1
|
Neves SP, Bomfim LM, Kataura T, Carvalho SG, Nogueira ML, Dias RB, Valverde LDF, Gurgel Rocha CA, Soares MBP, Silva MMD, Batista AA, Korolchuk VI, Bezerra DP. Ruthenium complex containing 1,3-thiazolidine-2-thione inhibits hepatic cancer stem cells by suppressing Akt/mTOR signalling and leading to apoptotic and autophagic cell death. Biomed Pharmacother 2024; 177:117059. [PMID: 38955086 DOI: 10.1016/j.biopha.2024.117059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/16/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024] Open
Abstract
Hepatic cancer is one of the main causes of cancer-related death worldwide. Cancer stem cells (CSCs) are a unique subset of cancer cells that promote tumour growth, maintenance, and therapeutic resistance, leading to recurrence. In the present work, the ability of a ruthenium complex containing 1,3-thiazolidine-2-thione (RCT), with the chemical formula [Ru(tzdt)(bipy)(dppb)]PF6, to inhibit hepatic CSCs was explored in human hepatocellular carcinoma HepG2 cells. RCT exhibited potent cytotoxicity to solid and haematological cancer cell lines and reduced the clonogenic potential, CD133+ and CD44high cell percentages and tumour spheroid growth of HepG2 cells. RCT also inhibited cell motility, as observed in the wound healing assay and transwell cell migration assay. RCT reduced the levels of Akt1, phospho-Akt (Ser473), phospho-Akt (Thr308), phospho-mTOR (Ser2448), and phospho-S6 (Ser235/Ser236) in HepG2 cells, indicating that interfering with Akt/mTOR signalling is a mechanism of action of RCT. The levels of active caspase-3 and cleaved PARP (Asp214) were increased in RCT-treated HepG2 cells, indicating the induction of apoptotic cell death. In addition, RCT modulated the autophagy markers LC3B and p62/SQSTM1 in HepG2 cells and increased mitophagy in a mt-Keima-transfected mouse embryonic fibroblast (MEF) cell model, and RCT-induced cytotoxicity was partially prevented by autophagy inhibitors. Furthermore, mutant Atg5-/- MEFs and PentaKO HeLa cells (human cervical adenocarcinoma with five autophagy receptor knockouts) were less sensitive to RCT cytotoxicity than their parental cell lines, indicating that RCT induces autophagy-mediated cell death. Taken together, these data indicate that RCT is a novel potential anti-liver cancer drug with a suppressive effect on CSCs.
Collapse
Affiliation(s)
- Sara P Neves
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Larissa M Bomfim
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Tetsushi Kataura
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Sabrine G Carvalho
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Mateus L Nogueira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil; Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador, Bahia, 40110-909, Brazil; Department of Biological Sciences, State University of Feira de Santana, Feira de Santana, Bahia, 44036-900, Brazil
| | - Ludmila de F Valverde
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil; Department of Dentistry, Federal University of Sergipe, Lagarto, Sergipe, 49400-000, Brazil
| | - Clarissa A Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil; Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador, Bahia, 40110-909, Brazil; Center for Biotechnology and Cell Therapy, D'Or Institute for Research and Education (IDOR), Salvador, Bahia, 41253-190, Brazil
| | - Milena B P Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil; SENAI Institute of Innovation (ISI) in Health Advanced Systems, University Center SENAI/CIMATEC, Salvador, Bahia, 41650-010, Brazil
| | - Monize M da Silva
- Department of Chemistry, Federal University of São Carlos, São Carlos, São Paulo, 13561-901 Brazil
| | - Alzir A Batista
- Department of Chemistry, Federal University of São Carlos, São Carlos, São Paulo, 13561-901 Brazil
| | - Viktor I Korolchuk
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil.
| |
Collapse
|
2
|
Yamashita N, Uchiyama M, Yamagata R, Hwang GW. Methylmercury Induces Apoptosis in Mouse C17.2 Neural Stem Cells through the Induction of OSGIN1 Expression by NRF2. Int J Mol Sci 2024; 25:3886. [PMID: 38612696 PMCID: PMC11011283 DOI: 10.3390/ijms25073886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/24/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
Methylmercury is a known environmental pollutant that exhibits severe neurotoxic effects. However, the mechanism by which methylmercury causes neurotoxicity remains unclear. To date, we have found that oxidative stress-induced growth inhibitor 1 (OSGIN1), which is induced by oxidative stress and DNA damage, is also induced by methylmercury. Therefore, in this study, we investigated the relationship between methylmercury toxicity and the induction of OSGIN1 expression using C17.2 cells, which are mouse brain neural stem cells. Methylmercury increased both OSGIN1 mRNA and protein levels in a time- and concentration-dependent manner. Moreover, these increases were almost entirely canceled out by pretreatment with actinomycin D, a transcription inhibitor. Furthermore, similar results were obtained from cells in which expression of the transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) was suppressed, indicating that methylmercury induces OSGIN1 expression via NRF2. Methylmercury causes neuronal cell death by inducing apoptosis. Therefore, we next investigated the role of OSGIN1 in methylmercury-induced neuronal cell death using the activation of caspase-3, which is involved in apoptosis induction, as an indicator. As a result, the increase in cleaved caspase-3 (activated form) induced by methylmercury exposure was decreased by suppressing OSGIN1, and the overexpression of OSGIN1 further promoted the increase in cleaved caspase-3 caused by methylmercury. These results suggest, for the first time, that OSGIN1 is a novel factor involved in methylmercury toxicity, and methylmercury induces apoptosis in C17.2 cells through the induction of OSGIN1 expression by NRF2.
Collapse
Affiliation(s)
| | | | | | - Gi-Wook Hwang
- Laboratory of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Miyagi, Japan; (N.Y.); (R.Y.)
| |
Collapse
|
3
|
Miyazaki I, Asanuma M. Multifunctional Metallothioneins as a Target for Neuroprotection in Parkinson's Disease. Antioxidants (Basel) 2023; 12:antiox12040894. [PMID: 37107269 PMCID: PMC10135286 DOI: 10.3390/antiox12040894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 03/27/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Parkinson's disease (PD) is characterized by motor symptoms based on a loss of nigrostriatal dopaminergic neurons and by non-motor symptoms which precede motor symptoms. Neurodegeneration accompanied by an accumulation of α-synuclein is thought to propagate from the enteric nervous system to the central nervous system. The pathogenesis in sporadic PD remains unknown. However, many reports indicate various etiological factors, such as oxidative stress, inflammation, α-synuclein toxicity and mitochondrial impairment, drive neurodegeneration. Exposure to heavy metals contributes to these etiopathogenesis and increases the risk of developing PD. Metallothioneins (MTs) are cysteine-rich metal-binding proteins; MTs chelate metals and inhibit metal-induced oxidative stress, inflammation and mitochondrial dysfunction. In addition, MTs possess antioxidative properties by scavenging free radicals and exert anti-inflammatory effects by suppression of microglial activation. Furthermore, MTs recently received attention as a potential target for attenuating metal-induced α-synuclein aggregation. In this article, we summarize MTs expression in the central and enteric nervous system, and review protective functions of MTs against etiopathogenesis in PD. We also discuss neuroprotective strategies for the prevention of central dopaminergic and enteric neurodegeneration by targeting MTs. This review highlights multifunctional MTs as a target for the development of disease-modifying drugs for PD.
Collapse
Affiliation(s)
- Ikuko Miyazaki
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Masato Asanuma
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
4
|
Ruczaj A, Brzóska MM. Environmental exposure of the general population to cadmium as a risk factor of the damage to the nervous system: A critical review of current data. J Appl Toxicol 2023; 43:66-88. [PMID: 35304765 PMCID: PMC10084305 DOI: 10.1002/jat.4322] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/14/2022] [Accepted: 03/15/2022] [Indexed: 12/16/2022]
Abstract
Nowadays, more and more attention has been focused on the risk of the neurotoxic action of cadmium (Cd) under environmental exposure. Due to the growing incidence of nervous system diseases, including neurodegenerative changes, and suggested involvement of Cd in their aetiopathogenesis, this review aimed to discuss critically this element neurotoxicity. Attempts have been made to recognize at which concentrations in the blood and urine Cd may increase the risk of damage to the nervous system and compare it to the risk of injury of other organs and systems. The performed overview of the available literature shows that Cd may have an unfavourable impact on the human's nervous system at the concentration >0.8 μg Cd/L in the urine and >0.6 μg Cd/L in the blood. Because such concentrations are currently noted in the general population of industrialized countries, it can be concluded that environmental exposure to this xenobiotic may create a risk of damage to the nervous system and be involved in the aetiopathogenesis of neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease, as well as worsening cognitive and behavioural functions. The potential mechanism of Cd neurotoxicity consists in inducing oxidative stress, disrupting the activity of enzymes essential to the proper functioning of the nervous system and destroying the homoeostasis of bioelements in the brain. Thus, further studies are necessary to recognize accurately both the risk of nervous system damage in the general population due to environmental exposure to Cd and the mechanism of this action.
Collapse
Affiliation(s)
- Agnieszka Ruczaj
- Department of ToxicologyMedical University of BialystokBialystokPoland
| | | |
Collapse
|
5
|
KARPUZ B, ÇAKIR Ö. Effect of proteasome inhibitor MG132 on the expression of oxidative metabolism related genes in tomato. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.52420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
6
|
Chang CH, Wei CC, Ho CT, Liao VHC. N-γ-(L-glutamyl)-L-selenomethionine shows neuroprotective effects against Parkinson's disease associated with SKN-1/Nrf2 and TRXR-1 in Caenorhabditis elegans. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 92:153733. [PMID: 34537465 DOI: 10.1016/j.phymed.2021.153733] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 07/08/2021] [Accepted: 09/01/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Parkinson's disease (PD) is a common neurodegenerative disease, yet fundamental treatments for the disease remain sparse. Thus, the search for potentially efficacious compounds from medicinal plants that can be used in the treatment of PD has gained significant interest. PURPOSE In many medicinal plants, selenium is primarily found in an organic form. We investigated the neuroprotective potential of an organic form of selenium, N-γ-(L-glutamyl)-L-selenomethionine (Glu-SeMet) in a Caenorhabditis elegans PD model and its possible molecular mechanisms. METHODS We used a C. elegans pharmacological PD strain (BZ555) that specifically expresses green fluorescent protein (GFP) in dopaminergic neurons and a transgenic PD strain (NL5901) that expresses human α-synuclein (α-syn) in muscle cells to investigate the neuroprotective potential of Glu-SeMet against PD. RESULTS We found that Glu-SeMet significantly ameliorated 6-hydroxydopamine (6-OHDA)-induced dopaminergic neuron damage in the transgenic BZ555 strain, with corresponding improvements in slowing behavior and intracellular ROS levels. In addition, compared with clinical PD drugs (L-DOPA and selegiline), Glu-SeMet demonstrated stronger ameliorated effects on 6-OHDA-induced toxicity. Glu-SeMet also triggered the nuclear translocation of SKN-1/Nrf2 and significantly increased SKN-1, GST-4, and GCS-1 mRNA levels in the BZ555 strain. However, Glu-SeMet did not increase mRNA levels or ameliorate the damage to dopaminergic neurons when the BZ555 strain was subjected to skn-1 RNA interference (RNAi). Glu-SeMet also upregulated the mRNA levels of the selenoprotein TRXR-1 in both the BZ555 and BZ555; skn-1 RNAi strains and significantly decreased α-syn accumulation in the NL5901 strain, although this was not observed in the NL5901; trxr-1 strain. CONCLUSION We found that Glu-SeMet has a neuroprotective effect against PD in a C. elegans PD model and that the anti-PD effects of Glu-SeMet were associated with SKN-1/Nrf2 and TRXR-1. Glu-SeMet may thus have the potential for use in therapeutic applications or supplements to slow the progression of PD.
