1
|
Xiao C, Lai D. Impact of oxidative stress induced by heavy metals on ovarian function. J Appl Toxicol 2025; 45:107-116. [PMID: 38938153 PMCID: PMC11634564 DOI: 10.1002/jat.4664] [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: 03/12/2024] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 06/29/2024]
Abstract
As a crucial organ of the female reproductive system, the ovary has both reproductive and endocrine functions. Oxidative stress refers to an increase in intracellular reactive oxygen species (ROS), which play a role in the normal physiological activity of the ovary. However, excessive ROS can cause damage to the ovary. With the advancement of human industrial activities, heavy metal pollution has become increasingly severe. Heavy metals cause oxidative stress through both direct and indirect mechanisms, leading to changes in signal transduction pathways that damage the ovaries. This review aims to outline the adverse effects of oxidative stress on the ovaries triggered by heavy metals such as copper, arsenic, cadmium, mercury, and lead. The detrimental effects of heavy metals on ovaries include follicular atresia and decreased estrogen production in experimental animals, and they also cause premature ovarian insufficiency in women. Additionally, this review discusses the role of antioxidants, provides some treatment methods, summarizes the limitations of current research, and offers perspectives for future research directions.
Collapse
Affiliation(s)
- Chengqi Xiao
- The International Peace Maternity and Child Health Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Embryo Original DiseasesShanghaiChina
| | - Dongmei Lai
- The International Peace Maternity and Child Health Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Embryo Original DiseasesShanghaiChina
| |
Collapse
|
2
|
Wang C, Wang B, Wei Y, Li S, Ren J, Dai Y, Liu G. Effect of Gentianella acuta (Michx.) Hulten against the arsenic-induced development hindrance of mouse oocytes. Biometals 2024; 37:1411-1430. [PMID: 38814492 DOI: 10.1007/s10534-024-00613-1] [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: 10/02/2023] [Accepted: 05/18/2024] [Indexed: 05/31/2024]
Abstract
The current study was designed to investigate the alleviative effect of Gentianella acuta (Michx.) Hulten (G. acuta) against the sodium arsenite (NaAsO2)-induced development hindrance of mouse oocytes. For this purpose, the in vitro maturation (IVM) of mouse cumulus-oocyte complexes (COCs) was conducted in the presence of NaAsO2 and G. acuta, followed by the assessments of IVM efficiency including oocyte maturation, spindle organization, chromosome alignment, cytoskeleton assembly, cortical granule (CGs) dynamics, redox regulation, epigenetic modification, DNA damage, and apoptosis. Subsequently, the alleviative effect of G. acuta intervention on the fertilization impairments of NaAsO2-exposed oocytes was confirmed by the assessment of in vitro fertilization (IVF). The results showed that the G. acuta intervention effectively ameliorated the decreased maturation potentials and fertilization deficiency of NaAsO2-exposed oocytes but also significantly inhibited the DNA damages, apoptosis, and altered H3K27me3 expression level in the NaAsO2-exposed oocytes. The effective effects of G. acuta intervention against redox dysregulation including mitochondrial dysfunctions, accumulated reactive oxygen species (ROS) generation, glutathione (GSH) deficiency, and decreased adenosine triphosphate (ATP) further confirmed that the ameliorative effects of G. acuta intervention against the development hindrance of mouse oocytes were positively related to the antioxidant capacity of G. acuta. Evidenced by these abovementioned results, the present study provided fundamental bases for the ameliorative effect of G. acuta intervention against the meiotic defects caused by the NaAsO2 exposure, benefiting the future application potentials of G. acuta intervention in these nutritional and therapeutic research for attenuating the outcomes of arseniasis.
Collapse
Affiliation(s)
- Chunyu Wang
- Key Laboratory of Medical Cell Biology, Clinical Medicine Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010030, Inner Mongolia, China
- Department of Environmental Science and Engineering, Inner Mongolia University of Technology, Hohhot, 010051, Inner Mongolia, China
| | - Biao Wang
- Animal Husbandry Institute, Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, Hohhot, 010031, Inner Mongolia, China
| | - Ying Wei
- Key Laboratory of Medical Cell Biology, Clinical Medicine Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010030, Inner Mongolia, China
| | - Shubin Li
- Department of Geriatric Medical Center, Inner Mongolia People's Hospital, Hohhot, 010010, Inner Mongolia, China
| | - Jingyu Ren
- College of Life Science, Inner Mongolia University, Hohhot, 010070, Inner Mongolia, China
| | - Yanfeng Dai
- College of Life Science, Inner Mongolia University, Hohhot, 010070, Inner Mongolia, China
| | - Gang Liu
- Key Laboratory of Medical Cell Biology, Clinical Medicine Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010030, Inner Mongolia, China.
| |
Collapse
|
3
|
Han S, Yu C, Qiu M, Xiong X, Peng H, Song X, Hu C, Zhang Z, Xia B, Yang L, Chen J, Zhu S, Li W, Yang C. USP13 regulates ferroptosis in chicken follicle granulosa cells by deubiquitinating ATG7. Poult Sci 2024; 103:104209. [PMID: 39214053 PMCID: PMC11402030 DOI: 10.1016/j.psj.2024.104209] [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: 06/27/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
The development and maturation of follicles are intricately linked to egg production and reproductive performance of chickens. Granulosa cells death directly affects the development and maturation of follicles, thereby impacting the reproductive performance of hens. Ferroptosis is a new type of cell death, it is unknown how it affects the growth and development of chicken follicles. In this study, RNA-seq analysis revealed significant differences in the expression of ferroptosis-related genes between normal follicles and atretic follicles, suggesting a potential role for ferroptosis in follicle growth and development. In addition, we found that ubiquitin-specific protease 13 (USP13) was significantly upregulated in atrophic follicles. Overexpression of USP13 results in depletion of glutathione (GSH), peroxidation of lipids, accumulation of iron, and activation of ferroptosis in chicken granulosa cells. In contrast, USP13 knockdown significantly inhibited ferroptosis events. Mechanistically, USP13 prevents the degradation of autophagy related 7 (ATG7) by deubiquitinating it, thereby enhancing the stability of ATG7 protein and ultimately promoting ferroptosis. In conclusion, this study elucidates the crucial role of the USP13-ATG7 axis in regulating ferroptosis in chicken follicle granulosa cells, thereby presenting a novel avenue for molecular breeding research in chickens.
Collapse
Affiliation(s)
- Shunshun Han
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Chunlin Yu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Mohan Qiu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Xia Xiong
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Han Peng
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Xiaoyan Song
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Chenming Hu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Zengrong Zhang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Bo Xia
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Li Yang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Jialei Chen
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Shiliang Zhu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Wen Li
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Chaowu Yang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China.
| |
Collapse
|
4
|
Ding N, Wang X, Harlow SD, Randolph JF, Gold EB, Park SK. Heavy Metals and Trajectories of Anti-Müllerian Hormone During the Menopausal Transition. J Clin Endocrinol Metab 2024; 109:e2057-e2064. [PMID: 38271266 DOI: 10.1210/clinem/dgad756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Indexed: 01/27/2024]
Abstract
BACKGROUND Experimental and epidemiological studies have linked metals with women's reproductive aging, but the mechanisms are not well understood. Disrupted ovarian folliculogenesis and diminished ovarian reserve could be a pathway through which metals impact reproductive hormones and outcomes. OBJECTIVE The study aimed to evaluate the associations of heavy metals with anti-Müllerian hormone (AMH), a marker of ovarian reserve. METHODS The study included 549 women from the Study of Women's Health Across the Nation with 2252 repeated AMH measurements from 10 to 0 years before the final menstrual period (FMP). Serum AMH concentrations were measured using picoAMH ELISA. Urinary concentrations of arsenic, cadmium, mercury, and lead were measured using high-resolution inductively coupled plasma mass spectrometry. Multivariable linear mixed regressions modeled AMH as a function of time before the FMP interaction terms between metals and time to the FMP were also included. RESULTS Adjusting for confounders, compared with those in the lowest tertile, women in the highest tertile of urinary arsenic or mercury concentrations had lower AMH concentrations at the FMP (percent change: -32.1%; 95% CI, -52.9 to -2.2, P-trend = .03 for arsenic; percent change: -40.7%; 95% CI, -58.9 to -14.5, P-trend = .005 for mercury). Higher cadmium and mercury were also associated with accelerated rates of decline in AMH over time (percent change per year: -9.0%; 95% CI, -15.5 to -1.9, P-trend = .01 for cadmium; -7.3%; 95% CI, -14.0 to -0.1, P-trend = .04 for mercury). CONCLUSION Heavy metals including arsenic, cadmium, and mercury may act as ovarian toxicants by diminishing ovarian reserve in women approaching the FMP.
Collapse
Affiliation(s)
- Ning Ding
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xin Wang
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Siobán D Harlow
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - John F Randolph
- Department of Obstetrics and Gynecology, School of Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ellen B Gold
- Department of Public Health Sciences, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| | - Sung Kyun Park
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
5
|
Adeogun AE, Ogunleye OD, Akhigbe TM, Oyedokun PA, Adegbola CA, Saka WA, Afolabi OA, Akhigbe RE. Impact of arsenic on male and female reproductive function: a review of the pathophysiology and potential therapeutic strategies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03452-6. [PMID: 39287676 DOI: 10.1007/s00210-024-03452-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
Arsenic is a ubiquitous metalloid and heavy metal that contributes to the global decline in human fertility. Humans are constantly exposed to arsenic through biotic and abiotic sources, especially ingestion of arsenic-contaminated food and water. Its exposure is associated with several adverse health challenges, including reproductive toxicity. In spite of its reported adverse effects, arsenic exposure remains a global challenge. Hence, this study provides a comprehensive review of the literature on the impact and mechanism of arsenic on male and female reproductive function. Additionally, a review of the potential therapeutic strategies is presented. Evidence from the literature reveals that arsenic upregulates reactive oxygen species (ROS) generation which mediates arsenic-induced suppression of the hypothalamic-pituitary-gonadal axis and inactivation of 3β-HSD and 17β-HSD activities, leading to reduced gonadal steroidogenesis. Through several oxidative stress-dependent signaling, arsenic induces the apoptosis of the germ cells, thus contributing to the development of infertility. At the moment, there is no specific treatment for arsenic-induced reproductive toxicity. However, increasing data form the scientific literature reveals the benefits of antioxidants in ameliorating arsenic-induced reproductive toxicity. These molecules suppress ROS generation and maintain optimal activities of the hypothalamic-pituitary-gonadal axis, leading to optimal steroidogenesis and gametogenesis as well as improved germ cells. Overall, this study revealed the impact and associated mechanism of arsenic-induced reproductive toxicity. It also provides evidence from the literature demonstrating potential therapeutic measures in managing arsenic-induced reproductive toxicity.
Collapse
Affiliation(s)
- A E Adeogun
- Department of Physiology, Babcock University, Ilishan Remo, Ogun State, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
| | - O D Ogunleye
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - T M Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
- Department of Agronomy, Osun State University, Ejigbo Campus, Osogbu, Osun State, Nigeria
| | - P A Oyedokun
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - C A Adegbola
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - W A Saka
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - O A Afolabi
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - R E Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria.
