1
|
Aboubaker DH, Shaffie NA, Shabana MF, Abd Elghafour A, Ibrahim BM. Protective role of savory essential oil on vital organs in rats against deleterious effects induced by lead acetate. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2025; 45:e00871. [PMID: 39811400 PMCID: PMC11732139 DOI: 10.1016/j.btre.2024.e00871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025]
Abstract
The damaging effects of heavy metal exposure on vital organs like the heart, liver, kidneys, and brain can be lowered by natural compounds' anti-inflammatory and anti-oxidant capacity. In the current investigation, the protective potential of savory(Saturejahortensis)essential oil (EO) against lead acetate-induced multi-organ damage in rats was evaluated. Thirty female Wister Albino rats were divided into the following groups: normal, positive control given lead acetate without concomitant treatment, reference given ethylene-diamine-tetra-acetic acid and groups treated with savory EO (0.5 and 1 ml/rat), treatments were administered concomitantly with lead acetate for ten successive days. Electrocardiogram (ECG), open field, and rota-rod tests for evaluation of behaviour, as well as TNF-α, R-GSH & MDA were measured in brain and liver homogenates respectively, CRP, liver, kidney, and heart function tests, were measured in serum. Histopathology of vital organs was conducted at the end of the experiment.A high dose of savory EO, significantly improved the cardiac electrophysiology, psychological state, and locomotor activity of rats; moreover, it lowered inflammatory, oxidative biomarkers, liver, kidney, and cardiac function tests of the rats. Savory EO protects vital organs against heavy metal damage with the superiority of the high dose.
Collapse
Affiliation(s)
- Doha H. Aboubaker
- Medicinal and Aromatic Plants Department, National Research Centre, Dokki, Giza, Egypt
| | | | - Mona F. Shabana
- Medical Pharmacology Department, Faculty of Medicine-Fayoum University, Fayoum, Egypt
| | - Ahmed Abd Elghafour
- Medicinal and Aromatic Plants Department, National Research Centre, Dokki, Giza, Egypt
| | | |
Collapse
|
2
|
Lewandowska MA, Różycka A, Grzelak T, Kempisty B, Jagodziński PP, Lianeri M, Dorszewska J. Expression of Neuronal Nicotinic Acetylcholine Receptor and Early Oxidative DNA Damage in Aging Rat Brain-The Effects of Memantine. Int J Mol Sci 2025; 26:1634. [PMID: 40004097 PMCID: PMC11855568 DOI: 10.3390/ijms26041634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/04/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Aging and age-related neurodegenerative disorders are characterized by the dysfunction or loss of brain nicotinic acetylcholine receptors (nAChRs), and these changes may be related to other senescence markers, such as oxidative stress and DNA repair dysfunction. However, the mechanism of nAChR loss in the aging brain and the modification of this process by drugs (e.g., memantine, Mem) are not yet fully understood. To study whether the differences in nAChR expression in the rat brain occur due to aging or oxidative stress and are modulated by Mem, we analyzed nAChR subunits (at RNA and protein levels) and other biomarkers by real-time quantitative polymerase chain reaction (RQ-PCR) and Western blot validation. Twenty-one female Wistar rats were divided into four groups, depending on age, and the oldest group received injections of Mem or water with the use of intragastric catheters. We studied the cerebral grey matter (CGM), subcortical white matter (SCWM), and cerebellum (Ce). Results showed an age-related decrease of α7 nAChR mRNA level in SCWM. The α7 nAChR mRNA loss was accompanied by reduced expression of 8-oxoguanine DNA glycosylase 1 (OGG1) and an increased tumor necrosis factor alpha (TNFα) level. In the water group, we observed a higher level of α7 nAChR protein in the SCWM and Ce. Biomarker levels changed, but to a different extent depending on the brain area. Importantly, the dysfunction in antioxidative status was stopped and even regressed under Mem treatment. After two weeks of treatment, an increase in TP53 protein level and a decrease in 8-oxo-2'deoxyguanosine (8-oxo-2'dG) level were observed. We conclude that Mem administration may be protective against the senescence process by antioxidative mechanisms.
Collapse
Affiliation(s)
- Małgorzata Anna Lewandowska
- Faculty of Medicine, Poznan Medical University, 55 Bulgarska St., 60-320 Poznan, Poland;
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 6 Świecickiego St., 60-781 Poznan, Poland; (P.P.J.); (M.L.)
| | - Agata Różycka
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 6 Świecickiego St., 60-781 Poznan, Poland; (P.P.J.); (M.L.)
| | - Teresa Grzelak
- Department of Physiology, Poznan University of Medical Sciences, 6 Świecickiego St., 60-781 Poznan, Poland
| | - Bartosz Kempisty
- Department of Human Morphology and Embryology, Division of Anatomy, Wrocław Medical University, 50-368 Wroclaw, Poland;
- Institute of Veterinary Medicine, Nicolaus Copernicus University, 87-100 Torun, Poland
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27695, USA
- Center of Assisted Reproduction, Department of Obstetrics and Gynecology, University Hospital and Masaryk University, 625 00 Brno, Czech Republic
| | - Paweł Piotr Jagodziński
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 6 Świecickiego St., 60-781 Poznan, Poland; (P.P.J.); (M.L.)
| | - Margarita Lianeri
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 6 Świecickiego St., 60-781 Poznan, Poland; (P.P.J.); (M.L.)