Collapse
Affiliation(s)
- Chun-Han Chang
- Department of Bioenvironmental Systems Engineering, National Taiwan University, No. 1 Roosevelt Road, Sec. 4, Taipei 106, Taiwan
| | - Chia-Cheng Wei
- Institute of Food Safety and Health, National Taiwan University, No. 17, Xuzhou Rd., Taipei 100, Taiwan; Department of Public Health, National Taiwan University, No. 17, Xuzhou Rd., Taipei, 100, Taiwan
| | - Chi-Tang Ho
- Department of Food Science, School of Environmental and Biological Sciences, Rutgers, the State University of New Jersey, 65 Dudley Rd., New Brunswick, NJ 08901-8520, United States
| | - Vivian Hsiu-Chuan Liao
- Department of Bioenvironmental Systems Engineering, National Taiwan University, No. 1 Roosevelt Road, Sec. 4, Taipei 106, Taiwan.
| |
Collapse
|
7
|
Abbott LC, Nigussie F. Mercury Toxicity and Neurogenesis in the Mammalian Brain. Int J Mol Sci 2021; 22:ijms22147520. [PMID: 34299140 PMCID: PMC8305137 DOI: 10.3390/ijms22147520] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/12/2021] [Accepted: 07/05/2021] [Indexed: 01/21/2023] Open
Abstract
The mammalian brain is formed from billions of cells that include a wide array of neuronal and glial subtypes. Neural progenitor cells give rise to the vast majority of these cells during embryonic, fetal, and early postnatal developmental periods. The process of embryonic neurogenesis includes proliferation, differentiation, migration, the programmed death of some newly formed cells, and the final integration of differentiated neurons into neural networks. Adult neurogenesis also occurs in the mammalian brain, but adult neurogenesis is beyond the scope of this review. Developing embryonic neurons are particularly susceptible to neurotoxicants and especially mercury toxicity. This review focused on observations concerning how mercury, and in particular, methylmercury, affects neurogenesis in the developing mammalian brain. We summarized information on models used to study developmental mercury toxicity, theories of pathogenesis, and treatments that could be used to reduce the toxic effects of mercury on developing neurons.
Collapse
Affiliation(s)
- Louise C. Abbott
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, 4458 TAMU, College Station, TX 77843-4458, USA
- Correspondence: ; Tel.: +1-541-254-0779
| | - Fikru Nigussie
- College of Veterinary Medicine, Oregon State University, 700 SW 30th Street, Corvallis, OR 97331, USA;
| |
Collapse
|
8
|
Ijomone OM, Ijomone OK, Iroegbu JD, Ifenatuoha CW, Olung NF, Aschner M. Epigenetic influence of environmentally neurotoxic metals. Neurotoxicology 2020; 81:51-65. [PMID: 32882300 PMCID: PMC7708394 DOI: 10.1016/j.neuro.2020.08.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/25/2020] [Accepted: 08/25/2020] [Indexed: 02/08/2023]
Abstract
Continuous globalization and industrialization have ensured metals are an increasing aspect of daily life. Their usefulness in manufacturing has made them vital to national commerce, security and global economy. However, excess exposure to metals, particularly as a result of environmental contamination or occupational exposures, has been detrimental to overall health. Excess exposure to several metals is considered environmental risk in the aetiology of several neurological and neurodegenerative diseases. Metal-induced neurotoxicity has been a major health concern globally with intensive research to unravel the mechanisms associated with it. Recently, greater focus has been directed at epigenetics to better characterize the underlying mechanisms of metal-induced neurotoxicity. Epigenetic changes are those modifications on the DNA that can turn genes on or off without altering the DNA sequence. This review discusses how epigenetic changes such as DNA methylation, post translational histone modification and noncoding RNA-mediated gene silencing mediate the neurotoxic effects of several metals, focusing on manganese, arsenic, nickel, cadmium, lead, and mercury.
Collapse
Affiliation(s)
- Omamuyovwi M Ijomone
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria.
| | - Olayemi K Ijomone
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria; Department of Anatomy, University of Medical Sciences, Ondo, Nigeria
| | - Joy D Iroegbu
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Chibuzor W Ifenatuoha
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Nzube F Olung
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Michael Aschner
- Departments of Molecular Pharmacology and Neurosciences, Albert Einstein College of Medicine, NY, USA.
| |
Collapse
|
9
|
Abdelnour SA, Swelum AA, Abd El-Hack ME, Khafaga AF, Taha AE, Abdo M. Cellular and functional adaptation to thermal stress in ovarian granulosa cells in mammals. J Therm Biol 2020; 92:102688. [PMID: 32888576 DOI: 10.1016/j.jtherbio.2020.102688] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/21/2020] [Accepted: 08/05/2020] [Indexed: 01/06/2023]
Abstract
Climate change represents a significant environmental challenge to human welfare. One of many negative impacts may be on animal reproduction. Elevated ambient temperature unfavourably influences reproductive processes in mammals. High temperature can affect reproductive processes such as follicle development and may alter follicular fluid concentrations of amino acids, fatty acids, minerals, enzymes, antioxidants defence and growth factors. These impacts may lead to inferior oocyte competence and abnormal granulosa cell (GCs) function. Mammalian oocytes are enclosed by GCs that secret hormones and signalling molecules to promote oocyte competence. GCs are essential for proper follicular development, oocyte maturation, ovulation, and luteinization. Many environmental stressors, including thermal stress, affect GC function and alter oocyte development and growth. Several studies documented a link between elevated ambient temperature and increased generation of cellular reactive oxygen species (ROS). ROS can damage DNA, reduce cell proliferation, and induce apoptosis in GCs, thus altering oocyte development. Additionally, thermal stress induces upregulation of thermal shock proteins, such as HSP70 and HSP90. This review provides an update on the influence of thermal stress on GCs of mammals. Discussions include impacts to steroidogenesis (estradiol and progesterone), proliferation and cell cycle transition, apoptosis, oxidative stress (ROS), antioxidants related genes, heat shock proteins (HSPs) and endoplasmic reticulum responses.
Collapse
Affiliation(s)
- Sameh A Abdelnour
- Animal Production Department, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Ayman A Swelum
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, P.O. Box 2460, Riyadh, 11451, Saudi Arabia; Department of Theriogenology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt.
| | - Mohamed E Abd El-Hack
- Department of Poultry, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt.
| | - Asmaa F Khafaga
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina, 22758, Egypt
| | - Ayman E Taha
- Department of Animal Husbandry and Animal Wealth Development, Faculty of Veterinary Medicine, Alexandria University, Edfina, 22578, Egypt
| | - Mohamed Abdo
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, 32897, Egypt
| |
Collapse
|
10
|
Yang L, Zhang Y, Wang F, Luo Z, Guo S, Strähle U. Toxicity of mercury: Molecular evidence. CHEMOSPHERE 2020; 245:125586. [PMID: 31881386 DOI: 10.1016/j.chemosphere.2019.125586] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/28/2019] [Accepted: 12/08/2019] [Indexed: 05/25/2023]
Abstract
Minamata disease in Japan and the large-scale poisoning by methylmercury (MeHg) in Iraq caused wide public concerns about the risk emanating from mercury for human health. Nowadays, it is widely known that all forms of mercury induce toxic effects in mammals, and increasing evidence supports the concern that environmentally relevant levels of MeHg could impact normal biological functions in wildlife. The information of mechanism involved in mercurial toxicity is growing but knowledge gaps still exist between the adverse effects and mechanisms of action, especially at the molecular level. A body of data obtained from experimental studies on mechanisms of mercurial toxicity in vivo and in vitro points to that disruption of the antioxidant system may play an important role in the mercurial toxic effects. Moreover, the accumulating evidence indicates that signaling transduction, protein or/and enzyme activity, and gene regulation are involving in mediating toxic and adaptive response to mercury exposure. We conducted here a comprehensive review of mercurial toxic effects on wildlife and human, in particular synthesized key findings of molecular pathways involved in mercurial toxicity from the cells to human. We discuss the molecular evidence related mercurial toxicity to the adverse effects, with particular emphasis on the gene regulation. The further studies relying on Omic analysis connected to adverse effects and modes of action of mercury will aid in the evaluation and validation of causative relationship between health outcomes and gene expression.
Collapse
Affiliation(s)
- Lixin Yang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China.
| | - Yuanyuan Zhang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Feifei Wang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Zidie Luo
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Shaojuan Guo
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Uwe Strähle
- Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
11
|
Jiménez-Vidal L, Espitia-Pérez P, Torres-Ávila J, Ricardo-Caldera D, Salcedo-Arteaga S, Galeano-Páez C, Pastor-Sierra K, Espitia-Pérez L. Nuclear factor erythroid 2 - related factor 2 and its relationship with cellular response in nickel exposure: a systems biology analysis. BMC Pharmacol Toxicol 2019; 20:78. [PMID: 31852525 PMCID: PMC6921378 DOI: 10.1186/s40360-019-0360-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Nickel and nickel-containing compounds (NCC) are known human carcinogens. However, the precise molecular mechanisms of nickel-induced malignant transformation remain unknown. Proposed mechanisms suggest that nickel and NCC may participate in the dual activation/inactivation of enzymatic pathways involved in cell defenses against oxidative damage, where Nuclear factor-erythroid 2 related factor 2 (Nrf2) plays a central role. Methods For assessing the potential role of proteins involved in the Nrf2-mediated response to nickel and NCC exposure, we designed an interactome network using the STITCH search engine version 5.0 and the STRING software 10.0. The major NCC-protein interactome (NCPI) generated was analyzed using the MCODE plugin, version 1.5.1 for the detection of interaction modules or subnetworks. Main centralities of the NCPI were determined with the CentiScape 2.2 plugin of Cytoscape 3.4.0 and main biological processes associated with each cluster were assessed using the BiNGO plugin of Cytoscape 3.4.0. Results Water-soluble NiSO4 and insoluble Ni3S2 were the most connected to proteins involved in the NCPI network. Nfr2 was detected as one of the most relevant proteins in the network, participating in several multifunctional protein complexes in clusters 1, 2, 3 and 5. Ontological analysis of cluster 3 revealed several processes related to unfolded protein response (UPR) and response to endoplasmic reticulum (ER) stress. Conclusions Cellular response to NCC exposure was very comparable, particularly concerning oxidative stress response, inflammation, cell cycle/proliferation, and apoptosis. In this cellular response, Nfr2 was highly centralized and participated in several multifunctional protein complexes, including several related to ER-stress. These results add evidence on the possible Ni2+ induced – ER stress mainly associated with insoluble NCC. In this scenario, we also show how protein degradation mediated by ubiquitination seems to play key roles in cellular responses to Ni.