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria.
| |
Collapse
|
6
|
Liao T, Ni F, Yang X, Liu J, Xia L, Yang Q, Gao X, Li C, Wang X, Wu C, Wang L, Bao S, Pan G, Liang C, Jiang H, Tao F, Shao S. Couples' preconception urinary essential trace elements concentration and spontaneous abortion risk: A nested case-control study in a community population. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116764. [PMID: 39067081 DOI: 10.1016/j.ecoenv.2024.116764] [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: 04/18/2024] [Revised: 07/13/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Previous studies have indicated a correlation between maternal imbalances in essential trace elements during pregnancy and the occurrence of spontaneous abortion (SA). Nonetheless, the impact of these elements from both partners and during the preconception period remains unexplored. OBJECTIVE This study sought to evaluate the relationship between preconception essential trace elements and spontaneous abortion (SA) based on husband-wife dyads. METHODS This study selected 390 couples with spontaneous abortion (SA) and 390 matched couples with live births from a preconception cohort of 33,687 couples. Urine samples collected prior to pregnancy were analyzed for ten essential trace elements (Se, Cr, Mo, Cu, Zn, Fe, Mn, V, Co, and Ni) using inductively coupled plasma mass spectrometry (ICP-MS). RESULTS Multivariate conditional logistic regression analysis identified that elevated concentrations of Zn (OR = 0.73) and Ni (OR = 0.69) in couples were associated with a reduced risk of SA, whereas elevated levels of Cr (OR = 1.30) and Mn (OR = 1.39) were linked to an increased risk. Restricted cubic spline models suggested a U-shaped association between couples' Cu and Co concentrations and SA. Bayesian Kernel Machine Regression further supported a U-shaped relationship between the mixture of ten elements and SA, showing significant protection at the 50th and 55th percentiles compared to the 10th percentile. Additionally, the effects of Cr, Zn, Mn, and Ni on SA varied when the concentrations of the other nine elements were held constant at their 25th, 50th, and 75th percentiles. Stratified analysis revealed that maternal Cu (OR = 0.43) and Fe (OR = 0.63) reduced the risk of SA when paternal Cu and Fe were in the lower quartile. Conversely, maternal Cu (OR = 2.03) and Fe (OR = 1.77) increased the risk of SA when paternal concentrations were in the higher quartile. Similar patterns were observed for Cr, Mn, Co, and Zn. CONCLUSION Elevated urinary concentrations of Zn and Ni in couples were associated with a reduced risk of SA, while higher levels of Cr and Mn were linked to an increased risk. Cu, Co, and a mixture of ten essential trace elements exhibited a U-shaped relationship with SA. The impact of certain essential trace elements (Cu, Fe, Cr, Mn, Co, and Zn) on SA in one partner was influenced by their concentrations in the other partner.
Collapse
Affiliation(s)
- Tierong Liao
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui, China
| | - Feng Ni
- Reproductive Medicine Center, the 901th Hospital of the Joint Logistics Support Force of People's Liberation Army, Hefei, Anhui, China
| | - Xinliu Yang
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui, China
| | - Junjun Liu
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui, China
| | - Luobin Xia
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui, China
| | - Qianhui Yang
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui, China
| | - Xin Gao
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui, China
| | - Chaojie Li
- NHC Key Laboratory of Birth Defects Prevention, Zhenzhou, Henan, China; Henan Key Laboratory of Population Defects Prevention, Zhenzhou, Henan, China
| | - Xuemei Wang
- Reproductive Medicine Center, the 901th Hospital of the Joint Logistics Support Force of People's Liberation Army, Hefei, Anhui, China
| | - Caiyun Wu
- Reproductive Medicine Center, the 901th Hospital of the Joint Logistics Support Force of People's Liberation Army, Hefei, Anhui, China
| | - Liuchang Wang
- School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Shuangshuang Bao
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui, China
| | - Guixia Pan
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui, China
| | - Chunmei Liang
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui, China
| | - Hong Jiang
- Reproductive Medicine Center, the 901th Hospital of the Joint Logistics Support Force of People's Liberation Army, Hefei, Anhui, China.
| | - Fangbiao Tao
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui, China.
| | - Shanshan Shao
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; NHC Key Laboratory of Birth Defects Prevention, Zhenzhou, Henan, China; Henan Key Laboratory of Population Defects Prevention, Zhenzhou, Henan, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui Province, China.
| |
Collapse
|
7
|
Eslami H, Askari FR, Mahdavi M, Taghavi M, Ghaseminasab-Parizi M. Environmental arsenic exposure and reproductive system toxicity in male and female and mitigatory strategies: a review. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2024; 46:420. [PMID: 39269655 DOI: 10.1007/s10653-024-02197-8] [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: 03/11/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
Environmental Arsenic (As) exposure is one of the main health challenges in different area of the world. As is a significant factor responsible to the reproductive system toxicity in both male and female. In this study, the most important effects mechanisms and biomarkers related to environmental exposure to As and the reproductive system toxicity, and infertility risk are reviewed in male and female. The results showed that the most important As-induced reproductive system toxicity in the male were alteration in the quantity and quality of semen, testicular toxicity, oxidative stress, testosterone reduction, and sperm apoptosis. For female were oxidative stress, spontaneous miscarriage, reproductive cycle disruption, decrease in the estradiol, progesterone, and testosterone levels and impair fecundity. The main mechanisms of reproductive system toxicity caused by As exposure in male were, genotoxic effects, reduction of glutathione, disruption of sex hormones, sperm flagellum formation impairment, inhibition of spermatogenesis, disruption of cell signaling pathways, and metabolites disruption. For female were abnormal signaling in gene expression, hormonal homeostasis, As-accumulation in placental tissue and creation of reactive oxygen, disruption in the neurotransmitters balance, and sex hormones disruption. The suitable biomarkers for As-induced reproductive toxicity in male were changes in testosterone, one-carbon and lipid metabolism, noncoding RNAs, and steroid hormone homeostasis, and for female was human chorionic gonadotropin (hCG) changes. Finaly, taking selenium, zinc, silymarin, vitamins (C and E) and phytonutrients can be effective in reducing the As-induced reproductive system toxicity and infertility risk.
Collapse
Affiliation(s)
- Hadi Eslami
- Occupational Environment Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
- Department of Environmental Health Engineering, School of Health, Occupational Safety and Health Research Center, NICICO, World Safety Organization and Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Foad Ranjbar Askari
- Student Research Committee, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Masoumeh Mahdavi
- Student Research Committee, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mahmoud Taghavi
- Department of Environment Health Engineering, School of Health, Social Determinates of Health Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Maryam Ghaseminasab-Parizi
- Department of Health Education and Health Promotion, School of Health, Occupational Environment Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
8
|
K N, P J, Nalla SV, Dubey I, Kushwaha S. Arsenic-Induced Thyroid Hormonal Alterations and Their Putative Influence on Ovarian Follicles in Balb/c Mice. Biol Trace Elem Res 2024; 202:4087-4100. [PMID: 38093019 DOI: 10.1007/s12011-023-03988-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/28/2023] [Indexed: 07/18/2024]
Abstract
Thyroid issues are common among women in their reproductive years, and women with thyroid dysfunction often encounter challenges with fertility. Arsenic is known for its toxic effects on the thyroid and ovaries, investigated independently. However, there is no known study directly or indirectly addressing the association between arsenic, thyroid function, and ovarian reserve. This study aims to investigate the effect of arsenic on thyroid function and its possible implications on ovarian follicular reserve. Female Balb/c mice were given sodium arsenite (0.2 ppm, 2 ppm, and 20 ppm) via drinking water for 30 days. Findings showed that arsenic decreased thyroid hormone levels (fT3 and fT4) while increasing TSH levels, which might have led to elevated levels of FSH and LH. Furthermore, arsenic treatment not only decreased thyroid follicle sizes but also altered the ovarian follicular count. The finding demonstrates that arsenic significantly reduced the expression of LAMP1, a lysosomal marker protein. This reduction leads to increased lysosomal permeability in the thyroid, resulting in a significant release of cathepsin B. These changes led to hypothyroidism, which might indirectly affect the ovaries. Also, the elevated levels of growth differentiation factor-8 in arsenic-treated ovaries indicate impaired folliculogenesis and ovulation. Furthermore, arsenic significantly increased the expressions of pAkt and pFoxo3a, implying that arsenic accelerated the activation of the primordial follicular pools. In conclusion, arsenic disrupts lysosomal stabilization, potentially leading to a decline in circulating fT3 and fT4 levels. This disturbance could, in turn, affect the estrous cycle and may alter the pattern of follicular development.
Collapse
Affiliation(s)
- Nandheeswari K
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, Uttar Pradesh, 226002, India
| | - Jayapradha P
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, Uttar Pradesh, 226002, India
| | - Sree Vaishnavi Nalla
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, Uttar Pradesh, 226002, India
| | - Itishree Dubey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, Uttar Pradesh, 226002, India
| | - Sapana Kushwaha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, Uttar Pradesh, 226002, India.
| |
Collapse
|
9
|
Ommati MM, Nozhat Z, Sabouri S, Kong X, Retana-Márquez S, Eftekhari A, Ma Y, Evazzadeh F, Juárez-Rojas L, Heidari R, Wang HW. Pesticide-Induced Alterations in Locomotor Activity, Anxiety, and Depression-like Behavior Are Mediated through Oxidative Stress-Related Autophagy: A Persistent Developmental Study in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:11205-11220. [PMID: 38708789 DOI: 10.1021/acs.jafc.4c02299] [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: 05/07/2024]
Abstract
Chlorpyrifos (CPF), dichlorvos (DDV), and cypermethrin (CP), as commonly used pesticides, have been implicated in inducing neuropsychiatric disorders, such as anxiety, depression-like behaviors, and locomotor activity impairment. However, the exact molecular mechanisms of these adverse effects, particularly in both sexes and their next-generation effects, remain unclear. In this study, we conducted behavioral analysis, along with cellular assays (monodansylcadaverine staining) and molecular investigations (qRT-PCR and western blotting of mTOR, P62, and Beclin-1) to clear the potential role of autophagy in pesticide-induced behavioral alterations. For this purpose, 42 adult female and 21 male inbred ICR mice (F0) were distributed into seven groups. Maternal mice (F0) and 112 F1 offspring were exposed to 0.5 and 1 ppm of CPF, DDV, and CP through drinking water. F1 male and female animals were studied to assess the sex-specific effects of pesticides on brain tissue. Our findings revealed pronounced anxiogenic effects and impaired locomotor activity in mice. F1 males exposed to CPF (1 ppm) exhibited significantly elevated depression-like behaviors compared to other groups. Moreover, pesticide exposure reduced mTOR and P62 levels, while enhancing the Beclin-1 gene and protein expression. These changes in autophagy signaling pathways, coupled with oxidative and neurogenic damage in the cerebral cortex and hippocampus, potentially contribute to heightened locomotor activity, anxiety, and depression-like behaviors following pesticide exposure. This study underscores the substantial impact of pesticides on both physiological and behavioral aspects, emphasizing the necessity for comprehensive assessments and regulatory considerations for pesticide use. Additionally, the identification of sex-specific responses presents a crucial dimension for pharmaceutical sciences, highlighting the need for tailored therapeutic interventions and further research in this field.
Collapse
Affiliation(s)
- Mohammad Mehdi Ommati
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471000, China
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz 71468 64685, Iran
| | - Zahra Nozhat
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Samira Sabouri
- College of Animal Science and Veterinary, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Xiangdong Kong
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Socorro Retana-Márquez
- Department of Reproductive Biology, Universidad Autónoma Metropolitana-Iztapalapa, México City 09340, Mexico
| | - Aziz Eftekhari
- Department of Biochemistry, Faculty of Science, Ege University, Izmir 35100, Turkey
| | - Yanqin Ma
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Fatemeh Evazzadeh
- Department of Psychology, Science & Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | - Lizbeth Juárez-Rojas
- Department of Reproductive Biology, Universidad Autónoma Metropolitana-Iztapalapa, México City 09340, Mexico
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz 71468 64685, Iran
| | - Hong-Wei Wang
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471000, China
| |
Collapse
|
10
|
Xie P, Li P, Zhu X, Chen D, Ommati MM, Wang H, Han L, Xu S, Sun P. Hepatotoxic of polystyrene microplastics in aged mice: Focus on the role of gastrointestinal transformation and AMPK/FoxO pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 917:170471. [PMID: 38296072 DOI: 10.1016/j.scitotenv.2024.170471] [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: 10/12/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/05/2024]
Abstract
Microplastic (MP) toxicity has attracted widespread attention, whereas before triggering hepatotoxicity, ingested MPs first undergo transportation and digestion processes in the gastrointestinal tract, possibly interacting with the gastrointestinal contents (GIC). More alarming is the need for more understanding of how this process may impact the liver health of aged animals. This study selected old mice. Firstly, we incubated polystyrene microplastics (PS-MPs, 1 μm) with GIC extract. The results of SEM/EDS indicated a structural alteration in PS-MPs. Additionally, impurities resembling corona, rich in heteroatoms (O, N, and S), were observed. This resulted in an enhanced aggregating phenomenon of MPs. We conducted a 10-day experiment exposing aged mice to four concentrations of PS-MPs, ranging from 1 × 103 to 1 × 1012 particles/L. Subsequent measurements of tissue pathology and body and organ weights were conducted, revealing alterations in liver structure. In the liver, 12 crucial metabolites were found by LC-MS technology, including purines, lipids, and amino acids. The AMPK/FoxO pathway was enriched, activated, and validated in western blotting results. We also comprehensively examined the innate immune system, inflammatory factors, and oxidative stress indicators. The results indicated decreased C3 levels, stable C4 levels, inflammatory factors (IL-6 and IL-8), and antioxidant enzymes were increased to varying degrees. PS-MPs also caused DNA oxidative damage. These toxic effects exhibited a specific dose dependence. Overall, after the formation of the gastrointestinal corona, PS-MPs subsequently impact various cellular processes, such as cycle arrest (p21), leading to hepatic and health crises in the elderly. The presence of gastrointestinal coronas also underscores the MPs' morphology and characteristics, which should be distinguished after ingestion.
