1
|
Eriten B, Caglayan C, Gür C, Küçükler S, Diril H. Hepatoprotective effects of zingerone on sodium arsenite-induced hepatotoxicity in rats: Modulating the levels of caspase-3/Bax/Bcl-2, NLRP3/NF-κB/TNF-α and ATF6/IRE1/PERK/GRP78 signaling pathways. Biochem Biophys Res Commun 2024; 725:150258. [PMID: 38897041 DOI: 10.1016/j.bbrc.2024.150258] [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: 04/03/2024] [Revised: 05/29/2024] [Accepted: 06/11/2024] [Indexed: 06/21/2024]
Abstract
OBJECTIVE Long-term exposure to arsenic has been linked to several illnesses, including hypertension, diabetes, hepatic and renal diseases and cardiovascular malfunction. The aim of the current investigation was to determine whether zingerone (ZN) could shield rats against the hepatotoxicity that sodium arsenite (SA) causes. METHODS The following five groups of thirty-five male Sprague Dawley rats were created: I) Control; received normal saline, II) ZN; received ZN, III) SA; received SA, IV) SA + ZN 25; received 10 mg/kg body weight SA + 25 mg/kg body weight ZN, and V) SA + ZN 50; received 10 mg/kg body weight SA + 50 mg/kg body weight ZN. The experiment lasted 14 days, and the rats were sacrificed on the 15th day. While oxidative stress parameters were studied by spectrophotometric method, apoptosis, inflammation and endoplasmic reticulum stress parameters were measured by RT-PCR method. RESULTS The SA disrupted the histological architecture and integrity of the liver and enhanced oxidative damage by lowering antioxidant enzyme activity, such as those of glutathione peroxidase (GPx), catalase (CAT), superoxide dismutase (SOD), glutathione (GSH) level and increasing malondialdehyde (MDA) level in the liver tissue. Additionally, SA increased the mRNA transcript levels of Bcl2 associated x (Bax), caspases (-3, -6, -9), apoptotic protease-activating factor 1 (Apaf-1), p53, tumor necrosis factor-α (TNF-α), nuclear factor kappa B (NF-κB), interleukin-1β (IL-1β), interleukin-6 (IL-6), c-Jun NH2-terminal kinase (JNK), mitogen-activated protein kinase 14 (MAPK14), MAPK15, receptor for advanced glycation endproducts (RAGE) and nod-like receptor family pyrin domain-containing 3 (NLRP3) in the liver tissue. Also produced endoplasmic reticulum stress by raising the mRNA transcript levels of activating transcription factor 6 (ATF-6), protein kinase RNA-like ER kinase (PERK), inositol-requiring enzyme 1 (IRE1), and glucose-regulated protein 78 (GRP-78). These factors together led to inflammation, apoptosis, and endoplasmic reticulum stress. On the other hand, liver tissue treated with ZN at doses of 25 and 50 mg/kg showed significant improvement in oxidative stress, inflammation, apoptosis and endoplasmic reticulum stress. CONCLUSIONS Overall, the study's data suggest that administering ZN may be able to lessen the liver damage caused by SA toxicity.
Collapse
Affiliation(s)
- Berna Eriten
- Department of Pathology, Sancaktepe Sehit Prof. Dr. Ilhan Varank Training and Research Hospital, Istanbul, Turkey.
| | - Cuneyt Caglayan
- Department of Medical Biochemistry, Faculty of Medicine, Bilecik Seyh Edebali University, Bilecik, Turkey.