| | - Jolanta Dorszewska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355 Poznan, Poland;
| |
Collapse
|
3
|
Flores-Montoya G, Tian Z, Michii A, Chan SY, Sanchez N. The effects of developmental sub-chronic low-level lead exposure on microglia and a test of possible mitigation by apigenin in C57BL/6J young mice. Neurotoxicol Teratol 2025; 107:107406. [PMID: 39638160 DOI: 10.1016/j.ntt.2024.107406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
Developmental chronic low-level lead (Pb) exposure disrupts central nervous system function and diminishes neurocognition. Microglial cell activation might contribute to these deficits. The present study evaluated the effects of developmental sub-chronic low-level lead exposure on microglial cells and the possible effectiveness of a natural anti-inflammatory intervention with apigenin to mitigate these effects. From PND 0 to 28, 87 C57BL/6 J mice were exposed to one of six treatment conditions: 0 ppm Pb; 30 ppm Pb; 430 ppm Pb; 30 ppm Pb + 400 ppm apigenin; 430 ppm Pb + 400 ppm apigenin; or 400 ppm apigenin, via dams' drinking water. Following sacrifice, brain tissue was harvested and microglial cells were labeled via immunohistochemistry and counted within the dentate gyrus (DG) using unbiased stereology methods. It was hypothesized that developmental sub-chronic low-level lead exposure would increase microglial cell numbers within the DG and that apigenin treatment may mitigate these effects. A significant effect of treatment group was found and post-hoc analyses revealed that Pb-exposure generated an increased number of microglial cells as compared to controls. Interestingly, the 30 ppm Pb with apigenin treatment group did not generate microglial cell numbers different from the control group unexposed to Pb. These results suggested that developmental sub-chronic low-level lead exposure increased microglial cell activation within the DG and that, at low-levels of Pb exposure, apigenin treatment may mitigate these effects. These results provided the groundwork for studies that could identify an effective intervention to alleviate the effects of developmental chronic low-level lead exposure in child neurocognition.
Collapse
Affiliation(s)
- Gisel Flores-Montoya
- Laboratory of Neuroimmunology and Neurotoxicology, Carleton College, Psychology Department, United States of America.
| | - Zichen Tian
- Laboratory of Neuroimmunology and Neurotoxicology, Carleton College, Psychology Department, United States of America
| | - Ayasa Michii
- Laboratory of Neuroimmunology and Neurotoxicology, Carleton College, Psychology Department, United States of America
| | - Sze Ying Chan
- Laboratory of Neuroimmunology and Neurotoxicology, Carleton College, Psychology Department, United States of America
| | - Natalie Sanchez
- Laboratory of Neuroimmunology and Neurotoxicology, Carleton College, Psychology Department, United States of America
| |
Collapse
|
4
|
Wei R, Wei P, Yuan H, Yi X, Aschner M, Jiang YM, Li SJ. Inflammation in Metal-Induced Neurological Disorders and Neurodegenerative Diseases. Biol Trace Elem Res 2024; 202:4459-4481. [PMID: 38206494 DOI: 10.1007/s12011-023-04041-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024]
Abstract
Essential metals play critical roles in maintaining human health as they participate in various physiological activities. Nonetheless, both excessive accumulation and deficiency of these metals may result in neurotoxicity secondary to neuroinflammation and the activation of microglia and astrocytes. Activation of these cells can promote the release of pro-inflammatory cytokines. It is well known that neuroinflammation plays a critical role in metal-induced neurotoxicity as well as the development of neurological disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). Initially seen as a defense mechanism, persistent inflammatory responses are now considered harmful. Astrocytes and microglia are key regulators of neuroinflammation in the central nervous system, and their excessive activation may induce sustained neuroinflammation. Therefore, in this review, we aim to emphasize the important role and molecular mechanisms underlying metal-induced neurotoxicity. Our objective is to raise the awareness on metal-induced neuroinflammation in neurological disorders. However, it is not only just neuroinflammation that different metals could induce; they can also cause harm to the nervous system through oxidative stress, apoptosis, and autophagy, to name a few. The primary pathophysiological mechanism by which these metals induce neurological disorders remains to be determined. In addition, given the various pathways through which individuals are exposed to metals, it is necessary to also consider the effects of co-exposure to multiple metals on neurological disorders.
Collapse
Affiliation(s)
- Ruokun Wei
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Peiqi Wei
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Haiyan Yuan
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Xiang Yi
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Michael Aschner
- The Department of Molecular Pharmacology at Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Yue-Ming Jiang
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China.
| | - Shao-Jun Li
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China.
| |
Collapse
|
5
|
Walker KA, Rhodes ST, Liberman DA, Gore AC, Bell MR. Microglial responses to inflammatory challenge in adult rats altered by developmental exposure to polychlorinated biphenyls in a sex-specific manner. Neurotoxicology 2024; 104:95-115. [PMID: 39038526 PMCID: PMC11548868 DOI: 10.1016/j.neuro.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
Polychlorinated biphenyls are ubiquitous environmental contaminants linkedc with peripheral immune and neural dysfunction. Neuroimmune signaling is critical to brain development and later health; however, effects of PCBs on neuroimmune processes are largely undescribed. This study extends our previous work in neonatal or adolescent rats by investigating longer-term effects of perinatal PCB exposure on later neuroimmune responses to an inflammatory challenge in adulthood. Male and female Sprague-Dawley rats were exposed to a low-dose, environmentally relevant, mixture of PCBs (Aroclors 1242, 1248, and 1254, 1:1:1, 20 μg / kg dam BW per gestational day) or oil control during gestation and via lactation. Upon reaching adulthood, rats were given a mild inflammatory challenge with lipopolysaccharide (LPS, 50 μg / kg BW, ip) or saline control and then euthanized 3 hours later for gene expression analysis or 24 hours later for immunohistochemical labeling of Iba1+ microglia. PCB exposure did not alter gene expression or microglial morphology independently, but instead interacted with the LPS challenge in brain region- and sex-specific ways. In the female hypothalamus, PCB exposure blunted LPS responses of neuroimmune and neuromodulatory genes without changing microglial morphology. In the female prefrontal cortex, PCBs shifted Iba1+ cells from reactive to hyperramified morphology in response to LPS. Conversely, in the male hypothalamus, PCBs shifted cell phenotypes from hyperramified to reactive morphologies in response to LPS. The results highlight the potential for long-lasting effects of environmental contaminants that are differentially revealed over a lifetime, sometimes only after a secondary challenge. These neuroimmune endpoints are possible mechanisms for PCB effects on a range of neural dysfunction in adulthood, including mental health and neurodegenerative disorders. The findings suggest possible interactions with other environmental challenges that also influence neuroimmune systems.
