1
|
Du W, Zeng W, Wang Z, Zhu F, Zheng Y, Hu H, Zhuang W, Quan R, Ruan H. Morroniside repairs atrazine-induced skin damage by ameliorating lipid metabolism disorders and inhibiting ferroptosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117047. [PMID: 39321526 DOI: 10.1016/j.ecoenv.2024.117047] [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: 06/27/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 09/27/2024]
Abstract
Morroniside (MOR) has shown great potential in treating atrazine (ATZ)-induced skin damage. This study aims to elucidate MOR's mechanism of action in mitigating lipid metabolism disorders and inhibiting ferroptosis to repair ATZ-induced skin damage. Twenty C57BL/6 mice were divided into four groups: the control group, the ATZ group, the MOR-H group and the MOR-L group, each comprising five mice. Following a one-month intervention, mouse skin tissues were harvested for untargeted lipid metabolomics analysis. Subsequently, the samples were assessed for indices related to ferroptosis. Untargeted lipid metabolomics analysis showed 127 differential metabolites in the ATZ vs. Ctrl group. There were 57 differential metabolites in the MOR-L vs. ATZ group. 34 differential metabolites in the MOR-H vs. ATZ group. the most obvious lipid reversal occurred after MOR-L administration, which primarily involved phospholipids, ceramides, and sphingomyelins. The levels of GPX4, Ferritin, MDA, SOD and GSH-PX, ferroptosis-related indicators, and the levels of p21 and p53, apoptosis-related indicators, were most significantly regressed in the MOR-L group (all P < 0.05). MOR may delay cellular aging and correct skin damage by reversing ATZ-induced lipid metabolism disorders, inhibiting ferroptosis and excessive oxidative stress occurrence.
Collapse
Affiliation(s)
- Weibin Du
- Research Institute of Orthopedics, the Jiangnan Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Hangzhou Xiaoshan Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China.
| | - Wenxiang Zeng
- Research Institute of Orthopedics, the Jiangnan Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Hangzhou Xiaoshan Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Zhenwei Wang
- Research Institute of Orthopedics, the Jiangnan Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Hangzhou Xiaoshan Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Fangbing Zhu
- Research Institute of Orthopedics, the Jiangnan Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Hangzhou Xiaoshan Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Yang Zheng
- Research Institute of Orthopedics, the Jiangnan Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Hangzhou Xiaoshan Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Huahui Hu
- Research Institute of Orthopedics, the Jiangnan Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Hangzhou Xiaoshan Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Wei Zhuang
- Research Institute of Orthopedics, the Jiangnan Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Hangzhou Xiaoshan Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Renfu Quan
- Research Institute of Orthopedics, the Jiangnan Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Hangzhou Xiaoshan Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Hongfeng Ruan
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Li J, Fang X, Cui D, Ma Z, Yang J, Niu Y, Liu H, Xiang P. Mechanistic insights into cadmium exacerbating 2-Ethylhexyl diphenyl phosphate-induced human keratinocyte toxicity: Oxidative damage, cell apoptosis, and tight junction disruption. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116858. [PMID: 39137464 DOI: 10.1016/j.ecoenv.2024.116858] [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: 05/25/2024] [Revised: 07/19/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024]
Abstract
Organophosphate flame retardants 2-ethylhexyldiphenyl phosphate (EHDPP) and cadmium (Cd) are ubiquitous in environmental matrices, and dermal absorption is a major human exposure pathway. However, their detrimental effects on the human epidermis remain largely unknown. In this study, human keratinocytes (HaCaT cells) were employed to examine the toxicity and underlying mechanisms of co-exposure to EHDPP and Cd. Their influence on cell morphology and viability, oxidative damage, apoptosis, and tight junction were determined. The results showed that co-exposure decreased cell viability by >40 %, induced a higher level of oxidative damage by increasing the generation of reactive oxygen species (1.3 folds) and inhibited CAT (79 %) and GPX (90 %) activities. Moreover, Cd exacerbated EHDPP-induced mitochondrial disorder and cellular apoptosis, which was evidenced by a reduction in mitochondrial membrane potential and an elevation of cyt-c and Caspase-3 mRNA expression. In addition, greater loss of ZO-1 immunoreactivity at cellular boundaries was observed after co-exposure, indicating skin epithelial barrier function disruption, which may increase the human bioavailability of contaminants via the dermal absorption pathway. Taken together, oxidative damage, cell apoptosis, and tight junction disruption played a crucial role in EHDPP + Cd triggered cytotoxicity in HaCaT cells. The detrimental effects of EHDPP + Cd co-exposure were greater than individual exposure, suggesting the current health risk assessment or adverse effects evaluation of individual exposure may underestimate their perniciousness. Our data imply the importance of considering the combined exposure to accurately assess their health implication.