| | - Cihan Gür
- Department of Medical Laboratory Techniques, Vocational School of Health Services, Atatürk University, Erzurum, Turkey
| | - Sefa Küçükler
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Halit Diril
- Medical Biochemistry Laboratory, Dursun Odabaş Medical Center, Van Yüzüncü Yıl University, Van, Turkey
| |
Collapse
|
2
|
Wang Y, Yin D, Sun X, Zhang W, Ma H, Huang J, Yang C, Wang J, Geng Q. Perfluoroalkyl sulfonate induces cardiomyocyte apoptosis via endoplasmic reticulum stress activation and autophagy flux inhibition. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 930:172582. [PMID: 38649052 DOI: 10.1016/j.scitotenv.2024.172582] [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: 02/22/2024] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024]
Abstract
Perfluoroalkyl sulfonate (PFOS) is a commonly used chemical compound that often found in materials such as waterproofing agents, food packaging, and fire retardants. Known for its stability and persistence in the environment, PFOS can enter the human body through various pathways, including water and the food chain, raising concerns about its potential harm to human health. Previous studies have suggested a cardiac toxicity of PFOS, but the specific cellular mechanisms remained unclear. Here, by using AC16 cardiomyocyte as a model to investigate the molecular mechanisms potential the cardiac toxicity of PFOS. Our findings revealed that PFOS exposure reduced cell viability and induces apoptosis in human cardiomyocyte. Proteomic analysis and molecular biological techniques showed that the Endoplasmic Reticulum (ER) stress-related pathways were activated, while the cellular autophagy flux was inhibited in PFOS-exposed cells. Subsequently, we employed strategies such as autophagy activation and ER stress inhibition to alleviate the PFOS-induced apoptosis in AC16 cells. These results collectively suggest that PFOS-induced ER stress activation and autophagy flux inhibition contribute to cardiomyocyte apoptosis, providing new insights into the mechanisms of PFOS-induced cardiomyocyte toxicity.
Collapse
Affiliation(s)
- Yuanhao Wang
- Department of Geriatrics, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Da Yin
- Department of Cardiology, Shenzhen Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen People's Hospital, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Xin Sun
- Department of Cardiology, Shenzhen Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen People's Hospital, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Wei Zhang
- Department of Geriatrics, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Huan Ma
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No.106 Zhongshan Er Road, Guangzhou, Guangdong, China
| | - Jingnan Huang
- Department of Geriatrics, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Chuanbin Yang
- Department of Geriatrics, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China.
| | - Jigang Wang
- Department of Geriatrics, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China; State Key Laboratory for Quality Esurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Qingshan Geng
- Department of Geriatrics, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China.
| |
Collapse
|
3
|
Hou L, Dong H, Zhang E, Lu H, Zhang Y, Zhao H, Xing M. A new insight into fluoride induces cardiotoxicity in chickens: Involving the regulation of PERK/IRE1/ATF6 pathway and heat shock proteins. Toxicology 2024; 501:153688. [PMID: 38036095 DOI: 10.1016/j.tox.2023.153688] [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: 10/03/2023] [Revised: 11/24/2023] [Accepted: 11/25/2023] [Indexed: 12/02/2023]
Abstract
Fluorosis poses a significant threat to human and animal health and is an urgent public safety concern in various countries. Subchronic exposure to fluoride has the potential to result in pathological damage to the heart, but its potential mechanism requires further investigation. This study investigated the effects of long-term exposure to sodium fluoride (0, 500, 1000, and 2000 mg/kg) on the hearts of chickens were investigated. The results showed that an elevated exposure dose of sodium fluoride led to congested cardiac tissue and disrupted myofiber organisation. Sodium fluoride exposure activated the ERS pathways of PERK, IRE1, and ATF6, increasing HSP60 and HSP70 and decreasing HSP90. The NF-κB pathway and the activation of TNF-α and iNOS elicited an inflammatory response. BAX, cytc, and cleaved-caspase3 were increased, triggering apoptosis and leading to cardiac injury. The abnormal expression of HSP90 and HSP70 affected the stability and function of RIPK1, RIPK3, and MLKL, which are crucial necroptosis markers. HSPs inhibited TNF-α-mediated necroptosis and apoptosis of the death receptor pathway. Sodium fluoride resulted in heart injury in chickens because of the ERS and variations in HSPs, inducing inflammation and apoptosis. Cardiac-adapted HSPs impeded the activation of necroptosis. This paper may provide a reference for examining the potential cardiotoxic effects of sodium fluoride.