Collapse
Affiliation(s)
- Katherine A Walker
- Departments of Biological Sciences and Health Sciences, DePaul University, Chicago, IL 60614, USA.
| | - Simone T Rhodes
- Departments of Biological Sciences and Health Sciences, DePaul University, Chicago, IL 60614, USA.
| | - Deborah A Liberman
- Departments of Biological Sciences and Health Sciences, DePaul University, Chicago, IL 60614, USA.
| | - Andrea C Gore
- Division of Pharmacology and Toxicology, College of Pharmacy and Department of Psychology, University of Texas at Austin, Austin, TX 78712, USA.
| | - Margaret R Bell
- Departments of Biological Sciences and Health Sciences, DePaul University, Chicago, IL 60614, USA; Division of Pharmacology and Toxicology, College of Pharmacy and Department of Psychology, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
6
|
Tang W, Peng J, Chen L, Yu C, Wang Y, Zou F, Zheng G, Meng X. Lead inhibits microglial cell migration via suppression of store-operated calcium entry. Toxicol Lett 2024; 393:69-77. [PMID: 38281554 DOI: 10.1016/j.toxlet.2024.01.011] [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: 01/24/2023] [Revised: 01/05/2024] [Accepted: 01/22/2024] [Indexed: 01/30/2024]
Abstract
Lead (Pb) is a non-biodegradable environmental pollutant that can lead to neurotoxicity by inducing neuroinflammation. Microglial activation plays a key role in neuroinflammation, and microglial migration is one of its main features. However, whether Pb affects microglial migration has not yet been elucidated. Herein, the effect of Pb on microglial migration was investigated using BV-2 microglial cells and primary microglial cells. The results showed that cell activation markers (TNF-α and CD206) in BV-2 cells were increased after Pb treatment. The migration ability of microglia was inhibited by Pb. Both store-operated calcium entry (SOCE) and the Ca2+ release-activated Ca2+ (CRAC) current were downregulated by microglia treatment with Pb in a dose-dependent manner. However, there was no statistical difference in the protein levels of stromal interaction molecule (STIM) 1, STIM2, or Ca2+ release-activated Ca2+ channel protein (Orai) 1 in microglia. The external Ca2+ influx and cell migration ability were restored to a certain extent after overexpression of either STIM1 or its CRAC activation domain in microglia. These results indicated that Pb inhibits microglial migration by downregulation of SOCE and impairment of the function of STIM1.
Collapse
Affiliation(s)
- Wei Tang
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China; Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Jiawen Peng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China; Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Lixuan Chen
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China; Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Changhui Yu
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China; Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Yuhao Wang
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China; Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Fei Zou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China; Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Gang Zheng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China; Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Xiaojing Meng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China; Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
7
|
Huang D, Chen L, Ji Q, Xiang Y, Zhou Q, Chen K, Zhang X, Zou F, Zhang X, Zhao Z, Wang T, Zheng G, Meng X. Lead aggravates Alzheimer's disease pathology via mitochondrial copper accumulation regulated by COX17. Redox Biol 2024; 69:102990. [PMID: 38091880 PMCID: PMC10716782 DOI: 10.1016/j.redox.2023.102990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/03/2023] [Accepted: 12/06/2023] [Indexed: 01/23/2024] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease that is associated with multiple environmental risk factors, including heavy metals. Lead (Pb) is a heavy metal contaminant, which is closely related to the incidence of AD. However, the research on the role of microglia in Pb-induced AD-like pathology is limited. To determine the mechanism by which Pb exposure aggravates AD progression and the role of microglial activation, we exposed APP/PS1 mice and Aβ1-42-treated BV-2 cells to Pb. Our results suggested that chronic Pb exposure exacerbated learning and memory impairments in APP/PS1 mice. Pb exposure increased the activation of microglia in the hippocampus of APP/PS1 mice, which was associated with increased deposition of Aβ1-42, and induced hippocampal neuron damage. Pb exposure upregulated copper transporter 1 (CTR1) and downregulated copper P-type ATPase transporter (ATP7A) in the hippocampus of APP/PS1 mice and Aβ1-42-treated BV-2 cells. Moreover, Pb enhanced mitochondrial translocation of the mitochondrial copper transporter COX17, leading to an increase in mitochondrial copper concentration and mitochondrial damage. This could be reversed by copper-chelating agents or by inhibiting the mitochondrial translocation of COX17. The increased mitochondrial copper concentration caused by increased mitochondrial translocation of COX17 after Pb exposure may be related to the enhanced mitochondrial import pathway of AIF/CHCHD4. These results indicate that Pb induces the activation of microglia by increasing the concentration of copper in the mitochondria of microglia, and microglia release inflammatory factors to promote neuroinflammation, thus aggravating the pathology of AD. The present study provides new ideas for the prevention of Pb-induced AD.
Collapse
Affiliation(s)
- Dingbang Huang
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Lixuan Chen
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Qiuyi Ji
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Yang Xiang
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Qin Zhou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Kaiju Chen
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoshun Zhang
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Fei Zou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Xingmei Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zaihua Zhao
- Department of Occupational and Environmental Health and the Ministry of Education's Key Laboratory of Hazard Assessment and Control in Special Operational Environment, School of Military Preventive Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Tao Wang
- Department of Occupational and Environmental Health and the Ministry of Education's Key Laboratory of Hazard Assessment and Control in Special Operational Environment, School of Military Preventive Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Gang Zheng
- Department of Occupational and Environmental Health and the Ministry of Education's Key Laboratory of Hazard Assessment and Control in Special Operational Environment, School of Military Preventive Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Xiaojing Meng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
8
|
Jiang C, Li X, Xiang C, Ye F. Pb induces the release of CXCL10 and CCL2 chemokines via mtROS/NF-κB activation in BV-2 cells. Toxicol Lett 2024; 391:62-70. [PMID: 38061439 DOI: 10.1016/j.toxlet.2023.12.001] [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: 05/28/2023] [Revised: 11/19/2023] [Accepted: 12/04/2023] [Indexed: 12/18/2023]
Abstract
Lead (Pb), a well-known environmental pollutant, could cause damage of microglia, the resident macrophages vitally regulating inflammation in brain. Previous studies have found that Pb exposure induces typical pro-inflammatory factors release, such as tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), but what effects of Pb treatment below the dose causing these factors release are unknown. Thus, cytokines assay was performed to identify the factors released from Pb-treated BV-2 cells at 2.5 μM, causing no effects on TNF-α, IL-1β, and IL-6 release and cell death. Cytokines assay identified low doses of Pb exposure mainly induce an increase in specific chemokines, including CXCL10, CCL2, and CXCL2, which were confirmed by ELISA. Subsequent assessment found Pb could damage mitochondria function and generate mitochondrial reactive oxygen species (mtROS), and Mito TEMPO, a specific inhibitor of mtROS, suppressed Pb-caused upregulation of CXCL10 and CCL2, but not CXCL2. Finally, we determined that mtROS mediated Pb-induced activation of NF-κB pathway, as Mito TEMPO treatment inhibited P-p65/p65 escalation during Pb treatment. Inhibition of NF-κB pathway by Bay11-7821 suppressed the release of CXCL10 and CCL2. Collectively, low dose of Pb induces the release of CXCL10 and CCL2 chemokines, but not TNF-α and IL-1β, via mtROS/NF-κB activation in BV-2 cells.