Collapse
Affiliation(s)
- Pengfei Xie
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Pengcheng Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Xiaoshan Zhu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Deshan Chen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Mohammad Mehdi Ommati
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Hongwei Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Lei Han
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Shixiao Xu
- Northwest Institute of Plateau Biology Chinese Academy of Sciences, Xining, Qinghai 810008, China
| | - Ping Sun
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China.
| |
Collapse
|
11
|
Yan M, Wang H, Wei R, Li W. Arsenic trioxide: applications, mechanisms of action, toxicity and rescue strategies to date. Arch Pharm Res 2024; 47:249-271. [PMID: 38147202 DOI: 10.1007/s12272-023-01481-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 12/15/2023] [Indexed: 12/27/2023]
Abstract
Arsenical medicine has obtained its status in traditional Chinese medicine for more than 2,000 years. In the 1970s, arsenic trioxide was identified to have high efficacy and potency for the treatment of acute promyelocytic leukemia, which promoted many studies on the therapeutic effects of arsenic trioxide. Currently, arsenic trioxide is widely used to treat acute promyelocytic leukemia and various solid tumors through various mechanisms of action in clinical practice; however, it is accompanied by a series of adverse reactions, especially cardiac toxicity. This review presents a comprehensive overview of arsenic trioxide from preclinical and clinical efficacy, potential mechanisms of action, toxicities, and rescue strategies for toxicities to provide guidance or assistance for the clinical application of arsenic trioxide.
Collapse
Affiliation(s)
- Meng Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China.
| | - Hao Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Rui Wei
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Pharmacy Department, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenwen Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
12
|
Ommati MM, Sabouri S, Sun Z, Zamiri MJ, Retana-Marquez S, Nategh Ahmadi H, Zuo Q, Eftekhari A, Juárez-Rojas L, Asefi Y, Lei L, Cui SG, Jadidi MH, Wang HW, Heidari R. Inactivation of Mst/Nrf2/Keap1 signaling flexibly mitigates MAPK/NQO-HO1 activation in the reproductive axis of experimental fluorosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 271:115947. [PMID: 38215664 DOI: 10.1016/j.ecoenv.2024.115947] [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: 09/12/2023] [Revised: 12/30/2023] [Accepted: 01/03/2024] [Indexed: 01/14/2024]
Abstract
Fluoride induced reprotoxicity through oxidative stress-mediated reproductive cell death. Hence, the current study evaluated the importance of the MST/Nrf2/MAPK/NQO-HO1 signaling pathway in fluorosis-induced reproductive toxicity. For this purpose, the reproductive toxicity of sodium fluoride (NaF) at physiological, biochemical, and intracellular levels was evaluated. In-vivo, NaF at 100 mg/L instigated physiological dysfunction, morphological, stereological, and structural injuries in the gut-gonadal axis of fluorosis mice through weakening the antioxidant signaling, Nrf2/HO-1/NQO1signaling pathway, causing the gut-gonadal barrier disintegrated via oxidative stress-induced inflammation, mitochondrial damage, apoptosis, and autophagy. Similar trends were also observed in-vitro in the isolated Leydig cells (LCs) challenging with 20 mg/L NaF. Henceforth, activating the cellular antioxidant signaling pathway, Nrf2/HO-1/NQO1, inactivating autophagy and apoptosis, or attenuating lipopolysaccharide (LPS) can be the theoretical basis and valuable therapeutic targets for coping with NaF-induced reproductive toxicity.
Collapse
Affiliation(s)
- Mohammad Mehdi Ommati
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471000, China; Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Samira Sabouri
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Zilong Sun
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, China
| | | | - Socorro Retana-Marquez
- Department of Biology of Reproduction, Autonomous Metropolitan University-Iztapalapa, Mexico City, Mexico
| | - Hassan Nategh Ahmadi
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, China; College of Animal Science and Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Qiyong Zuo
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Aziz Eftekhari
- Department of Biochemistry, Faculty of Science, Ege University, Izmir, Turkey; Nanotechnology and Biochemical Toxicology (NBT) Center, Azerbaijan State University of Economics (UNEC), Baku AZ1001, Azerbaijan
| | - Lizbeth Juárez-Rojas
- Department of Biology of Reproduction, Autonomous Metropolitan University-Iztapalapa, Mexico City, Mexico
| | - Yaser Asefi
- Department of Genetics, Ahar Branch, Islamic Azad University, Ahar, Iran
| | - Lina Lei
- The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Shu-Gang Cui
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Mohammad Hasan Jadidi
- Comparative Medicine and Animal Resources Centre, McGill University, Montreal, Canada
| | - Hong-Wei Wang
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471000, China.
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
13
|
Singh C, Saini M, Jain A, Lad D, Prakash G, Khadwal A, Naseem S, Malhotra P. Fertility and parenthood in patients with acute promyelocytic leukemia treated with Arsenic Trioxide and All-Trans retinoic acid. Blood Cancer J 2024; 14:14. [PMID: 38238308 PMCID: PMC10796907 DOI: 10.1038/s41408-024-00984-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 01/22/2024] Open
Affiliation(s)
- Charanpreet Singh
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Manpreet Saini
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arihant Jain
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Deepesh Lad
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Gaurav Prakash
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Alka Khadwal
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Shano Naseem
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pankaj Malhotra
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
14
|
Ommati MM, Rezaei H, Socorro RM, Tian W, Zhao J, Rouhani A, Sabouri S, Ghaderi F, Niknahad AM, Najibi A, Mazloomi S, Safipour M, Honarpishefard Z, Wang HW, Niknahad H, Heidari R. Pre/postnatal taurine supplementation improves neurodevelopment and brain function in mice offspring: A persistent developmental study from puberty to maturity. Life Sci 2024; 336:122284. [PMID: 38008208 DOI: 10.1016/j.lfs.2023.122284] [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: 10/06/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 11/28/2023]
Abstract
Taurine (TAU) is a sulfur-containing amino acid abundantly found in the human body. Endogenously, TAU is synthesized from cysteine in the liver. However, newborns rely entirely on TAU's dietary supply (milk). There is no investigation on the effect of long-term TAU administration on next-generation neurological development. The current study evaluated the effect of long-term TAU supplementation during the maternal gestational and litter weaning time on several neurological parameters in mice offspring. Moreover, the effects of TAU on mitochondrial function and oxidative stress biomarkers as plausible mechanisms of its action in the whole brain and hippocampus have been evaluated. TAU (0.5 % and 1 % w/v) was dissolved in the drinking water of pregnant mice (Day one of pregnancy), and amino acid supplementation was continued during the weaning time (post-natal day; PND = 21) until litters maturity (PND = 65). It was found that TAU significantly improved cognitive function, memory performance, reflexive motor activity, and emotional behaviors in F1-mice generation. TAU measurement in the brain and hippocampus revealed higher levels of this amino acid. TAU and ATP levels were also significantly higher in the mitochondria isolated from the whole brain and hippocampus. Based on these data, TAU could be suggested as a supplement during pregnancy or in pediatric formula. The effects of TAU on cellular mitochondrial function and energy metabolism might play a fundamental role in the positive effects of this amino acid observed in this investigation.
Collapse
Affiliation(s)
- Mohammad Mehdi Ommati
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, Henan, China; Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Heresh Rezaei
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Retana-Márquez Socorro
- Department of Reproductive Biology, Universidad Autónoma Metropolitana-Iztapalapa, México City, Mexico
| | - Weishun Tian
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, Henan, China
| | - Jing Zhao
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, Henan, China
| | - Ayeh Rouhani
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sabouri
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, Henan, China; College of Animal Science and Veterinary, Shanxi agricultural University, Taigu, Shanxi, China
| | - Fatemeh Ghaderi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Mohammad Niknahad
- Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asma Najibi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Chemistry and Biochemistry, Miami University, 244 Hughes Laboratories, 651 E. High Street, Oxford, OH 45056, USA
| | - Sahra Mazloomi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Moslem Safipour
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Honarpishefard
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hong-Wei Wang
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, Henan, China.
| | - Hossein Niknahad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Reproductive Biology, Universidad Autónoma Metropolitana-Iztapalapa, México City, Mexico.
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
15
|
Guo Z, Zhang Y, Huang A, Ni Q, Zeng C. Phenylbutyrate and Dichloroacetate Enhance the Liquid-Stored Boar Sperm Quality via PDK1 and PDK3. Int J Mol Sci 2023; 24:17091. [PMID: 38069413 PMCID: PMC10707026 DOI: 10.3390/ijms242317091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Artificial insemination (AI) with liquid-stored semen is the most prevalent and efficient assisted reproduction technique in the modern pork industry. Pyruvate dehydrogenase complex component X (PDHX) was demonstrated to be associated with sperm metabolism and affected the boar sperm viability, motility, and fertility. Pyruvate Dehydrogenase Kinases (PDKs) are the key metabolic enzymes that regulate pyruvate dehydrogenase complex (PDHC) activity and also the conversion from glycolysis to oxidative phosphorylation. In the present study, two PDK inhibitors, Dichloroacetate (DCA) and Phenylbutyrate (4-PBA), were added to an extender and investigated to determine their regulatory roles in liquid-stored boar sperm at 17 °C. The results indicated that PDK1 and PDK3 were predominantly located at the head and flagella of the boar sperm. The addition of 2 mM DCA and 0.5 mM 4-PBA significantly enhanced the sperm motility, plasma membrane integrity (PMI), mitochondrial membrane potential (MMP), and ATP content. In addition, DCA and 4-PBA exerted their effects by inhibiting PDK1 and PDK3, respectively. In conclusion, DCA and 4-PBA were found to regulate the boar sperm metabolic activities via PDK1 and PDK3. These both can improve the quality parameters of liquid-stored boar sperm, which will help to improve and optimize liquid-stored boar semen after their addition in the extender.
Collapse
Affiliation(s)
- Zhihua Guo
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China; (Z.G.); (Y.Z.); (Q.N.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
| | - Yan Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China; (Z.G.); (Y.Z.); (Q.N.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
| | - Anqi Huang
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China;
| | - Qingyong Ni
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China; (Z.G.); (Y.Z.); (Q.N.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
| | - Changjun Zeng
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China; (Z.G.); (Y.Z.); (Q.N.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
| |
Collapse
|
16
|
Mentese A, Demir S, Mungan SA, Alemdar NT, Demir EA, Aliyazicioglu Y. Gentisic acid ameliorates cisplatin-induced reprotoxicity through suppressing endoplasmic reticulum stress and upregulating Nrf2 pathway. Tissue Cell 2023; 85:102256. [PMID: 37918215 DOI: 10.1016/j.tice.2023.102256] [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: 07/17/2023] [Revised: 10/13/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023]
Abstract
Reproductive toxicity is a serious side effect of cisplatin (CP) chemotherapy. Gentisic acid (GTA) is a phenolic acid with strong antioxidant properties. Here, we aimed to determine therapeutic effect of GTA against CP-induced testicular toxicity in rats for the first time. Male Sprague-Dawley rats received a single dose of CP (5 mg/kg; intraperitoneal) and treated with GTA (1.5 and 3 mg/kg; intraperitoneal; 3 consecutive days). The levels of oxidative stress (OS), inflammation, endoplasmic reticulum stress (ERS) and apoptosis biomarkers were assessed in the testicular tissue of rats. In addition, how CP affects the nuclear factor erythroid-2-related factor 2 (Nrf2) pathway and the effect of GTA on this situation were also addressed in the testicular tissue. CP administration induced histopathological changes in testicular tissue of rats with a significant increase in OS, inflammation, ERS and apoptosis biomarkers and a decrease in antioxidant capacity and Nrf2 expression levels. Administrations of GTA resulted in an amelioration of these altered parameters. These data suggest that GTA may be a potential therapeutic agent against CP-induced testicular toxicity. Activation of the Nrf2 pathway plays a key role of this therapeutic effect of GTA.