Collapse
Affiliation(s)
- Lulu Hou
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China
| | - Haiyan Dong
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China
| | - Enyu Zhang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China
| | - Hongmin Lu
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China
| | - Yue Zhang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China
| | - Hongjing Zhao
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China.
| | - Mingwei Xing
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China.
| |
Collapse
|
4
|
Zhu Z, Pu J, Li Y, Chen J, Ding H, Zhou A, Zhang X. RBM25 regulates hypoxic cardiomyocyte apoptosis through CHOP-associated endoplasmic reticulum stress. Cell Stress Chaperones 2023; 28:861-876. [PMID: 37736860 PMCID: PMC10746693 DOI: 10.1007/s12192-023-01380-7] [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: 05/30/2023] [Revised: 08/15/2023] [Accepted: 09/10/2023] [Indexed: 09/23/2023] Open
Abstract
Ischemic heart failure (HF) is one of the leading causes of global morbidity and mortality; blocking the apoptotic cascade could help improve adverse outcomes of it. RNA-binding motif protein 25 (RBM25) is an RNA-binding protein related to apoptosis; however, its role remains unknown in ischemic HF. The main purpose of this study is to explore the mechanism of RBM25 in ischemic HF. Establishing an ischemic HF model and oxygen-glucose deprivation (OGD) model. ELISA was performed to evaluate the BNP level in the ischemic HF model. Echocardiography and histological analysis were performed to assess cardiac function and infarct size. Proteins were quantitatively and locationally analyzed by western blotting and immunofluorescence. The morphological changes of endoplasmic reticulum (ER) were observed with ER-tracker. Cardiac function and myocardial injury were observed in ischemic HF rats. RBM25 was elevated in cardiomyocytes of hypoxia injury hearts and localized in nucleus both in vitro and in vivo. In addition, cell apoptosis was significantly increased when overexpressed RBM25. Moreover, ER stress stimulated upregulation of RBM25 and promoted cell apoptosis through the CHOP related pathway. Finally, inhibiting the expression of RBM25 could ameliorate the apoptosis and improve cardiac function through blocking the activation of CHOP signaling pathway. RBM25 is significantly upregulated in ischemic HF rat heart and OGD model, which leads to apoptosis by modulating the ER stress through CHOP pathway. Knockdown of RBM25 could reverse apoptosis-mediated cardiac dysfunction. RBM25 may be a promising target for the treatment of ischemic HF.
Collapse
Affiliation(s)
- Ziwei Zhu
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Jie Pu
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Yongnan Li
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Jianshu Chen
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Hong Ding
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Anyu Zhou
- Department of Cardiology, Warren Alpert School of Medicine at Brown University, Providence, RI, USA
| | - XiaoWei Zhang
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou, 730000, China.
| |
Collapse
|
5
|
Zhao Q, Pan W, Li J, Yu S, Liu Y, Zhang X, Qu R, Zhang Q, Li B, Yan X, Ren X, Qiu Y. Effects of neuron autophagy induced by arsenic and fluoride on spatial learning and memory in offspring rats. CHEMOSPHERE 2022; 308:136341. [PMID: 36087721 DOI: 10.1016/j.chemosphere.2022.136341] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 08/25/2022] [Accepted: 09/02/2022] [Indexed: 06/15/2023]
Abstract
There are numerous studies showing that exposure to arsenic (As) or fluoride (F) damages the nervous system, but there is no literature investigating the effects of combined As and F exposure to induce autophagy on neurotoxicity in the offspring. In this study, we developed a rat model of As and/or F exposure through drinking water from before pregnancy to 90 days postnatal. The offspring rats were randomly divided into nine groups. Sodium arsenite (NaAsO2) (0, 35, 70 mg/L) and Sodium fluoride (NaF) (0, 50, 100 mg/L) were designed according to 3 × 3 factorial design. Our results suggested that the presence of F might antagonize the excretion of total As in urine, and As-F co-exposure led to severe pathological damage in brain tissue and reduced spatial learning and memory ability. At the same time, the experiments showed that As and F increased Beclin1 expression and LC3B ratio to activate autophagy; both P62 and Lamp2 expression were increased, suggesting that autophagy lysosomal degradation was blocked; SYN and JIP1 expression were significantly decreased, disrupting synaptic structure and function. Axonal autophagosome reverse transport regulation might be affected by combined As-F exposure, exacerbating neuronal synaptic damage and inducing neurotoxicity. Further analysis showed that there was an interaction between As and F exposure-induced changes in autolysosome-related proteins in the hippocampus, which showed antagonism, and the antagonism of the high As combined exposure groups were stronger than that of the low As combined exposure groups. In conclusion, our study showed that combined As and F exposure might induce reverse transport impairment of autophagy on axons, leading to autophagy defects, which in turn led to disruption of synaptic morphology and function, induced neurotoxicity, and there was an interaction between As and F, the type of its combined effect was antagonism.
