1
|
Chlubek M, Baranowska-Bosiacka I. Selected Functions and Disorders of Mitochondrial Metabolism under Lead Exposure. Cells 2024; 13:1182. [PMID: 39056765 PMCID: PMC11275214 DOI: 10.3390/cells13141182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Mitochondria play a fundamental role in the energy metabolism of eukaryotic cells. Numerous studies indicate lead (Pb) as a widely occurring environmental factor capable of disrupting oxidative metabolism by modulating the mitochondrial processes. The multitude of known molecular targets of Pb and its strong affinity for biochemical pathways involving divalent metals suggest that it may pose a health threat at any given dose. Changes in the bioenergetics of cells exposed to Pb have been repeatedly demonstrated in research, primarily showing a reduced ability to synthesize ATP. In addition, lead interferes with mitochondrial-mediated processes essential for maintaining homeostasis, such as apoptosis, mitophagy, mitochondrial dynamics, and the inflammatory response. This article describes selected aspects of mitochondrial metabolism in relation to potential mechanisms of energy metabolism disorders induced by Pb.
Collapse
Affiliation(s)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| |
Collapse
|
2
|
Farag MR, El-Kassas S, Attia YA, Alhotan RA, Mahmoud MA, Di Cerbo A, Alagawany M. Yucca schidigera Extract Mitigates the Oxidative Damages, Inflammation, and Neurochemical Impairments in the Brains of Quails Exposed to Lead. Biol Trace Elem Res 2024; 202:713-724. [PMID: 37171738 DOI: 10.1007/s12011-023-03696-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/03/2023] [Indexed: 05/13/2023]
Abstract
The present study explored the neurotoxic impacts of lead (Pb) and the potential alleviating effect of Yucca schidigera extract (YSE) in Japanese quails. About 360 adult Japanese quails (8 weeks old) were used. Quails were randomly distributed to six groups with 4 replicates each: the control group (fed basal diet, BD), the BD + YSE1 and BD + YSE2 groups (BD + 100 and 200 mg/kg diet of YSE, respectively), the Pb group (BD + 100 mg/kg Pb), and the Pb + YSE1 and Pb + YSE2 groups (BD + Pb + 100 and 200 mg/kg YSE, respectively). This feeding trial lasted for 8 weeks. The exposure to Pb in the diet induced oxidative damage stress in the brain of exposed quails reflected by the significant increase in the oxidative markers including malonaldehyde (MDA) and protein carbonyl (PC) and the significant reduction in the activities of antioxidants including catalase (CAT), superoxide dismutase (SOD), and the reduced glutathione (GSH). Brain neurochemistry and enzyme activities were also altered following Pb exposure. Pb significantly reduced serotonin, dopamine, norepinephrine, GABA, Ach, and Na + /K + -ATPase activities. Pb dietary intoxication markedly increased brain inflammatory biomarkers, including tumor necrosis factor (TNF-α), myeloperoxidase, and nitric oxide. Peripherally, Pb toxicity decreased the amino acid neurotransmitters (glutamic acid, glycine, and aspartic acid) in the serum of birds. At the transcriptomic level, Pb exposure upregulated the transcription patterns of CASP3, TNF-α, HSP70, and IL-1β. The single effect of YSE maintained that all the assessed parameters were not changed compared to the control. Interestingly, the YSE co-supplementation with Pb alleviated the Pb-induced neuro-oxidative damages by lowering the lipid, protein, and DNA damage, and the inflammatory biomarkers.
Collapse
Affiliation(s)
- Mayada R Farag
- Forensic Medicine and Toxicology Department, Veterinary Medicine Faculty, Zagazig University, Zagazig, 44111, Egypt
| | - Seham El-Kassas
- Animal, Poultry, and Fish Breeding and Production, Department of Animal Wealth Development, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El Sheikh, 33516, Egypt
| | - Youssef A Attia
- Department of Agriculture, Faculty of Environmental Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Rashed A Alhotan
- Department of Animal Production, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed A Mahmoud
- Department of Physiology, Faculty of Veterinary Medicine, New Valley University, Kharga, New Valley, Egypt
| | - Alessandro Di Cerbo
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024, Matelica, Italy
| | - Mahmoud Alagawany
- Poultry Department, Agriculture Faculty, Zagazig University, Zagazig, 44511, Egypt.
| |
Collapse
|
3
|
Bjørklund G, Tippairote T, Hangan T, Chirumbolo S, Peana M. Early-Life Lead Exposure: Risks and Neurotoxic Consequences. Curr Med Chem 2024; 31:1620-1633. [PMID: 37031386 DOI: 10.2174/0929867330666230409135310] [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: 08/14/2022] [Revised: 02/10/2023] [Accepted: 02/16/2023] [Indexed: 04/10/2023]
Abstract
BACKGROUND Lead (Pb) does not have any biological function in a human, and it is likely no safe level of Pb in the human body. The Pb exposure impacts are a global concern for their potential neurotoxic consequences. Despite decreasing both the environmental Pb levels and the average blood Pb levels in the survey populations, the lifetime redistribution from the tissues-stored Pb still poses neurotoxic risks from the low-level exposure in later life. The growing fetus and children hold their innate high-susceptible to these Pb-induced neurodevelopmental and neurobehavioral effects. OBJECTIVE This article aims to evaluate cumulative studies and insights on the topic of Pb neurotoxicology while assessing the emerging trends in the field. RESULTS The Pb-induced neurochemical and neuro-immunological mechanisms are likely responsible for the high-level Pb exposure with the neurodevelopmental and neurobehavioral impacts at the initial stages. Early-life Pb exposure can still produce neurodegenerative consequences in later life due to the altered epigenetic imprints and the ongoing endogenous Pb exposure. Several mechanisms contribute to the Pb-induced neurotoxic impacts, including the direct neurochemical effects, the induction of oxidative stress and inflammation through immunologic activations, and epigenetic alterations. Furthermore, the individual nutritional status, such as macro-, micro-, or antioxidant nutrients, can significantly influence the neurotoxic impacts even at low-level exposure to Pb. CONCLUSION The prevention of early-life Pb exposure is, therefore, the critical determinant for alleviating various Pb-induced neurotoxic impacts across the different age groups.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Toften 24, Mo i Rana, 8610, Norway
| | - Torsak Tippairote
- Department of Nutritional and Environmental Medicine, HP Medical Center, Bangkok 10540, Thailand
| | - Tony Hangan
- Faculty of Medicine, Ovidius University of Constanta, Constanta, 900470, Romania
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, 37134, Italy
- CONEM Scientific Secretary, Strada Le Grazie 9, 37134, Verona, Italy
| | - Massimiliano Peana
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, Via Vienna 2, Sassari, 07100, Italy
| |
Collapse
|
4
|
Caetano ELA, Frattes CDC, Segato TCM, Leite FG, Pickler TB, de Oliveira Junior JM, Jozala AF, Grotto D. Protective effect of Agaricus bisporus mushroom against maternal and fetal damage induced by lead administration during pregnancy in rats. Birth Defects Res 2023; 115:1424-1437. [PMID: 37421350 DOI: 10.1002/bdr2.2218] [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: 04/04/2023] [Revised: 05/26/2023] [Accepted: 06/16/2023] [Indexed: 07/10/2023]
Abstract
INTRODUCTION Lead (Pb) is a toxic pollutant, which can affect different tissues of the human body. The use of natural elements, as medicinal mushroom can reduce the toxic effects of Pb. OBJECTIVE We evaluated, through preclinical tests, the oral co exposures to mushroom Agaricus bisporus (Ab) by gavage and Pb in drinking water, and the capability of Ab be a protective agent for both pregnant rats and their fetuses. METHODS Female Wistar rats were divided into four groups (n = 5/group): Group I-Control; Group II-Ab 100 mg/kg; Group III-Pb 100 mg/L; Group IV-Ab +Pb -100 mg/kg +100 mg/L. Exposure was performed until the 19th day of gestation. On the 20th day, pregnant rats were euthanized, and the outcomes evaluated were weight gain; hematological profile; biochemical markers; oxidative stress markers; reproductive capacity; and embryo fetal development. RESULTS The characterization of mushrooms reveals them to be a valuable source of nutrients. However, Pb ingestion resulted in reduced weight gain and negative impacts on hematological and biochemical parameters. Fortunately, co administration of mushrooms helped to mitigate these negative effects and promote recovery. The mushroom also showed antioxidant activity, improving parameters of oxidative stress. In addition, Ab partially recovered the damage in fetal morphology and bone parameters. CONCLUSION Our findings indicated that the co administration of Ab improved the toxicity caused by Pb, and the mushroom could be used as a natural alternative as a protective/chelator agent.
Collapse
Affiliation(s)
| | | | | | - Fernanda Gomes Leite
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Programa de Pós-Graduação em Toxicologia, Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
5
|
Wang H, Wang H, Guan J, Guan W, Liu Z. Lead induces mouse skin fibroblast apoptosis by disrupting intracellular homeostasis. Sci Rep 2023; 13:9670. [PMID: 37316700 DOI: 10.1038/s41598-023-36835-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 06/10/2023] [Indexed: 06/16/2023] Open
Abstract
Lead (Pb) is a critical industrial and environmental contaminant that can cause pathophysiological changes in several cellular and organ systems and their processes, including cell proliferation, differentiation, apoptosis, and survival. The skin is readily exposed to and damaged by Pb, but the mechanisms through which Pb damages cells are not fully understood. We examined the apoptotic properties of Pb in mouse skin fibroblast (MSF) in vitro. Treatment of fibroblasts with 40, 80, and 160 μM Pb for 24 h revealed morphological alterations, DNA damage, enhanced caspase-3, -8, and -9 activities, and apoptotic cell population. Furthermore, apoptosis was dosage (0-160 μM) and time (12-48 h) dependent. Concentrations of intracellular calcium (Ca2+) and reactive oxygen species were increased, and the mitochondrial membrane potential was decreased in exposed cells. Cell cycle arrest was evident at the G0/G1 phase. The Bax, Fas, caspase-3 and -8, and p53 transcript levels were increased, whereas Bcl-2 gene expression was decreased. Based on our analysis, Pb triggers MSF apoptosis bydisrupting intracellular homeostasis. Our findings enrich the knowledge about the mechanistic function of Pb-induced cytotoxicity on human skin fibroblasts and could potentially guide future Pb health risk assessments.