Collapse
Affiliation(s)
- Chenghao Jiang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xintong Li
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Cui Xiang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Fang Ye
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
9
|
Ji X, Ma Q, Wang X, Ming H, Bao G, Fu M, Wei C. Digeda-4 decoction and its disassembled prescriptions improve dyslipidemia and apoptosis by regulating AMPK/SIRT1 pathway on tyloxapol-induced nonalcoholic fatty liver disease in mice. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116827. [PMID: 37348794 DOI: 10.1016/j.jep.2023.116827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/17/2023] [Accepted: 06/19/2023] [Indexed: 06/24/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Nonalcoholic fatty liver disease (NAFLD) is a manifestation of metabolic syndrome in the liver and the leading cause of chronic liver disease worldwide. Digeda-4 decoction (DGD-4) is a commonly prescribed Mongolian herbal drug for treating acute and chronic liver injury and fatty liver. However, the mechanisms underlying the improvement of dislipidemia and liver injury via treatment with DGD-4 remain unclear. Disassembling a prescription is an effective approach to studying the effects and mechanisms underlying Mongolian medicine prescriptions. By disassembling a prescription, it is feasible to discover effective combinations of individual herbs to optimize a given prescription. Accordingly, we disassembled DGD-4 into two groups: the single Lomatogonium rotatum (L.) Fries ex Nym (LR) (DGD-1) and non-LR (DGD-3). AIM OF THIS STUDY To study whether DGD-4 and its disassembled prescriptions have protective effects against tyloxapol (TY)-induced NAFLD and to explore the underlying mechanisms of action and compatibility of prescriptions. MATERIAL AND METHODS NAFLD mice were developed by TY induction. Biochemical horizontal analyses, enzyme-linked immunosorbent assay, and liver histological staining were performed to explore the protective effects of DGD-4 and its disassembled prescriptions DGD-3 and DGD-1. Furthermore, we performed immunohistochemical analyses and Western blotting to further explore the expression of target proteins. RESULTS DGD-4 and its disassembled prescriptions could inhibit TY-induced dislipidemia and liver injury. In addition, DGD-4 and its disassembled prescriptions increased the levels of p-AMPKα and p-ACC, but decreased the levels of SREBP1c, SCD-1, SREBP-2, and HMGCS1 proteins. The activation of lipid metabolic pathways SIRT1, PGC-1α, and PPARα improved lipid accumulation in the liver. Moreover, DGD-4 could inhibit hepatocyte apoptosis and treat TY-induced liver injury by upregulating the Bcl-2 expression, downregulating the expression of Bax, caspase-3, caspase-8, and the ratio of Bax/Bcl-2, and positively regulating the imbalance of oxidative stress (OxS) markers (such as superoxide dismutase [SOD], catalase [CAT], malondialdehyde [MDA], and myeloperoxidase [MPO]). DGD-1 was superior to DGD-3 in regulating lipid synthesis-related proteins such as SREBP1c, SCD-1, SREBP-2, and HMGCS1. DGD-3 significantly affected the expression of lipid metabolic proteins SIRT1, PGC-1α, PPARα, apoptotic proteins Bcl-2, Bax, caspase-3, caspase-8, and the regulation of Bax/Bcl-2 ratio. However, DGD-1 showed no regulatory effects on Bax and Bcl-2 proteins. CONCLUSION This study demonstrates the protective effects of DGD-4 in the TY-induced NAFLD mice through a mechanism involving improvement of dyslipidemia and apoptosis by regulating the AMPK/SIRT1 pathway. Although the Monarch drug DGD-1 reduces lipid accumulation and DGD-3 inhibits apoptosis and protects the liver from injury, DGD-4 can be more effective overall as a therapy when compared to DGD-1 and DGD-3.
Collapse
Affiliation(s)
- Xiaoping Ji
- School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao, 028000, China; Institute of Pharmaceutical Chemistry and Pharmacology, Inner Mongolia Minzu University, Tongliao, 028000, China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Inner Mongolia Minzu University, Tongliao, 028000, China.
| | - Qianqian Ma
- Institute of Pharmaceutical Chemistry and Pharmacology, Inner Mongolia Minzu University, Tongliao, 028000, China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Inner Mongolia Minzu University, Tongliao, 028000, China.
| | - Xuan Wang
- Institute of Pharmaceutical Chemistry and Pharmacology, Inner Mongolia Minzu University, Tongliao, 028000, China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Inner Mongolia Minzu University, Tongliao, 028000, China.
| | - Hui Ming
- Institute of Pharmaceutical Chemistry and Pharmacology, Inner Mongolia Minzu University, Tongliao, 028000, China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Inner Mongolia Minzu University, Tongliao, 028000, China.
| | - Guihua Bao
- School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao, 028000, China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Inner Mongolia Minzu University, Tongliao, 028000, China.
| | - Minghai Fu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China.
| | - Chengxi Wei
- School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao, 028000, China; Institute of Pharmaceutical Chemistry and Pharmacology, Inner Mongolia Minzu University, Tongliao, 028000, China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Inner Mongolia Minzu University, Tongliao, 028000, China.