Collapse
Affiliation(s)
- Luisa Jiménez-Vidal
- Facultad de Ciencias de la Salud, Grupo de Investigación Biomédica y Biología Molecular, Universidad del Sinú, Calle 38 Cra 1W, Barrio Juan XXIII, Montería, Córdoba, Colombia
| | - Pedro Espitia-Pérez
- Facultad de Ciencias de la Salud, Grupo de Investigación Biomédica y Biología Molecular, Universidad del Sinú, Calle 38 Cra 1W, Barrio Juan XXIII, Montería, Córdoba, Colombia
| | - José Torres-Ávila
- Unit for Development and Innovation in Genetics and Molecular Biology, Universidad Simón Bolívar, Barranquilla, Atlántico, Colombia
| | - Dina Ricardo-Caldera
- Facultad de Ciencias de la Salud, Grupo de Investigación en Enfermedades Tropicales y Resistencia Bacteriana, Universidad del Sinú, Montería, Córdoba, Colombia
| | - Shirley Salcedo-Arteaga
- Facultad de Ciencias de la Salud, Grupo de Investigación Biomédica y Biología Molecular, Universidad del Sinú, Calle 38 Cra 1W, Barrio Juan XXIII, Montería, Córdoba, Colombia
| | - Claudia Galeano-Páez
- Facultad de Ciencias de la Salud, Grupo de Investigación Biomédica y Biología Molecular, Universidad del Sinú, Calle 38 Cra 1W, Barrio Juan XXIII, Montería, Córdoba, Colombia
| | - Karina Pastor-Sierra
- Facultad de Ciencias de la Salud, Grupo de Investigación Biomédica y Biología Molecular, Universidad del Sinú, Calle 38 Cra 1W, Barrio Juan XXIII, Montería, Córdoba, Colombia
| | - Lyda Espitia-Pérez
- Facultad de Ciencias de la Salud, Grupo de Investigación Biomédica y Biología Molecular, Universidad del Sinú, Calle 38 Cra 1W, Barrio Juan XXIII, Montería, Córdoba, Colombia.
| |
Collapse
|
12
|
Sudo K, VAN Dao C, Miyamoto A, Shiraishi M. Comparative analysis of in vitro neurotoxicity of methylmercury, mercury, cadmium, and hydrogen peroxide on SH-SY5Y cells. J Vet Med Sci 2019; 81:828-837. [PMID: 30996207 PMCID: PMC6612504 DOI: 10.1292/jvms.19-0059] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mercury (Hg) and cadmium (Cd) are the major toxic heavy metals and are known to induce
neurotoxicity. Although many studies have shown that several heavy metals have neurotoxic
effects, the cellular and molecular mechanisms thereof are still not clear. Oxidative
stress is reported to be a common and important mechanism in cytotoxicity induced by heavy
metals. However, the assays for identifying toxic mechanisms were not performed under the
same experimental conditions, making it difficult to compare toxic properties of the heavy
metals. In this study, we investigated the mechanisms underlying neurotoxicity induced by
heavy metals and H2O2, focusing on cell death, cell proliferation,
and oxidative stress under the same experimental condition. Our results showed that MeHg
caused lactate dehydrogenase (LDH) release, caspase activation and cell-cycle alteration,
and ROS generation in accordance with decreased cell viability. HgCl2 caused
LDH release and cell-cycle alteration, but not caspase activation. CdCl2 had a
remarkable effect on the cell cycle profiles without induction of LDH release, caspase
activation, or ROS generation. Pretreatment with N-acetyl-l-cysteine (NAC) prevented the
decrease in cell viability induced by MeHg and HgCl2, but not CdCl2.
Our results demonstrate a clear difference in neurotoxic mechanisms induced by MeHg,
HgCl2, CdCl2 or H2O2 in SH-SY5Y cells.
Elucidating the characteristics and mechanisms of each heavy metal under the same
experimental conditions will be helpful to understand the effect of heavy metals on health
and to develop a more effective therapy for heavy metal poisoning.
Collapse
Affiliation(s)
- Kasumi Sudo
- Department of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan.,Present address: Assay Division I, National Veterinary Assay Laboratory, Ministry of Agriculture, Forestry and Fisheries, 1-15-1 Tokura, Kokubunji, Tokyo 185-8511, Japan
| | - Cuong VAN Dao
- Department of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan.,Department of Veterinary Pharmacology, Faculty of Animal Husbandry and Veterinary Medicine, Thai Nguyen University of Agriculture and Forestry, Group 10, Quyet Thang Commune, Thai Nguyen City, Thai Nguyen, Vietnam
| | - Atsushi Miyamoto
- Department of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Mitsuya Shiraishi
- Department of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| |
Collapse
|
13
|
Wang P, Gong P, Wang W, Li J, Ai Y, Zhang X. An Eimeria acervulina OTU protease exhibits linkage-specific deubiquitinase activity. Parasitol Res 2018; 118:47-55. [PMID: 30415394 DOI: 10.1007/s00436-018-6113-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 10/07/2018] [Indexed: 02/03/2023]
Abstract
Ubiquitination is an important post-translational modification process that regulates many cellular processes. Proteins can be modified at single or multiple lysine residues by a single ubiquitin protein or by ubiquitin oligomers. It is important to note that the type of ubiquitin chains determines the functional outcome of the modification. Ubiquitin or ubiquitin chains can be removed by deubiquitinases (DUBs). In our previous study, the Eimeria tenella ovarian tumour (Et-OTU) DUB was shown to regulate the telomerase activity of E. tenella and affect E. tenella proliferation. The amino acid sequences of Et-OTU (GenBank: XP_013229759.1) and Eimeria acervulina (E. acervulina) ovarian tumour (Ea-OTUD3) DUB (XP_013250378.1) are 74% identical. Although Et-OTU may regulate E. tenella telomerase activity, whether Ea-OTUD3 affects E. acervulina growth and reproduction remains unclear. We show here that Ea-OTUD3 belongs to the OTU domain class of cysteine protease deubiquitinating enzymes. Ea-OTUD3 is highly linkage-specific, cleaving K48 (Lys48)-, K63-, and K6-linked diubiquitin but not K29-, K33-, and K11-linked diubiquitin. The precise linkage preference of Ea-OTUD3 among these three nonlinear diubiquitin chains is K6 > K48 > K63. Recombinant Ea-OTUD3, but not its catalytic-site mutant Ea-OTUD3 (C247A), exhibits activity against diubiquitin. Ea-OTUD3 removes ubiquitin from the K48-, but to a lesser extent from the K63-linked ubiquitinated E. acervulina proteins of the modified target protein, thereby exhibiting the characteristics of deubiquitinase. This study reveals that the Ea-OTUD3 is a novel functional deubiquitinating enzyme. Furthermore, the Ea-OTUD3 protein may regulate the stability of some K48-linked ubiquitinated E. acervulina proteins.
Collapse
Affiliation(s)
- Pu Wang
- College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Pengtao Gong
- College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Weirong Wang
- College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Jianhua Li
- College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Yongxing Ai
- College of Animal Science, Jilin University, Changchun, 130062, China.
| | - Xichen Zhang
- College of Veterinary Medicine, Jilin University, Changchun, 130062, China.
| |
Collapse
|
14
|
Arsenic-induced apoptosis in the p53-proficient and p53-deficient cells through differential modulation of NFkB pathway. Food Chem Toxicol 2018; 118:849-860. [PMID: 29944914 DOI: 10.1016/j.fct.2018.06.053] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/21/2018] [Accepted: 06/22/2018] [Indexed: 12/31/2022]
Abstract
Arsenic is a well-known environmental carcinogen and an effective chemotherapeutic agent. The underlying mechanism of this dual-effect, however, is not fully understood. In this study, we applied mouse p53+/+ and p53-/- cells to examine the NFκB pathway and proinflammatory cytokines after arsenic treatment. Arsenic reduced cell viability and increased more apoptosis in the p53-/- cells as compared to p53+/+ cells, which was correlated with activation of SAPK/JNK, p38 MAPK, and AKT pathways. A transcriptional regulatory network analysis revealed that arsenic activated transcription regulatory elements E2F, Egr1, Trp53, Stat6, Bcl6, Creb2 and ATF4 in the p53+/+ cells, while in the p53-/- cells, arsenic treatment altered transcription factors NFκB, Pparg, Creb2, ATF4, and Egr1. We observed dynamic changes in phosphorylated NFκB p65 (p-NFκB p65) and phosphorylated IKKαβ (p-IKKαβ) in both genotypes from 4 h to 24 h after treatment, significant decreases of p-NFκB p65 and p-IKKαβ in the p53-/- cells, whereas increases of p-NFκB p65 and p-IKKαβ were observed in the p53+/+ cells. Our study confirmed the differential modulation of NFκB pathway by arsenic in the p53+/+ or p53-/- cells and this observation of the differential mechanism of cell death between the p53+/+ and p53-/- cells might be linked to the unique ability of arsenic to act as both a carcinogen and a chemotherapeutic agent.
Collapse
|
15
|
Mohammadi S, Gholamin M, Mohammadi M, Mansouri A, Mahmoodian R, Attari S, Kebriaei SM, Zibaei B, Roshanaei M, Daneshvar F, Khandehro M, Khodadadegan MA, Delshad A, Mohammadzadeh F, Peyvandi M, Ghayour-Mobarhan M, Tavallaie S, Boroumand-Noughabi S, Ferns GAA. Down-regulation of CatSper 1 and CatSper 2 genes by lead and mercury. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 59:82-86. [PMID: 29549816 DOI: 10.1016/j.etap.2018.03.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 06/29/2017] [Accepted: 03/05/2018] [Indexed: 06/08/2023]
Abstract
In the study of the expression of CatSper genes, consideration of the effects of environmental metal toxicity is very important. Therefore, in this study, the effects of lead acetate and mercury chloride exposure on expression of CatSper genes, sperm parameters, histology of testis and prooxidant antioxidant balance (PAB) values of serum were investigated. A total of 28 mice was divided into four groups. The control group did not receive injections. The sham group received normal saline intraperitoneally. Lead and mercury groups were injected 60 and 1.25 mg/kg/daily lead acetate and mercury chloride respectively intraperitoneally for 2 weeks. After 35 days, the sperm analysis and histology of left testis were performed. In addition, serum was obtained to measure the PAB values. The right testis was used for molecular analysis of real-time PCR. Administration with either lead acetate or mercury caused significant damage to the seminiferous tubules as well as a reduction in sperm parameters compared to the control group. The relative expression of CatSper 1 and CatSper 2 in the lead group was lower than that of the control group (-0.01 ± 0.24, -0.007 ± 0.52 vs. 1 ± 0.50, P = 0.34). The relative expression of CatSper 1 and CatSper 2 was significantly lower in the mercury group compared to the control ones (-0.24 ± 2.28, -4.49 ± 4.86 vs. 1 ± 0.50, P = 0.21). PAB values significantly increased in lead or mercury exposed- mice compared to the control ones (0.93 ± 0.17, 1.54 ± 0.17 vs. 0.51 ± 0.11; P ≤ 0.000). The results of this study showed that administration with either lead acetate or mercury chloride caused degenerative damage in seminiferous tubules and reduction in sperm quality and expression of CatSper 1, 2 genes in mice. Therefore, it is possible in infertile men who have had exposure to lead acetate or mercury chloride. Owing to structural similarities, these metals are substitutes for calcium ions and have effects on calcium channels. These cause immobility in sperm by blocking CatSper-specific calcium channels. However, more studies are required to elucidate the mechanism underlying the impact of different doses of heavy metals on CatSper genes expression.