Collapse
Affiliation(s)
- Hui Wang
- Jinzhou Medical University, Jinzhou, 121001, China
- Meat Processing and Safety Control Engineering Technology Research Center of Liaoning Province, Jinzhou, 121001, China
| | - Huinuan Wang
- Jinzhou Medical University, Jinzhou, 121001, China
| | - Jiawen Guan
- Jinzhou Medical University, Jinzhou, 121001, China
- Meat Processing and Safety Control Engineering Technology Research Center of Liaoning Province, Jinzhou, 121001, China
| | - Weijun Guan
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - Zheng Liu
- Jinzhou Medical University, Jinzhou, 121001, China.
- Meat Processing and Safety Control Engineering Technology Research Center of Liaoning Province, Jinzhou, 121001, China.
| |
Collapse
|
6
|
Gąssowska-Dobrowolska M, Chlubek M, Kolasa A, Tomasiak P, Korbecki J, Skowrońska K, Tarnowski M, Masztalewicz M, Baranowska-Bosiacka I. Microglia and Astroglia-The Potential Role in Neuroinflammation Induced by Pre- and Neonatal Exposure to Lead (Pb). Int J Mol Sci 2023; 24:9903. [PMID: 37373050 DOI: 10.3390/ijms24129903] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/01/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Neuroinflammation is one of the postulated mechanisms for Pb neurotoxicity. However, the exact molecular mechanisms responsible for its pro-inflammatory effect are not fully elucidated. In this study, we examined the role of glial cells in neuroinflammation induced by Pb exposure. We investigated how microglia, a type of glial cell, responded to the changes caused by perinatal exposure to Pb by measuring the expression of Iba1 at the mRNA and protein levels. To assess the state of microglia, we analyzed the mRNA levels of specific markers associated with the cytotoxic M1 phenotype (Il1b, Il6, and Tnfa) and the cytoprotective M2 phenotype (Arg1, Chi3l1, Mrc1, Fcgr1a, Sphk1, and Tgfb1). Additionally, we measured the concentration of pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α). To assess the reactivity and functionality status of astrocytes, we analyzed the GFAP (mRNA expression and protein concentration) as well as glutamine synthase (GS) protein level and activity. Using an electron microscope, we assessed ultrastructural abnormalities in the examined brain structures (forebrain cortex, cerebellum, and hippocampus). In addition, we measured the mRNA levels of Cxcl1 and Cxcl2, and their receptor, Cxcr2. Our data showed that perinatal exposure to Pb at low doses affected both microglia and astrocyte cells' status (their mobilization, activation, function, and changes in gene expression profile) in a brain-structure-specific manner. The results suggest that both microglia and astrocytes represent a potential target for Pb neurotoxicity, thus being key mediators of neuroinflammation and further neuropathology evoked by Pb poisoning during perinatal brain development.
Collapse
Affiliation(s)
- Magdalena Gąssowska-Dobrowolska
- Department of Cellular Signalling, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland
| | - Mikołaj Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Agnieszka Kolasa
- Department of Histology and Embryology, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Patrycja Tomasiak
- Department of Physiology in Health Sciences, Pomeranian Medical University in Szczecin, Żołnierska 54, 70-210 Szczecin, Poland
| | - Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Góra, Zyty 28 St., 65-046 Zielona Góra, Poland
| | - Katarzyna Skowrońska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Maciej Tarnowski
- Department of Physiology in Health Sciences, Pomeranian Medical University in Szczecin, Żołnierska 54, 70-210 Szczecin, Poland
| | - Marta Masztalewicz
- Department of Neurology, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252 Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| |
Collapse
|
7
|
Bandaru LJM, Murumulla L, C BL, D KP, Challa S. Exposure of combination of environmental pollutant, lead (Pb) and β-amyloid peptides causes mitochondrial dysfunction and oxidative stress in human neuronal cells. J Bioenerg Biomembr 2023; 55:79-89. [PMID: 36637735 DOI: 10.1007/s10863-023-09956-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 01/06/2023] [Indexed: 01/14/2023]
Abstract
Exposure to the environmental pollutant lead (Pb) has been linked to Alzheimer's disease (AD), in which mitochondrial dysfunction is a pathological consequence of neuronal degeneration. The toxicity of Pb in combination with β-amyloid peptides (1-40) and (25-35) causes selective death in neuronal cells. However, the precise mechanism through which Pb induces Alzheimer's disease, particularly mitochondrial damage, is unknown. Changes in mitochondrial mass, membrane potential, mitochondrial complex activities, mitochondrial DNA and oxidative stress were examined in neuronal cells of human origin exposed to Pb and β-amyloid peptides (1-40) and (25-35) individually and in different combinations. The results showed depolarization of mitochondrial membrane potential, decrease in mitochondrial mass, ATP levels and mtDNA copy number in Pb and β-amyloid peptides (1-40) and (25-35) exposed cells. Also, significant reductions in the expression of mitochondrial electron transport chain (ETC) complex proteins (ATP5A, COXIV, UQCRC2, SDHB, NDUFS3), as well as down regulation of ETC complex gene expressions such as COXIV, ATP5F1 and NDUFS3 and antioxidant gene expressions like MnSOD and Gpx4 were observed in exposed cells. Furthermore, Pb and β-amyloid peptides exposure resulted in elevated mitochondrial malondialdehyde levels and a decrease in mitochondrial GSH levels. Our findings suggest that Pb toxicity could be one of the causative factors for the mitochondrial dysfunction and oxidative stress in Alzheimer's disease progression.
Collapse
Affiliation(s)
- Lakshmi Jaya Madhuri Bandaru
- Cell Biology Division, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India
| | - Lokesh Murumulla
- Cell Biology Division, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India
| | - Bindu Lasya C
- Department of Pharmacology, Anurag University, Hyderabad, India
| | | | - Suresh Challa
- Cell Biology Division, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India.
| |
Collapse
|
8
|
Bandaru LJM, Ayyalasomayajula N, Murumulla L, Challa S. Mechanisms associated with the dysregulation of mitochondrial function due to lead exposure and possible implications on the development of Alzheimer's disease. Biometals 2022; 35:1-25. [PMID: 35048237 DOI: 10.1007/s10534-021-00360-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/09/2021] [Indexed: 01/17/2023]
Abstract
Lead (Pb) is a multimedia contaminant with various pathophysiological consequences, including cognitive decline and neural abnormalities. Recent findings have reported an association of Pb toxicity with Alzheimer's disease (AD). Studies have revealed that mitochondrial dysfunction is a pathological characteristic of AD. According to toxicology reports, Pb promotes mitochondrial oxidative stress by lowering complex III activity in the electron transport chain, boosting reactive oxygen species formation, and reducing the cell's antioxidant defence system. Here, we review recent advances in the role of mitochondria in Pb-induced AD pathology, as well as the mechanisms associated with the mitochondrial dysfunction, such as the depolarisation of the mitochondrial membrane potential, mitochondrial permeability transition pore opening; mitochondrial biogenesis, bioenergetics and mitochondrial dynamics alterations; and mitophagy and apoptosis. We also discuss possible therapeutic options for mitochondrial-targeted neurodegenerative disease (AD).
Collapse
Affiliation(s)
- Lakshmi Jaya Madhuri Bandaru
- Department of Cell Biology, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India
| | - Neelima Ayyalasomayajula
- Department of Cell Biology, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India
| | - Lokesh Murumulla
- Department of Cell Biology, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India
| | - Suresh Challa
- Department of Cell Biology, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India.
| |
Collapse
|
9
|
Wang K, Bao G, Fan Q, Zhu L, Yang L, Liu T, Zhang Z, Li G, Chen X, Xu X, Xu X, He B, Zheng Y. Feasibility evaluation of a Cu-38 Zn alloy for intrauterine devices: In vitro and in vivo studies. Acta Biomater 2022; 138:561-575. [PMID: 34774783 DOI: 10.1016/j.actbio.2021.11.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/29/2021] [Accepted: 11/04/2021] [Indexed: 11/01/2022]
Abstract
The existing adverse effects of copper in copper-containing intrauterine devices (Cu-IUDs) have raised concerns regarding their use. These adverse effects include burst release of cupric ions (Cu2+) at the initial stage and an increasingly rough surface of the Cu-IUDs. In this study, we investigated the use of two copper alloys, Cu-38 Zn and H62 as the new upgrading or alternative material for IUDs. Their corrosive properties were studied in simulated uterine fluid (SUF) by using electrochemical methods, with pure Cu as a control. We studied the in vitro long-term corrosion behaviors in SUF, cytotoxicity to uterine cells (human endometrial epithelial cells and human endometrial stromal cells), in vivo biocompatibility and contraceptive efficacy of pure Cu, H62, and Cu-38 Zn. In the first month, the burst release rate of Cu2+ in the Cu-38 Zn group was significantly lower than those in the pure Cu and H62 groups. The in vitro cytocompatibility Cu-38 Zn was better than that of pure Cu and H62. Moreover, Cu-38 Zn showed improved tissue biocompatibility in vivo experiments. Therefore, the contraceptive efficacy of the Cu-38 Zn is still maintained as high as the pure Cu while the adverse effects are significantly eased, suggesting that Cu-38 Zn can be a suitable potential candidate material for IUDs. STATEMENT OF SIGNIFICANCE: The existing adverse effects associated with the intrinsic properties of copper materials for copper-containing intrauterine devices (Cu-IUD) are of concern in their employment. Such as, burst release of cupric ions (Cu2+) at the initial stage and an increasingly rough surface of the Cu-IUD. In this work, Cu alloyed with a high amount of bioactive Zn was used for a Cu-IUD. The Cu-38 Zn alloy exhibited reduced burst release of Cu2+ within the first month compared with the pure Cu and H62. Furthermore, the Cu-38 Zn alloy displayed significantly improved biocompatibility and a much smoother surface. Therefore, high antifertility efficacy of the Cu-38 Zn alloy was well maintained, while the adverse effects are significantly eased, suggesting that the Cu-38 Zn alloy is promising for a Cu-IUD.