| |
Collapse
|
10
|
Xu Y, Peng T, Zhou Q, Zhu J, Liao G, Zou F, Meng X. Evaluation of the oxidative toxicity induced by lead, manganese, and cadmium using genetically modified nrf2a-mutant zebrafish. Comp Biochem Physiol C Toxicol Pharmacol 2023; 266:109550. [PMID: 36717045 DOI: 10.1016/j.cbpc.2023.109550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 01/03/2023] [Accepted: 01/15/2023] [Indexed: 01/30/2023]
Abstract
Heavy metal pollution has become a serious environmental concern and a threat to public health. Three of the most common heavy metals are cadmium (Cd), lead (Pb), and manganese (Mn). Nuclear factor erythroid 2-related factor 2 (Nrf2) is an important transcription factor activated in the response to oxidative stress. In this study, mutant zebrafish with an nrf2a deletion of 7 bp were constructed by the CRISPR/Cas9 system to investigate the oxidative toxicity of these three heavy metals. The results of general toxicity tests showed that Pb exposure did not cause significant damage to mutant zebrafish compared with wild-type (WT) zebrafish. However, high Mn exposure increased mortality and malformation rates in mutant zebrafish. Of concern, Cd exposure caused significant toxic damage, including increased mortality and malformation rates, apoptosis of brain neurons, and severe locomotor behavior aberration in mutant zebrafish. The results of qRT-PCR indicated that Cd exposure could induce the activation of genes related to oxidative stress resistance in WT zebrafish, while the expression of these genes was inhibited in mutant zebrafish. This study showed that of the three heavy metals, Cd had the strongest oxidative toxicity, Mn had medium toxicity, and Pb had the weakest toxicity.
Collapse
Affiliation(s)
- Yongjie Xu
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Tao Peng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Qin Zhou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiawei Zhu
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Gengze Liao
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Fei Zou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| | - Xiaojing Meng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
11
|
Comparative analysis of Acomys cahirinus and Mus musculus responses to genotoxicity, oxidative stress, and inflammation. Sci Rep 2023; 13:3989. [PMID: 36894692 PMCID: PMC9998436 DOI: 10.1038/s41598-023-31143-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
The Egyptian spiny mouse, Acomys cahirinus, is a recently described model organism for regeneration studies. It has surprising powers of regeneration with relatively fast repairing mechanisms and reduced inflammation form compared to other mammals. Although several studies have documented the exceptional capabilities of Acomys to regenerate different tissues after injury, its response to different cellular and genetic stresses is not yet investigated. Therefore, the current study aimed to investigate Acomys abilities to resist genotoxicity, oxidative stress and inflammation induced by acute and subacute treatments with lead acetate. Responses of Acomys were compared with those of the lab mouse (Mus musculus), which displays signatures of the "typical" mammalian response to various stressors. Cellular and genetic stresses were induced by using acute and subacute doses of Lead acetate (400 mg/kg and 50 mg/kg for 5 days, respectively). The assessment of genotoxicity was carried out by using comet assay, while oxidative stress was evaluated by measuring the biomarkers; MDA, GSH and antioxidant enzymes CAT and SOD. Moreover, inflammation was assessed by analyzing the expression of some inflammatory-regeneration-related genes: CXCL1, IL1-β, and Notch 2 and immunohistochemical staining of TNF-α protein in brain tissue, in addition to histopathological examination of brain, liver, and kidneys. The obtained results revealed a unique resistance potency of Acomys to genotoxicity, oxidative stress, and inflammation in certain tissues in comparison to Mus. Altogether, the results revealed an adaptive and protective response to cellular and genetic stresses in Acomys.
Collapse
|
12
|
Shvachiy L, Amaro-Leal Â, Outeiro TF, Rocha I, Geraldes V. Intermittent Lead Exposure Induces Behavioral and Cardiovascular Alterations Associated with Neuroinflammation. Cells 2023; 12:cells12050818. [PMID: 36899953 PMCID: PMC10000953 DOI: 10.3390/cells12050818] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023] Open
Abstract
The nervous system is the primary target for lead exposure and the developing brain appears to be especially susceptible, namely the hippocampus. The mechanisms of lead neurotoxicity remain unclear, but microgliosis and astrogliosis are potential candidates, leading to an inflammatory cascade and interrupting the pathways involved in hippocampal functions. Moreover, these molecular changes can be impactful as they may contribute to the pathophysiology of behavioral deficits and cardiovascular complications observed in chronic lead exposure. Nevertheless, the health effects and the underlying influence mechanism of intermittent lead exposure in the nervous and cardiovascular systems are still vague. Thus, we used a rat model of intermittent lead exposure to determine the systemic effects of lead and on microglial and astroglial activation in the hippocampal dentate gyrus throughout time. In this study, the intermittent group was exposed to lead from the fetal period until 12 weeks of age, no exposure (tap water) until 20 weeks, and a second exposure from 20 to 28 weeks of age. A control group (without lead exposure) matched in age and sex was used. At 12, 20 and 28 weeks of age, both groups were submitted to a physiological and behavioral evaluation. Behavioral tests were performed for the assessment of anxiety-like behavior and locomotor activity (open-field test), and memory (novel object recognition test). In the physiological evaluation, in an acute experiment, blood pressure, electrocardiogram, and heart and respiratory rates were recorded, and autonomic reflexes were evaluated. The expression of GFAP, Iba-1, NeuN and Synaptophysin in the hippocampal dentate gyrus was assessed. Intermittent lead exposure induced microgliosis and astrogliosis in the hippocampus of rats and changes in behavioral and cardiovascular function. We identified increases in GFAP and Iba1 markers together with presynaptic dysfunction in the hippocampus, concomitant with behavioral changes. This type of exposure produced significant long-term memory dysfunction. Regarding physiological changes, hypertension, tachypnea, baroreceptor reflex impairment and increased chemoreceptor reflex sensitivity were observed. In conclusion, the present study demonstrated the potential of lead intermittent exposure inducing reactive astrogliosis and microgliosis, along with a presynaptic loss that was accompanied by alterations of homeostatic mechanisms. This suggests that chronic neuroinflammation promoted by intermittent lead exposure since fetal period may increase the susceptibility to adverse events in individuals with pre-existing cardiovascular disease and/or in the elderly.