Collapse
Affiliation(s)
- Sh Mohammadi
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - M Gholamin
- Bou-ali Research Institute, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - M Mohammadi
- Department of Public Health, School of Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - A Mansouri
- Department of Nanomedicine, Avicenna Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - R Mahmoodian
- Bou-ali Research Institute, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - S Attari
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - S M Kebriaei
- Student Research Committee, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
| | - B Zibaei
- Student Research Committee, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - M Roshanaei
- Student Research Committee, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - F Daneshvar
- Student Research Committee, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - M Khandehro
- Student Research Committee, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - M A Khodadadegan
- Student Research Committee, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - A Delshad
- Student Research Committee, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - F Mohammadzadeh
- Student Research Committee, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Mostafa Peyvandi
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Ghayour-Mobarhan
- Biochemistry of Nutritional Research Center, Mashhad University of Medical Science, Mashhad, Iran
| | - Shima Tavallaie
- Biochemistry of Nutritional Research Center, Mashhad University of Medical Science, Mashhad, Iran
| | | | - Gordon A A Ferns
- Institute for Science and Technology in Medicine, University of Keele, Guy Hilton Research Center, Thornburrow Drive, Stoke on Trent, Staffordshire ST4 7QB, UK
| |
Collapse
|
16
|
Alemu TW, Pandey HO, Salilew Wondim D, Gebremedhn S, Neuhof C, Tholen E, Holker M, Schellander K, Tesfaye D. Oxidative and endoplasmic reticulum stress defense mechanisms of bovine granulosa cells exposed to heat stress. Theriogenology 2018; 110:130-141. [PMID: 29396041 DOI: 10.1016/j.theriogenology.2017.12.042] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 12/15/2017] [Accepted: 12/29/2017] [Indexed: 12/20/2022]
Abstract
In most mammalian species including cattle, heat stress has detrimental effects on ovarian function through disturbing estradiol production and viability of granulosa cells. However, effect of heat stress and underlying cellular defense mechanisms of bovine granulosa cells is not fully understood. Here, we aimed to investigate the effect of heat stress on granulosa cells function and the associated defense mechanism. For this an in vitro granulosa cell model was used to investigate the role of elevated temperature (41 °C) on granulosa cell functions at 24 h and 48 h exposure compared to the control cultured at 37 °C. The results showed that reactive oxygen species level was higher in cells under 41 °C at 24 h compared to control. In response to increased reactive oxygen species level, the expression of NRF2 and its antioxidant genes, CAT and PRDX1 were higher in bovine granulosa cells exposed to heat stress. Interestingly, heat stress markedly increased expression of endoplasmic reticulum stress marker genes; GRP78 and GRP94, in cultured bovine granulosa cells at 24 h, and higher protein accumulation of GRP78 accompanied by increased expression of apoptotic genes, BAX and CASPASE-3. Moreover, heat stress significantly decreased the bovine granulosa cells proliferation, which was supported by decreased in the expression of proliferation marker gene PCNA. All in all heat stress induce reactive oxygen species accumulation, apoptosis and reduced proliferation, which trigger the NRF2 mediated oxidative stress and endoplasmic reticulum stress response by bovine granulosa cells.
Collapse
Affiliation(s)
- Teshome Wondie Alemu
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, 53115, Bonn, Germany
| | - Hari Om Pandey
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, 53115, Bonn, Germany
| | - Dessie Salilew Wondim
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, 53115, Bonn, Germany
| | - Samuel Gebremedhn
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, 53115, Bonn, Germany
| | - Christiane Neuhof
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, 53115, Bonn, Germany
| | - Ernst Tholen
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, 53115, Bonn, Germany
| | - Michael Holker
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, 53115, Bonn, Germany
| | - Karl Schellander
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, 53115, Bonn, Germany
| | - Dawit Tesfaye
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, 53115, Bonn, Germany.
| |
Collapse
|
17
|
Gleixner AM, Hutchison DF, Sannino S, Bhatia TN, Leak LC, Flaherty PT, Wipf P, Brodsky JL, Leak RK. N-Acetyl-l-Cysteine Protects Astrocytes against Proteotoxicity without Recourse to Glutathione. Mol Pharmacol 2017; 92:564-575. [PMID: 28830914 DOI: 10.1124/mol.117.109926] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/17/2017] [Indexed: 02/06/2023] Open
Abstract
N-acetyl-l-cysteine (NAC) exhibits protective properties in brain injury models and has undergone a number of clinical trials. Most studies of NAC have focused on neurons. However, neuroprotection may be complemented by the protection of astrocytes because healthier astrocytes can better support the viability of neurons. Here, we show that NAC can protect astrocytes against protein misfolding stress (proteotoxicity), the hallmark of neurodegenerative disorders. Although NAC is thought to be a glutathione precursor, NAC protected primary astrocytes from the toxicity of the proteasome inhibitor MG132 without eliciting any increase in glutathione. Furthermore, glutathione depletion failed to attenuate the protective effects of NAC. MG132 elicited a robust increase in the folding chaperone heat shock protein 70 (Hsp70), and NAC mitigated this effect. Nevertheless, three independent inhibitors of Hsp70 function ablated the protective effects of NAC, suggesting that NAC may help preserve Hsp70 chaperone activity and improve protein quality control without need for Hsp70 induction. Consistent with this view, NAC abolished an increase in ubiquitinated proteins in MG132-treated astrocytes. However, NAC did not affect the loss of proteasome activity in response to MG132, demonstrating that it boosted protein homeostasis and cell viability without directly interfering with the efficacy of this proteasome inhibitor. The thiol-containing molecules l-cysteine and d-cysteine both mimicked the protective effects of NAC, whereas the thiol-lacking molecule N-acetyl-S-methyl-l-cysteine failed to exert protection or blunt the rise in ubiquitinated proteins. Collectively, these findings suggest that the thiol group in NAC is required for its effects on glial viability and protein quality control.
Collapse
Affiliation(s)
- Amanda M Gleixner
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Daniel F Hutchison
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Sara Sannino
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Tarun N Bhatia
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Lillian C Leak
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Patrick T Flaherty
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Peter Wipf
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Jeffrey L Brodsky
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| |
Collapse
|
18
|
Abstract
BACKGROUND Recent evidence highlights the reality of unprecedented human exposure to toxic chemical agents found throughout our environment - in our food and water supply, in the air we breathe, in the products we apply to our skin, in the medical and dental materials placed into our bodies, and even within the confines of the womb. With biomonitoring confirming the widespread bioaccumulation of myriad toxicants among population groups, expanding research continues to explore the pathobiological impact of these agents on human metabolism. METHODS This review was prepared by assessing available medical and scientific literature from Medline as well as by reviewing several books, toxicology journals, government publications, and conference proceedings. The format of a traditional integrated review was chosen. RESULTS Toxicant exposure and accrual has been linked to numerous biochemical and pathophysiological mechanisms of harm. Some toxicants effect metabolic disruption via multiple mechanisms. CONCLUSIONS As a primary causative determinant of chronic disease, toxicant exposures induce metabolic disruption in myriad ways, which consequently result in varied clinical manifestations, which are then categorized by health providers into innumerable diagnoses. Chemical disruption of human metabolism has become an etiological determinant of much illness throughout the lifecycle, from neurodevelopmental abnormalities in-utero to dementia in the elderly.
Collapse
Affiliation(s)
- Stephen J Genuis
- a Faculty of Medicine, University of Alberta , Edmonton , Alberta , Canada
| | - Edmond Kyrillos
- b Department of Family Medicine , Faculty of Medicine, University of Ottawa , Ottawa , Ontario , Canada
| |
Collapse
|
19
|
Upadhyay A, Joshi V, Amanullah A, Mishra R, Arora N, Prasad A, Mishra A. E3 Ubiquitin Ligases Neurobiological Mechanisms: Development to Degeneration. Front Mol Neurosci 2017; 10:151. [PMID: 28579943 PMCID: PMC5437216 DOI: 10.3389/fnmol.2017.00151] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 05/04/2017] [Indexed: 01/08/2023] Open
Abstract
Cells regularly synthesize new proteins to replace old or damaged proteins. Deposition of various aberrant proteins in specific brain regions leads to neurodegeneration and aging. The cellular protein quality control system develop various defense mechanisms against the accumulation of misfolded and aggregated proteins. The mechanisms underlying the selective recognition of specific crucial protein or misfolded proteins are majorly governed by quality control E3 ubiquitin ligases mediated through ubiquitin-proteasome system. Few known E3 ubiquitin ligases have shown prominent neurodevelopmental functions, but their interactions with different developmental proteins play critical roles in neurodevelopmental disorders. Several questions are yet to be understood properly. How E3 ubiquitin ligases determine the specificity and regulate degradation of a particular substrate involved in neuronal proliferation and differentiation is certainly the one, which needs detailed investigations. Another important question is how neurodevelopmental E3 ubiquitin ligases specifically differentiate between their versatile range of substrates and timing of their functional modulations during different phases of development. The premise of this article is to understand how few E3 ubiquitin ligases sense major molecular events, which are crucial for human brain development from its early embryonic stages to throughout adolescence period. A better understanding of these few E3 ubiquitin ligases and their interactions with other potential proteins will provide invaluable insight into disease mechanisms to approach toward therapeutic interventions.
Collapse
Affiliation(s)
- Arun Upadhyay
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology JodhpurJodhpur, India
| | - Vibhuti Joshi
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology JodhpurJodhpur, India
| | - Ayeman Amanullah
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology JodhpurJodhpur, India
| | - Ribhav Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology JodhpurJodhpur, India
| | - Naina Arora
- School of Basic Sciences, Indian Institute of Technology MandiMandi, India
| | - Amit Prasad
- School of Basic Sciences, Indian Institute of Technology MandiMandi, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology JodhpurJodhpur, India
| |
Collapse
|
20
|
Schumacher L, Abbott LC. Effects of methyl mercury exposure on pancreatic beta cell development and function. J Appl Toxicol 2016; 37:4-12. [PMID: 27594070 DOI: 10.1002/jat.3381] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 07/29/2016] [Accepted: 07/31/2016] [Indexed: 12/31/2022]
Abstract
Methyl mercury is an environmental contaminant of worldwide concern. Since the discovery of methyl mercury exposure due to eating contaminated fish as the underlying cause of the Minamata disaster, the scientific community has known about the sensitivity of the developing central nervous system to mercury toxicity. Warnings are given to pregnant women and young children to limit consumption of foods containing methyl mercury to protect the embryonic, fetal and postnatally developing central nervous system. However, evidence also suggests that exposure to methyl mercury or various forms of inorganic mercury may also affect development and function of other organs. Numerous reports indicate a worldwide increase in diabetes, particularly type 2 diabetes. Quite recently, methyl mercury has been shown to have adverse effects on pancreatic beta (β) cell development and function, resulting in insulin resistance and hyperglycemia and may even lead to the development of diabetes. This review discusses possible mechanisms by which methyl mercury exposure may adversely affect pancreatic β cell development and function, and the role that methyl mercury exposure may have in the reported worldwide increase in diabetes, particularly type 2 diabetes. While additional information is needed regarding associations between mercury exposure and specific mechanisms of the pathogenesis of diabetes in the human population, methyl mercury's adverse effects on the body's natural sources of antioxidants suggest that one possible therapeutic strategy could involve supplementation with antioxidants. Thus, it is important that additional investigation be undertaken into the role of methyl mercury exposure and reduced pancreatic β cell function. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Lauren Schumacher
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, TX, 77843-4458, USA
| | - Louise C Abbott
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, TX, 77843-4458, USA
| |
Collapse
|
21
|
Shao Y, Figeys D, Ning Z, Mailloux R, Chan HM. Methylmercury can induce Parkinson's-like neurotoxicity similar to 1-methyl-4- phenylpyridinium: a genomic and proteomic analysis on MN9D dopaminergic neuron cells. J Toxicol Sci 2016; 40:817-28. [PMID: 26558463 DOI: 10.2131/jts.40.817] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Exposure to environmental chemicals has been implicated as a possible risk factor for the development of neurodegenerative diseases. Our previous study showed that methylmercury (MeHg) exposure can disrupt synthesis, uptake and metabolism of dopamine similar to 1-methyl-4-phenylpyridinium (MPP(+)). The objective of this study was to investigate the effects of MeHg exposure on gene and protein profiles in a dopaminergic MN9D cell line. MN9D cells were treated with MeHg (1-5 μM) and MPP(+) (10-40 μM) for 48 hr. Real-time PCR Parkinson's disease (PD) arrays and high-performance liquid chromatography/electrospray ionization tandem mass spectrometry (HPLC-ESI-MS/MS) were performed for the analysis. PD PCR array results showed that 19% genes were significantly changed in the 2.5 μM MeHg treated cells, and 39% genes were changed in the 5 μM MeHg treated cells. In comparison, MPP(+) treatment (40 µM) resulted in significant changes in 25% genes. A total of 15 common genes were altered by both MeHg and MPP(+), and dopaminergic signaling transduction was the most affected pathway. Proteomic analysis identified a total of 2496 proteins, of which 188, 233 and 395 proteins were differentially changed by 1 μM and 2.5 μM MeHg, and MPP(+) respectively. A total of 61 common proteins were changed by both MeHg and MPP(+) treatment. The changed proteins were mainly involved in energetic generation-related metabolism pathway (propanoate metabolism, pyruvate metabolism and fatty acid metabolism), oxidative phosphorylation, proteasome, PD and other neurodegenerative disorders. A total of 7 genes/proteins including Ube2l3 (Ubiquitin-conjugating enzyme E2 L3) and Th (Tyrosine 3-monooxygenase) were changed in both genomic and proteomic analysis. These results suggest that MeHg and MPP(+) share many similar signaling pathways leading to the pathogenesis of PD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Yueting Shao
- Natural Resources and Environmental Studies Program, University of Northern British Columbia, Canada
| | | | | | | | | |
Collapse
|
22
|
Coherent and Contradictory Facts, Feats and Fictions Associated with Metal Accumulation in Parkinson's Disease: Epicenter or Outcome, Yet a Demigod Question. Mol Neurobiol 2016; 54:4738-4755. [PMID: 27480264 DOI: 10.1007/s12035-016-0016-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 07/12/2016] [Indexed: 01/30/2023]
Abstract
Unwarranted exposure due to liberal use of metals for maintaining the lavish life and to achieve the food demand for escalating population along with an incredible boost in the average human life span owing to orchestrated progress in rejuvenation therapy have gradually increased the occurrence of Parkinson's disease (PD). Etiology is albeit elusive; association of PD with metal accumulation has never been overlooked due to noteworthy similitude between metal-exposure symptoms and a few cardinal features of disease. Even though metals are entailed in the vital functions, a hysterical shift, primarily augmentation, escorts the stern nigrostriatal dopaminergic neurodegeneration. An increase in the passage of metals through the blood brain barrier and impaired metabolic activity and elimination system could lead to metal accumulation in the brain, which eventually makes dopaminergic neurons quite susceptible. In the present article, an update on implication of metal accumulation in PD/Parkinsonism has been provided. Moreover, encouraging and paradoxical facts and fictions associated with metal accumulation in PD/Parkinsonism have also been compiled. Systematic literature survey of PD is performed to describe updated information if metal accumulation is an epicenter or merely an outcome. Finally, a perspective on the association of metal accumulation with pesticide-induced Parkinsonism has been explained to unveil the likely impact of the former in the latter.