Collapse
|
10
|
Metryka E, Kupnicka P, Kapczuk P, Aszakiewicz B, Piotrowska K, Tkacz M, Gutowska I, Chlubek D, Baranowska-Bosiacka I. Lead (Pb) Accumulation in Human THP-1 Monocytes/Macrophages In Vitro and the Influence on Cell Apoptosis. Biol Trace Elem Res 2021; 199:955-967. [PMID: 32557104 PMCID: PMC7813697 DOI: 10.1007/s12011-020-02215-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 05/20/2020] [Indexed: 12/22/2022]
Abstract
In this study, we investigated the ability of THP-1 monocytes and macrophages to accumulate lead (Pb) in vitro, relative to Pb concentration and length of exposure. Moreover, we also evaluated the effect of Pb accumulation on cell viability and apoptosis. THP-1 monocytes and macrophages were cultured in the presence of Pb at 1.25 μg/dL, 2.5 μg/dL, 5 μg/dL, and 10 μg/dL. Pb accumulation was examined by inductively coupled plasma and confocal microscopy. The influence of Pb on cell viability, apoptosis, and necrosis was assessed using flow cytometry. The results showed that Pb was toxic to THP-1 monocytes/macrophages even at very low environmental concentrations. Despite the use of low concentrations, both monocytes and macrophages showed dose-dependent and time-dependent decreases in viability, with a simultaneous increase in the percentage of early and late apoptotic cells. Macrophages reacted more strongly to Pb than monocytes. When exposed to the same Pb concentrations, they showed lower viability and a higher percentage of necrotic cells. The incubation time positively correlated with Pb accumulation in a dose-dependent manner. The obtained results indicate that environmental exposure to low Pb concentrations may significantly impair the function of macrophages, with the increased number of apoptotic cells potentially contributing to the development of many pathologies in the brain and whole body.
Collapse
Affiliation(s)
- Emilia Metryka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Patrycja Kapczuk
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Beata Aszakiewicz
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Katarzyna Piotrowska
- Department of Physiology, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Marta Tkacz
- Department of Physiology, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Izabela Gutowska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland.
| |
Collapse
|
11
|
Abstract
Lead (Pb2+) is a non-essential metal with numerous industrial applications that have led to ts ubiquity in the environment. Thus, not only occupational-exposed individuals' health is compromised, but also that of the general population and in particular children. Notably, although the central nervous system is particularly susceptible to Pb2+, other systems are affected as well. The present study focuses on molecular mechanisms that underlie the effects that arise from the presence of Pb2+ in situ in the brain, and the possible toxic effects that follows. As the brain barriers represent the first target of systemic Pb2+, mechanisms of Pb2+ entry into the brain are discussed, followed by a detailed discussion on neurotoxic mechanisms, with special emphasis on theories of ion mimicry, mitochondrial dysfunction, redox imbalance, and neuroinflammation. Most importantly, the confluence and crosstalk between these events is combined into a cogent mechanism of toxicity, by intertwining recent and old evidences from humans, in vitro cell culture and experimental animals. Finally, pharmacological interventions, including chelators, antioxidants substances, anti-inflammatory drugs, or their combination are reviewed as integrated approaches to ameliorate Pb2+ harmful effects in both developing or adult organisms.
Collapse
Affiliation(s)
- Miriam B. Virgolini
- IFEC CONICET. IFEC-CONICET. Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba. Haya de la Torre y Medina Allende, Ciudad Universitaria, 5016, Córdoba, Argentina
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA and IM Sechenov First Moscow State Medical University (Sechenov University), 119146, Moscow, Russia
| |
Collapse
|
12
|
Cognitive Impairment Induced by Lead Exposure during Lifespan: Mechanisms of Lead Neurotoxicity. TOXICS 2021; 9:toxics9020023. [PMID: 33525464 PMCID: PMC7912619 DOI: 10.3390/toxics9020023] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/19/2021] [Accepted: 01/25/2021] [Indexed: 12/25/2022]
Abstract
Lead (Pb) is considered a strong environmental toxin with human health repercussions. Due to its widespread use and the number of people potentially exposed to different sources of this heavy metal, Pb intoxication is recognized as a public health problem in many countries. Exposure to Pb can occur through ingestion, inhalation, dermal, and transplacental routes. The magnitude of its effects depends on several toxicity conditions: lead speciation, doses, time, and age of exposure, among others. It has been demonstrated that Pb exposure induces stronger effects during early life. The central nervous system is especially vulnerable to Pb toxicity; Pb exposure is linked to cognitive impairment, executive function alterations, abnormal social behavior, and fine motor control perturbations. This review aims to provide a general view of the cognitive consequences associated with Pb exposure during early life as well as during adulthood. Additionally, it describes the neurotoxic mechanisms associated with cognitive impairment induced by Pb, which include neurochemical, molecular, and morphological changes that jointly could have a synergic effect on the cognitive performance.
Collapse
|
13
|
Kupnicka P, Kojder K, Metryka E, Kapczuk P, Jeżewski D, Gutowska I, Goschorska M, Chlubek D, Baranowska-Bosiacka I. Morphine-element interactions - The influence of selected chemical elements on neural pathways associated with addiction. J Trace Elem Med Biol 2020; 60:126495. [PMID: 32179426 DOI: 10.1016/j.jtemb.2020.126495] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 02/17/2020] [Accepted: 03/05/2020] [Indexed: 02/06/2023]
Abstract
Addiction is a pressing social problem worldwide and opioid dependence can be considered the strongest and most difficult addiction to treat. Mesolimbic and mesocortical dopaminergic pathways play an important role in modulation of cognitive processes and decision making and, therefore, changes in dopamine metabolism are considered the central basis for the development of dependence. Disturbances caused by excesses or deficiency of certain elements have a significant impact on the functioning of the central nervous system (CNS) both in physiological conditions and in pathology and can affect the cerebral reward system and therefore, may modulate processes associated with the development of addiction. In this paper we review the mechanisms of interactions between morphine and zinc, manganese, chromium, cadmium, lead, fluoride, their impact on neural pathways associated with addiction, and on antinociception and morphine tolerance and dependence.
Collapse
Affiliation(s)
- Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Klaudyna Kojder
- Department of Anaesthesiology and Intensive Care, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252, Szczecin, Poland.
| | - Emilia Metryka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Patrycja Kapczuk
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Dariusz Jeżewski
- Department of Applied Neurocognitive Science, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252, Szczecin, Poland
| | - Izabela Gutowska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460, Szczecin, Poland
| | - Marta Goschorska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| |
Collapse
|
14
|
Ahmad F, Haque S, Ravinayagam V, Ahmad A, Kamli MR, Barreto GE, Ghulam Md Ashraf. Developmental lead (Pb)-induced deficits in redox and bioenergetic status of cerebellar synapses are ameliorated by ascorbate supplementation. Toxicology 2020; 440:152492. [PMID: 32407874 DOI: 10.1016/j.tox.2020.152492] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/08/2020] [Accepted: 05/07/2020] [Indexed: 10/24/2022]
Abstract
Neurotoxicity induced by exposure to heavy metal lead (Pb) is a concern of utmost importance particularly for countries with industrial-based economies. The developing brain is especially sensitive to exposure to even minute quantities of Pb which can alter neurodevelopmental trajectory with irreversible effects on motor, emotive-social and cognitive attributes even into later adulthood. Chemical synapses form the major pathway of inter-neuronal communications and are prime candidates for higher order brain (motor, memory and behavior) functions and determine the resistance/susceptibility for neurological disorders, including neuropsychopathologies. The synaptic pathways and mechanisms underlying Pb-mediated alterations in neuronal signaling and plasticity are not completely understood. Employing a biochemically isolated synaptosomal fraction which is enriched in synaptic terminals and synaptic mitochondria, this study aimed to analyze the alterations in bioenergetic and redox/antioxidant status of cerebellar synapses induced by developmental exposure to Pb (0.2 %). Moreover, we test the efficacy of vitamin C (ascorbate; 500 mg/kg body weight), a neuroprotective and neuromodulatory antioxidant, in mitigation of Pb-induced neuronal deficits. Our results implicate redox and bioenergetic disruptions as an underlying feature of the synaptic dysfunction observed in developmental Pb neurotoxicity, potentially contributing to consequent deficits in motor, behavioral and psychological attributes of the organisms. In addition, we establish ascorbate as a key ingredient for therapeutic approach against Pb induced neurotoxicity, particularly for early-life exposures.
Collapse
Affiliation(s)
- Faraz Ahmad
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand.
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, 45142, Saudi Arabia
| | - Vijaya Ravinayagam
- Deanship of Scientific Research, Department of Nano-medicine Research, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, 31441, Saudi Arabia
| | - Aqeel Ahmad
- Department of Medical Biochemistry, College of Medicine, Shaqra University, Shaqra, 11961, Saudi Arabia
| | - Majid Rasool Kamli
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, 21589, Saudi Arabia; Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute, University of Limerick, Limerick, Ireland
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
15
|
Mitochondrial Functions, Energy Metabolism and Protein Glycosylation are Interconnected Processes Mediating Resistance to Bortezomib in Multiple Myeloma Cells. Biomolecules 2020; 10:biom10050696. [PMID: 32365811 PMCID: PMC7277183 DOI: 10.3390/biom10050696] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 03/31/2020] [Accepted: 04/28/2020] [Indexed: 12/18/2022] Open
Abstract
The proteasome inhibitor bortezomib (BTZ) has emerged as an effective drug for the treatment of multiple myeloma even though many patients relapse from BTZ therapy. The present study investigated the metabolic pathways underlying the acquisition of bortezomib resistance in multiple myeloma. We used two different clones of multiple myeloma cell lines exhibiting different sensitivities to BTZ (U266 and U266-R) and compared them in terms of metabolic profile, mitochondrial fitness and redox balance homeostasis capacity. Our results showed that the BTZ-resistant clone (U266-R) presented increased glycosylated UDP-derivatives when compared to BTZ-sensitive cells (U266), thus also suggesting higher activities of the hexosamine biosynthetic pathway (HBP), regulating not only protein O- and N-glycosylation but also mitochondrial functions. Notably, U266-R displayed increased mitochondrial biogenesis and mitochondrial dynamics associated with stronger antioxidant defenses. Furthermore, U266-R maintained a significantly higher concentration of substrates for protein glycosylation when compared to U266, particularly for UDP-GlcNac, thus further suggesting the importance of glycosylation in the BTZ pharmacological response. Moreover, BTZ-treated U266-R showed significantly higher ATP/ADP ratios and levels of ECP and also exhibited increased mitochondrial fitness and antioxidant response. In conclusions, our findings suggest that the HBP may play a major role in mitochondrial fitness, driving BTZ resistance in multiple myeloma and thus representing a possible target for new drug development for BTZ-resistant patients.