Collapse
Affiliation(s)
- Liana Shvachiy
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany
- Cardiovascular Centre of the University of Lisbon, 1649-028 Lisbon, Portugal
- Institute of Physiology, Faculty of Medicine of the University of Lisbon, 1649-028 Lisbon, Portugal
| | - Ângela Amaro-Leal
- Cardiovascular Centre of the University of Lisbon, 1649-028 Lisbon, Portugal
- Institute of Physiology, Faculty of Medicine of the University of Lisbon, 1649-028 Lisbon, Portugal
| | - Tiago F. Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany
- Max Planck Institute for Natural Science, 37075 Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK
- Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 37073 Göttingen, Germany
| | - Isabel Rocha
- Cardiovascular Centre of the University of Lisbon, 1649-028 Lisbon, Portugal
- Institute of Physiology, Faculty of Medicine of the University of Lisbon, 1649-028 Lisbon, Portugal
| | - Vera Geraldes
- Cardiovascular Centre of the University of Lisbon, 1649-028 Lisbon, Portugal
- Institute of Physiology, Faculty of Medicine of the University of Lisbon, 1649-028 Lisbon, Portugal
- Correspondence: ; Tel.: +351-217999435
| |
Collapse
|
13
|
Guo J, Zhang J, Liu Q, Yang N, Huang Y, Hu T, Rao C. Research progress on components and mechanisms of neurotoxicity induced by traditional Chinese medicine. J Appl Toxicol 2023; 43:338-349. [PMID: 36148542 DOI: 10.1002/jat.4396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/17/2022] [Accepted: 09/17/2022] [Indexed: 11/08/2022]
Abstract
Over the years, the safety of traditional Chinese medicine (TCM) has received widespread attention, especially the central nervous system-related adverse reactions. Indeed, the complexity of TCM has limited the widespread application of TCM. The article summarizes the main components associated with neurotoxicity, including alkaloids, terpenes, flavonoids, saponins, proteins, and heavy metals, by reviewing the literature on the neurotoxicity of TCM. It has been established that the neurotoxicity mechanisms mainly include mitochondrial damage, oxidative damage, inhibition of cell proliferation (including transcriptional and DNA damage), changes in cell membrane permeability, and apoptosis. By reviewing the latest literature, this paper provides the foothold for follow-up studies and can assist clinicians in preventing neurotoxicity via rational and safe TCM drug use.
Collapse
Affiliation(s)
- Jiafu Guo
- R&D Center for Efficiency, Safety and Application in Chinese Materia Medica With Medical and Edible Values, School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jian Zhang
- R&D Center for Efficiency, Safety and Application in Chinese Materia Medica With Medical and Edible Values, School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiuyan Liu
- R&D Center for Efficiency, Safety and Application in Chinese Materia Medica With Medical and Edible Values, School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Nannan Yang
- R&D Center for Efficiency, Safety and Application in Chinese Materia Medica With Medical and Edible Values, School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Huang
- R&D Center for Efficiency, Safety and Application in Chinese Materia Medica With Medical and Edible Values, School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tingting Hu
- R&D Center for Efficiency, Safety and Application in Chinese Materia Medica With Medical and Edible Values, School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chaolong Rao
- R&D Center for Efficiency, Safety and Application in Chinese Materia Medica With Medical and Edible Values, School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
14
|
Yen JS, Wu YC, Yen JC, Wang IK, Fu JF, Cheng CM, Yen TH. Immune Responses to COVID-19 Vaccines in Patients with Chronic Kidney Disease and Lead Exposure. Int J Mol Sci 2022; 23:15003. [PMID: 36499330 PMCID: PMC9736384 DOI: 10.3390/ijms232315003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/17/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Literature data regarding the response rate to COVID-19 vaccination in chronic kidney disease (CKD) patients remain inconclusive. Furthermore, studies have reported a relationship between lead exposure and susceptibility to viral infections. This study examined immune responses to COVID-19 vaccines in patients with CKD and lead exposure. Between October and December 2021, 50 lead-exposed CKD patients received two doses of vaccination against COVID-19 at Chang Gung Memorial Hospital. Patients were stratified into two groups based on the median blood lead level (BLL): upper (≥1.30 μg/dL, n = 24) and lower (<1.30 μg/dL, n = 26) 50th percentile. The patients were aged 65.9 ± 11.8 years. CKD stages 1, 2, 3, 4 and 5 accounted for 26.0%, 20.0%, 22.0%, 8.0% and 24.0% of the patients, respectively. Patients in the lower 50th percentile of BLL had a lower proportion of CKD stage 5 than patients in the upper 50th percentile BLL group (p = 0.047). The patients in the lower 50th percentile BLL group also received a higher proportion of messenger RNA vaccines and a lower proportion of adenovirus-vectored vaccines than the patients in the upper 50th percentile BLL group (p = 0.031). Notably, the neutralizing antibody titers were higher in the lower 50th percentile than in the upper 50th percentile BLL group. Furthermore, the circulating levels of granulocyte-colony stimulating factor, interleukin-8, monocyte chemoattractant protein-1 and macrophage inflammatory protein-1α were higher in the upper 50th percentile than in the lower 50th percentile BLL group. Therefore, it was concluded that lead-exposed CKD patients are characterized by an impaired immune response to COVID-19 vaccination with diminished neutralizing antibodies and augmented inflammatory reactions.