Collapse
Affiliation(s)
- Qiuyi Zhao
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Weizhe Pan
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Jia Li
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Shengnan Yu
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Yan Liu
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Xiaoli Zhang
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China; Department of Microbiology Laboratory, Linfen Central Hospital, Linfen, China.
| | - Ruodi Qu
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Qian Zhang
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Ben Li
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Xiaoyan Yan
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Xuefeng Ren
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, USA; Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| | - Yulan Qiu
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
6
|
Xu Q, Zhao T, Ri H, Ye J, Zhao W, Zhang Y, Ye L. Di(2-ethylhexyl) phthalate induced thyroid toxicity via endoplasmic reticulum stress: In vivo and in vitro study. ENVIRONMENTAL TOXICOLOGY 2022; 37:2924-2936. [PMID: 36005737 DOI: 10.1002/tox.23648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/03/2022] [Accepted: 08/15/2022] [Indexed: 06/15/2023]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) could induce thyroid injury but the mechanism was unclear. This study combined in vivo and in vitro experiments to clarify the mechanism. In vivo, the offspring of Sprague Dawley rats were gavaged with different doses of DEHP (5, 50, and 250 mg/[kg⋅d]) from in utero to 12 weeks-old. Transcriptome sequencing was used to detect the mRNA expression profile of the offspring's thyroids. Differentially expressed genes were identified, followed by Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. In vitro, Nthy-ori 3-1 cells were exposed to DEHP's metabolite mono (2-ethylhexyl) phthalate (MEHP) to verify the pathway we found by KEGG analysis. The results indicated that DEHP could disorder the thyroid hormones. Compared with the offspring in control group, the mRNA levels of 656 genes were upregulated in the offspring exposed to 50 mg/(kg⋅d) DEHP. The upregulated genes were enriched in the pathway of "protein processing in the endoplasmic reticulum (ER)." It indicated that the ER stress might play significant role in the thyroid toxicity induced by DEHP. In vitro, the mitochondrial membrane potential (ΔΨm) level of cells was decreased while the reactive oxygen species level was increased after MEHP exposure. MEHP increased the intracellular Ca2+ level and induced ER stress. After ER stress was inhibited by the 4-phenylbutyric acid, the thyroid toxicity caused by MEHP was alleviated. Taken together, our results indicated that DEHP could induce thyroid toxicity by activating ER stress.