Collapse
Affiliation(s)
- Jingya Li
- Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming 650224, China
| | - Xianlei Fang
- School of Basic Medical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Daolei Cui
- Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming 650224, China
| | - Ziya Ma
- Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming 650224, China
| | - Ji Yang
- Affiliated Hospital of Yunnan University, Eye Hospital of Yunnan Province, Kunming 650224, China
| | - Youya Niu
- School of Basic Medical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Hai Liu
- Affiliated Hospital of Yunnan University, Eye Hospital of Yunnan Province, Kunming 650224, China.
| | - Ping Xiang
- Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming 650224, China.
| |
Collapse
|
3
|
Maliyam P, Laphookhieo S, Koedrith P, Puttarak P. Antioxidative and anti-cytogenotoxic potential of Lysiphyllum strychnifolium (Craib) A. Schmitz extracts against cadmium-induced toxicity in human embryonic kidney (HEK293) and dermal fibroblast (HDF) cells. Heliyon 2024; 10:e34480. [PMID: 39130464 PMCID: PMC11315074 DOI: 10.1016/j.heliyon.2024.e34480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/12/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024] Open
Abstract
Exposure to cadmium (Cd) results in bioaccumulation and irreversible damage; this encourages an investigation of alternatives to address Cd toxicity, using natural compounds. Lysiphyllum strychnifolium, a well-known Thai medicinal plant, was investigated for its phytochemical compounds and corresponding bioactivities, including antioxidant and anti-cytogenotoxic effects against Cd toxicity in HEK293 renal and HDF dermal cell models. The crude extract of L. strychnifolium (LsCrude) was partitioned into four fractions, using sequential polarity solvents (hexane, dichloromethane, ethyl acetate, and water, denoted as LsH, LsD, LsE, and LsW, respectively). The extraction yields were 1.79 %, 5.08 %, 8.53 %, and 70.25 % (w/w), respectively. Phytochemical screening revealed the presence of tannins, alkaloids, and flavonoids in LsCrude and its fractions, except for LsH. LsE exhibited the highest concentrations of phenolics (286.83 ± 6.83 mg GAE/g extract) and flavonoids (86.36 ± 1.29 mg QE/g extract). Subsequent 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical-scavenging and ferric-reducing ability of plasma (FRAP) reducing powder assays demonstrated the high antioxidant capacity of LsCrude and its fractions. The lowest IC50 value (9.11 ± 0.43 μg/mL) in the DPPH assay corresponded to LsW, whereas the highest total FRAP value (6.06 ± 0.70 mg QE Eq./g dry mass) corresponded to LsE. MTT and alkaline comet assays revealed the lack of toxicity of the extracts, which were considered safe. Upon exposure to Cd at the CC50 level, HEK293 cells treated with LsE suppressed Cd-induced damage. HDF cells treated with LsCrude, LsD, or LsE attenuated Cd-induced damage. In the pre-treatment, LsD protected the HDF cells against Cd-mediated cytogenotoxicity. These anti-cytogenotoxic potentials are likely due to the antioxidant properties of the phytochemicals. Our findings highlight the cyto-geno-protective properties of L. strychnifolium stem extracts against Cd toxicity in HEK293 and HDF cells, and provide a novel approach for combating oxidative stress and DNA damage caused by environmental pollutants.
Collapse
Affiliation(s)
- Pattaravan Maliyam
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai District, Songkhla, 90112, Thailand
| | - Surat Laphookhieo
- Center of Chemical Innovation for Sustainability (CIS) and School of Science, Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Preeyaporn Koedrith
- Faculty of Environment and Resource Studies, Mahidol University, Phuttamonthon District, NakhonPathom, 73170, Thailand
| | - Panupong Puttarak
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai District, Songkhla, 90112, Thailand
- Phytomedicine and Pharmaceutical Biotechnology Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai District, Songkhla, 90112, Thailand
| |
Collapse
|
4
|
Romashin D, Arzumanian V, Poverennaya E, Varshaver A, Luzgina N, Rusanov A. Evaluation of Cd-induced cytotoxicity in primary human keratinocytes. Hum Exp Toxicol 2024; 43:9603271231224458. [PMID: 38174414 DOI: 10.1177/09603271231224458] [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] [Indexed: 01/05/2024]
Abstract
An increasing number of studies have investigated the effects of Cd on human health. Cd-induced dermatotoxicity is an important field of research, but numerous studies have focused on the effects of Cd on the human skin. Moreover, most studies have been performed using HaCaT cells but not primary keratinocytes. In this study, we provide the results describing the cytotoxic effects of Cd exposure on primary human epidermal keratinocytes obtained from different donors. The subtoxic concentration of cadmium chloride was determined via MTT assay, and transcriptomic analysis of the cells exposed to this concentration (25 µM) was performed. As in HaCaT cells, Cd exposure resulted in increased ROS levels, cell cycle arrest, and induction of apoptosis. In addition, we report that exposure to Cd affects zinc and copper homeostasis, induces metallothionein expression, and activates various signaling pathways, including Nrf2, NF-kB, TRAIL, and PI3K. Cd induces the secretion of various cytokines (IL-1, IL-6, IL-10, and PGE2) and upregulates the expression of several cytokeratins, such as KRT6B, KRT6C, KRT16, and KRT17. The results provide a better understanding of the mechanisms of cadmium-induced cytotoxicity and its effect on human epidermal skin cells.