Collapse
Affiliation(s)
- Ju-Shao Yen
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan
| | - Yao-Cheng Wu
- Department of Nephrology, Clinical Poison Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan
| | - Ju-Ching Yen
- School of Medicine, College of Medicine, China Medical University, Taichung 406, Taiwan
| | - I-Kuan Wang
- School of Medicine, College of Medicine, China Medical University, Taichung 406, Taiwan
- Department of Nephrology, China Medical University Hospital, Taichung 404, Taiwan
| | - Jen-Fen Fu
- Department of Medical Research, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chao-Min Cheng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Tzung-Hai Yen
- Department of Nephrology, Clinical Poison Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
15
|
Majerczyk D, Ayad E, Brewton K, Saing P, Hart P. Systemic maternal inflammation promotes ASD via IL-6 and IFN-γ. Biosci Rep 2022; 42:BSR20220713. [PMID: 36300375 PMCID: PMC9670245 DOI: 10.1042/bsr20220713] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 09/30/2022] [Accepted: 10/26/2022] [Indexed: 07/25/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurological disorder that manifests during early development, impacting individuals through their ways of communicating, social behaviors, and their ability to perform day-to-day activities. There have been different proposed mechanisms on how ASD precipitates within a patient, one of which being the impact cytokines have on fetal development once a mother's immune system has been activated (referred to as maternal immune activation, MIA). The occurrence of ASD has long been associated with elevated levels of several cytokines, including interleukin-6 (IL-6) and interferon gamma (IFN-γ). These proinflammatory cytokines can achieve high systemic levels in response to immune activating pathogens from various extrinsic sources. Transfer of cytokines such as IL-6 across the placental barrier allows accumulation in the fetus, potentially inducing neuroinflammation and consequently altering neurodevelopmental processes. Individuals who have been later diagnosed with ASD have been observed to have elevated levels of IL-6 and other proinflammatory cytokines during gestation. Moreover, the outcome of MIA has been associated with neurological effects such as impaired social interaction and an increase in repetitive behavior in animal models, supporting a mechanistic link between gestational inflammation and development of ASD-like characteristics. The present review attempts to provide a concise overview of the available preclinical and clinical data that suggest cross-talk between IL-6 and IFN-γ through both extrinsic and intrinsic factors as a central mechanism of MIA that may promote the development of ASD.
Collapse
Affiliation(s)
- Daniel Majerczyk
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
- Loyola Medicine, Berwyn, Illinois 60402, U.S.A
| | - Elizabeth G. Ayad
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| | - Kari L. Brewton
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| | - Pichrasmei Saing
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| | - Peter C. Hart
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| |
Collapse
|
16
|
Islam F, Shohag S, Akhter S, Islam MR, Sultana S, Mitra S, Chandran D, Khandaker MU, Ashraf GM, Idris AM, Emran TB, Cavalu S. Exposure of metal toxicity in Alzheimer's disease: An extensive review. Front Pharmacol 2022; 13:903099. [PMID: 36105221 PMCID: PMC9465172 DOI: 10.3389/fphar.2022.903099] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Metals serve important roles in the human body, including the maintenance of cell structure and the regulation of gene expression, the antioxidant response, and neurotransmission. High metal uptake in the nervous system is harmful because it can cause oxidative stress, disrupt mitochondrial function, and impair the activity of various enzymes. Metal accumulation can cause lifelong deterioration, including severe neurological problems. There is a strong association between accidental metal exposure and various neurodegenerative disorders, including Alzheimer's disease (AD), the most common form of dementia that causes degeneration in the aged. Chronic exposure to various metals is a well-known environmental risk factor that has become more widespread due to the rapid pace at which human activities are releasing large amounts of metals into the environment. Consequently, humans are exposed to both biometals and heavy metals, affecting metal homeostasis at molecular and biological levels. This review highlights how these metals affect brain physiology and immunity and their roles in creating harmful proteins such as β-amyloid and tau in AD. In addition, we address findings that confirm the disruption of immune-related pathways as a significant toxicity mechanism through which metals may contribute to AD.
Collapse
Affiliation(s)
- Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Sheikh Shohag
- Department of Genetic Engineering and Biotechnology, Faculty of Earth and Ocean Science, Bangabandhu Sheikh Mujibur Rahman Maritime University, Dhaka, Bangladesh
| | - Shomaya Akhter
- Department of Genetic Engineering and Biotechnology, Faculty of Earth and Ocean Science, Bangabandhu Sheikh Mujibur Rahman Maritime University, Dhaka, Bangladesh
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Sharifa Sultana
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Deepak Chandran
- Department of Veterinary Sciences and Animal Husbandry, Amrita School of Agricultural Sciences, Amrita Vishwa Vidyapeetham University, Coimbatore, India
| | - Mayeen Uddin Khandaker
- Centre for Applied Physics and Radiation Technologies, School of Engineering and Technology, Sunway University, Subang Jaya, Malaysia
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, 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
| | - Abubakr M. Idris
- Department of Chemistry, College of Science, King Khalid University, Abha, Saudi Arabia
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
17
|
Zhou CC, Wang XJ, Li ZC, Lu WJ, Zhang YT, Shen FM, Li DJ. Lead Exposure in Developmental Ages Promotes Aβ Accumulation by Disturbing Aβ Transportation in Blood-Cerebrospinal Fluid Barrier/Blood-Brain Barriers and Impairing Aβ Clearance in the Liver. Biol Trace Elem Res 2022; 200:3702-3711. [PMID: 34787833 DOI: 10.1007/s12011-021-02969-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/12/2021] [Indexed: 12/24/2022]
Abstract
Environmental lead exposure is closely related to the progression of Alzheimer's disease (AD). Our previous study has shown that exposure to lead could result in the cholesterol unbalance and increase amyloid-beta (Aβ) generation in the brain. However, the potential effect of lead exposure on Aβ transportation is poorly reported. In this study, we sought to explore whether lead exposure in developmental ages impaired the integrity of BCSFB and BBB, two highly vascularized structures in the brain in a rat model. The Aβ clearance in the liver was also assessed. Our results showed that lead treatment in developmental ages increased the number of TUNEL-positive apoptotic cells in rat choroid plexus and microvessels. Moreover, lead exposure markedly increased pro-inflammatory factors expression including TNF-α and IL-1β in rat choroid plexus and microvessels. Interestingly, lead treatment increased the expression of AQP-1 and reduced the expression of TTR, two key proteins associated with the functions of choroid plexus and microvessels. Additionally, the expressions of ABCB1, LRP-1, and RAGE, three major receptors responsible for Aβ transportation, were disturbed by developmental lead exposure. All these pathologies resulted in Aβ1-40 deposition within BCSFB and BBB and malfunctions of these two vascularized structures. Finally, we found that lead treatment remarkably inhibited the gene expression of LRP-1, which is responsible for Aβ endocytosis, in the liver tissue of the rat model. Collectively, our results provide the first evidence that developmental lead exposure induces Aβ deposition in BCSFB and BBB and impairs Aβ clearance in the liver, which would ultimately disturb Aβ transportation via choroid plexus/brain microvessels and facilitate Aβ deposition in the brain.