Collapse
Affiliation(s)
- Ahmet Mentese
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, 61080 Trabzon, Turkey
| | - Selim Demir
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Karadeniz Technical University, 61080 Trabzon, Turkey.
| | - Sevdegul Aydin Mungan
- Department of Medical Pathology, Faculty of Medicine, Karadeniz Technical University, 61080 Trabzon, Turkey
| | - Nihal Turkmen Alemdar
- Department of Medical Biochemistry, Graduate School of Health Sciences, Karadeniz Technical University, 61080 Trabzon, Turkey; Department of Medical Services and Techniques, Vocational School of Health Services, Recep Tayyip Erdogan University, 53100 Rize, Turkey
| | - Elif Ayazoglu Demir
- Department of Chemistry and Chemical Processing Technologies, Macka Vocational School, Karadeniz Technical University, 61750 Trabzon, Turkey
| | - Yuksel Aliyazicioglu
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, 61080 Trabzon, Turkey
| |
Collapse
|
17
|
Niknahad H, Mobasheri A, Arjmand A, Rafiei E, Alidaee S, Razavi H, Bagheri S, Rezaei H, Sabouri S, Najibi A, Khodaei F, Kashani SMA, Ommati MM, Heidari R. Hepatic encephalopathy complications are diminished by piracetam via the interaction between mitochondrial function, oxidative stress, inflammatory response, and locomotor activity. Heliyon 2023; 9:e20557. [PMID: 37810869 PMCID: PMC10551565 DOI: 10.1016/j.heliyon.2023.e20557] [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: 04/12/2023] [Revised: 09/01/2023] [Accepted: 09/28/2023] [Indexed: 10/10/2023] Open
Abstract
Background of the study: Hepatic encephalopathy (HE) is a complication in which brain ammonia (NH4+) levels reach critically high concentrations because of liver failure. HE could lead to a range of neurological complications from locomotor and behavioral disturbances to coma. Several tactics have been established for subsiding blood and brain NH4+. However, there is no precise intervention to mitigate the direct neurological complications of NH4+. Purpose It has been found that oxidative stress, mitochondrial damage, and neuro-inflammation play a fundamental role in NH4+ neurotoxicity. Piracetam is a drug used clinically in neurological complications such as stroke and head trauma. Piracetam could significantly diminish oxidative stress and improve brain mitochondrial function. Research methods In the current study, piracetam (100 and 500 mg/kg, oral) was used in a mice model of HE induced by thioacetamide (TA, 800 mg/kg, single dose, i.p). Results Significant disturbances in animals' locomotor activity, along with increased oxidative stress biomarkers, including reactive oxygen species formation, protein carbonylation, lipid peroxidation, depleted tissue glutathione, and decreased antioxidant capacity, were evident in the brain of TA-treated mice. Meanwhile, mitochondrial permeabilization, mitochondrial depolarization, suppression of dehydrogenases activity, and decreased ATP levels were found in the brain of the TA group. The level of pro-inflammatory cytokines was also significantly high in the brain of HE animals. Conclusion It was found that piracetam significantly enhanced mice's locomotor activity, blunted oxidative stress biomarkers, decreased inflammatory cytokines, and improved mitochondrial indices in hyperammonemic mice. These data suggest piracetam as a neuroprotective agent which could be repurposed for the management of HE.
Collapse
Affiliation(s)
- Hossein Niknahad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics, And Technology, Faculty of Medicine, University of Oulu, FI-90014, Oulu, Finland
- University Medical Center Utrecht, Departments of Orthopedics Rheumatology and Clinical Immunology, 3508, GA, Utrecht, the Netherlands
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406, Vilnius, Lithuania
| | - Abdollah Arjmand
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elahe Rafiei
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sepideh Alidaee
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hadi Razavi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Bagheri
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Heresh Rezaei
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sabouri
- Shanxi Key Laboratory of Ecological, Animal Sciences, And Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Asma Najibi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Forouzan Khodaei
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyyed Mohammad Amin Kashani
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Mehdi Ommati
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Shanxi Key Laboratory of Ecological, Animal Sciences, And Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
18
|
Mondal R, Pal P, Biswas S, Chattopadhyay A, Bandyopadhyay A, Mukhopadhyay A, Mukhopadhyay PK. Attenuation of sodium arsenite mediated ovarian DNA damage, follicular atresia, and oxidative injury by combined application of vitamin E and C in post pubertal Wistar rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2701-2720. [PMID: 37129605 DOI: 10.1007/s00210-023-02491-9] [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: 01/12/2023] [Accepted: 04/11/2023] [Indexed: 05/03/2023]
Abstract
Arsenic being a toxic metalloid ubiquitously persists in environment and causes several health complications including female reproductive anomalies. Epidemiological studies documented birth anomalies due to arsenic exposure. Augmented reactive oxygen species (ROS) generation and quenched antioxidant pool are foremost consequences of arsenic threat. On the contrary, Vitamin E (VE) and C (VC) are persuasive antioxidants and conventionally used in toxicity management. Present study was designed to explore the extent of efficacy of combined VE and VC (VEC) against Sodium arsenite (NaAsO2) mediated ovarian damage. Thirty-six female Wistar rats were randomly divided into three groups (Grs) and treated for consecutive 30 days; Gr I (control) was vehicle fed, Gr II (treated) was gavaged with NaAsO2 (3 mg/kg/day), Gr III (supplement) was provided with VE (400 mg/kg/day) & VC (200 mg/kg/day) along with NaAsO2. Marked histological alterations were evidenced by disorganization in oocyte, granulosa cells and zona pellucida layers in treated group. Considerable reduction of different growing follicles along with increased atretic follicles was noted in treated group. Altered activities ofΔ5 3β-Hydroxysteroid dehydrogenase and 17β-Hydroxysteroid dehydrogenase accompanied by reduced luteinizing hormone, follicle-stimulating hormone and estradiol levels were observed in treated animals. Irregular estrous cyclicity pattern was also observed due to NaAsO2 threat. Surplus ROS production affected ovarian antioxidant strata as evidenced by altered oxidative stress markers. Provoked oxidative strain further affects DNA status of ovary. However, supplementation with VEC caused notable restoration from such disparaging effects of NaAsO2 toxicities. Antioxidant and antiapoptotic attributes of those vitamins might be liable for such restoration.
Collapse
Affiliation(s)
- Rubia Mondal
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Priyankar Pal
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Sagnik Biswas
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Alok Chattopadhyay
- Department of Physiology, Harimohan Ghose College, Affiliated to University of Calcutta, Kolkata, India
| | - Amit Bandyopadhyay
- Sports and Exercise Physiology Laboratory, Department of Physiology, University Colleges of Science & Technology, University of Calcutta, Kolkata, India
| | | | | |
Collapse
|
19
|
Yao X, Liu W, Xie Y, Xi M, Xiao L. Fertility loss: negative effects of environmental toxicants on oogenesis. Front Physiol 2023; 14:1219045. [PMID: 37601637 PMCID: PMC10436557 DOI: 10.3389/fphys.2023.1219045] [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: 05/08/2023] [Accepted: 07/27/2023] [Indexed: 08/22/2023] Open
Abstract
There has been a global decline in fertility rates, with ovulatory disorders emerging as the leading cause, contributing to a global lifetime infertility prevalence of 17.5%. Formation of the primordial follicle pool during early and further development of oocytes after puberty is crucial in determining female fertility and reproductive quality. However, the increasing exposure to environmental toxins (through occupational exposure and ubiquitous chemicals) in daily life is a growing concern; these toxins have been identified as significant risk factors for oogenesis in women. In light of this concern, this review aims to enhance our understanding of female reproductive system diseases and their implications. Specifically, we summarized and categorized the environmental toxins that can affect oogenesis. Here, we provide an overview of oogenesis, highlighting specific stages that may be susceptible to the influence of environmental toxins. Furthermore, we discuss the genetic and molecular mechanisms by which various environmental toxins, including metals, cigarette smoke, and agricultural and industrial toxins, affect female oogenesis. Raising awareness about the potential risks associated with toxin exposure is crucial. However, further research is needed to fully comprehend the mechanisms underlying these effects, including the identification of biomarkers to assess exposure levels and predict reproductive outcomes. By providing a comprehensive overview, this review aims to contribute to a better understanding of the impact of environmental toxins on female oogenesis and guide future research in this field.
Collapse
Affiliation(s)
- Xiaoxi Yao
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Weijing Liu
- Breast Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yidong Xie
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Mingrong Xi
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Li Xiao
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| |
Collapse
|
20
|
Hu X, Yuan X, Yang M, Han M, Ommati MM, Ma Y. Arsenic exposure induced anxiety-like behaviors in male mice via influencing the GABAergic Signaling in the prefrontal cortex. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:86352-86364. [PMID: 37402917 DOI: 10.1007/s11356-023-28426-8] [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: 07/27/2022] [Accepted: 06/21/2023] [Indexed: 07/06/2023]
Abstract
Arsenic contamination in drinking water causes a global public health problem. Emerging evidence suggests that arsenic may act as an environmental risk factor for anxiety disorders. However, the exact mechanism underlying the adverse effects has not been fully elucidated. This study aimed to evaluate the anxiety-like behaviors of mice exposed to arsenic trioxide (As2O3), to observe the neuropathological changes, and to explore the link between the GABAergic system and behavioral manifestations. For this purpose, male C57BL/6 mice were exposed to various doses of As2O3 (0, 0.15, 1.5, and 15 mg/L) through drinking water for 12 weeks. Anxiety-like behaviors were assessed using the open field test (OFT), light/dark choice test, and elevated zero maze (EZM). Neuronal injuries in the cerebral cortex and hippocampus were assessed by light microscopy with H&E and Nissl staining. Ultrastructural alteration in the cerebral cortex was assessed by transmission electron microscope (TEM). The expression levels of GABAergic system-related molecules (i.e., glutamate decarboxylase, GABA transporter, and GABAB receptor subunits) in the prefrontal cortex (PFC) were determined by qRT-PCR and western blotting. Arsenic exposure showed a striking anxiogenic effect on mice, especially in the group exposed to 15 mg/L As2O3. Light microscopy showed neuron necrosis and reduced cell counts. TEM revealed marked ultrastructural changes, including the vacuolated mitochondria, disrupted Nissl bodies, an indentation in the nucleus membrane, and delamination of myelin sheath in the cortex. In addition, As2O3 influenced the GABAergic system in the PFC by decreasing the expression of the glutamate decarboxylase 1 (GAD1) and the GABAB2 receptor subunit, but not the GABAB1 receptor subunit. To sum up, sub-chronic exposure to As2O3 is associated with increased anxiety-like behaviors, which may be mediated by altered GABAergic signaling in the PFC. These findings shed light on the mechanisms responsible for the neurotoxic effects of arsenic and therefore more cautions should be taken.
Collapse
Affiliation(s)
- Xin Hu
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Xiaohong Yuan
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Mingyu Yang
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Mingsheng Han
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Mohammad Mehdi Ommati
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Yanqin Ma
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China.
| |
Collapse
|
21
|
Li Q, Feng Y, Wang R, Liu R, Ba Y, Huang H. Recent insights into autophagy and metals/nanoparticles exposure. Toxicol Res 2023; 39:355-372. [PMID: 37398566 PMCID: PMC10313637 DOI: 10.1007/s43188-023-00184-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 03/08/2023] [Accepted: 04/04/2023] [Indexed: 07/04/2023] Open
Abstract
Some anthropogenic pollutants, such as heavy metals and nanoparticles (NPs), are widely distributed and a major threat to environmental safety and public health. In particular, lead (Pb), cadmium (Cd), chromium (Cr), arsenic (As), and mercury (Hg) have systemic toxicity even at extremely low concentrations, so they are listed as priority metals in relation to their significant public health burden. Aluminum (Al) is also toxic to multiple organs and is linked to Alzheimer's disease. As the utilization of many metal nanoparticles (MNPs) gradually gain traction in industrial and medical applications, they are increasingly being investigated to address potential toxicity by impairing certain biological barriers. The dominant toxic mechanism of these metals and MNPs is the induction of oxidative stress, which subsequently triggers lipid peroxidation, protein modification, and DNA damage. Notably, a growing body of research has revealed the linkage between dysregulated autophagy and some diseases, including neurodegenerative diseases and cancers. Among them, some metals or metal mixtures can act as environmental stimuli and disturb basal autophagic activity, which has an underlying adverse health effect. Some studies also revealed that specific autophagy inhibitors or activators could modify the abnormal autophagic flux attributed to continuous exposure to metals. In this review, we have gathered recent data about the contribution of the autophagy/mitophagy mediated toxic effects and focused on the involvement of some key regulatory factors of autophagic signaling during exposure to selected metals, metal mixtures, as well as MNPs in the real world. Besides this, we summarized the potential significance of interactions between autophagy and excessive reactive oxygen species (ROS)-mediated oxidative damage in the regulation of cell survival response to metals/NPs. A critical view is given on the application of autophagy activators/inhibitors to modulate the systematic toxicity of various metals/MNPs.