Collapse
Affiliation(s)
- Qi Xu
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Tianyang Zhao
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Hyonju Ri
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
- Faculty of Public Health, Pyongyang Medical University, Pyongyang, North Korea
| | - Jiaming Ye
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Weisen Zhao
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Yuezhu Zhang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Lin Ye
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
7
|
Zhang J, Tang Y, Xu W, Hu Z, Xu S, Niu Q. Fluoride-Induced Cortical Toxicity in Rats: the Role of Excessive Endoplasmic Reticulum Stress and Its Mediated Defective Autophagy. Biol Trace Elem Res 2022:10.1007/s12011-022-03463-5. [PMID: 36327065 DOI: 10.1007/s12011-022-03463-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
Abstract
The cerebral cortex is closely associated with learning and memory, and fluoride is capable of inducing cortical toxicity, but its mechanism is unclear. This study aimed to investigate the role of endoplasmic reticulum stress and autophagy in fluoride-induced cortical toxicity. Rats exposed to sodium fluoride (NaF) were used as an in vivo model. The results showed that NaF exposure impaired the learning and memory capacities and increased urinary fluoride levels in rats. In addition, NaF exposure induced excessive endoplasmic reticulum stress and associated apoptosis, as evidenced by elevated IRE1α, GRP78, cleaved caspase-12, and cleaved caspase-3, as well as defective autophagy, as evidenced by increased expression of Beclin1, LC3-II, and p62 in cortical areas. Importantly, the endoplasmic reticulum stress inhibitor 4-phenylbutyric acid (4-PBA) alleviated endoplasmic reticulum stress as well as defective autophagy, thus confirming the critical role of endoplasmic reticulum stress and autophagy in fluoride-induced cortical toxicity. Taken together, these results suggest that excessive endoplasmic reticulum stress and its mediated defective autophagy lead to fluoride-induced cortical toxicity. This provides new insights into the mechanisms of fluoride-induced neurotoxicity and a new theoretical basis for the prevention and treatment of fluoride-induced neurotoxicity.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Preventive Medicine, School of Medicine, Shihezi University, North 2th Road, Shihezi, Xinjiang, 832000, People's Republic of China
- Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
| | - Yanling Tang
- Department of Preventive Medicine, School of Medicine, Shihezi University, North 2th Road, Shihezi, Xinjiang, 832000, People's Republic of China
- Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
| | - Wanjing Xu
- Department of Preventive Medicine, School of Medicine, Shihezi University, North 2th Road, Shihezi, Xinjiang, 832000, People's Republic of China
- Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
| | - Zeyu Hu
- Department of Preventive Medicine, School of Medicine, Shihezi University, North 2th Road, Shihezi, Xinjiang, 832000, People's Republic of China
- Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
| | - Shangzhi Xu
- Department of Preventive Medicine, School of Medicine, Shihezi University, North 2th Road, Shihezi, Xinjiang, 832000, People's Republic of China
- Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
| | - Qiang Niu
- Department of Preventive Medicine, School of Medicine, Shihezi University, North 2th Road, Shihezi, Xinjiang, 832000, People's Republic of China.
- Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China.
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China.
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China.
| |
Collapse
|
8
|
Orta Yilmaz B, Aydin Y. Disruption of Leydig cell steroidogenic function by sodium arsenite and/or sodium fluoride. Theriogenology 2022; 193:146-156. [PMID: 36182826 DOI: 10.1016/j.theriogenology.2022.09.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 09/08/2022] [Accepted: 09/19/2022] [Indexed: 11/18/2022]
Abstract
Arsenite (As) and fluoride (F), both of which are linked to a variety of human ailments, are regularly found in underground drinking water. Numerous studies have shown that As and/or F have negative impacts on testicular function and fertility. For this purpose, mouse Leydig cells, the main cells responsible for the generation and regulation of steroid hormones such as testosterone, were used to reveal the effects of individual and combined exposure of As and F on the steroidogenic pathway in the male reproductive system. Leydig cells were treated with 0.39 μM (50 ppb) As and 0.0476 mM (2 ppm) F alone and in combination for 24 h. The findings revealed that As and/or F exposure induced oxidative stress and apoptosis in Leydig cells and altered antioxidant equilibrium of the cells by reducing superoxide dismutase, catalase, glutathione peroxidase. Additionally, individual and combined administration of As and/or F significantly supressed the expression of both steroidogenic enzymes and the genes encoding these enzymes. In conclusion, this study showed that exposure to As and F at environmentally relevant concentrations dispersed by water decreased testosterone production in Leydig cells, an important cell of the male reproductive system. The deleterious effects of even the lowest concentrations of As and F elements that can reach humans from the environment on the Leydig cell, and therefore on male infertility, emphasize necessity new safe limits for these elements.