Collapse
|
23
|
Fleming DS, Koltes JE, Markey AD, Schmidt CJ, Ashwell CM, Rothschild MF, Persia ME, Reecy JM, Lamont SJ. Genomic analysis of Ugandan and Rwandan chicken ecotypes using a 600 k genotyping array. BMC Genomics 2016; 17:407. [PMID: 27230772 PMCID: PMC4882793 DOI: 10.1186/s12864-016-2711-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 05/06/2016] [Indexed: 02/07/2023] Open
Abstract
Background Indigenous populations of animals have developed unique adaptations to their local environments, which may include factors such as response to thermal stress, drought, pathogens and suboptimal nutrition. The survival and subsequent evolution within these local environments can be the result of both natural and artificial selection driving the acquisition of favorable traits, which over time leave genomic signatures in a population. This study’s goals are to characterize genomic diversity and identify selection signatures in chickens from equatorial Africa to identify genomic regions that may confer adaptive advantages of these ecotypes to their environments. Results Indigenous chickens from Uganda (n = 72) and Rwanda (n = 100), plus Kuroilers (n = 24, an Indian breed imported to Africa), were genotyped using the Axiom® 600 k Chicken Genotyping Array. Indigenous ecotypes were defined based upon location of sampling within Africa. The results revealed the presence of admixture among the Ugandan, Rwandan, and Kuroiler populations. Genes within runs of homozygosity consensus regions are linked to gene ontology (GO) terms related to lipid metabolism, immune functions and stress-mediated responses (FDR < 0.15). The genes within regions of signatures of selection are enriched for GO terms related to health and oxidative stress processes. Key genes in these regions had anti-oxidant, apoptosis, and inflammation functions. Conclusions The study suggests that these populations have alleles under selective pressure from their environment, which may aid in adaptation to harsh environments. The correspondence in gene ontology terms connected to stress-mediated processes across the populations could be related to the similarity of environments or an artifact of the detected admixture. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2711-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - J E Koltes
- Iowa State University, Ames, IA, USA.,University of Arkansas, Fayetteville, AR, USA
| | | | | | - C M Ashwell
- North Carolina State University, Raleigh, NC, USA
| | | | - M E Persia
- Virginia Polytechnic University, Blacksburg, VA, USA
| | - J M Reecy
- Iowa State University, Ames, IA, USA
| | | |
Collapse
|
24
|
Guan D, Mo F, Han Y, Gu W, Zhang M. Digital gene expression profiling (DGE) of cadmium-treated Drosophila melanogaster. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 39:300-6. [PMID: 25543212 DOI: 10.1016/j.etap.2014.11.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 11/28/2014] [Accepted: 11/29/2014] [Indexed: 05/06/2023]
Abstract
Cadmium is highly toxic and can cause oxidative damage, metabolic disorders, and reduced lifespan and fertility in animals. In this study, we investigated the effects of cadmium in Drosophila melanogaster, performing transcriptome analysis by using tag-based digital gene expression (DGE) profiling. Among 1970 candidate genes, 1443 were up-regulated and 527 were down-regulated following cadmium exposure. Using Gene Ontology analysis, we found that cadmium stress affects three processes: transferase activity, stress response, and the cell cycle. Furthermore, we identified five differentially expressed genes (confirmed by real-time PCR) involved in all three processes: Ald, Cdc2, skpA, tefu, and Pvr. Pathway analysis revealed that these genes were involved in the cell cycle pathway and fat digestion and absorption pathway. This study reveals the gene expression response to cadmium stress in Drosophila, it provides insights into the mechanisms of this response, and it could contribute to our understanding of cadmium toxicity in humans.
Collapse
Affiliation(s)
- Delong Guan
- College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China
| | - Fei Mo
- College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China
| | - Yan Han
- College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China
| | - Wei Gu
- College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China
| | - Min Zhang
- College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China.
| |
Collapse
|
25
|
Götze S, Bose A, Sokolova IM, Abele D, Saborowski R. The proteasomes of two marine decapod crustaceans, European lobster (Homarus gammarus) and Edible crab (Cancer pagurus), are differently impaired by heavy metals. Comp Biochem Physiol C Toxicol Pharmacol 2014; 162:62-9. [PMID: 24721378 DOI: 10.1016/j.cbpc.2014.03.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 02/23/2014] [Accepted: 03/31/2014] [Indexed: 01/19/2023]
Abstract
The intracellular ubiquitin-proteasome system is a key regulator of cellular processes involved in the controlled degradation of short-living or malfunctioning proteins. Certain diseases and cellular dysfunctions are known to arise from the disruption of proteasome pathways. Trace metals are recognized stressors of the proteasome system in vertebrates and plants, but their effects on the proteasome of invertebrates are not well understood. Since marine invertebrates, and particularly benthic crustaceans, can be exposed to high metal levels, we studied the effects of in vitro exposure to Hg(2+), Zn(2+), Cu(2+), and Cd(2+) on the activities of the proteasome from the claw muscles of lobsters (Homarus gammarus) and crabs (Cancer pagurus). The chymotrypsin like activity of the proteasome of these two species showed different sensitivity to metals. In lobsters the activity was significantly inhibited by all metals to a similar extent. In crabs the activities were severely suppressed only by Hg(2+) and Cu(2+) while Zn(2+) had only a moderate effect and Cd(2+) caused almost no inhibition of the crab proteasome. This indicates that the proteasomes of both species possess structural characteristics that determine different susceptibility to metals. Consequently, the proteasome-mediated protein degradation in crab C. pagurus may be less affected by metal pollution than that of the lobster H. gammarus.
Collapse
Affiliation(s)
- Sandra Götze
- Alfred Wegener Institute, Helmholtz Centre for Polar and Marine Research, Functional Ecology, 27570 Bremerhaven, Germany
| | - Aneesh Bose
- Alfred Wegener Institute, Helmholtz Centre for Polar and Marine Research, Functional Ecology, 27570 Bremerhaven, Germany
| | - Inna M Sokolova
- Department of Biology, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Doris Abele
- Alfred Wegener Institute, Helmholtz Centre for Polar and Marine Research, Functional Ecology, 27570 Bremerhaven, Germany
| | - Reinhard Saborowski
- Alfred Wegener Institute, Helmholtz Centre for Polar and Marine Research, Functional Ecology, 27570 Bremerhaven, Germany.
| |
Collapse
|
26
|
Ivanina AV, Hawkins C, Sokolova IM. Immunomodulation by the interactive effects of cadmium and hypercapnia in marine bivalves Crassostrea virginica and Mercenaria mercenaria. FISH & SHELLFISH IMMUNOLOGY 2014; 37:299-312. [PMID: 24594010 DOI: 10.1016/j.fsi.2014.02.016] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 02/17/2014] [Accepted: 02/20/2014] [Indexed: 06/03/2023]
Abstract
Estuarine organisms are exposed to multiple stressors including large fluctuations in partial pressure of carbon dioxide (P2CO) and concentrations of trace metals such as cadmium (Cd) that can affect their survival and fitness. Ocean acidification due to the increasing atmospheric (P2CO) leads to a decrease in pH and shifts in the carbonate chemistry of seawater which can change bioavailability and toxicity of metals. We studied the interactive effects of (P2CO) and Cd exposure on metal levels, metabolism and immune-related functions in hemocytes of two ecologically and economically important bivalve species, Mercenaria mercenaria (hard shell clam) and Crassostrea virginica (Eastern oyster). Clams and oysters were exposed to combinations of three (P2CO) levels (∼400, 800 and 2000 μatm (P2CO), corresponding to the present day conditions and the projections for the years 2100 and 2250, respectively) and two Cd concentrations (0 and 50 μg l(-1)) in seawater. Following four weeks of exposure to Cd, hemolymph of both species contained similar Cd levels (50-70 μg l(-1)), whereas hemocytes accumulated intracellular Cd burdens up to 15-42 mg l(-1), regardless of the exposure P2CO. Clam hemocytes had considerably lower Cd burdens than those of oysters (0.7-1 ng 10(-6) cells vs. 4-6 ng 10(-6) cells, respectively). Cd exposure suppressed hemocyte metabolism and increased the rates of mitochondrial proton leak in normocapnia indicating partial mitochondrial uncoupling. This Cd-induced mitochondrial uncoupling was alleviated in hypercapnia. Cd exposure suppressed immune-related functions in hemocytes of clams and oysters, and these effects were exacerbated at elevated (P2CO). Thus, elevated (P2CO) combined with Cd exposure resulted in decrease in phagocytic activity and adhesion capacity as well as lower expression of mRNA for lectin and heat shock protein (HSP70) in clam and oyster hemocytes. In oysters, combined exposure to elevated (P2CO) and Cd also led to reduced activity of lysozyme in hemocytes and hemolymph. Overall, our study shows that moderately elevated (P2CO) (∼800-2000 μatm P2CO) potentiates the negative effects of Cd on immunity and thus may sensitize clams and oysters to pathogens and diseases during seasonal hypercapnia and/or ocean acidification in polluted estuaries.