Collapse
|
16
|
Owsianowska J, Kamińska MS, Bosiacki M, Chlubek D, Karakiewicz B, Jurczak A, Stanisławska M, Barczak K, Grochans E. Depression, changes in peripheral blood cell count, and changes in selected biochemical parameters related to lead concentration in whole blood (Pb-B) of women in the menopausal period. J Trace Elem Med Biol 2020; 61:126501. [PMID: 32289550 DOI: 10.1016/j.jtemb.2020.126501] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 01/24/2020] [Accepted: 03/18/2020] [Indexed: 11/24/2022]
Abstract
THE AIM The aim of this study was to assess the severity of depression, vasomotor symptoms, changes in peripheral blood cell count, and selected biochemical parameters in relation to the concentration of lead in whole blood of women in the perimenopausal period. METHODS The study sample consisted of 233 women from the general population of the West Pomeranian Province (Poland) in age between 44-65 years. The intensity of menopausal symptoms was examined using the Blatt-Kupperman Index, and the severity of depression using the Beck Depression Inventory. The following biochemical data were evaluated: concentrations of glucose, triglycerides, HDL, C-reactive protein, glycated haemoglobin, cortisol, insulin, blood cell count, and lead concentration in whole blood (Pb-B). RESULTS A whole blood Pb concentration below 5 μg/dl was found in 55 subjects (23.61 %), in 142 women (60.94 %) it ranged from 5 to 10 μg/dl, while in 36 women (15.45 %) was higher than 10 μg/dl. There was a strong positive correlation between Pb concentration in the blood of the examined women and the severity of depressive symptoms (Rs=+0.60, p = 0.001). The lowest mean values for total leukocytes (5.07 ± 1.22 thousand/μl) and neutrophils (2.76 ± 0.86 thousand/μl) were found in women with Pb concentration above 10 μg/dl (p < 0.05). There was a significant negative correlation between the number of total leukocytes (r=-0.45, p = 0.002) and neutrophils (r=-0.50, p = 0.001) and blood Pb concentration. Analysis showed statistically significant differences in glucose concentration (p < 0.05) between groups. Blood glucose was higher in women with Pb-B <5 and between 5-10 μg/dl than in women with Pb-B >10 μg/dl. CONCLUSION Exposure to Pb may be a factor playing a significant role in the development of depressive symptoms in menopausal women. It may also be associated with glucose metabolism disorders and immunosuppression in women during this period of life.
Collapse
Affiliation(s)
- Joanna Owsianowska
- Department of Specialized Nursing, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 48 Żołnierska St., 71-210, Szczecin, Poland.
| | - Magdalena Sylwia Kamińska
- Subdepartment of Long-Term Care, Department of Social Medicine, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 48 Żołnierska St., 71-210, Szczecin, Poland.
| | - Mateusz Bosiacki
- Department of Functional Diagnostics and Physical Medicine, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 56 Żołnierska St., 71-210, Szczecin, Poland.
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, 72 Powstańców Wielkopolskich St., 70-111, Szczecin, Poland.
| | - Beata Karakiewicz
- Subdepartment of Social Medicine and Public Health, Department of Social Medicine, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 48 Żołnierska St., 71-210, Szczecin, Poland.
| | - Anna Jurczak
- Department of Specialized Nursing, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 48 Żołnierska St., 71-210, Szczecin, Poland.
| | - Marzanna Stanisławska
- Department of Nursing, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 48 Żołnierska St., 71-210, Szczecin, Poland.
| | - Katarzyna Barczak
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University, 72 Powstańców Wielkopolskich St., 70-111, Szczecin, Poland.
| | - Elżbieta Grochans
- Department of Nursing, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 48 Żołnierska St., 71-210, Szczecin, Poland.
| |
Collapse
|
17
|
Pre- and Neonatal Exposure to Lead (Pb) Induces Neuroinflammation in the Forebrain Cortex, Hippocampus and Cerebellum of Rat Pups. Int J Mol Sci 2020; 21:ijms21031083. [PMID: 32041252 PMCID: PMC7037720 DOI: 10.3390/ijms21031083] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/01/2020] [Accepted: 02/03/2020] [Indexed: 12/11/2022] Open
Abstract
Lead (Pb) is a heavy metal with a proven neurotoxic effect. Exposure is particularly dangerous to the developing brain in the pre- and neonatal periods. One postulated mechanism of its neurotoxicity is induction of inflammation. This study analyzed the effect of exposure of rat pups to Pb during periods of brain development on the concentrations of selected cytokines and prostanoids in the forebrain cortex, hippocampus and cerebellum. Methods: Administration of 0.1% lead acetate (PbAc) in drinking water ad libitum, from the first day of gestation to postnatal day 21, resulted in blood Pb in rat pups reaching levels below the threshold considered safe for humans by the Centers for Disease Control and Prevention (10 µg/dL). Enzyme-linked immunosorbent assay (ELISA) method was used to determine the levels of interleukins IL-1β, IL-6, transforming growth factor-β (TGF-β), prostaglandin E2 (PGE2) and thromboxane B2 (TXB2). Western blot and quantitative real-time PCR were used to determine the expression levels of cyclooxygenases COX-1 and COX-2. Finally, Western blot was used to determine the level of nuclear factor kappa B (NF-κB). Results: In all studied brain structures (forebrain cortex, hippocampus and cerebellum), the administration of Pb caused a significant increase in all studied cytokines and prostanoids (IL-1β, IL-6, TGF-β, PGE2 and TXB2). The protein and mRNA expression of COX-1 and COX-2 increased in all studied brain structures, as did NF-κB expression. Conclusions: Chronic pre- and neonatal exposure to Pb induces neuroinflammation in the forebrain cortex, hippocampus and cerebellum of rat pups.
Collapse
|
18
|
Troisi J, Giugliano L, Sarno L, Landolfi A, Richards S, Symes S, Colucci A, Maruotti G, Adair D, Guida M, Martinelli P, Guida M. Serum metallome in pregnant women and the relationship with congenital malformations of the central nervous system: a case-control study. BMC Pregnancy Childbirth 2019; 19:471. [PMID: 31805895 PMCID: PMC6896487 DOI: 10.1186/s12884-019-2636-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/26/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Congenital malformations of the central nervous system (CNS) consist of a wide range of birth defects of multifactorial origin. METHODS Concentrations of 44 metals were determined by Inductively Coupled Plasma Mass Spectrometry in serum of 111 mothers in the second trimester of pregnancy who carried a malformed fetus and compared them with serum concentrations of the same metals in 90 mothers with a normally developed fetus at the same week of pregnancy. Data are reported as means ± standard deviations. RESULTS We found a direct relationship between congenital defects of the CNS and maternal serum concentration of aluminum: it was statistically higher in women carrying a fetus with this class of malformation, compared both to mothers carrying a fetus with another class of malformation (6.45 ± 15.15 μg/L Vs 1.44 ± 4.21 μg/L, p < 0.0006) and to Controls (i.e. mothers carrying a normally-developed fetus) (6.45 ± 15.15 μg/L Vs 0.11 ± 0.51 μg/L, p < 0.0006). Moreover, Aluminum abundances were below the limit of detection in the majority of control samples. CONCLUSION CAluminum may play a role in the onset of central nervous system malformations, although the exact Aluminum species and related specific type of malformation needs further elucidation.
Collapse
Affiliation(s)
- Jacopo Troisi
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, Salerno, Italy.,THEOREO Srl Spin-off Company of the University of Salerno, Salerno, Italy
| | - Luigi Giugliano
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, Salerno, Italy
| | - Laura Sarno
- Department of Neurosciences, Dentistry and Reproductive Sciences, University of Naples "Federico II", Naples, Italy.
| | - Annamaria Landolfi
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, Salerno, Italy
| | - Sean Richards
- Department of Biology, Geology and Environmental Sciences, University of Tennessee at Chattanooga, 615 McCallie Ave, Chattanooga, TN, 37403, USA.,Department of Obstetrics and Gynecology, University of Tennessee College of Medicine, Chattanooga, TN, USA
| | - Steven Symes
- Department of Chemistry and Physics, University of Tennessee at Chattanooga, 615 McCallie Ave, Chattanooga, TN, 37403, USA.,Department of Obstetrics and Gynecology, University of Tennessee College of Medicine, Chattanooga, TN, USA
| | - Angelo Colucci
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, Salerno, Italy.,THEOREO Srl Spin-off Company of the University of Salerno, Salerno, Italy
| | - Giuseppe Maruotti
- Department of Neurosciences, Dentistry and Reproductive Sciences, University of Naples "Federico II", Naples, Italy
| | - David Adair
- Department of Chemistry and Physics, University of Tennessee at Chattanooga, 615 McCallie Ave, Chattanooga, TN, 37403, USA
| | - Marco Guida
- Department of Biology, University of Naples "Federico II", Naples, Italy
| | - Pasquale Martinelli
- Department of Neurosciences, Dentistry and Reproductive Sciences, University of Naples "Federico II", Naples, Italy
| | - Maurizio Guida
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, Salerno, Italy.,THEOREO Srl Spin-off Company of the University of Salerno, Salerno, Italy
| |
Collapse
|
19
|
Khedr LH, Nassar NN, Rashed L, El-Denshary ED, Abdel-Tawab AM. TLR4 signaling modulation of PGC1-α mediated mitochondrial biogenesis in the LPS-Chronic mild stress model: Effect of fluoxetine and pentoxiyfylline. Life Sci 2019; 239:116869. [PMID: 31678277 DOI: 10.1016/j.lfs.2019.116869] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 09/10/2019] [Accepted: 09/10/2019] [Indexed: 11/15/2022]
Abstract
AIM The addition of repeated lipopolysaccharide (LPS) to chronic mild stress was recently proposed in our lab as an alternative model of depression, highlighting the possible interaction between stress and immune-inflammatory pathways in predisposing depression. Given that CMS-induced depressive behavior was previously related to impaired hippocampal energy metabolism and mitochondrial dysfunction, our current study aimed to investigate the interplay between toll-like receptor 4 (TLR4) signaling and peroxisome proliferator-activated receptor gamma coactivators-1-alpha (PGC1-α) as a physiological regulator of energy metabolism and mitochondrial biogenesis in the combined LPS/CMS model. MAIN METHODS Male Wistar rats were exposed to either LPS (50 μg/kg i.p.) over 2 weeks, CMS protocol for 4 weeks or LPS over 2 weeks followed by 4 weeks of CMS (LPS/CMS). Three additional groups of rats were exposed to LPS/CMS protocol and treated with either pentoxifylline (PTX), fluoxetine (FLX) or a combination of both. Rats were examined for behavioral, neurochemical, gene expression and mitochondrial ultra-structural changes. KEY FINDINGS LPS/CMS increased the expression of TLR4 and its downstream players; MyD88, NFκB and TNF-α along with an escalation in hippocampal-energy metabolism and p-AMPK. Simultaneously LPS/CMS attenuated the expression of PGC1-α/NRF1/Tfam and mt-DNA. The antidepressant (AD) 'FLX', the TNF-α inhibitor 'PTX' and their combination ameliorated the LPS/CMS-induced changes. Interestingly, all the aforementioned changes induced by the LPS/CMS combined model were significantly less than those induced by CMS alone. SIGNIFICANCE Blocking the TLR4/NFκB signaling enhanced the activation of the PGC1-α/NRF1/Tfam and mt-DNA content independent on the activation of the energy-sensing kinase AMPK.