Collapse
Affiliation(s)
- Qiong Li
- Department of Environmental Health and Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Yajing Feng
- Department of Environmental Health and Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Ruike Wang
- Department of Environmental Health and Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Rundong Liu
- Department of Environmental Health and Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Yue Ba
- Department of Environmental Health and Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Hui Huang
- Department of Environmental Health and Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| |
Collapse
|
22
|
Li X, Wang W, Hou Y, Li G, Yi H, Cui S, Zhang J, He X, Zhao H, Yang Z, Qiu Y, Liu Z, Xie J. Arsenic interferes with spermatogenesis involving Rictor/mTORC2-mediated blood-testis barrier disruption in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 257:114914. [PMID: 37084658 DOI: 10.1016/j.ecoenv.2023.114914] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/15/2023] [Accepted: 04/12/2023] [Indexed: 05/03/2023]
Abstract
Ingestion of arsenic interferes with spermatogenesis and increases the risk of male infertility, but the underlying mechanism remines unclear. In this study, we investigated spermatogenic injury with a focus on blood-testis barrier (BTB) disruption by administrating 5 mg/L and 15 mg/L arsenic orally to adult male mice for 60 d. Our results showed that arsenic exposure reduced sperm quality, altered testicular architecture, and impaired Sertoli cell junctions at the BTB. Analysis of BTB junctional proteins revealed that arsenic intake downregulated Claudin-11 expression and increased protein levels of β-catenin, N-cadherin, and Connexin-43. Aberrant localization of these membrane proteins was also observed in arsenic-treated mice. Meanwhile, arsenic exposure altered the components of Rictor/mTORC2 pathway in mouse testis, including inhibition of Rictor expression, reduced phosphorylation of protein kinase Cα (PKCα) and protein kinase B (PKB), and elevated matrix metalloproteinase-9 (MMP-9) levels. Furthermore, arsenic also induced testicular lipid peroxidative damage, inhibited antioxidant enzyme (T-SOD) activity, and caused glutathione (GSH) depletion. Our findings suggest that disruption of BTB integrity is one of the main factors responsible for the decline in sperm quality caused by arsenic. PKCα-mediated rearrangement of actin filaments and PKB/MMP-9-increased barrier permeability jointly contribute to arsenic-induced BTB disruption.
Collapse
Affiliation(s)
- Xiujuan Li
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, China, Shanxi Medical University, Taiyuan 030001, China
| | - Wenting Wang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, China, Shanxi Medical University, Taiyuan 030001, China
| | - Yue Hou
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, China, Shanxi Medical University, Taiyuan 030001, China; Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Gexuan Li
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, China, Shanxi Medical University, Taiyuan 030001, China; Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Huilan Yi
- School of Life Science, Shanxi University, Taiyuan 030006, China
| | - Shuo Cui
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, China, Shanxi Medical University, Taiyuan 030001, China; Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Juan Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, China, Shanxi Medical University, Taiyuan 030001, China
| | - Xiaohong He
- Taiyuan Hospital of Integrated Traditional Chinese and Western Medicine, Taiyuan 030003, China
| | - Hong Zhao
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, China, Shanxi Medical University, Taiyuan 030001, China
| | - Zeyu Yang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, China, Shanxi Medical University, Taiyuan 030001, China
| | - Yulan Qiu
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan 030001, China.
| | - Zhizhen Liu
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, China, Shanxi Medical University, Taiyuan 030001, China.
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, China, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
23
|
Hu Y, Yan Z, He Y, Li Y, Li M, Li Y, Zhang D, Zhao Y, Ommati MM, Wang J, Huo M, Wang J. Ameliorative effects of different doses of selenium against fluoride-triggered apoptosis and oxidative stress-mediated renal injury in rats through the activation of Nrf2/HO-1/NQO1 signaling pathway. Food Chem Toxicol 2023; 174:113647. [PMID: 36736877 DOI: 10.1016/j.fct.2023.113647] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/04/2023]
Abstract
Excess fluoride (F) exposure can cause oxidative stress in the kidney. As an antioxidant, selenium (Se) can potentially protect the kidney from F-induced injury in rats. Hence, the histopathological, renal biochemical, oxidative stress, and apoptotic-related indices upon exposure to 100 mg/L sodium fluoride (NaF) and various doses of sodium selenite (Na2SeO3; 0.5, 1, and 2 mg/L) were assessed. Our results demonstrated that F-mediated renal structural damage and apoptosis elevated the content of serum creatinine (SCr), inhibited the activity of catalase (CAT) in serum, and increased the production of reactive oxygen species (ROS) in kidney and malondialdehyde (MDA) in serum. Interestingly, 1 mg/L dietary supplementation of Se tangibly mitigated these injuries. Furthermore, F could also change the gene and protein expression of the nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), and NAD(P)H quinone oxidoreductase1 (NQO1). Concomitantly, the different concentrations of Se notably alleviated their expression. Taken together, 1-2 mg/L Se ameliorated F-induced renal injury through oxidative stress and apoptosis-related routes. The recorded ameliorative effects might be related to the activation of the Nrf2/HO-1/NQO1 signaling pathway.
Collapse
Affiliation(s)
- Yingjun Hu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, PR China
| | - Zipeng Yan
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, PR China
| | - Yang He
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, PR China
| | - Yan Li
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, PR China
| | - Meng Li
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, PR China
| | - Yuanyuan Li
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, PR China
| | - DingLi Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, PR China
| | - Yangfei Zhao
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, PR China
| | - Mohammad Mehdi Ommati
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, PR China
| | - Jundong Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, PR China
| | - Meijun Huo
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, PR China.
| | - Jinming Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, PR China.
| |
Collapse
|
24
|
Pulmonary inflammation, oxidative stress, and fibrosis in a mouse model of cholestasis: the potential protective properties of the dipeptide carnosine. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:1129-1142. [PMID: 36651945 DOI: 10.1007/s00210-023-02391-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 01/09/2023] [Indexed: 01/19/2023]
Abstract
Cholestasis is a clinical complication that primarily influences the liver. However, it is well known that many other organs could be affected by cholestasis. Lung tissue is a major organ influenced during cholestasis. Cholestasis-induced lung injury could induce severe complications such as respiratory distress, serious pulmonary infections, and tissue fibrosis. Unfortunately, there is no specific pharmacological intervention against this complication. Several studies revealed that oxidative stress and inflammatory response play a role in cholestasis-induced lung injury. Carnosine (CARN) is a dipeptide found at high concentrations in different tissues of humans. CARN's antioxidant and antiinflammatory properties are repeatedly mentioned in various experimental models. This study aimed to assess the role of CARN on cholestasis-induced lung injury. Rats underwent bile duct ligation (BDL) to induce cholestasis. Broncho-alveolar lavage fluid (BALF) levels of inflammatory cells, pro-inflammatory cytokines, and immunoglobulin were monitored at scheduled intervals (7, 14, and 28 days after BDL). Moreover, lung tissue histopathological alterations and biomarkers of oxidative stress were evaluated. A significant increase in BALF inflammatory cells, TNF-α, IL-1β, IL-6, and immunoglobulin-G (IgG) was detected in the BALF of BDL rats. Moreover, lung tissue histopathological changes, collagen deposition, increased TGF-β, and elevated levels of oxidative stress biomarkers were evident in cholestatic animals. It was found that CARN (100 and 500 mg/kg, i.p.) significantly alleviated lung oxidative stress biomarkers, inflammatory response, tissue fibrosis, and histopathological alterations. These data indicate the potential protective properties of CARN in the management of cholestasis-induced pulmonary damage. The effects of CARN on inflammatory response and oxidative stress biomarkers seems to play a crucial role in its protective properties in the lung of cholestatic animals.
Collapse
|
25
|
Zhu S, Liu J, Zhao J, Zhou B, Zhang Y, Wang H. HIF-1α-mediated autophagy and canonical Wnt/β-catenin signalling activation are involved in fluoride-induced osteosclerosis in rats. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 315:120396. [PMID: 36220573 DOI: 10.1016/j.envpol.2022.120396] [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: 06/15/2022] [Revised: 09/23/2022] [Accepted: 10/05/2022] [Indexed: 06/16/2023]
Abstract
Fluoride (F) exposure can cause osteosclerosis, which is characterised by a high bone mass, but its mechanism is not fully illustrated. Here, we aimed to evaluate the effects of excessive F exposure on the bone lesion by treating female Sprague-Dawley rats with different concentrations of sodium fluoride (NaF) (0, 55, 110 and 221 mg/L) for 90 days and the corresponding concentrations of fluorine ion (0, 25, 50 and 100 mg/L, respectively). Histopathological results showed that excessive F exposure caused the enlargement of trabeculae and their integration into one large piece, growth plate thickening, articular cartilage impairment and bone collagen abnormality. Meanwhile, F promoted calcium deposition and bone mineralisation, and induced abnormal osteogenesis increased. The results of micro-computed tomography also confirmed that excessive F destroyed the bone microstructure and induced a high-bone-mass phenotype, consistent with the results of pathomorphology. Mechanistically, excessive amounts of F led to angiogenesis inhibition and HIF-1α signalling enhancement. Subsequently, F induced autophagy and canonical Wnt/β-catenin signalling pathway activation. Collectively, these results manifested that F enhanced the hypoxia inducible factor-1α signalling, which in turn triggered autophagy and canonical Wnt/β-catenin signalling activation, ultimately leading to osteosclerosis in the rats.
Collapse
Affiliation(s)
- Shiquan Zhu
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, 471000, Henan, People's Republic of China.
| | - Jing Liu
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, 471000, Henan, People's Republic of China.
| | - Jing Zhao
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, 471000, Henan, People's Republic of China.
| | - Bianhua Zhou
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, 471000, Henan, People's Republic of China.
| | - Yuling Zhang
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, 471000, Henan, People's Republic of China.
| | - Hongwei Wang
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, 471000, Henan, People's Republic of China.
| |
Collapse
|
26
|
Taurine Improves Sperm Mitochondrial Indices, Blunts Oxidative Stress Parameters, and Enhances Steroidogenesis and Kinematics of Sperm in Lead-Exposed Mice. Reprod Sci 2022; 30:1891-1910. [DOI: 10.1007/s43032-022-01140-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022]
|
27
|
Ommati MM, Ahmadi HN, Sabouri S, Retana-Marquez S, Abdoli N, Rashno S, Niknahad H, Jamshidzadeh A, Mousavi K, Rezaei M, Akhlagh A, Azarpira N, Khodaei F, Heidari R. Glycine protects the male reproductive system against lead toxicity via alleviating oxidative stress, preventing sperm mitochondrial impairment, improving kinematics of sperm, and blunting the downregulation of enzymes involved in the steroidogenesis. ENVIRONMENTAL TOXICOLOGY 2022; 37:2990-3006. [PMID: 36088639 DOI: 10.1002/tox.23654] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/22/2022] [Accepted: 08/27/2022] [Indexed: 06/15/2023]
Abstract
Lead (Pb) is a highly toxic heavy metal widely dispersed in the environment because of human industrial activities. Many studies revealed that Pb could adversely affect several organs, including the male reproductive system. Pb-induced reproductive toxicity could lead to infertility. Thus, finding safe and clinically applicable protective agents against this complication is important. It has been found that oxidative stress plays a fundamental role in the pathogenesis of Pb-induced reprotoxicity. Glycine is the simplest amino acid with a wide range of pharmacological activities. It has been found that glycine could attenuate oxidative stress and mitochondrial impairment in various experimental models. The current study was designed to evaluate the role of glycine in Pb-induced reproductive toxicity in male mice. Male BALB/c mice received Pb (20 mg/kg/day; gavage; 35 consecutive days) and treated with glycine (250 and 500 mg/kg/day; gavage; 35 consecutive days). Then, reproductive system weight indices, biomarkers of oxidative stress in the testis and isolated sperm, sperm kinetic, sperm mitochondrial indices, and testis histopathological alterations were monitored. A significant change in testis, epididymis, and Vas deferens weight was evident in Pb-treated animals. Markers of oxidative stress were also significantly increased in the testis and isolated sperm of the Pb-treated group. A significant disruption in sperm kinetic was also evident when mice received Pb. Moreover, Pb exposure caused significant deterioration in sperm mitochondrial indices. Tubular injury, tubular desquamation, and decreased spermatogenic index were histopathological alterations detected in Pb-treated mice. It was found that glycine significantly blunted oxidative stress markers in testis and sperm, improved sperm mitochondrial parameters, causing considerable higher velocity-related indices (VSL, VCL, and VAP) and percentages of progressively motile sperm, and decreased testis histopathological changes in Pb-exposed animals. These data suggest glycine as a potential protective agent against Pb-induced reproductive toxicity. The effects of glycine on oxidative stress markers and mitochondrial function play a key role in its protective mechanism.