Collapse
Affiliation(s)
- Anna V Ivanina
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Chelsea Hawkins
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Inna M Sokolova
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA.
| |
Collapse
|
27
|
Tokumoto M, Lee JY, Fujiwara Y, Satoh M. DNA microarray expression analysis of mouse kidney following cadmium exposure for 12 months. J Toxicol Sci 2014; 38:799-802. [PMID: 24067728 DOI: 10.2131/jts.38.799] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Cadmium (Cd) is a toxic heavy metal and chronic exposure causes kidney injury. This study used DNA microarray analysis to examine gene expression in the kidney of mice chronically exposed to Cd. Female C57BL/6J mice were fed a 300 ppm Cd-containing diet or a control diet for 12 months. In comparison with control mice, the expression levels of 32 genes, including Hmox1 and Mt2, were elevated more than 2.0-fold, whereas 113 genes, including transport- and ubiquitination-related genes, were reduced less than 0.5-fold.
Collapse
Affiliation(s)
- Maki Tokumoto
- Laboratory of Chemical Toxicology and Environmental Health, Showa Pharmaceutical University
| | | | | | | |
Collapse
|
28
|
Environmental pollutants and lifestyle factors induce oxidative stress and poor prenatal development. Reprod Biomed Online 2014; 29:17-31. [PMID: 24813750 DOI: 10.1016/j.rbmo.2014.03.002] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/13/2013] [Accepted: 03/06/2014] [Indexed: 01/20/2023]
Abstract
Developmental toxicity caused by exposure to a mixture of environmental pollutants has become a major health concern. Human-made chemicals, including xenoestrogens, pesticides and heavy metals, as well as unhealthy lifestyle behaviours, mainly tobacco smoking, alcohol consumption and medical drug abuse, are major factors that adversely influence prenatal development and increase susceptibility of offspring to diseases. There is evidence to suggest that the developmental toxicological mechanisms of chemicals and lifestyle factors involve the generation of reactive oxygen species (ROS) and cellular oxidative damage. Overproduction of ROS induces oxidative stress, a state where increased ROS generation overwhelms antioxidant protection and subsequently leads to oxidative damage of cellular macromolecules. Data on the involvement of oxidative stress in the mechanism of developmental toxicity following exposure to environmental pollutants are reviewed in an attempt to provide an updated basis for future studies on the toxic effect of such pollutants, particularly the notion of increased risk for developmental toxicity due to combined and cumulative exposure to various environmental pollutants. The aims of such studies are to better understand the mechanisms by which environmental pollutants adversely affect conceptus development and to elucidate the impact of cumulative exposures to multiple pollutants on post-natal development and health outcomes. Developmental toxicity caused by exposure to mixture of environmental pollutants has become a major health concern. Human-made chemicals, including xenoestrogens, pesticides and heavy metals, as well as unhealthy lifestyle behaviors, mainly tobacco smoking, alcohol consumption and medical drug abuse, are major factors that adversely influence prenatal development and increase the susceptibility of offspring to development complications and diseases. There is evidence to suggest that the developmental toxicological mechanisms of human-made chemicals and unhealthy lifestyle factors involve the generation of reactive oxygen species (ROS) and cellular oxidative damage. Overproduction of ROS induces oxidative stress, a state where increased generation of ROS overwhelms antioxidant protection and subsequently leads to oxidative damage of cellular macromolecules. Exposure to various environmental pollutants induces synergic and cumulative dose-additive adverse effects on prenatal development, pregnancy outcomes and neonate health. Data from the literature on the involvement of oxidative stress in the mechanism of developmental toxicity following in vivo exposure to environmental pollutants will be reviewed in an attempt to provide an updated basis for future studies on the toxic effect of such pollutants, particularly the notion of increased risk for developmental toxicity due to combined and cumulative exposure to various environmental pollutants. The aims of such studies are to better understand the mechanisms by which environmental pollutants adversely affect conceptus development and to elucidate the impact of cumulative exposures to multiple pollutants on postnatal development and health outcomes.
Collapse
|
29
|
VanDuyn N, Nass R. The putative multidrug resistance protein MRP-7 inhibits methylmercury-associated animal toxicity and dopaminergic neurodegeneration in Caenorhabditis elegans. J Neurochem 2013; 128:962-74. [PMID: 24266639 DOI: 10.1111/jnc.12515] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 10/17/2013] [Accepted: 10/28/2013] [Indexed: 10/26/2022]
Abstract
Parkinson's disease (PD) is the most prevalent neurodegenerative motor disorder worldwide, and results in the progressive loss of dopamine (DA) neurons in the substantia nigra pars compacta. Gene-environment interactions are believed to play a significant role in the vast majority of PD cases, yet the toxicants and the associated genes involved in the neuropathology are largely ill-defined. Recent epidemiological and biochemical evidence suggests that methylmercury (MeHg) may be an environmental toxicant that contributes to the development of PD. Here, we report that a gene coding for the putative multidrug resistance protein MRP-7 in Caenorhabditis elegans modulates whole animal and DA neuron sensitivity to MeHg. In this study, we demonstrate that genetic knockdown of MRP-7 results in a twofold increase in Hg levels and a dramatic increase in stress response proteins associated with the endoplasmic reticulum, golgi apparatus, and mitochondria, as well as an increase in MeHg-associated animal death. Chronic exposure to low concentrations of MeHg induces MRP-7 gene expression, while exposures in MRP-7 genetic knockdown animals results in a loss of DA neuron integrity without affecting whole animal viability. Furthermore, transgenic animals expressing a fluorescent reporter behind the endogenous MRP-7 promoter indicate that the transporter is expressed in DA neurons. These studies show for the first time that a multidrug resistance protein is expressed in DA neurons, and its expression inhibits MeHg-associated DA neuron pathology.
Collapse
Affiliation(s)
- Natalia VanDuyn
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | |
Collapse
|
30
|
McElwee MK, Ho LA, Chou JW, Smith MV, Freedman JH. Comparative toxicogenomic responses of mercuric and methyl-mercury. BMC Genomics 2013; 14:698. [PMID: 24118919 PMCID: PMC3870996 DOI: 10.1186/1471-2164-14-698] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 09/13/2013] [Indexed: 01/16/2023] Open
Abstract
Background Mercury is a ubiquitous environmental toxicant that exists in multiple chemical forms. A paucity of information exists regarding the differences or similarities by which different mercurials act at the molecular level. Results Transcriptomes of mixed-stage C. elegans following equitoxic sub-, low- and high-toxicity exposures to inorganic mercuric chloride (HgCl2) and organic methylmercury chloride (MeHgCl) were analyzed. In C. elegans, the mercurials had highly different effects on transcription, with MeHgCl affecting the expression of significantly more genes than HgCl2. Bioinformatics analysis indicated that inorganic and organic mercurials affected different biological processes. RNAi identified 18 genes that were important in C. elegans response to mercurial exposure, although only two of these genes responded to both mercurials. To determine if the responses observed in C. elegans were evolutionarily conserved, the two mercurials were investigated in human neuroblastoma (SK-N-SH), hepatocellular carcinoma (HepG2) and embryonic kidney (HEK293) cells. The human homologs of the affected C. elegans genes were then used to test the effects on gene expression and cell viability after using siRNA during HgCl2 and MeHgCl exposure. As was observed with C. elegans, exposure to the HgCl2 and MeHgCl had different effects on gene expression, and different genes were important in the cellular response to the two mercurials. Conclusions These results suggest that, contrary to previous reports, inorganic and organic mercurials have different mechanisms of toxicity. The two mercurials induced disparate effects on gene expression, and different genes were important in protecting the organism from mercurial toxicity.
Collapse
Affiliation(s)
- Matthew K McElwee
- Laboratory of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, NIH, 111 T,W Alexander Drive, Research Triangle Park, P,O, Box 12233, 27709 Durham, NC, USA.
| | | | | | | | | |
Collapse
|
31
|
Khamis I, Heikkila JJ. Enhanced HSP30 and HSP70 accumulation in Xenopus cells subjected to concurrent sodium arsenite and cadmium chloride stress. Comp Biochem Physiol C Toxicol Pharmacol 2013; 158:165-72. [PMID: 23919948 DOI: 10.1016/j.cbpc.2013.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 07/26/2013] [Accepted: 07/26/2013] [Indexed: 01/31/2023]
Abstract
Heat shock proteins (HSPs) are molecular chaperones that aid in protein folding, translocation and in preventing stress-induced protein aggregation. The present study examined the effect of simultaneous sodium arsenite and cadmium chloride treatment on the pattern of HSP30 and HSP70 accumulation in A6 kidney epithelial cells of the frog, Xenopus laevis. Immunoblot analysis revealed that HSP30 and HSP70 accumulation in concurrent stressor treatments were significantly higher than the sum of HSP30 or HSP70 accumulation in individual treatments. This finding suggested a synergistic action between sodium arsenite and cadmium chloride. KNK437 inhibitor studies indicated that the combined stressor-induced accumulation of HSPs may be regulated, at least in part, at the level of transcription. Immunocytochemistry revealed that simultaneous treatment of cells with the two stressors induced HSP30 accumulation primarily in the cytoplasm in a punctate pattern with some dysregulation of F-actin structure. Increased ubiquitinated protein accumulation was observed with combined sodium arsenite and cadmium chloride treatment compared to individual stressors suggesting an impairment of the ubiquitin proteasome degradation system. The addition of a mild heat shock further enhanced the accumulation of HSP30 and HSP70 in response to relatively low concentrations of sodium arsenite plus cadmium chloride.
Collapse
Affiliation(s)
- Imran Khamis
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | | |
Collapse
|
32
|
Simon DF, Domingos RF, Hauser C, Hutchins CM, Zerges W, Wilkinson KJ. Transcriptome sequencing (RNA-seq) analysis of the effects of metal nanoparticle exposure on the transcriptome of Chlamydomonas reinhardtii. Appl Environ Microbiol 2013; 79:4774-85. [PMID: 23728819 PMCID: PMC3754720 DOI: 10.1128/aem.00998-13] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 05/29/2013] [Indexed: 01/04/2023] Open
Abstract
The widespread use of nanoparticles (NPs) raises concern over their potential toxicological effects in humans and ecosystems. Here we used transcriptome sequencing (RNA-seq) to evaluate the effects of exposure to four different metal-based NPs, nano-Ag (nAg), nano-TiO2 (nTiO2), nano-ZnO (nZnO), and CdTe/CdS quantum dots (QDs), in the eukaryotic green alga Chlamydomonas reinhardtii. The transcriptome was characterized before and after exposure to each NP type. Specific toxicological effects were inferred from the functions of genes whose transcripts either increased or decreased. Data analysis resulted in important differences and also similarities among the NPs. Elevated levels of transcripts of several marker genes for stress were observed, suggesting that only nZnO caused nonspecific global stress to the cells under environmentally relevant conditions. Genes with photosynthesis-related functions were decreased drastically during exposure to nTiO2 and slightly during exposures to the other NP types. This pattern suggests either toxicological effects in the chloroplast or effects that mimic a transition from low to high light. nAg exposure dramatically elevated the levels of transcripts encoding known or predicted components of the cell wall and the flagella, suggesting that it damages structures exposed to the external milieu. Exposures to nTiO2, nZnO, and QDs elevated the levels of transcripts encoding subunits of the proteasome, suggesting proteasome inhibition, a phenomenon believed to underlie the development and progression of several major diseases, including Alzheimer's disease, and used in chemotherapy against multiple myeloma.