Collapse
Affiliation(s)
- L H Khedr
- Departmment of Pharmacology, Faculty of Pharmacy, Misr International University, Cairo, Egypt.
| | - N N Nassar
- Department of Pharmacology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Laila Rashed
- Department of Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - E D El-Denshary
- Department of Pharmacology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - A M Abdel-Tawab
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
20
|
Sanchez-Guerra M, Peng C, Trevisi L, Cardenas A, Wilson A, Osorio-Yáñez C, Niedzwiecki MM, Zhong J, Svensson K, Acevedo MT, Solano-Gonzalez M, Amarasiriwardena CJ, Estrada-Gutierrez G, Brennan KJM, Schnaas L, Just AC, Laue HE, Wright RJ, Téllez-Rojo MM, Wright RO, Baccarelli AA. Altered cord blood mitochondrial DNA content and pregnancy lead exposure in the PROGRESS cohort. ENVIRONMENT INTERNATIONAL 2019; 125:437-444. [PMID: 30753999 PMCID: PMC6391888 DOI: 10.1016/j.envint.2019.01.077] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 05/23/2023]
Abstract
INTRODUCTION Lead (Pb) crosses the placenta and can cause oxidative stress, reduced fetal growth and neurological problems. The principal source of oxidative stress in human cells is mitochondria. Therefore, disruption of normal mitochondrial function during pregnancy may represent a primary mechanism behind the adverse effects of lead. We sought to assess the association of Pb exposure during pregnancy with mitochondrial DNA (mtDNA) content, a sensitive marker of mitochondrial function, in cord blood. MATERIALS AND METHODS This study comprised mother-infant pairs from the Programming Research in Obesity, Growth, Environment and Social Stressors (PROGRESS) study, a prospective birth-cohort that enrolled 1050 pregnant women from Mexico City who were receiving prenatal care between December 2007 and July 2011. Quantitative PCR was used to calculate relative MtDNA content (mitochondrial-to-nuclear DNA ratio (mtDNA/nDNA)) in cord blood. Lead concentrations in both maternal blood (2nd and 3rd trimester and at delivery day) and in cord blood were measured by ICP-MS. Multivariable regression models adjusting for multiple confounders were fitted with 410 mother-infant pairs for whom complete data for mtDNA content, lead levels, and covariates were available. RESULTS Maternal blood Pb measured in the second (mean 3.79 μg/dL, SD 2.63; β = 0.059, 95% CI 0.008, 0.111) and third trimester (mean 3.90 μg/dL; SD 2.84; β = 0.054, 95% CI 0.002, 0.107) during pregnancy and PB in cord blood (mean 3.50 μg/dL, SD 2.59; β = 0.050, 95% CI 0.004; 0.096) were associated with increased cord blood mtDNA content (mean 1.46, SD 0.44). In two-way interaction analyses, cord blood Pb marginally interacted with gestational age leading to an increase in mtDNA content for pre-term births (Benjamini-Hochberg False Discovery Rate correction; BH-FDR = 0.08). CONCLUSION This study shows that lead exposure in pregnancy alters mtDNA content in cord blood; therefore, alteration of mtDNA content might be a mechanism underlying the toxicity of lead.
Collapse
Affiliation(s)
- Marco Sanchez-Guerra
- Department of Developmental Neurobiology, National Institute of Perinatology, Montes Urales 800, Lomas Virreyes, Mexico City 11000, Mexico.
| | - Cheng Peng
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Letizia Trevisi
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, MA, USA
| | - Andres Cardenas
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim HealthCare Institute, Boston, MA, USA
| | - Ander Wilson
- Department of Statistics, Colorado State University, Fort Collins, CO 80523, USA
| | - Citlalli Osorio-Yáñez
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Center for Nutrition and Health Research, National Institute of Public Health, Ministry of Health, Cuernavaca, Morelos, Mexico
| | - Megan M Niedzwiecki
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia Zhong
- Department of Environmental Health Sciences, Columbia University, Mailman School of Public Health, New York, NY, USA
| | - Katherine Svensson
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria Teresa Acevedo
- Department of Developmental Neurobiology, National Institute of Perinatology, Montes Urales 800, Lomas Virreyes, Mexico City 11000, Mexico
| | - Maritsa Solano-Gonzalez
- Center for Nutrition and Health Research, National Institute of Public Health, Ministry of Health, Cuernavaca, Morelos, Mexico
| | | | - Guadalupe Estrada-Gutierrez
- Department of Developmental Neurobiology, National Institute of Perinatology, Montes Urales 800, Lomas Virreyes, Mexico City 11000, Mexico
| | - Kasey J M Brennan
- Department of Environmental Health Sciences, Columbia University, Mailman School of Public Health, New York, NY, USA
| | - Lourdes Schnaas
- Department of Developmental Neurobiology, National Institute of Perinatology, Montes Urales 800, Lomas Virreyes, Mexico City 11000, Mexico
| | - Allan C Just
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hannah E Laue
- Department of Environmental Health Sciences, Columbia University, Mailman School of Public Health, New York, NY, USA
| | - Rosalind J Wright
- Kravis Children's Hospital, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Martha Maria Téllez-Rojo
- Center for Nutrition and Health Research, National Institute of Public Health, Ministry of Health, Cuernavaca, Morelos, Mexico
| | - Robert O Wright
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Columbia University, Mailman School of Public Health, New York, NY, USA.
| |
Collapse
|
21
|
Baranowska-Bosiacka I, Olszowski T, Gutowska I, Korbecki J, Rębacz-Maron E, Barczak K, Lubkowska A, Chlubek D. Fatty acid levels alterations in THP-1 macrophages cultured with lead (Pb). J Trace Elem Med Biol 2019; 52:222-231. [PMID: 30732887 DOI: 10.1016/j.jtemb.2019.01.003] [Citation(s) in RCA: 2] [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: 10/23/2018] [Revised: 12/16/2018] [Accepted: 01/04/2019] [Indexed: 10/27/2022]
Abstract
OBJECTIVE As cardiovascular events are one of the main causes of death in developed countries, each factor potentially increasing the risk of cardiovascular disease deserves special attention. One such factor is the potentially atherogenic effect of lead (Pb) on lipid metabolism, and is significant in view of the still considerable Pb environmental pollution and the non-degradability of Pb compounds. METHODS Analysis of saturated fatty acids (SFA) (caprylic acid (C8:0), decanoic acid (C10:0), lauric acid (C12:0), tridecanoic acid (C13:0), myristic acid (C14:0), pentadecanoic acid (C15:0), palmitic acid (C16:0), heptadecanoic acid (C17:0), stearic acid (C18:0), and behenic acid (C22:0)), monounsaturated fatty acid (MUFA) (palmitoleic acid (C16:1), oleic acid (18:1w9), trans-vaccenic acid (C18:1 trans11)), and polyunsaturated fatty acid (PUFA) (linoleic acid (C18:2n6), gamma-linolenic acid (C18:3n6), arachidonic acid (C20:4n6)), was conducted by gas chromatography. Analysis of stearoyl-CoA desaturase (SCD), fatty acid desaturase 1 (FADS1) and fatty acid desaturase 2 (FADS2) expression was performed using qRT-PCR. Oxidative stress intensity (malondialdehyde - MDA concentration) was measured using spectrophotometric method. Intracellular generation of reactive oxygen species (ROS) in macrophages was visualized by fluorescence microscopy and quantitatively measured by plate reader. RESULTS Pb caused quantitative alterations in FAs profile in macrophages; the effect was Pb-concentration dependent and selective (i.e. concerned only selected FAs). In general, the effect of Pb was biphasic, with Pb levels of 1.25 μg/dL and 2.5 μg/dL being stimulatory, and 10 μg/dL being inhibitory on concentrations of selected FAs. The most potent Pb concentration, resulting in increase in levels of 9 FAs, was 2.5 μg/dL, the Pb-level corresponding to the mean blood Pb concentrations of people living in urban areas not contaminated by Pb. Pb was found to exert similar, biphasic effect on the expression of FADS1. However, Pb decreased, in a concentration-dependent manner, the expression of SCD and FADS2. Pb significantly increased MDA and ROS concentration in macrophages. CONCLUSION Environmental Pb exposure might be a risk factor resulting in alterations in FAs levels, oxidative stress and increased MDA concentration in macrophages, which might lead to the formation of foam cells and to inflammatory reactions.
Collapse
Affiliation(s)
- Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72 St., 70-111, Szczecin, Poland.
| | - Tomasz Olszowski
- Department of Hygiene and Epidemiology, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72 St., 70-111, Szczecin, Poland
| | - Izabela Gutowska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, Broniewskiego 24 St., 71-460, Szczecin, Poland
| | - Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72 St., 70-111, Szczecin, Poland
| | - Ewa Rębacz-Maron
- University of Szczecin, Department of Vertebrate Zoology and Anthropology, Institute for Research on Biodiversity, Faculty of Biology, University of Szczecin, Wąska 13 St., 71-415, Szczecin, Poland
| | - Katarzyna Barczak
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Anna Lubkowska
- Department of Functional Diagnostics and Physical Medicine, Pomeranian Medical University in Szczecin, 71-210, Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72 St., 70-111, Szczecin, Poland
| |
Collapse
|
22
|
Influence of Acetylcholinesterase Inhibitors Used in Alzheimer's Disease Treatment on the Activity of Antioxidant Enzymes and the Concentration of Glutathione in THP-1 Macrophages under Fluoride-Induced Oxidative Stress. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 16:ijerph16010010. [PMID: 30577562 PMCID: PMC6339019 DOI: 10.3390/ijerph16010010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/09/2018] [Accepted: 12/17/2018] [Indexed: 01/03/2023]
Abstract
It has been reported that donepezil and rivastigmine, the acetylcholinesterase (AchE) inhibitors commonly used in the treatment of Alzheimer’s disease (AD), do not only inhibit AChE but also have antioxidant properties. As oxidative stress is involved in AD pathogenesis, in our study we attempted to examine the influence of donepezil and rivastigmine on the activity of antioxidant enzymes and glutathione concentration in macrophages—an important source of reactive oxygen species and crucial for oxidative stress progression. The macrophages were exposed to sodium fluoride induced oxidative stress. The antioxidant enzymes activity and concentration of glutathione were measured spectrophotometrically. The generation of reactive oxygen species was visualized by confocal microscopy. The results of our study showed that donepezil and rivastigmine had a stimulating effect on catalase activity. However, when exposed to fluoride-induced oxidative stress, the drugs reduced the activity of some antioxidant enzymes (Cat, SOD, GR). These observations suggest that the fluoride-induced oxidative stress may suppress the antioxidant action of AChE inhibitors. Our results may have significance in the clinical practice of treatment of AD and other dementia diseases.