Collapse
Affiliation(s)
- Mohammad Mehdi Ommati
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hassan Nategh Ahmadi
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- College of Animal Science and Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Samira Sabouri
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Socorro Retana-Marquez
- Department of Biology of Reproduction, Autonomous Metropolitan University-Iztapalapa, Mexico City, Mexico
| | - Narges Abdoli
- Food and Drug Administration, Iran Ministry of Health and Medical Education, Tehran, Iran
| | - Sajjad Rashno
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Niknahad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Akram Jamshidzadeh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Khadijeh Mousavi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Rezaei
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Akhlagh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Forouzan Khodaei
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
28
|
Ommati MM, Mobasheri A, Ma Y, Xu D, Tang Z, Manthari RK, Abdoli N, Azarpira N, Lu Y, Sadeghian I, Mousavifaraz A, Nadgaran A, Nikoozadeh A, Mazloomi S, Mehrabani PS, Rezaei M, Xin H, Mingyu Y, Niknahad H, Heidari R. Taurine mitigates the development of pulmonary inflammation, oxidative stress, and histopathological alterations in a rat model of bile duct ligation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:1557-1572. [PMID: 36097067 DOI: 10.1007/s00210-022-02291-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022]
Abstract
Lung injury is a significant complication associated with cholestasis/cirrhosis. This problem significantly increases the risk of cirrhosis-related morbidity and mortality. Hence, finding effective therapeutic options in this field has significant clinical value. Severe inflammation and oxidative stress are involved in the mechanism of cirrhosis-induced lung injury. Taurine (TAU) is an abundant amino acid with substantial anti-inflammatory and antioxidative properties. The current study was designed to evaluate the role of TAU in cholestasis-related lung injury. For this purpose, bile duct ligated (BDL) rats were treated with TAU (0.5 and 1% w: v in drinking water). Significant increases in the broncho-alveolar lavage fluid (BALF) level of inflammatory cells (lymphocytes, neutrophils, basophils, monocytes, and eosinophils), increased IgG, and TNF-α were detected in the BDL animals (14 and 28 days after the BDL surgery). Alveolar congestion, hemorrhage, and fibrosis were the dominant pulmonary histopathological changes in the BDL group. Significant increases in the pulmonary tissue biomarkers of oxidative stress, including reactive oxygen species formation, lipid peroxidation, increased oxidized glutathione levels, and decreased reduced glutathione, were also detected in the BDL rats. Moreover, significant myeloperoxidase activity and nitric oxide levels were seen in the lung of BDL rats. It was found that TAU significantly blunted inflammation, alleviated oxidative stress, and mitigated lung histopathological changes in BDL animals. These data suggest TAU as a potential protective agent against cholestasis/cirrhosis-related lung injury.
Collapse
Affiliation(s)
- Mohammad Mehdi Ommati
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mobasheri
- Physics, and Technology, Faculty of Medicine, Research Unit of Medical Imaging, University of Oulu, 90014, Oulu, Finland
- Departments of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, 3508 GA, Utrecht, The Netherlands
- Department of Regenerative Medicine, State Research Institute Center for Innovative Medicine, 08406, Vilnius, Lithuania
| | - Yanqin Ma
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Dongmei Xu
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Zhongwei Tang
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Ram Kumar Manthari
- Department of Biotechnology, GITAM Institute of Science, Gandhi Institute of Technology and Management, Visakhapatnam-530045, Andhra Pradesh, India
| | - Narges Abdoli
- Food and Drug Administration, Iran Ministry of Health and Medical Education, Tehran, Iran
| | - Negar Azarpira
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yu Lu
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Issa Sadeghian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abolghasem Mousavifaraz
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Nadgaran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Nikoozadeh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sahra Mazloomi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pooria Sayar Mehrabani
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Rezaei
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hu Xin
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Yang Mingyu
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Hossein Niknahad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
29
|
Rachamalla M, Chinthada J, Kushwaha S, Putnala SK, Sahu C, Jena G, Niyogi S. Contemporary Comprehensive Review on Arsenic-Induced Male Reproductive Toxicity and Mechanisms of Phytonutrient Intervention. TOXICS 2022; 10:toxics10120744. [PMID: 36548577 PMCID: PMC9784647 DOI: 10.3390/toxics10120744] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 05/26/2023]
Abstract
Arsenic (As) is a poisonous metalloid that is toxic to both humans and animals. Drinking water contamination has been linked to the development of cancer (skin, lung, urinary bladder, and liver), as well as other disorders such as diabetes and cardiovascular, gastrointestinal, neurological, and developmental damage. According to epidemiological studies, As contributes to male infertility, sexual dysfunction, poor sperm quality, and developmental consequences such as low birth weight, spontaneous abortion, and small for gestational age (SGA). Arsenic exposure negatively affected male reproductive systems by lowering testicular and accessory organ weights, and sperm counts, increasing sperm abnormalities and causing apoptotic cell death in Leydig and Sertoli cells, which resulted in decreased testosterone synthesis. Furthermore, during male reproductive toxicity, several molecular signalling pathways, such as apoptosis, inflammation, and autophagy are involved. Phytonutrient intervention in arsenic-induced male reproductive toxicity in various species has received a lot of attention over the years. The current review provides an in-depth summary of the available literature on arsenic-induced male toxicity, as well as therapeutic approaches and future directions.
Collapse
Affiliation(s)
- Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | - Joshi Chinthada
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S Nagar 160062, India
| | - Sapana Kushwaha
- Department of Pharmacology and Toxicology, Transit Campus, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Sravan Kumar Putnala
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | - Chittaranjan Sahu
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S Nagar 160062, India
| | - Gopabandhu Jena
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S Nagar 160062, India
| | - Som Niyogi
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| |
Collapse
|
30
|
Kang HG, Jeong PS, Kim MJ, Joo YE, Gwon MA, Jeon SB, Song BS, Kim SU, Lee S, Sim BW. Arsenic exposure during porcine oocyte maturation negatively affects embryonic development by triggering oxidative stress-induced mitochondrial dysfunction and apoptosis. Toxicology 2022; 480:153314. [PMID: 36084880 DOI: 10.1016/j.tox.2022.153314] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/16/2022] [Accepted: 09/04/2022] [Indexed: 11/15/2022]
Abstract
Arsenic (AS), an environmental contaminant, is a known human carcinogen that can cause cancer of the lung, liver, and skin. Furthermore, AS induces oxidative stress and mitochondrial impairments in mammalian cells. However, limited information is available on the effect of AS exposure on oocyte maturation of porcine, whose anatomy, physiology, and metabolism are similar to those of human. Therefore, we examined the effect of AS exposure on the in vitro maturation (IVM) of porcine oocytes and the possible underlying mechanisms. Cumulus-cell enclosed oocytes were cultured with or without AS for maturation, and then were used for analyses. This study indicated that AS under a concentration of 1 μM significantly increased the abnormal expansion of cumulus cells and the number of oocytes maintained in meiotic arrest. In addition, AS exposure significantly reduced subsequent development of embryos and increased the rate of apoptosis of blastocysts following parthenogenetic activation (PA) and in vitro fertilization (IVF). Moreover, AS exposure induced oxidative stress with increased reactive oxygen species (ROS), and decreased glutathione (GSH), leading to reduced mitochondrial membrane potential, mitochondrial quantity, DNA damage, excessive autophagy activity, and early apoptosis in porcine oocytes. Taken together, the results demonstrated that AS exposure exerts several negative effects, such as meiotic defects and embryo developmental arrest by causing mitochondrial dysfunction and apoptosis via inducing oxidative stress.
Collapse
Affiliation(s)
- Hyo-Gu Kang
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, South Korea; Laboratory of Animal Reproduction and Physiology, Department of Animal Science and Biotechnology, College of Agriculture and Life Science, Chungnam National University, Daejeon 34134, Korea
| | - Pil-Soo Jeong
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, South Korea
| | - Min Ju Kim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, South Korea
| | - Ye Eun Joo
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, South Korea
| | - Min-Ah Gwon
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, South Korea
| | - Se-Been Jeon
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, South Korea
| | - Bong-Seok Song
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, South Korea
| | - Sun-Uk Kim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, South Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, South Korea
| | - Sanghoon Lee
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, South Korea; Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, South Korea.
| | - Bo-Woong Sim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, South Korea.
| |
Collapse
|
31
|
Melatonin improves arsenic-induced hypertension through the inactivation of the Sirt1/autophagy pathway in rat. Biomed Pharmacother 2022; 151:113135. [PMID: 35598369 DOI: 10.1016/j.biopha.2022.113135] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/12/2022] [Accepted: 05/15/2022] [Indexed: 11/20/2022] Open
Abstract
Arsenic (As), a metalloid chemical element, is classified as heavy metal. Previous studies proposed that As induces vascular toxicity by inducing autophagy, apoptosis, and oxidative stress. It has been shown that melatonin (Mel) can decrease oxidative stress and apoptosis, and modulate autophagy in different pathological situations. Hence, this study aimed to investigate the Mel effect on As-induced vascular toxicity through apoptosis and autophagy regulation. Forty male rats were treated with As (15 mg/kg; oral gavage) and Mel (10 and 20 mg/kg, intraperitoneally; i.p.) for 28 days. The systolic blood pressure (SBP) changes, oxidative stress markers, the aorta histopathological injuries, contractile and relaxant responses, the level of apoptosis (Bnip3 and caspase-3) and autophagy (Sirt1, Beclin-1 and LC3 II/I ratio) proteins were determined in rats aorta. The As exposure significantly increased SBP and enhanced MDA level while reduced GSH content. The exposure to As caused substantial histological damage in aorta tissue and changed vasoconstriction and vasorelaxation responses to KCl, PE, and Ach in isolated rat aorta. The levels of HO-1 and Nrf-2, apoptosis markers, Sirt1, and autophagy proteins also enhanced in As group. Interestingly, Mel could reduce changes in oxidative stress, blood pressure, apoptosis, and autophagy induced by As. On the other hand, Mel led to more increased the levels of Nrf-2 and HO-1 proteins compared with the As group. In conclusion, our findings showed that Mel could have a protective effect against As-induced vascular toxicity by inhibiting apoptosis and the Sirt1/autophagy pathway.