Collapse
Affiliation(s)
- Banu Orta Yilmaz
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey.
| | - Yasemin Aydin
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey
| |
Collapse
|
9
|
Wang H, Yang L, Gao P, Deng P, Yue Y, Tian L, Xie J, Chen M, Luo Y, Liang Y, Qing W, Zhou Z, Pi H, Yu Z. Fluoride exposure induces lysosomal dysfunction unveiled by an integrated transcriptomic and metabolomic study in bone marrow mesenchymal stem cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 239:113672. [PMID: 35617906 DOI: 10.1016/j.ecoenv.2022.113672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 06/15/2023]
Abstract
Fluoride has received much attention for its predominant bone toxicity in the human body. However, the toxic mechanism of bone injury caused by fluoride exposure remains largely unclear. Bone marrow mesenchymal stem cells (BMSCs) are widely used as model cells for evaluating bone toxicity after environmental toxicant exposure. In this study, BMSCs were exposed to fluoride at 1, 2, and 4 mM for 24 h, and fluoride significantly inhibited cell viability at 2 and 4 mM. A multiomics analysis combining transcriptomics with metabolomics was employed to detect alterations in genes and metabolites in BMSCs treated with 2 mM fluoride. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of transcriptomics profiles identified "lysosomes" as the top enriched pathway, which was severely damaged by fluoride exposure. Lysosomal damage was indicated by decreases in the expression of lysosomal associated membrane protein 2 (LAMP 2) and cathepsin B (CTSB) as well as an increase in pH. Upregulation of the lysosome-related genes Atp6v0b and Gla was observed, which may be attributed to a compensatory lysosomal biogenesis transcriptional response. Interestingly, inhibition of glutathione metabolism was observed in fluoride-treated BMSCs at the metabolomic level. Moreover, an integrative analysis between altered genes, metabolites and lysosome signaling pathways was conducted. Palmitic acid, prostaglandin C2, and prostaglandin B2 metabolites were positively associated with Atp6v0b, a lysosome-related gene. Overall, our results provide novel insights into the mechanism responsible for fluoride-induced bone toxicity.
Collapse
Affiliation(s)
- Hui Wang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Lu Yang
- Hunan Province Prevention and Treatment Hospital for Occupational Diseases, Hunan, China
| | - Peng Gao
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Ping Deng
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Yang Yue
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Li Tian
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Jia Xie
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Mengyan Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Yan Luo
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Yidan Liang
- School of Medicine, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Weijia Qing
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China; The 63710th Military Hospital of PLA, Xinzhou, Shanxi, China
| | - Zhou Zhou
- Department of Environmental Medicine, School of Public Health, and Department of Emergency Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huifeng Pi
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China.
| | - Zhengping Yu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China.
| |
Collapse
|
10
|
Aydin Y, Orta-Yilmaz B. Synergistic effects of arsenic and fluoride on oxidative stress and apoptotic pathway in Leydig and Sertoli cells. Toxicology 2022; 475:153241. [PMID: 35714946 DOI: 10.1016/j.tox.2022.153241] [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: 03/30/2022] [Revised: 06/08/2022] [Accepted: 06/11/2022] [Indexed: 11/15/2022]
Abstract
Excessive intake of arsenic (As) and fluoride (F), which are present in underground drinking water, have adverse effects on human health, and especially on the male reproductive system. In this regard, it's critical to figure out how As and F affect Leydig and Sertoli cells, which are key cells in the male reproductive system. The goal of this study was to determine the synergistic effects of co-exposure of As and F, via drinking water, on Leydig and Sertoli cells, which are models for the male reproductive system, as well as the mechanisms underlying these effects in terms of oxidative damage and apoptosis. Leydig and Sertoli cells were exposed to concentrations of 7.7 µM (0.57 ppm) As and 0.4 mM (7.24 ppm) F based on the highest daily intake of drinking water for 24 h. The present results revealed that As and/or F treatment reduced cell viability and proliferation in Leydig and Sertoli cells, elevated lactate dehydrogenase, a cytotoxicity marker, and triggered oxidative stress and apoptosis. Furthermore, it has been proven that when As and F are exposed in combination, they have a synergistic effect. In conclusion, by revealing the harmful effects of As and F on Leydig and Sertoli cells, and thus on male infertility, it is possible to reduce As and F exposure to prevent infertility after exposure to these molecules not only separately but also together. It will be considered to determine new action and action plans to reduce As and F exposure.
Collapse
Affiliation(s)
- Yasemin Aydin
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey.
| | - Banu Orta-Yilmaz
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey
| |
Collapse
|