Collapse
|
23
|
Zhou CC, Gao ZY, Wang J, Wu MQ, Hu S, Chen F, Liu JX, Pan H, Yan CH. Lead exposure induces Alzheimers's disease (AD)-like pathology and disturbes cholesterol metabolism in the young rat brain. Toxicol Lett 2018; 296:173-183. [PMID: 29908845 DOI: 10.1016/j.toxlet.2018.06.1065] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/30/2018] [Accepted: 06/12/2018] [Indexed: 02/07/2023]
Abstract
Lead exposure has been evidenced as a risk factor for Alzheimer's disease (AD), mainly affecting the ageing. However, the early manifestation and mechanisms of AD-like pathology induced by lead exposure remains to be elucidated. Considering the fact that impaired cholesterol metabolism is associated with many neurodegenerative disorders including AD, in this study we focused on the role of cholesterol metabolism in lead induced premature AD-like pathology. We treated weaning rats with lead at different concentrations for 4 weeks. We found that developmental lead exposure increased amyloid-beta (Aβ) accumulation and amyloid plaque deposition in the cortex and hippocampus. Lead exposure increased amyloid precursor protein (APP) expression and activated the sterol regulatory element binding protein 2 (SREBP2)-beta secretase (BACE1) pathway. In addition, we found that lead exposure decreased cholesterol levels by upregulating the expression of liver X receptor-a (LXR-a) and ATP-binding cassette transporter protein family member A1 (ABCA1) and decreasing the expression of 3-hydroxy-3-methylglutaryl-CoA reductase (HMG-CR) and low density lipoprotein receptor (LDL-R) in young rat brain tissues. Taken together, our data demonstrated that developmental lead exposure induced early manifestation of AD-like pathology and disturbed cholesterol metabolism in young rat brains.
Collapse
Affiliation(s)
- Can-Can Zhou
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China
| | - Zhen-Yan Gao
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China
| | - Ju Wang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China
| | - Mei-Qin Wu
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China
| | - Shuang Hu
- East China University of Science and Technology, Shanghai, 200237, PR China
| | - Fei Chen
- East China University of Science and Technology, Shanghai, 200237, PR China
| | - Jun-Xia Liu
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China
| | - Hui Pan
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China
| | - Chong-Huai Yan
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China.
| |
Collapse
|
24
|
Monteiro C, Ferreira de Oliveira JMP, Pinho F, Bastos V, Oliveira H, Peixoto F, Santos C. Biochemical and transcriptional analyses of cadmium-induced mitochondrial dysfunction and oxidative stress in human osteoblasts. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2018; 81:705-717. [PMID: 29913117 DOI: 10.1080/15287394.2018.1485122] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Cadmium (Cd) accumulation is known to occur predominantly in kidney and liver; however, low-level long-term exposure to Cd may also result in bone damage. Few studies have addressed Cd-induced toxicity in osteoblasts, particularly upon cell mitochondrial energy processing and putative associations with oxidative stress in bone. To assess the influence of Cd treatment on mitochondrial function and oxidative status in osteoblast cells, human MG-63 cells were treated with Cd (up to 65 μM) for 24 or 48 h. Intracellular reactive oxygen species (ROS), lipid and protein oxidation and antioxidant defense mechanisms such as total antioxidant activity (TAA) and gene expression of antioxidant enzymes were analyzed. In addition, Cd-induced effects on mitochondrial function were assessed by analyzing the activity of enzymes involved in mitochondrial respiration, membrane potential (ΔΨm), mitochondrial morphology and adenylate energy charge. Treatment with Cd increased oxidative stress, concomitantly with lipid and protein oxidation. Real-time polymerase chain reaction (qRT-PCR) analyses of antioxidant genes catalase (CAT), glutathione peroxidase 1 (GPX1), glutathione S-reductase (GSR), and superoxide dismutase (SOD1 and SOD2) exhibited a trend toward decrease in transcripts in Cd-stressed cells, particularly a downregulation of GSR. Longer treatment with Cd (48 h) resulted in energy charge states significantly below those commonly observed in living cells. Mitochondrial function was affected by ΔΨm reduction. Inhibition of mitochondrial respiratory chain enzymes and citrate synthase also occurred following Cd treatment. In conclusion, Cd induced mitochondrial dysfunction which appeared to be associated with oxidative stress in human osteoblasts.
Collapse
Affiliation(s)
- Cristina Monteiro
- a Department of Biology & CESAM , University of Aveiro, Campus Universitário , Aveiro , Portugal
| | - José Miguel P Ferreira de Oliveira
- b LAQV/REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy , University of Porto , Porto , Portugal
| | - Francisco Pinho
- a Department of Biology & CESAM , University of Aveiro, Campus Universitário , Aveiro , Portugal
| | - Verónica Bastos
- c Department of Biology & LAQV/REQUIMTE , Faculty of Sciences of University of Porto , Porto , Portugal
| | - Helena Oliveira
- a Department of Biology & CESAM , University of Aveiro, Campus Universitário , Aveiro , Portugal
| | - Francisco Peixoto
- d Biology and Environment Department , Chemistry Research Center, University of Trás-os-Montes & Alto Douro , Portugal
| | - Conceição Santos
- c Department of Biology & LAQV/REQUIMTE , Faculty of Sciences of University of Porto , Porto , Portugal
| |
Collapse
|
25
|
Ahmad F, Salahuddin M, Alamoudi W, Acharya S. Dysfunction of cortical synapse-specific mitochondria in developing rats exposed to lead and its amelioration by ascorbate supplementation. Neuropsychiatr Dis Treat 2018; 14:813-824. [PMID: 29606875 PMCID: PMC5868605 DOI: 10.2147/ndt.s148248] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Lead (Pb) is a widespread environmental neurotoxin and its exposure even in minute quantities can lead to compromised neuronal functions. A developing brain is particularly vulnerable to Pb mediated toxicity and early-life exposure leads to permanent alterations in brain development and neuronal signaling and plasticity, culminating into cognitive and behavioral dysfunctions and elevated risk of neuropsychiatric disorders later in life. Nevertheless, the underlying biochemical mechanisms have not been completely discerned. METHODS Because of their ability to fulfill high energy needs and to act as calcium buffers in events of high intensity neuronal activity as well as their adaptive regulatory capability to match the requirements of the dynamicity of synaptic signaling, synapse-specific or synaptic mitochondria (SM) are critical for synaptic development, function and plasticity. Our aim for the present study hence was to characterize the effects of early-life Pb exposure on the functions of SM of prepubertal rats. For this purpose, employing a chronic model of Pb neurotoxicity, we exposed rat pups perinatally and postnatally to Pb and used a plethora of colorimetric and fluorometric assays for assessing redox and bioenergetic properties of SM. In addition, taking advantage of its ability as an antioxidant and as a metal chelator, we employed ascorbic acid (vitamin C) supplementation as an ameliorative therapeutic strategy against Pb-induced neurotoxicity and dysfunction of SM. RESULTS Our results suggest that early-life exposure to Pb leads to elevated oxidative stress in cortical SM with consequent compromises in its energy metabolism activity. Ascorbate supplementation resulted in significant recovery of Pb-induced oxidative stress and functional compromise of SM. CONCLUSION Alterations in redox status and bioenergetic properties of SM could potentially contribute to the synaptic dysfunction observed in events of Pb neurotoxicity. Additionally, our study provides evidence for suitability of ascorbate as a significant ameliorative agent in tacking Pb neurotoxicity.
Collapse
Affiliation(s)
- Faraz Ahmad
- Department of Public Health, College of Public Health, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.,Neuroscience Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Mohammad Salahuddin
- Animal House Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Widyan Alamoudi
- Neuroscience Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Sadananda Acharya
- Department of Public Health, College of Public Health, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
26
|
Glycogen metabolism in brain and neurons - astrocytes metabolic cooperation can be altered by pre- and neonatal lead (Pb) exposure. Toxicology 2017; 390:146-158. [PMID: 28916327 DOI: 10.1016/j.tox.2017.09.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/28/2017] [Accepted: 09/07/2017] [Indexed: 11/22/2022]
Abstract
Lead (Pb) is an environmental neurotoxin which particularly affects the developing brain but the molecular mechanism of its neurotoxicity still needs clarification. The aim of this paper was to examine whether pre- and neonatal exposure to Pb (concentration of Pb in rat offspring blood below the "threshold level") may affect the brain's energy metabolism in neurons and astrocytes via the amount of available glycogen. We investigated the glycogen concentration in the brain, as well as the expression of the key enzymes involved in glycogen metabolism in brain: glycogen synthase 1 (Gys1), glycogen phosphorylase (PYGM, an isoform active in astrocytes; and PYGB, an isoform active in neurons) and phosphorylase kinase β (PHKB). Moreover, the expression of connexin 43 (Cx43) was evaluated to analyze whether Pb poisoning during the early phase of life may affect the neuron-astrocytes' metabolic cooperation. This work shows for the first time that exposure to Pb in early life can impair brain energy metabolism by reducing the amount of glycogen and decreasing the rate of its metabolism. This reduction in brain glycogen level was accompanied by a decrease in Gys1 expression. We noted a reduction in the immunoreactivity and the gene expression of both PYGB and PYGM isoform, as well as an increase in the expression of PHKB in Pb-treated rats. Moreover, exposure to Pb induced decrease in connexin 43 immunoexpression in all the brain structures analyzed, both in astrocytes as well as in neurons. Our data suggests that exposure to Pb in the pre- and neonatal periods results in a decrease in the level of brain glycogen and a reduction in the rate of its metabolism, thereby reducing glucose availability, which as a further consequence may lead to the impairment of brain energy metabolism and the metabolic cooperation between neurons and astrocytes.