Collapse
|
5
|
Huertas-Abril PV, Jurado J, Prieto-Álamo MJ, García-Barrera T, Abril N. Proteomic analysis of the hepatic response to a pollutant mixture in mice. The protective action of selenium. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 903:166558. [PMID: 37633382 DOI: 10.1016/j.scitotenv.2023.166558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
Metals and pharmaceuticals contaminate water and food worldwide, forming mixtures where they can interact to enhance their individual toxicity. Here we use a shotgun proteomic approach to evaluate the toxicity of a pollutant mixture (PM) of metals (As, Cd, Hg) and pharmaceuticals (diclofenac, flumequine) on mice liver proteostasis. These pollutants are abundant in the environment, accumulate in the food chain, and are toxic to humans primarily through oxidative damage. Thus, we also evaluated the putative antagonistic effect of low-dose dietary supplementation with the antioxidant trace element selenium. A total of 275 proteins were affected by PM treatment. Functional analyses revealed an increased abundance of proteins involved in the integrated stress response that promotes translation, the inflammatory response, carbohydrate and lipid metabolism, and the sustained expression of the antioxidative response mediated by NRF2. As a consequence, a reductive stress situation arises in the cell that inhibits the RICTOR pathway, thus activating the early stage of autophagy, impairing xenobiotic metabolism, and potentiating lipid biosynthesis and steatosis. PM exposure-induced hepato-proteostatic alterations were significantly reduced in Se supplemented mice, suggesting that the use of this trace element as a dietary supplement may at least partially ameliorate liver damage caused by exposure to environmental mixtures.
Collapse
Affiliation(s)
- Paula V Huertas-Abril
- Department of Biochemistry and Molecular Biology, University of Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071 Córdoba, Spain
| | - Juan Jurado
- Department of Biochemistry and Molecular Biology, University of Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071 Córdoba, Spain
| | - María-José Prieto-Álamo
- Department of Biochemistry and Molecular Biology, University of Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071 Córdoba, Spain
| | - Tamara García-Barrera
- Research Center of Natural Resources, Health, and the Environment (RENSMA), Department of Chemistry, Faculty of Experimental Sciences, Campus El Carmen, University of Huelva, Fuerzas Armadas Ave., 21007 Huelva, Spain
| | - Nieves Abril
- Department of Biochemistry and Molecular Biology, University of Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071 Córdoba, Spain.
| |
Collapse
|
6
|
Chen Y, Zhou C, Bian Y, Fu F, Zhu B, Zhao X, Zhang M, Zhou C, Yao S, Zhang Z, Luo H, Ge Y, Wu C, Ruan H. Cadmium exposure promotes thyroid pyroptosis and endocrine dysfunction by inhibiting Nrf2/Keap1 signaling. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114376. [PMID: 36508821 DOI: 10.1016/j.ecoenv.2022.114376] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 06/17/2023]
Abstract
Cadmium (Cd) is a ubiquitous toxic metal and environmental pollutant. Increasing studies have shown that Cd exposure increases the incidence of various endocrine system diseases, including thyrotoxicity reflected by thyroid structural damage and endocrine toxicity. However, the observed outcomes are complex and conflicting, leading to the mechanism of Cd-induced thyrotoxicity remaining obscure. In this study, 4-week-old male C57BL/6 mice were given 2 or 7 mg/kg Cadmium Chloride (CdCl2) intragastrically for 4 and 8 weeks, and the Cd-mediated thyrotoxicity was evaluated by determining alterations in thyroid structure and endocrine function, and alterations of oxidant stress, apoptosis, and pyroptosis. Our data showed that Cd exposure could reduce body weight and induce thyrotoxicity by impairing thyroid follicular morphology and endocrine function, accompanied by elevated oxidative stress and apoptosis, macrophage infiltration, and inflammatory cytokine secretion. Importantly, Cd significantly promoted thyroid follicular cell pyroptosis by increasing Nlrp3, Asc, Caspase-1, Gsdmd, IL-1β, and IL-18 expression. Mechanistical analysis suggested that Cd treatment could inhibit antioxidant pathway by downregulating antioxidant response protein, Nrf2, and upregulating its negative feedback regulator, Keap1. Collectively, our in vivo findings suggest that Cd exposure could facilitate thyroid follicular cell pyroptosis by inhibiting Nrf2/Keap1 signaling, thereby disrupting thyroid tissue structure and endocrine function, which offers novel insights into the Cd-mediated detrimental consequences on thyroid homeostasis.
Collapse
Affiliation(s)
- Yuying Chen
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou 310053, China; The Fourth Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Chengcong Zhou
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou 310053, China
| | - Yishan Bian
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou 310053, China
| | - Fangda Fu
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou 310053, China
| | - Bian'an Zhu
- The Fourth Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xuyan Zhao
- The Fourth Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Muxin Zhang
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou 310053, China
| | - Chunyuan Zhou
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou 310053, China
| | - Sai Yao
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou 310053, China
| | - Zhiguo Zhang
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou 310053, China
| | - Huan Luo
- Department of Pharmacy, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China.
| | - Yuying Ge
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou 310053, China
| | - Chengliang Wu
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou 310053, China
| | - Hongfeng Ruan
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou 310053, China.
| |
Collapse
|