Collapse
|
32
|
Zhang Z, Wang H, Lei X, Mehdi Ommati M, Tang Z, Yuan J. Bisphenol a exposure decreases learning ability through the suppression of mitochondrial oxidative phosphorylation in the hippocampus of male mice. Food Chem Toxicol 2022; 165:113167. [DOI: 10.1016/j.fct.2022.113167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 11/25/2022]
|
33
|
Yang W, Liang C, Zhang X, Tian X, Ren C, Chen S, Wang J, Zhang J. Melamine induced changes in histopathology of the main organs and transcriptional levels of MAPK signaling genes in kidneys of female mice. ENVIRONMENTAL TOXICOLOGY 2022; 37:585-592. [PMID: 34842327 DOI: 10.1002/tox.23424] [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: 09/01/2021] [Revised: 11/10/2021] [Accepted: 11/20/2021] [Indexed: 06/13/2023]
Abstract
Melamine is an important chemical raw material used in industries, which has potential health risks to animals and humans. Current research mainly focuses on the toxic effects of high-dose melamine ingestion. However, there are few reports on whether melamine at the current limited standard dose has adverse effects on various tissues and organs, and whether there are sensitive target genes for risk evaluation. For this, 24 female Kunming mice were fed 0, 1.8-, 3.6-, and 7.2- mg/kg/d melamine via drinking water for consecutive 28 days, respectively. The morphological changes of the ovarian, hepatic, and renal tissues were firstly observed. The results demonstrated that the histopathology of ovary, liver, and especially in kidney had been altered by melamine intake in female. And then, the transcriptional levels of MAPK signaling genes including p38, ERK1, ERK2, JNK1, and JNK2 in kidneys were investigated by real-time PCR. The data showed that ERK1 and p38 mRNAs expressions were up-regulated significantly by melamine, suggesting that ERK1 and p38 transcriptional levels in the kidney might to be considered as candidate targets for lower-dose melamine toxicity. This study not only provides potential targets for the diagnosis and prevention of melamine damage, but also helps to assess the health risks of the current minimum allowable levels of melamine in food and environment.
Collapse
Affiliation(s)
- Wei Yang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, People's Republic of China
| | - Chen Liang
- College of Animal Science, Shanxi Agricultural University, Taigu, People's Republic of China
| | - Xiaoyan Zhang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, People's Republic of China
| | - Xiaohui Tian
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, People's Republic of China
| | - Chenxia Ren
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, People's Republic of China
| | - Shuming Chen
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, People's Republic of China
| | - Jundong Wang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, People's Republic of China
| | - Jianhai Zhang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, People's Republic of China
| |
Collapse
|
34
|
Ommati MM, Li H, Jamshidzadeh A, Khoshghadam F, Retana-Márquez S, Lu Y, Farshad O, Nategh Ahmadi MH, Gholami A, Heidari R. The crucial role of oxidative stress in non-alcoholic fatty liver disease-induced male reproductive toxicity: the ameliorative effects of Iranian indigenous probiotics. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:247-265. [PMID: 34994824 DOI: 10.1007/s00210-021-02177-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 11/01/2021] [Indexed: 02/06/2023]
Abstract
Several studies have focused on the high potential effects of probiotics on the reproductive system. However, there is a paucity of information regarding the ameliorative intracellular roles of indigenous Iranian yogurt-extracted/cultured probiotics on animals' reproductive health suffering from obesity and/or fatty liver disease, such as non-alcoholic fatty liver disease (NAFLD). For this purpose, simultaneously with the consumption of D-fructose (200 g/1000 mL water, induction of NAFLD model), all pubertal animals were also gavaged every day for 63 consecutive days with extracted probiotics, including 1 × 109 CFU/mL of Lactobacillus acidophilus (LA), Bifidobacterium spp. (BIF), Bacillus coagulans (BC), Lactobacillus rhamnosus (LR), and a mixture form (LA + BIF + BC + LR). At the end of the ninth week, the indices of epididymal sperm, and oxidative stress, as well as histopathological changes, were assessed. The results show that NAFLD could induce robust oxidative stress, highlighted as considerable increments in ROS level, TBARS content, total oxidized protein levels, along with severe decrements in reduced glutathione reservoirs, total antioxidant capacity in the hepatic and testicular tissues, as well as testicular and hepatic histopathological alterations. Moreover, a significant decrease in the percentage of sperm progressive motility, sperm count, and membrane integrity along with an increment in the percentage of sperm abnormality was detected in NAFLD animals. The observed adverse effects were significantly reversed upon probiotics treatment, especially in the group challenged with a mixture of all probiotics. Taken together, these findings indicate that the indigenous yogurt-isolated/cultured probiotics had a high potential antioxidant activity and the ameliorative effect against reprotoxicity and blood biochemical alterations induced by the NAFLD model. Highlights: 1. Reproductive indices could be reversely affected by xenobiotics and diseases. 2. NAFLD and cholestasis considerably affect the reproductive system in both genders. 3. NAFLD induced hepatic and testicular oxidative stress (OS). 4. NAFLD induced histopathological alterations and spermatotoxicity through OS. 5. The adverse effects were significantly reversed upon exposure to probiotics.
Collapse
Affiliation(s)
- Mohammad Mehdi Ommati
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi, 030801, People's Republic of China.
| | - Huifeng Li
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi, 030801, People's Republic of China
| | - Akram Jamshidzadeh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fereshteh Khoshghadam
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Socorro Retana-Márquez
- Department of Biology of Reproduction, Autonomous Metropolitan University-Iztapalapa, Mexico City, Mexico
| | - Yu Lu
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi, 030801, People's Republic of China
| | - Omid Farshad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- College of Pharmacy and Nutrition, School of Pharmacy, University of Saskatchewan, Saskatoon, Canada
| | - Mohammad Hasan Nategh Ahmadi
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, People's Republic of China
- Department of Clinical Studies, School of Veterinary Medicine, Shiraz University, 71345, Shiraz, Iran
| | - Ahmad Gholami
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
35
|
Juárez-Rojas L, Casillas F, López A, Betancourt M, Ommati MM, Retana-Márquez S. Physiological role of reactive oxygen species in testis and epididymal spermatozoa. Andrologia 2022; 54:e14367. [PMID: 35034376 DOI: 10.1111/and.14367] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/02/2021] [Accepted: 12/18/2021] [Indexed: 11/26/2022] Open
Abstract
The reactive oxygen species (ROS) play an important role in various aspects of male reproductive function, for spermatozoa to acquire the ability to fertilize. However, the increase in ROS generation, both due to internal and external factors, can induce oxidative stress, causing alterations in the structure and function of phospholipids and proteins. In the nucleus, ROS attack DNA, causing its fragmentation and activation of apoptosis, thus altering gene and protein expression. Accumulating evidence also reveals that endogenously produced ROS can act as second messengers in regulating cell signalling pathways and in the transduction of signals that are responsible for regulating spermatogonia self-renewal and proliferation. In the epididymis, they actively participate in the formation of disulphide bridges required for the final condensation of chromatin, as well as in the phosphorylation and dephosphorylation of proteins contained in the fibrous sheath of the flagellum, stimulating the activation of progressive motility in epididymal spermatozoa. In this review, the role of small amounts of ROS during spermatogenesis and epididymal sperm maturation was discussed.
Collapse
Affiliation(s)
- Lizbeth Juárez-Rojas
- Department of Biology of Reproduction, Autonomous Metropolitan University-Iztapalapa, Mexico City, Mexico
| | - Fahiel Casillas
- Department of Biology of Reproduction, Autonomous Metropolitan University-Iztapalapa, Mexico City, Mexico
| | - Alma López
- Department of Health Sciences, Autonomous Metropolitan University-Iztapalapa Campus, Mexico City, Mexico
| | - Miguel Betancourt
- Department of Health Sciences, Autonomous Metropolitan University-Iztapalapa Campus, Mexico City, Mexico
| | - Mohammad Mehdi Ommati
- Department of Bioinformatics, College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi, People's Republic of China
| | - Socorro Retana-Márquez
- Department of Biology of Reproduction, Autonomous Metropolitan University-Iztapalapa, Mexico City, Mexico
| |
Collapse
|
36
|
Wang X, Ding N, Harlow SD, Randolph JF, Mukherjee B, Gold EB, Park SK. Urinary metals and metal mixtures and timing of natural menopause in midlife women: The Study of Women's Health Across the Nation. ENVIRONMENT INTERNATIONAL 2021; 157:106781. [PMID: 34311223 PMCID: PMC8490279 DOI: 10.1016/j.envint.2021.106781] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/21/2021] [Accepted: 07/15/2021] [Indexed: 05/02/2023]
Abstract
BACKGROUND Exposure to metals and metal mixtures may influence ovarian aging. However, epidemiologic evidence of their potential impact is lacking. OBJECTIVE We prospectively examined the associations of 15 urinary metal concentrations and their mixtures with natural menopause in the Study of Women's Health Across the Nation Multi-Pollutant Study. METHODS The study population consisted of 1082 premenopausal women from multiple racial/ethnic groups, aged 45-56 years at baseline (1999-2000), with the median follow-up of 4.1 years. Urinary concentrations of 15 metals, including arsenic, barium, cadmium, cobalt, cesium, copper, mercury, manganese, molybdenum, nickel, lead, antimony, tin, thallium, and zinc, were measured at baseline. Natural menopause was defined as the final bleeding episode prior to at least 12 months of amenorrhea, not due to surgery or hormone therapy. Cox proportional hazards models were used to examine associations between individual metal concentrations and timing of natural menopause. The associations between metal mixtures and natural menopause were evaluated using elastic net penalized Cox regression, and an environmental risk score (ERS) was computed to represent individual risks of natural menopause related to metal mixtures. RESULTS The median age at natural menopause was 53.2 years. Using the Cox proportional hazards models, the adjusted hazard ratio (HR) (and its 95% confidence interval (CI)) for natural menopause was 1.32 (1.03, 1.67) for arsenic and 1.36 (1.05, 1.76) for lead, comparing the highest with the lowest quartiles of metal concentrations. The predicted ages at natural menopause in the highest and lowest quartiles were 52.7 and 53.5 years for arsenic; and 52.9 and 53.8 years for lead. A significant association between ERS and menopause was also observed. Women in the highest vs. the lowest quartiles of ERS had an HR of 1.71 (1.36, 2.15), equivalent to a 1.6 year earlier median time to natural menopause. CONCLUSION This study suggests that arsenic, lead, and metal mixtures are associated with earlier natural menopause, a risk factor for adverse health outcomes in later life.
Collapse
Affiliation(s)
- Xin Wang
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Ning Ding
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Siobán D Harlow
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - John F Randolph
- Department of Obstetrics and Gynecology, School of Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Bhramar Mukherjee
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Ellen B Gold
- Department of Public Health Sciences, University of California, Davis, School of Medicine, Davis, CA, United States
| | - Sung Kyun Park
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States; Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
37
|
Metformin alleviates cholestasis-associated nephropathy through regulating oxidative stress and mitochondrial function. LIVER RESEARCH 2021. [DOI: 10.1016/j.livres.2020.12.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
38
|
Ghanbarinejad V, Ommati MM, Jia Z, Farshad O, Jamshidzadeh A, Heidari R. Disturbed mitochondrial redox state and tissue energy charge in cholestasis. J Biochem Mol Toxicol 2021; 35:e22846. [PMID: 34250697 DOI: 10.1002/jbt.22846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 05/23/2021] [Accepted: 07/01/2021] [Indexed: 12/17/2022]
Abstract
The liver is the primary organ affected by cholestasis. However, the brain, skeletal muscle, heart, and kidney are also severely influenced by cholestasis/cirrhosis. However, little is known about the molecular mechanisms of organ injury in cholestasis. The current study was designed to evaluate the mitochondrial glutathione redox state as a significant index in cell death. Moreover, tissue energy charge (EC) was calculated. Rats underwent bile duct ligation (BDL) and the brain, heart, liver, kidney, and skeletal muscle mitochondria were assessed at scheduled time intervals (3, 7, 14, and 28 days after BDL). A significant decrease in mitochondrial glutathione redox state and EC was detected in BDL animals. Moreover, disturbed mitochondrial indices were evident in different organs of BDL rats. These data could offer new insight into the mechanisms of organ injury and the source of oxidative stress during cholestasis and might provide novel therapeutic strategies against these complications.