Collapse
Affiliation(s)
- Dana F. Simon
- Département de Chimie, Université de Montréal, Succursale Centre-Ville, Montréal, Quebec, Canada
| | - Rute F. Domingos
- Centro de Química Estrutural, Instituto Superior Técnico/Universidade Técnica de Lisboa, Lisbon, Portugal
| | - Charles Hauser
- Bioinformatics Program, St. Edward's University, Austin, Texas, USA
| | - Colin M. Hutchins
- Département de Chimie, Université de Montréal, Succursale Centre-Ville, Montréal, Quebec, Canada
| | - William Zerges
- Biology Department and Centre for Structural and Functional Genomics, Concordia University, Montreal, Quebec, Canada
| | - Kevin J. Wilkinson
- Département de Chimie, Université de Montréal, Succursale Centre-Ville, Montréal, Quebec, Canada
| |
Collapse
|
33
|
Yadetie F, Karlsen OA, Lanzén A, Berg K, Olsvik P, Hogstrand C, Goksøyr A. Global transcriptome analysis of Atlantic cod (Gadus morhua) liver after in vivo methylmercury exposure suggests effects on energy metabolism pathways. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2013; 126:314-325. [PMID: 23103053 DOI: 10.1016/j.aquatox.2012.09.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 09/17/2012] [Accepted: 09/23/2012] [Indexed: 06/01/2023]
Abstract
Methylmercury (MeHg) is a widely distributed contaminant polluting many aquatic environments, with health risks to humans exposed mainly through consumption of seafood. The mechanisms of toxicity of MeHg are not completely understood. In order to map the range of molecular targets and gain better insights into the mechanisms of toxicity, we prepared Atlantic cod (Gadus morhua) 135k oligonucleotide arrays and performed global analysis of transcriptional changes in the liver of fish treated with MeHg (0.5 and 2 mg/kg of body weight) for 14 days. Inferring from the observed transcriptional changes, the main pathways significantly affected by the treatment were energy metabolism, oxidative stress response, immune response and cytoskeleton remodeling. Consistent with known effects of MeHg, many transcripts for genes in oxidative stress pathways such as glutathione metabolism and Nrf2 regulation of oxidative stress response were differentially regulated. Among the differentially regulated genes, there were disproportionate numbers of genes coding for enzymes involved in metabolism of amino acids, fatty acids and glucose. In particular, many genes coding for enzymes of fatty acid beta-oxidation were up-regulated. The coordinated effects observed on many transcripts coding for enzymes of energy pathways may suggest disruption of nutrient metabolism by MeHg. Many transcripts for genes coding for enzymes in the synthetic pathways of sulphur containing amino acids were also up-regulated, suggesting adaptive responses to MeHg toxicity. By this toxicogenomics approach, we were also able to identify many potential biomarker candidate genes for monitoring environmental MeHg pollution. These results based on changes on transcript levels, however, need to be confirmed by other methods such as proteomics.
Collapse
Affiliation(s)
- Fekadu Yadetie
- Department of Molecular Biology, University of Bergen, Norway.
| | | | | | | | | | | | | |
Collapse
|
34
|
Low level prenatal exposure to methylmercury disrupts neuronal migration in the developing rat cerebral cortex. Toxicology 2012; 304:57-68. [PMID: 23220560 DOI: 10.1016/j.tox.2012.11.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 11/11/2012] [Accepted: 11/15/2012] [Indexed: 11/22/2022]
Abstract
We determined the effects of low-level prenatal MeHg exposure on neuronal migration in the developing rat cerebral cortex using in utero electroporation. We used offspring rats born to dams that had been exposed to saline or various doses of MeHg (0.01 mg/kg/day, 0.1 mg/kg/day, and 1 mg/kg/day) from gestational day (GD) 11-21. Immunohistochemical examination of the brains of the offspring was conducted on postnatal day (PND) 0, PND3, and PND7. Our results showed that prenatal exposure to low levels of MeHg (0.1 mg/kg/day or 1 mg/kg/day) during the critical stage in neuronal migration resulted in migration defects of the cerebrocortical neurons in offspring rats. Importantly, our data revealed that the abnormal neuronal distribution induced by MeHg was not caused by altered proliferation of neural progenitor cells (NPCs), induction of apoptosis of NPCs and/or newborn neurons, abnormal differentiation of NPCs, and the morphological changes of radial glial scaffold, indicating that the defective neuronal positioning triggered by exposure to low-dose of MeHg is due to the impacts of MeHg on the process of neuronal migration itself. Moreover, we demonstrated that in utero exposure to low-level MeHg suppresses the expression of Rac1, Cdc42, and RhoA, which play key roles in the migration of cerebrocortical neurons during the early stage of brain development, suggesting that the MeHg-induced migratory disturbance of cerebrocortical neurons is likely associated with the Rho GTPases signal pathway. In conclusion, our results provide a novel perspective on clarifying the mechanisms underlying the impairment of neuronal migration induced by MeHg.
Collapse
|
35
|
Hossain MB, Vahter M, Concha G, Broberg K. Low-level environmental cadmium exposure is associated with DNA hypomethylation in Argentinean women. ENVIRONMENTAL HEALTH PERSPECTIVES 2012; 120:879-84. [PMID: 22382075 PMCID: PMC3385444 DOI: 10.1289/ehp.1104600] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 03/01/2012] [Indexed: 05/11/2023]
Abstract
BACKGROUND Cadmium, a common food pollutant, alters DNA methylation in vitro. Epigenetic effects might therefore partly explain cadmium's toxicity, including its carcinogenicity; however, human data on epigenetic effects are lacking. OBJECTIVE We evaluated the effects of dietary cadmium exposure on DNA methylation, considering other environmental exposures, genetic predisposition, and gene expression. METHODS Concentrations of cadmium, arsenic, selenium, and zinc in blood and urine of nonsmoking women (n = 202) from the northern Argentinean Andes were measured by inductively coupled mass spectrometry. Methylation in CpG islands of LINE-1 (long interspersed nuclear element-1; a proxy for global DNA methylation) and promoter regions of p16 [cyclin-dependent kinase inhibitor 2A (CDKN2A)] and MLH1 (mutL homolog 1) in peripheral blood were measured by bisulfite polymerase chain reaction pyrosequencing. Genotyping (n = 172) for the DNA (cytosine-5-)-methyltransferase 1 gene (DNMT1 rs10854076 and rs2228611) and DNA (cytosine-5-)-methyltransferase 3 beta gene (DNMT3B rs2424913 and rs2424932) was performed with Sequenom iPLEX GOLD SNP genotyping; and gene expression (n = 90), with DirectHyb HumanHT-12 (version 3.0). RESULTS Cadmium exposure was low: median concentrations in blood and urine were 0.36 and 0.23 µg/L, respectively. Urinary cadmium (natural log transformed) was inversely associated with LINE-1 methylation (β = -0.50, p = 0.0070; β = -0.44, p = 0.026, adjusted for age and coca chewing) but not with p16 or MLH1 methylation. Both DNMT1 rs10854076 and DNMT1 rs2228611 polymorphisms modified associations between urinary cadmium and LINE-1 (p-values for interaction in adjusted models were 0.045 and 0.064, respectively). The rare genotypes demonstrated stronger hypomethylation with increasing urinary cadmium concentrations. Cadmium was inversely associated with DNMT3B (r(S) = -0.28, p = 0.0086) but not with DNMT1 expression (r(S) = -0.075, p = 0.48). CONCLUSION Environmental cadmium exposure was associated with DNA hypomethylation in peripheral blood, and DNMT1 genotypes modified this association. The role of epigenetic modifications in cadmium-associated diseases needs clarification.
Collapse
Affiliation(s)
- Mohammad Bakhtiar Hossain
- Department of Laboratory Medicine, Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden
| | | | | | | |
Collapse
|
36
|
Brunt JJ, Khan S, Heikkila JJ. Sodium arsenite and cadmium chloride induction of proteasomal inhibition and HSP accumulation in Xenopus laevis A6 kidney epithelial cells. Comp Biochem Physiol C Toxicol Pharmacol 2012; 155:307-17. [PMID: 21983225 DOI: 10.1016/j.cbpc.2011.09.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Revised: 09/21/2011] [Accepted: 09/25/2011] [Indexed: 01/03/2023]
Abstract
Sodium arsenite (NA) and cadmium chloride (CdCl(2)) are relatively abundant environmental toxicants that have multiple toxic effects including carcinogenesis, dysfunction of gene regulation and DNA and protein damage. In the present study, treatment of Xenopus laevis A6 kidney epithelial cells with concentrations of NA (20-30 μM) or CdCl(2) (100-200 μM) that induced HSP30 and HSP70 accumulation also produced an increase in the relative levels of ubiquitinated protein. Actin protein levels were unchanged in these experiments. In time course experiments, the levels of ubiquitinated protein and HSPs increased over a 24h exposure to NA or CdCl(2). Furthermore, treatment of cells with NA or CdCl(2) reduced the relative levels of proteasome chymotrypsin (CT)-like activity compared to control. Interestingly, pretreatment of cells with the HSP accumulation inhibitor, KNK437, prior to NA or CdCl(2) exposure decreased the relative levels of ubiquitinated protein as well as HSP30 and HSP70. A similar finding was made with ubiquitinated protein induced by proteasomal inhibitors, MG132 and celastrol, known to induce HSP accumulation in A6 cells. However, the NA- or CdCl(2)-induced decrease in proteasome CT-like activity was not altered by KNK437 pretreatment. This study has shown for the first time in poikilothermic vertebrates that NA and CdCl(2) can inhibit proteasomal activity and that there is a possible association between HSP accumulation and the mechanism of protein ubiquitination.
Collapse
Affiliation(s)
- Jara J Brunt
- Department of Biology, University of Waterloo, Waterloo, ON, Canada N2L 3G1
| | | | | |
Collapse
|
37
|
Robinson JF, Verhoef A, Piersma AH. Transcriptomic analysis of neurulation and early organogenesis in rat embryos: an in vivo and ex vivo comparison. Toxicol Sci 2012; 126:255-66. [PMID: 22262562 DOI: 10.1093/toxsci/kfr343] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cultured embryos mimic the morphological developmental progression of embryos (in vivo) undergoing neurulation and early organogenesis. Using available genomics technologies, comparative molecular-based assessments between cultured embryos and in vivo models may further clarify commonalities and dissimilarities, which contribute to differences between systems. Therefore, in this study, using a transcriptomic approach, we compared cultured whole rat embryos and embryos in vivo at comparable time points in development (gestational day (GD) 10 + 2-48 h, GD 0 = copulatory plug) to assess for commonalities and differences in gene expression in relation to morphology. We reveal strong parallels in time-dependent expression of genes in terms of magnitude, directionality, and functionality between whole embryo culture (WEC) and in vivo (rat). Genes changing in expression over time resemble previously hypothesized mechanisms underlying early development in mammalian systems. Furthermore, at the gene and functional level, we identify genes, which differ in expression between models, including genes related to development, oxygen transport, and metabolism. In summary, our results support the use of WEC for toxicological studies aimed at representing in vivo development during this time window at the molecular level. Additionally, we indicate genes, which differ in expression between models, providing possible insights for improvement of culture conditions.
