Qinhuangdao, Qinhuangdao 066000, PR China
| | - Wei Qiao
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Yang-Jie Xiao
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Li Cui
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Xin Wang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Wei-Dong Ren
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| |
Collapse
|