Collapse
Affiliation(s)
- Vahid Ghanbarinejad
- Toxicology Laboratory, Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad M Ommati
- Department of Bioinformatics, College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Zhipeng Jia
- Department of Veterinary Medicine, College of Animal Sciences and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Omid Farshad
- Toxicology Laboratory, Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Akram Jamshidzadeh
- Toxicology Laboratory, Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Toxicology Laboratory, Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
39
|
Xu L, Deng C, Zhang Y, Zhao L, Linghu Y, Yu Y. Expression of Autophagy-Related Factors LC3A and Beclin 1 and Apoptosis-Related Factors Bcl-2 and BAX in Osteoblasts Treated With Sodium Fluoride. Front Physiol 2021; 12:603848. [PMID: 34276389 PMCID: PMC8281676 DOI: 10.3389/fphys.2021.603848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 04/19/2021] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVE This study aims to analyze the expressions of autophagy-related factors light chain 3 alpha (LC3A) and Beclin 1 and apoptosis-related factors B-cell lymphoma 2 (Bcl-2) and Bcl-2-associated X (BAX) in primary osteoblasts treated with sodium fluoride (NaF). METHODS Osteoblasts were extracted from Sprague-Dawley rats and treated with 0, 2.5, 5, and 10 mg/L NaF solutions, followed by 10 mmol/L 3-methyladenine (3-MA) for 24 h. The apoptotic rate was determined by flow cytometry, and the expressions of the autophagy- and apoptosis-related factors were measured by western blotting and real-time quantitative polymerase chain reaction. RESULTS The mRNA expressions of LC3A, Beclin 1, and BAX in the NaF-treated osteoblast group were higher than those in the control group, while the protein expressions of these factors in the NaF-treated group were significantly higher than those in the control group. However, the Bcl-2 protein expression in the NaF-treated osteoblasts was significantly decreased compared to that in the control cells. After the 3-MA treatment, the protein expressions of LC3A, Beclin 1, and Bcl-2 were significantly decreased compared with those of the NaF-treated group, whereas the expression of BAX increased. Moreover, the apoptosis rate was increased after the addition of the 3-MA inhibitor. CONCLUSION NaF stimulation promoted autophagy and apoptosis of the osteoblasts, suggesting the involvement of fluoride damage in these processes.
Collapse
Affiliation(s)
- Lin Xu
- Guiyang Maternal and Child Health Care Hospital of Guiyang, Guiyang, China
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Chaonan Deng
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Pathology, Guizhou Medical University, Guiyang, China
| | - Ying Zhang
- Guiyang Maternal and Child Health Care Hospital of Guiyang, Guiyang, China
| | - Lina Zhao
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yan Linghu
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Pathology, Guizhou Medical University, Guiyang, China
| | - Yanni Yu
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Pathology, Guizhou Medical University, Guiyang, China
| |
Collapse
|
40
|
Ommati MM, Mobasheri A, Heidari R. Drug-induced organ injury in coronavirus disease 2019 pharmacotherapy: Mechanisms and challenges in differential diagnosis and potential protective strategies. J Biochem Mol Toxicol 2021; 35:e22795. [PMID: 33973313 PMCID: PMC8237057 DOI: 10.1002/jbt.22795] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/13/2021] [Accepted: 04/22/2021] [Indexed: 12/20/2022]
Abstract
The world is currently facing an unprecedented pandemic caused by a newly recognized and highly pathogenic coronavirus disease 2019 (COVID-19; induced by SARS-CoV-2 virus), which is a severe and ongoing threat to global public health. Since COVID-19 was officially declared a pandemic by the World Health Organization in March 2020, several drug regimens have rapidly undergone clinical trials for the management of COVID-19. However, one of the major issues is drug-induced organ injury, which is a prominent clinical challenge. Unfortunately, most drugs used against COVID-19 are associated with adverse effects in different organs, such as the kidney, heart, and liver. These side effects are dangerous and, in some cases, they can be lethal. More importantly, organ injury is also a clinical manifestation of COVID-19 infection. These adverse reactions are increasingly recognized as outcomes of COVID-19 infection. Therefore, the differential diagnosis of drug-induced adverse effects from COVID-19-induced organ injury is a clinical complication. This review highlights the importance of drug-induced organ injury, its known mechanisms, and the potential therapeutic strategies in COVID-19 pharmacotherapy. We review the potential strategies for the differential diagnosis of drug-induced organ injury. This information can facilitate the development of therapeutic strategies, not only against COVID-19 but also for future outbreaks of other emerging infectious diseases.
Collapse
Affiliation(s)
- Mohammad Mehdi Ommati
- College of Life SciencesShanxi Agricultural UniversityTaiguChina
- Pharmaceutical Sciences Research CenterShiraz University of Medical SciencesShirazIran
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of MedicineUniversity of OuluOuluFinland
- Department of Regenerative MedicineState Research Institute Centre for Innovative MedicineVilniusLithuania
- Departments of Orthopedics, Rheumatology and Clinical ImmunologyUniversity Medical Center UtrechtUtrechtThe Netherlands
- Department of Joint SurgerySun Yat‐sen UniversityGuangzhouChina
| | - Reza Heidari
- Pharmaceutical Sciences Research CenterShiraz University of Medical SciencesShirazIran
| |
Collapse
|
41
|
Ommati MM, Arabnezhad MR, Farshad O, Jamshidzadeh A, Niknahad H, Retana-Marquez S, Jia Z, Nateghahmadi MH, Mousavi K, Arazi A, Azmoon MR, Azarpira N, Heidari R. The Role of Mitochondrial Impairment and Oxidative Stress in the Pathogenesis of Lithium-Induced Reproductive Toxicity in Male Mice. Front Vet Sci 2021; 8:603262. [PMID: 33842567 PMCID: PMC8025583 DOI: 10.3389/fvets.2021.603262] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 01/29/2021] [Indexed: 11/18/2022] Open
Abstract
Lithium (Li+) is prescribed against a wide range of neurological disorders. Besides its excellent therapeutic properties, there are several adverse effects associated with Li+. The impact of Li+ on renal function and diabetes insipidus is the most common adverse effect of this drug. On the other hand, infertility and decreased libido is another complication associated with Li+. It has been found that sperm indices of functionality, as well as libido, is significantly reduced in Li+-treated men. These adverse effects might lead to drug incompliance and the cessation of drug therapy. Hence, the main aims of the current study were to illustrate the mechanisms of adverse effects of Li+ on the testis tissue, spermatogenesis process, and hormonal changes in two experimental models. In the in vitro experiments, Leydig cells (LCs) were isolated from healthy mice, cultured, and exposed to increasing concentrations of Li+ (0, 10, 50, and 100 ppm). In the in vivo section of the current study, mice were treated with Li+ (0, 10, 50, and 100 ppm, in drinking water) for five consecutive weeks. Testis and sperm samples were collected and assessed. A significant sign of cytotoxicity (LDH release and MTT assay), along with disrupted testosterone biosynthesis, impaired mitochondrial indices (ATP level and mitochondrial depolarization), and increased biomarkers of oxidative stress were detected in LCs exposed to Li+. On the other hand, a significant increase in serum and testis Li+ levels were detected in drug-treated mice. Moreover, ROS formation, LPO, protein carbonylation, and increased oxidized glutathione (GSSG) were detected in both testis tissue and sperm specimens of Li+-treated mice. Several sperm anomalies were also detected in Li+-treated animals. On the other hand, sperm mitochondrial indices (mitochondrial dehydrogenases activity and ATP levels) were significantly decreased in drug-treated groups where mitochondrial depolarization was increased dose-dependently. Altogether, these data mention oxidative stress and mitochondrial impairment as pivotal mechanisms involved in Li+-induced reproductive toxicity. Therefore, based on our previous publications in this area, therapeutic options, including compounds with high antioxidant properties that target these points might find a clinical value in ameliorating Li+-induced adverse effects on the male reproductive system.
Collapse
Affiliation(s)
- Mohammad Mehdi Ommati
- Department of Bioinformatics, College of Life Sciences, Shanxi Agricultural University, Taigu, China
| | - Mohammad Reza Arabnezhad
- Department of Toxicology and Pharmacology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Omid Farshad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Akram Jamshidzadeh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Toxicology and Pharmacology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Niknahad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Toxicology and Pharmacology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Socorro Retana-Marquez
- Department of Biology and Reproduction, Autonomous Metropolitan University, Mexico City, Mexico
| | - Zhipeng Jia
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | | | - Khadijeh Mousavi
- Department of Toxicology and Pharmacology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Aysooda Arazi
- Department of Toxicology and Pharmacology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Azmoon
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Toxicology and Pharmacology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
42
|
Abdoli N, Sadeghian I, Azarpira N, Ommati MM, Heidari R. Taurine mitigates bile duct obstruction-associated cholemic nephropathy: effect on oxidative stress and mitochondrial parameters. Clin Exp Hepatol 2021; 7:30-40. [PMID: 34027113 PMCID: PMC8122090 DOI: 10.5114/ceh.2021.104675] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 10/30/2020] [Indexed: 12/19/2022] Open
Abstract
AIM OF THE STUDY Cholestasis is a serious complication affecting other organs such as the liver and kidney. Oxidative stress and mitochondrial impairment are proposed as the primary mechanisms for cholestasis-induced organ injury. Taurine (TAU) is the most abundant free amino acid in the human body, which is not incorporated in the structure of proteins. Several pharmacological effects have been attributed to TAU. It has been reported that TAU effectively mitigated oxidative stress and modulated mitochondrial function. The current study aimed to evaluate the impact of TAU on oxidative stress biomarkers and mitochondrial parameters in the kidney of cholestatic animals. MATERIAL AND METHODS Bile duct ligated (BDL) rats were used as an antioxidant model of cholestasis. Animals were treated with TAU (500 and 1000 mg/kg, oral) for seven consecutive days. Animals were anesthetized (thiopental 80 mg/kg, i.p.), and kidney and blood specimens were collected. RESULTS Severe elevation in serum and urine biomarkers of renal injury was evident in the BDL group. Significant lipid peroxidation, reactive oxygen species (ROS) formation, and protein carbonylation were detected in the kidney of BDL animals. Furthermore, depleted glutathione reservoirs and a significant decrease in the antioxidant capacity of renal tissue were detected in cholestatic rats. Renal tubular atrophy and interstitial inflammation were evident in BDL animals. Cholestasis also caused significant mitochondrial dysfunction in the kidney. TAU significantly prevented cholestasis-induced renal injury by inhibiting oxidative stress and mitochondrial impairment. CONCLUSIONS These data indicate TAU as a potential therapeutic agent in the management of cholestasis-induced renal injury.
Collapse
Affiliation(s)
- Narges Abdoli
- Iran Food and Drug Administration, Ministry of Health, Tehran, Iran
| | - Issa Sadeghian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Mehdi Ommati
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
43
|
Wu L, Wei Y, Li H, Li W, Gu C, Sun J, Xia H, Zhang J, Chen F, Liu Q. The ubiquitination and acetylation of histones are associated with male reproductive disorders induced by chronic exposure to arsenite. Toxicol Appl Pharmacol 2020; 408:115253. [PMID: 32991915 DOI: 10.1016/j.taap.2020.115253] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022]
Abstract
Exposure to arsenic, which occurs via various routes, can cause reproductive toxicity. However, the mechanism for arsenic-induced reproductive disorders in male mice has not been extensively investigated. Here, 6-week-old male mice were dosed to 0, 5, 10, or 20 ppm sodium arsenite (NaAsO2), an active form of arsenic, in drinking water for six months. For male mice exposed to arsenite, fertility was lower compared to control mice. Moreover, for exposed mice, there were lower sperm counts, lower sperm motility, and higher sperm malformation ratios. Further, the mRNA and protein levels of the gonadotropin-regulated testicular RNA helicase (DDX25) and chromosome region maintenance-1 protein (CRM1), along with proteins associated with high mobility group box 2 (HMGB2), phosphoglycerate kinase 2 (PGK2), and testicular angiotensin-converting enzyme (tACE) were lower. Furthermore, chronic exposure to arsenite led to lower H2A ubiquitination (ubH2A); histone H3 acetylation K18 (H3AcK18); and histone H4 acetylations K5, K8, K12, and K16 (H4tetraAck) in haploid spermatids from testicular tissues. These alterations disrupted deposition of protamine 1 (Prm1) in testes. Overall, the present results indicate that the ubiquitination and acetylation of histones is involved in the spermiogenesis disorders caused by chronic exposure to arsenite, which points to a previously unknown connection between the modification of histones and arsenite-induced male reproductive toxicity.
Collapse
Affiliation(s)
- Lu Wu
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Yongyue Wei
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Han Li
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Wenqi Li
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Chenxi Gu
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Jing Sun
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Haibo Xia
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Jingshu Zhang
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; Jiangsu Safety Assessment and Research Center for Drug, Pesticide, and Veterinary Drug, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Feng Chen
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China.
| | - Qizhan Liu
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China.
| |
Collapse
|