Collapse
Affiliation(s)
- Joshua F Robinson
- Laboratory for Health Protection Research, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, the Netherlands
| | | | | |
Collapse
|
38
|
Farina M, Aschner M, Rocha JBT. Oxidative stress in MeHg-induced neurotoxicity. Toxicol Appl Pharmacol 2011; 256:405-17. [PMID: 21601588 PMCID: PMC3166649 DOI: 10.1016/j.taap.2011.05.001] [Citation(s) in RCA: 253] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 05/01/2011] [Accepted: 05/02/2011] [Indexed: 12/20/2022]
Abstract
Methylmercury (MeHg) is an environmental toxicant that leads to long-lasting neurological and developmental deficits in animals and humans. Although the molecular mechanisms mediating MeHg-induced neurotoxicity are not completely understood, several lines of evidence indicate that oxidative stress represents a critical event related to the neurotoxic effects elicited by this toxicant. The objective of this review is to summarize and discuss data from experimental and epidemiological studies that have been important in clarifying the molecular events which mediate MeHg-induced oxidative damage and, consequently, toxicity. Although unanswered questions remain, the electrophilic properties of MeHg and its ability to oxidize thiols have been reported to play decisive roles to the oxidative consequences observed after MeHg exposure. However, a close examination of the relationship between low levels of MeHg necessary to induce oxidative stress and the high amounts of sulfhydryl-containing antioxidants in mammalian cells (e.g., glutathione) have led to the hypothesis that nucleophilic groups with extremely high affinities for MeHg (e.g., selenols) might represent primary targets in MeHg-induced oxidative stress. Indeed, the inhibition of antioxidant selenoproteins during MeHg poisoning in experimental animals has corroborated this hypothesis. The levels of different reactive species (superoxide anion, hydrogen peroxide and nitric oxide) have been reported to be increased in MeHg-exposed systems, and the mechanisms concerning these increments seem to involve a complex sequence of cascading molecular events, such as mitochondrial dysfunction, excitotoxicity, intracellular calcium dyshomeostasis and decreased antioxidant capacity. This review also discusses potential therapeutic strategies to counteract MeHg-induced toxicity and oxidative stress, emphasizing the use of organic selenocompounds, which generally present higher affinity for MeHg when compared to the classically studied agents.
Collapse
Affiliation(s)
- Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Michael Aschner
- Department of Pediatrics and Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - João B. T. Rocha
- Departamento de Química, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| |
Collapse
|
39
|
Robinson JF, Theunissen PT, van Dartel DA, Pennings JL, Faustman EM, Piersma AH. Comparison of MeHg-induced toxicogenomic responses across in vivo and in vitro models used in developmental toxicology. Reprod Toxicol 2011; 32:180-8. [DOI: 10.1016/j.reprotox.2011.05.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 05/12/2011] [Accepted: 05/13/2011] [Indexed: 12/14/2022]
|
40
|
Yu X, Sidhu JS, Hong S, Robinson JF, Ponce RA, Faustman EM. Cadmium induced p53-dependent activation of stress signaling, accumulation of ubiquitinated proteins, and apoptosis in mouse embryonic fibroblast cells. Toxicol Sci 2011; 120:403-12. [PMID: 21252392 DOI: 10.1093/toxsci/kfr010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The tumor suppressor oncoprotein, p53, is a critical regulator of stress-induced growth arrest and apoptosis. p53 activity is regulated through the ubiquitin proteasome system (UPS) with stress-induced disruption leading to increased accumulation of p53, resulting in growth arrest. In the present study, we investigate the role of p53 to determine sensitivity to cadmium (Cd) and whether induction of stress signaling responses and perturbation of the UPS are involved in Cd-induced cytotoxicity and apoptosis. We treated synchronously cultured p53 transgenic mouse embryonic fibroblasts, both wild-type p53+/+ and knockout p53-/- cells, with cadmium chloride (Cd, 0.5-20μM) for 24 h. Cd-induced cytotoxicity was assessed by cellular morphology disruption and neutral red dye uptake assay. Proteins in the stress signaling pathway, including p38 mitogen-activated protein kinase (MAPK) and stress-activated protein kinase/c-Jun NH2-terminal kinase (SAPK/JNK); ubiquitination, such as high-molecular weight of polyubiquitinated proteins (HMW-polyUb); and apoptotic pathways, were all measured. We found that Cd induced p53-dependent cytotoxicity in the p53+/+ cells, which exhibited a twofold greater sensitivity. We observed a dose-dependent stimulation of p38 MAPK and SAPK/JNK phosphorylation that corresponded to accumulation of HMW-polyUb conjugates and lead to the induction of apoptosis, as evidenced by the elevation of cleaved caspase-3. Our study suggests that Cd-mediated cytotoxicity and induction of stress signaling responses, elevated accumulation of HMW-polyUb conjugates, and resulting apoptosis are all dependent on p53 status.
Collapse
Affiliation(s)
- Xiaozhong Yu
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98105, USA
| | | | | | | | | | | |
Collapse
|
41
|
Jones DP, Go YM. Mapping the cysteine proteome: analysis of redox-sensing thiols. Curr Opin Chem Biol 2011; 15:103-12. [PMID: 21216657 DOI: 10.1016/j.cbpa.2010.12.014] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 12/10/2010] [Accepted: 12/13/2010] [Indexed: 01/01/2023]
Abstract
The cysteine (Cys) proteome includes 214,000 Cys with thiol and other forms. A relatively small subset functions in cell signaling, while a larger number coordinate cell functions in response to redox state. The former are redox-signaling thiols while the latter are defined as redox-sensing thiols. Bulk measurements are not very informative for systems biology because reactivity of thiols in proteins differs by seven orders of magnitude. Proteomic databases contain annotation of Cys, for example, disulfides and zinc fingers, but do not include quantitative information necessary to develop functional models. Complementary databases and Cys proteome maps are needed to describe thiol redox circuits and connect these to functional redox-dependent pathways. This article summarizes progress in quantitative redox proteomics to develop such maps.
Collapse
Affiliation(s)
- Dean P Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA.
| | | |
Collapse
|
42
|
Robinson JF, Yu X, Moreira EG, Hong S, Faustman EM. Arsenic- and cadmium-induced toxicogenomic response in mouse embryos undergoing neurulation. Toxicol Appl Pharmacol 2010; 250:117-29. [PMID: 20883709 DOI: 10.1016/j.taap.2010.09.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 09/20/2010] [Accepted: 09/22/2010] [Indexed: 01/19/2023]
Abstract
Arsenic (As) and cadmium (Cd) are well-characterized teratogens in animal models inducing embryotoxicity and neural tube defects (NTDs) when exposed during neurulation. Toxicological research is needed to resolve the specific biological processes and associated molecular pathways underlying metal-induced toxicity during this timeframe in gestational development. In this study, we investigated the dose-dependent effects of As and Cd on gene expression in C57BL/6J mouse embryos exposed in utero during neurulation (GD8) to identify significantly altered genes and corresponding biological processes associated with embryotoxicity. We quantitatively examined the toxicogenomic dose-response relationship at the gene level. Our results suggest that As and Cd induce dose-dependent gene expression alterations representing shared (cell cycle, response to UV, glutathione metabolism, RNA processing) and unique (alcohol/sugar metabolism) biological processes, which serve as robust indicators of metal-induced developmental toxicity and indicate underlying embryotoxic effects. Our observations also correlate well with previously identified impacts of As and Cd on specific genes associated with metal-induced toxicity (Cdkn1a, Mt1). In summary, we have identified in a quantitative manner As and Cd induced dose-dependent effects on gene expression in mouse embryos during a peak window of sensitivity to embryotoxicity and NTDs in the sensitive C57BL/6J strain.
Collapse
Affiliation(s)
- Joshua F Robinson
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
43
|
Vanduyn N, Settivari R, Wong G, Nass R. SKN-1/Nrf2 inhibits dopamine neuron degeneration in a Caenorhabditis elegans model of methylmercury toxicity. Toxicol Sci 2010; 118:613-24. [PMID: 20855423 DOI: 10.1093/toxsci/kfq285] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Methylmercury (MeHg) exposure from occupational, environmental, and food sources is a significant threat to public health. MeHg poisonings in adults may result in severe psychological and neurological deficits, and in utero exposures can confer embryonic defects and developmental delays. Recent epidemiological and vertebrate studies suggest that MeHg exposure may also contribute to dopamine (DA) neuron vulnerability and the propensity to develop Parkinson's disease (PD). In this study, we describe a Caenorhabditis elegans model of MeHg toxicity that shows that low, chronic exposure confers embryonic defects, developmental delays, decreases in brood size and animal viability, and DA neuron degeneration. Toxicant exposure results in the robust induction of the glutathione-S-transferases (GSTs) gst-4 and gst-38 that are largely dependent on the PD-associated phase II antioxidant transcription factor SKN-1/Nrf2. We also demonstrate that the expression of SKN-1, a protein previously localized to a small subset of chemosensory neurons and intestinal cells in the nematode, is also expressed in the DA neurons, and a reduction in SKN-1 gene expression increases MeHg-induced animal vulnerability and DA neuron degeneration. These studies recapitulate fundamental hallmarks of MeHg-induced mammalian toxicity, identify a key molecular regulator of toxicant-associated whole-animal and DA neuron vulnerability, and suggest that the nematode will be a useful in vivo tool to identify and characterize mediators of MeHg-induced developmental and DA neuron pathologies.
Collapse
Affiliation(s)
- Natalia Vanduyn
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | |
Collapse
|
44
|
Sawle AD, Wit E, Whale G, Cossins AR. An Information-Rich Alternative, Chemicals Testing Strategy Using a High Definition Toxicogenomics and Zebrafish (Danio rerio) Embryos. Toxicol Sci 2010; 118:128-39. [DOI: 10.1093/toxsci/kfq237] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
45
|
Yu X, Robinson JF, Gribble E, Hong SW, Sidhu JS, Faustman EM. Gene expression profiling analysis reveals arsenic-induced cell cycle arrest and apoptosis in p53-proficient and p53-deficient cells through differential gene pathways. Toxicol Appl Pharmacol 2008; 233:389-403. [PMID: 18929588 PMCID: PMC2677763 DOI: 10.1016/j.taap.2008.09.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Revised: 09/08/2008] [Accepted: 09/09/2008] [Indexed: 12/30/2022]
Abstract
Arsenic (As) is a well-known environmental toxicant and carcinogen as well as an effective chemotherapeutic agent. The underlying mechanism of this dual capability, however, is not fully understood. Tumor suppressor gene p53, a pivotal cell cycle checkpoint signaling protein, has been hypothesized to play a possible role in mediating As-induced toxicity and therapeutic efficiency. In this study, we found that arsenite (As(3+)) induced apoptosis and cell cycle arrest in a dose-dependent manner in both p53(+/+) and p53(-/-) mouse embryonic fibroblasts (MEFs). There was, however, a distinction between genotypes in the apoptotic response, with a more prominent induction of caspase-3 in the p53(-/-) cells than in the p53(+/+) cells. To examine this difference further, a systems-based genomic analysis was conducted comparing the critical molecular mechanisms between the p53 genotypes in response to As(3+). A significant alteration in the Nrf2-mediated oxidative stress response pathway was found in both genotypes. In p53(+/+) MEFs, As(3+) induced p53-dependent gene expression alterations in DNA damage and cell cycle regulation genes. However, in the p53(-/-) MEFs, As(3+) induced a significant up-regulation of pro-apoptotic genes (Noxa) and down-regulation of genes in immune modulation. Our findings demonstrate that As-induced cell death occurs through a p53-independent pathway in p53 deficient cells while apoptosis induction occurs through p53-dependent pathway in normal tissue. This difference in the mechanism of apoptotic responses between the genotypes provides important information regarding the apparent dichotomy of arsenic's dual mechanisms, and potentially leads to further advancement of its utility as a chemotherapeutic agent.
Collapse
Affiliation(s)
- Xiaozhong Yu
- Institute for Risk Analysis and Risk Communication, Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA.
| | | | | | | | | | | |
Collapse
|