Collapse
|
27
|
|
28
|
Łukomska A, Baranowska-Bosiacka I, Budkowska M, Pilutin A, Tarnowski M, Dec K, Dołęgowska B, Metryka E, Chlubek D, Gutowska I. The effect of low levels of lead (Pb) in the blood on levels of sphingosine-1-phosphate (S1P) and expression of S1P receptor 1 in the brain of the rat in the perinatal period. CHEMOSPHERE 2017; 166:221-229. [PMID: 27697711 DOI: 10.1016/j.chemosphere.2016.09.067] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 08/31/2016] [Accepted: 09/15/2016] [Indexed: 06/06/2023]
Abstract
Sphingolipids are the main components of the lipid membrane. They also perform structural functions and participate in many signal transmission processes. One of the bioactive sphingolipids is sphingosine-1-phosphate (S1P), a ligand for five G protein-coupled receptors (S1PRs1-5), which can also act as an intracellular second messenger. S1P is responsible for the stimulation of progenitor cells in the brain, but it can also induce apoptosis of mature neurons. This study is aimed at assessing the effect of pre- and neonatal exposure to permissible Pb concentrations on S1P levels and S1PR1 (EDG1) expression in the prefrontal cortex, cerebellum, and hippocampus of rats. The concentrations of S1P were determined by RP-HPLC, S1PR1 expression was determined by RT PCR and Western Blot, and receptor immunolocalization was determined by immunohistochemistry method. Our results showed that even low blood Pb concentrations, i.e. within the acceptable limit of 10 μg/dL caused changes in the concentration of S1P in the cerebellum, prefrontal cortex, and hippocampus. Our data also showed a significant decrease in the level of S1PR1 in all studied part of brain, without significant changes in S1PR1 gene expression. Pre- and neonatal exposure to Pb also resulted in a decrease in the expression of S1PR1 in glial cells in all regions of the Cornu Ammonis (CA1-CA4) and Dentate Gyrus in the hippocampus, as well as in all layers of the cerebellum and prefrontal cortex, compared to the unexposed control group.
Collapse
Affiliation(s)
- A Łukomska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University, Broniewskiego 24, 71-460 Szczecin, Poland
| | - I Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland.
| | - M Budkowska
- Department of Microbiology and Immunology, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - A Pilutin
- Department of Histology and Embryology, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - M Tarnowski
- Department of Physiology, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - K Dec
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University, Broniewskiego 24, 71-460 Szczecin, Poland
| | - B Dołęgowska
- Department of Histology and Embryology, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - E Metryka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - D Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - I Gutowska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University, Broniewskiego 24, 71-460 Szczecin, Poland
| |
Collapse
|
29
|
Cyclosporin A protects against Lead neurotoxicity through inhibiting mitochondrial permeability transition pore opening in nerve cells. Neurotoxicology 2016; 57:203-213. [PMID: 27725305 DOI: 10.1016/j.neuro.2016.10.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 09/22/2016] [Accepted: 10/06/2016] [Indexed: 01/28/2023]
Abstract
Mitochondria play a key role in the process of lead (Pb)-induced impairment in nervous system. To further clarify the underlying mechanism of Pb neurotoxicity, this study was designed to investigate the role of mitochondrial permeability transition (MPT) and cyclophilin D (CyPD), a component of MPT pore (MPTP), in Pb-induced mitochondrial apoptosis in nerve cells. In SH-SY5Y and PC12 cells, Cyclosporin A (CSA), a special inhibitor of CyPD, could alleviate cell death, lactate dehydrogenase (LDH) leakage and adenosine 5 triphosphate (ATP) decrease caused by PbAc. In the following experiments, we found PbAc increased the protein level of CyPD and induced MPT pore (MPTP) opening. When cells were pretreated with CSA to inhibit MPTP opening, the Pb-induced impairment of mitochondrial morphology (swelling and rupture) and the loss of mitochondria were attenuated. In addition, CSA obviously ameliorated the Pb-induced damage of mitochondrial function, such as reactive oxygen species (ROS) boost and mitochondrial membrane potential (MMP) collapse, as well as the release of cytochrome C (Cyto C) and apoptosis-inducing factor (AIF) from mitochondria. These beneficial effects could finally result in cell survival under Pb-exposure conditions. Furthermore, scavenging ROS also significantly abrogated MPTP opening and attenuated Pb neurotoxicity. Therefore, we found that MPT played an important role in Pb-induced mitochondrial damage and, ultimately, cell death. Our results provided a potential strategy for inhibiting PbAc neurotoxicity. However, due to the high Pb concentrations used in this study further investigations at Pb concentrations closer to human exposure are needed to verify the results.
Collapse
|
30
|
Baranowska-Bosiacka I, Listos J, Gutowska I, Machoy-Mokrzyńska A, Kolasa-Wołosiuk A, Tarnowski M, Puchałowicz K, Prokopowicz A, Talarek S, Listos P, Wąsik A, Chlubek D. Effects of perinatal exposure to lead (Pb) on purine receptor expression in the brain and gliosis in rats tolerant to morphine analgesia. Toxicology 2016; 339:19-33. [DOI: 10.1016/j.tox.2015.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/08/2015] [Accepted: 10/12/2015] [Indexed: 02/08/2023]
|
31
|
Abstract
SIGNIFICANCE Mitochondria are structurally and biochemically diverse, even within a single type of cell. Protein complexes localized to the inner mitochondrial membrane synthesize ATP by coupling electron transport and oxidative phosphorylation. The organelles produce reactive oxygen species (ROS) from mitochondrial oxygen and ROS can, in turn, alter the function and expression of proteins used for aerobic respiration by post-translational and transcriptional regulation. RECENT ADVANCES New interest is emerging not only into the roles of mitochondria in disease development and progression but also as a target for environmental toxicants. CRITICAL ISSUES Dysregulation of respiration has been linked to cell death and is a major contributor to acute neuronal trauma, peripheral diseases, as well as chronic neurodegenerative diseases, such as Parkinson's disease and Alzheimer's disease. FUTURE DIRECTIONS Here, we discuss the mechanisms underlying the sensitivity of the mitochondrial respiratory complexes to redox modulation, as well as examine the effects of environmental contaminants that have well-characterized mitochondrial toxicity. The contaminants discussed in this review are some of the most prevalent and potent environmental contaminants that have been linked to neurological dysfunction, altered cellular respiration, and oxidation.
Collapse
Affiliation(s)
- Samuel W Caito
- Department of Molecular Pharmacology, Albert Einstein College of Medicine , Bronx, New York
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine , Bronx, New York
| |
Collapse
|
32
|
Lassiter MG, Owens EO, Patel MM, Kirrane E, Madden M, Richmond-Bryant J, Hines EP, Davis JA, Vinikoor-Imler L, Dubois JJ. Cross-species coherence in effects and modes of action in support of causality determinations in the U.S. Environmental Protection Agency's Integrated Science Assessment for Lead. Toxicology 2015; 330:19-40. [PMID: 25637851 DOI: 10.1016/j.tox.2015.01.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 12/24/2014] [Accepted: 01/26/2015] [Indexed: 01/26/2023]
Abstract
The peer-reviewed literature on the health and ecological effects of lead (Pb) indicates common effects and underlying modes of action across multiple organisms for several endpoints. Based on such observations, the United States (U.S.) Environmental Protection Agency (EPA) applied a cross-species approach in the 2013 Integrated Science Assessment (ISA) for Lead for evaluating the causality of relationships between Pb exposure and specific endpoints that are shared by humans, laboratory animals, and ecological receptors (i.e., hematological effects, reproductive and developmental effects, and nervous system effects). Other effects of Pb (i.e., cardiovascular, renal, and inflammatory responses) are less commonly assessed in aquatic and terrestrial wildlife limiting the application of cross-species comparisons. Determinations of causality in ISAs are guided by a framework for classifying the weight of evidence across scientific disciplines and across related effects by considering aspects such as biological plausibility and coherence. As illustrated for effects of Pb where evidence across species exists, the integration of coherent effects and common underlying modes of action can serve as a means to substantiate conclusions regarding the causal nature of the health and ecological effects of environmental toxicants.
Collapse
Affiliation(s)
- Meredith Gooding Lassiter
- National Center for Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Elizabeth Oesterling Owens
- National Center for Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Molini M Patel
- National Center for Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Ellen Kirrane
- National Center for Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Meagan Madden
- Oak Ridge Institute for Science and Education Research Participation Program, National Center for Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Jennifer Richmond-Bryant
- National Center for Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Erin Pias Hines
- National Center for Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - J Allen Davis
- National Center for Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Lisa Vinikoor-Imler
- National Center for Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Jean-Jacques Dubois
- Southern Region Integrated Pest Management Center, North Carolina State University, 1730 Varsity Drive, Suite 110, Raleigh, NC 27606, USA.
| |
Collapse
|
33
|
Abstract
Metals are frequently used in industry and represent a major source of toxin exposure for workers. For this reason governmental agencies regulate the amount of metal exposure permissible for worker safety. While essential metals serve physiologic roles, metals pose significant health risks upon acute and chronic exposure to high levels. The central nervous system is particularly vulnerable to metals. The brain readily accumulates metals, which under physiologic conditions are incorporated into essential metalloproteins required for neuronal health and energy homeostasis. Severe consequences can arise from circumstances of excess essential metals or exposure to toxic nonessential metal. Herein, we discuss sources of occupational metal exposure, metal homeostasis in the human body, susceptibility of the nervous system to metals, detoxification, detection of metals in biologic samples, and chelation therapeutic strategies. The neurologic pathology and physiology following aluminum, arsenic, lead, manganese, mercury, and trimethyltin exposures are highlighted as classic examples of metal-induced neurotoxicity.
Collapse
Affiliation(s)
- Samuel Caito
- Division of Clinical Pharmacology and Pediatric Toxicology, Vanderbilt University Medical Center, Nashville, TN, USA; The Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Michael Aschner
- Division of Clinical Pharmacology and Pediatric Toxicology, Vanderbilt University Medical Center, Nashville, TN, USA; The Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, Nashville, TN, USA; Center in Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN, USA; Center for Molecular Neuroscience, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
34
|
Khedr LH, Nassar NN, El-Denshary ES, Abdel-Tawab AM. Paroxetine ameliorates changes in hippocampal energy metabolism in chronic mild stress-exposed rats. Neuropsychiatr Dis Treat 2015; 11:2887-901. [PMID: 26622178 PMCID: PMC4654549 DOI: 10.2147/ndt.s87089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The molecular mechanisms underlying stress-induced depression have not been fully outlined. Hence, the current study aimed at testing the link between behavioral changes in chronic mild stress (CMS) model and changes in hippocampal energy metabolism and the role of paroxetine (PAROX) in ameliorating these changes. Male Wistar rats were divided into three groups: vehicle control, CMS-exposed rats, and CMS-exposed rats receiving PAROX (10 mg/kg/day intraperitoneally). Sucrose preference, open-field, and forced swimming tests were carried out. Corticosterone (CORT) was measured in serum, while adenosine triphosphate and its metabolites, cytosolic cytochrome-c (Cyt-c), caspase-3 (Casp-3), as well as nitric oxide metabolites (NOx) were measured in hippocampal tissue homogenates. CMS-exposed rats showed a decrease in sucrose preference as well as body weight compared to control, which was reversed by PAROX. The latter further ameliorated the CMS-induced elevation of CORT in serum (91.71±1.77 ng/mL vs 124.5±4.44 ng/mL, P<0.001) as well as the changes in adenos-ine triphosphate/adenosine diphosphate (3.76±0.02 nmol/mg protein vs 1.07±0.01 nmol/mg protein, P<0.001). Furthermore, PAROX reduced the expression of Cyt-c and Casp-3, as well as restoring NOx levels. This study highlights the role of PAROX in reversing depressive behavior associated with stress-induced apoptosis and changes in hippocampal energy metabolism in the CMS model of depression.
Collapse
Affiliation(s)
- Lobna H Khedr
- Department of Pharmacology, Faculty of Pharmacy, Misr International University, Cairo, Egypt
| | - Noha N Nassar
- Department of Pharmacology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | | | - Ahmed M Abdel-Tawab
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
35
|
Zhou F, Chen Y, Fan G, Feng C, Du G, Zhu G, Li Y, Jiao H, Guan L, Wang Z. Lead-induced iron overload and attenuated effects of ferroportin 1 overexpression in PC12 cells. Toxicol In Vitro 2014; 28:1339-48. [DOI: 10.1016/j.tiv.2014.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 05/12/2014] [Accepted: 07/11/2014] [Indexed: 01/21/2023]
|
36
|
Mousa AM, Al-Fadhli AS, Rao MS, Kilarkaje N. Gestational lead exposure induces developmental abnormalities and up-regulates apoptosis of fetal cerebellar cells in rats. Drug Chem Toxicol 2014; 38:73-83. [PMID: 24724870 DOI: 10.3109/01480545.2014.907578] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Lead (Pb), a known environmental toxicant, adversely affects almost all organ systems. In this study, we investigated the effects of maternal lead exposure on fetal rat cerebellum. Female Sprague-Dawley rats were given lead nitrate in drinking water (0, 0.5, and 1%) for two weeks before conception, and during pregnancy. Fetuses were collected by caesarian section on gestational day 21 and observed for developmental abnormalities. The fetal cerebellar sections from control and 1% lead group were stained with cresyl violet. Immunohistochemical expressions of p53, Bax, Bcl-2, and caspase 3 were quantified by AnalySIS image analyzer (Life Science, Germany). Lead exposure induced developmental abnormalities of eyes, ear, limbs, neck and ventral abdominal wall; however, these abnormalities were commonly seen in the 1% lead-treated group. In addition, lead also caused fetal mortality and reduced body growth in both dose groups and reduced brain weight in the 1% lead-treated group. The fetal cerebella from the 1% lead-treated group showed unorganized cerebellar cortical layers, and degenerative changes in granule and Purkinje cells such as the formation of clumps of Nissl granules. An increase in Bax and caspase 3, and a decrease in Bcl-2 (p < 0.05), but not in p53, showed apoptosis of the neurons. In conclusion, gestational lead exposure in rats induces fetal toxicity and developmental abnormalities. The lead exposure also impairs development of cerebellar layers, induces structural changes, and apoptosis in the fetal cerebellar cortex. These results suggest that lead exposure during gestation is extremely toxic to developing cerebellum in rats.
Collapse
Affiliation(s)
- Alyaa M Mousa
- Department of Anatomy, Faculty of Medicine, Kuwait University , Kuwait
| | | | | | | |
Collapse
|
37
|
Blaurock-Busch E, Busch YM, Friedle A, Buerner H, Parkash C, Kaur A. Comparing the metal concentration in the hair of cancer patients and healthy people living in the malwa region of punjab, India. Clin Med Insights Oncol 2014; 8:1-13. [PMID: 24453505 PMCID: PMC3891755 DOI: 10.4137/cmo.s13410] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Revised: 11/17/2013] [Accepted: 11/21/2013] [Indexed: 01/16/2023] Open
Abstract
The cancer prevalence in the Malwa region of Punjab (1089/million/year) is much higher than the national average cancer prevalence in India (800/million/year). The participants in the present study were 50 healthy individuals and 49 cancer patients all living in the Malwa region of Punjab, with the healthy people being selected from the same household as the cancer patients. High concentrations of several potentially toxic elements were found in hair samples from people living in Punjab. Compared to standard reference ranges, the metals in excess in both the control and patient groups were aluminium (Al), barium (Ba), manganese (Mn), strontium (Sr) and uranium (U). The most significant findings were high lead (Pb), U and Ba concentrations. The maximum values for Ba, Mn, Pb and U were found in hair from breast cancer patients. The mean concentration of U in hair from the breast cancer patients was 0.63 μg U/g, which is more than double the value found in the control group and over six times higher than the reference range of 0.1 μg U/g. Water, soil, and phosphate fertilizers all seem to play a potential role, causing an increased metal burden in Punjabi people living in the Malwa region. The present study indicates that metals, and especially U, may be a factor in the development of breast cancer among Punjabi women.
Collapse
Affiliation(s)
| | | | | | - Holger Buerner
- Trace Minerals International, Colorado, USA
- Micro Trace Minerals, Hersbruck, Germany
| | | | - Anudeep Kaur
- Punjab Technical University, Kapurthala, Punjab, India
| |
Collapse
|
38
|
Bornhorst J, Ebert F, Lohren H, Humpf HU, Karst U, Schwerdtle T. Effects of manganese and arsenic species on the level of energy related nucleotides in human cells. Metallomics 2012; 4:297-306. [PMID: 22266671 DOI: 10.1039/c2mt00164k] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Cellular adenine and pyridine nucleotides play important roles in the cellular energy and redox state. An imbalance in the cellular levels of these tightly regulated energy related nucleotides can lead to oxidative stress and thus is discussed to contribute to neurotoxic and carcinogenic processes. Here we established a reliable ion-pair reversed phase HPLC based method for the parallel quantification of six energy related nucleotides (ATP, ADP, ADP-ribose, AMP, NAD(+), NADH) in cells and subsequently applied it to determine effects of manganese and arsenic species in cultured human cells. In human lung cells, MnCl(2) (≥50 μM) decreased the levels of ATP, NAD(+) and NADH as well as the NAD(+)/NADH ratio. This reflects a decline in the cellular energy metabolism, most likely resulting from a disturbance of the mitochondrial function. In contrast, cultured astrocytes were more resistant towards manganese. Regarding the arsenicals, a disturbance of the cellular energy related nucleotides was detected in lung cells for arsenite (≥50 μM), monomethylarsonous (≥1 μM), dimethylarsinous (≥1 μM) and dimethylarsinic acid (≥100 μM). Thereby, the single arsenicals seem to disturb the cellular energy and redox state by different mechanisms. Taken together, this study provides further evidence that cellular energy related nucleotides serve as sensitive indicators for toxic species exposure. When searching for a molecular mechanism of toxic compounds, the data illustrate the necessity of quantifying several energy related nucleotides in parallel, especially since ATP depletion, redox state alterations and oxidative stress are known to potentiate each other.
Collapse
Affiliation(s)
- Julia Bornhorst
- Graduate School of Chemistry, Wilhelm-Klemm-Str. 10, 48149 Münster, Germany
| | | | | | | | | | | |
Collapse
|
39
|
Tchernitchin AN, Gaete L, Bustamante R, Báez A. Effect of prenatal exposure to lead on estrogen action in the prepubertal rat uterus. ISRN OBSTETRICS AND GYNECOLOGY 2012; 2011:329692. [PMID: 22263113 PMCID: PMC3255308 DOI: 10.5402/2011/329692] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 09/21/2011] [Indexed: 11/23/2022]
Abstract
Lead is a widely spread environmental pollutant known to affect both male and female reproductive systems in humans and experimental animals and causes infertility and other adverse effects. The present paper investigated the effects of prenatal exposure to lead on different parameters of estrogen stimulation in the uterus of the prepubertal rat. In prenatally and perinatally exposed rats, estrogen-induced endometrial eosinophilia, endometrial stroma edema, and eosinophil migration towards the endometrium, and uterine luminal epithelial hypertrophy are enhanced while several other responses to estrogen appear unchanged. These effects may contribute to decrease in fertility following prenatal exposure to lead. The striking difference between most of these effects of prenatal exposure and the previously reported effects of chronic exposure to lead suggests that prenatal exposure to lead may neutralize the effects of chronic exposure to lead, providing partial protection of cell function against the adverse effects of chronic exposure to lead. We propose that the mechanism involved, named imprinting or cell programming, persisted through evolution as a nongenetic adaptive mechanism to provide protection against long-term environmental variations that otherwise may cause the extinction of species not displaying this kind of adaptation.
Collapse
Affiliation(s)
- Andrei N Tchernitchin
- Laboratory of Experimental Endocrinology and Environmental Pathology (LEEPA), Institute of Biomedical Sciences (ICBM), University of Chile Medical School, P.O. Box 21104, Santiago 21, Chile
| | | | | | | |
Collapse
|
40
|
Disrupted pro- and antioxidative balance as a mechanism of neurotoxicity induced by perinatal exposure to lead. Brain Res 2012; 1435:56-71. [DOI: 10.1016/j.brainres.2011.11.062] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 11/10/2011] [Accepted: 11/30/2011] [Indexed: 11/17/2022]
|
41
|
Inorganic lead (Pb)- and mercury (Hg)-induced neuronal cell death involves cytoskeletal reorganization. Lab Anim Res 2011; 27:219-25. [PMID: 21998611 PMCID: PMC3188729 DOI: 10.5625/lar.2011.27.3.219] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 09/05/2011] [Accepted: 09/05/2011] [Indexed: 11/23/2022] Open
Abstract
Inorganic lead and mercury are widely spread xenobiotic neurotoxicants threatening public health. The exposure to inorganic lead and mercury results in adverse effects of poisoning including IQ deficit and peripheral neuropathy. Additionally, inorganic neurotoxicants have even more serious impact on earlier stages of embryonic development. This study was therefore initiated in order to determine the cytotoxic effects of lead and mercury in earlier developmental stages of chick embryo. Administration of inorganic lead and mercury into the chick embryo resulted in the prolonged accumulation of inorganics in the neonatal brain, with detrimental cytotoxicity on neuronal cells. Subsequent studies demonstrated that exposure of chick embryo to inorganic lead and mercury resulted in the reorganization of cytoskeletal proteins in the neonatal brain. These results therefore suggest that inorganics-mediated cytoskeletal reorganization of the structural proteins, resulting in neurocytotoxicity, is one of the underlying mechanisms by which inorganics transfer deleterious effects on central nervous system.
Collapse
|