Collapse
Affiliation(s)
- Can-Can Zhou
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Xu-Jie Wang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Zi-Chen Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Wen-Jie Lu
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yun-Ting Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Fu-Ming Shen
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| |
Collapse
|
18
|
Olude MA, Mouihate A, Mustapha OA, Farina C, Quintana FJ, Olopade JO. Astrocytes and Microglia in Stress-Induced Neuroinflammation: The African Perspective. Front Immunol 2022; 13:795089. [PMID: 35707531 PMCID: PMC9190229 DOI: 10.3389/fimmu.2022.795089] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Africa is laden with a youthful population, vast mineral resources and rich fauna. However, decades of unfortunate historical, sociocultural and leadership challenges make the continent a hotspot for poverty, indoor and outdoor pollutants with attendant stress factors such as violence, malnutrition, infectious outbreaks and psychological perturbations. The burden of these stressors initiate neuroinflammatory responses but the pattern and mechanisms of glial activation in these scenarios are yet to be properly elucidated. Africa is therefore most vulnerable to neurological stressors when placed against a backdrop of demographics that favor explosive childbearing, a vast population of unemployed youths making up a projected 42% of global youth population by 2030, repressive sociocultural policies towards women, poor access to healthcare, malnutrition, rapid urbanization, climate change and pollution. Early life stress, whether physical or psychological, induces neuroinflammatory response in developing nervous system and consequently leads to the emergence of mental health problems during adulthood. Brain inflammatory response is driven largely by inflammatory mediators released by glial cells; namely astrocytes and microglia. These inflammatory mediators alter the developmental trajectory of fetal and neonatal brain and results in long-lasting maladaptive behaviors and cognitive deficits. This review seeks to highlight the patterns and mechanisms of stressors such as poverty, developmental stress, environmental pollutions as well as malnutrition stress on astrocytes and microglia in neuroinflammation within the African context.
Collapse
Affiliation(s)
- Matthew Ayokunle Olude
- Vertebrate Morphology, Environmental Toxicology and Neuroscience Unit, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Nigeria
- *Correspondence: Matthew Ayokunle Olude,
| | - Abdeslam Mouihate
- Department of Physiology, Faculty of Medicine, Health Sciences Centre, Kuwait University, Kuwait City, Kuwait
| | - Oluwaseun Ahmed Mustapha
- Vertebrate Morphology, Environmental Toxicology and Neuroscience Unit, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Nigeria
| | - Cinthia Farina
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS) San Raffaele Scientific Institute, Institute of Experimental Neurology (INSPE) and Division of Neuroscience, Milan, Italy
| | - Francisco Javier Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - James Olukayode Olopade
- Neuroscience Unit, Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
19
|
Nabi M, Tabassum N. Role of Environmental Toxicants on Neurodegenerative Disorders. FRONTIERS IN TOXICOLOGY 2022; 4:837579. [PMID: 35647576 PMCID: PMC9131020 DOI: 10.3389/ftox.2022.837579] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/22/2022] [Indexed: 12/22/2022] Open
Abstract
Neurodegeneration leads to the loss of structural and functioning components of neurons over time. Various studies have related neurodegeneration to a number of degenerative disorders. Neurological repercussions of neurodegeneration can have severe impacts on the physical and mental health of patients. In the recent past, various neurodegenerative ailments such as Alzheimer’s and Parkinson’s illnesses have received global consideration owing to their global occurrence. Environmental attributes have been regarded as the main contributors to neural dysfunction-related disorders. The majority of neurological diseases are mainly related to prenatal and postnatal exposure to industrially produced environmental toxins. Some neurotoxic metals, like lead (Pb), aluminium (Al), Mercury (Hg), manganese (Mn), cadmium (Cd), and arsenic (As), and also pesticides and metal-based nanoparticles, have been implicated in Parkinson’s and Alzheimer’s disease. The contaminants are known for their ability to produce senile or amyloid plaques and neurofibrillary tangles (NFTs), which are the key features of these neurological dysfunctions. Besides, solvent exposure is also a significant contributor to neurological diseases. This study recapitulates the role of environmental neurotoxins on neurodegeneration with special emphasis on major neurodegenerative disorders such as Alzheimer’s and Parkinson’s disease.
Collapse
Affiliation(s)
- Masarat Nabi
- Department of Environmental Science, University of Kashmir, Srinagar, India
- *Correspondence: Masarat Nabi, , orcid.org/0000-0003-1677-6498; Nahida Tabassum,
| | - Nahida Tabassum
- Department of Pharmaceutical Sciences, University of Kashmir, Srinagar, India
- *Correspondence: Masarat Nabi, , orcid.org/0000-0003-1677-6498; Nahida Tabassum,
| |
Collapse
|
20
|
Yan L, Yang J, Yu M, Sun W, Han Y, Lu X, Jin C, Wu S, Cai Y. Lanthanum Impairs Learning and Memory by Activating Microglia in the Hippocampus of Mice. Biol Trace Elem Res 2022; 200:1640-1649. [PMID: 35178682 DOI: 10.1007/s12011-021-02637-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/10/2021] [Indexed: 12/20/2022]
Abstract
Lanthanum can induce neurotoxicity and impair cognitive function; therefore, research on the mechanism by which the ability to learning and memory is decreased by lanthanum is vitally important for protecting health. Microglia are a type of neuroglia located throughout the brain and spinal cord that play an important role in the central nervous system. When overactive, these cells can cause the excessive production of inflammatory cytokines that can damage neighboring neurons. The purpose of this study was to explore the effect of lanthanum in the form of lanthanum chloride (LaCl3) on learning and the memory of mice and determine whether there is a relationship between hippocampal neurons or learning and memory damage and excessive production of inflammatory cytokines. Four groups of pregnant Chinese Kun Ming mice were exposed to 0, 18, 36, or 72 mM LaCl3 in their drinking water during lactation. The offspring were then exposed to LaCl3 in the breast milk at birth until weaning and then exposed to these concentrations in their drinking water for 2 months after weaning. The results showed that LaCl3 impaired learning and memory in mice and injured their neurons, activated the microglia, and significantly overregulated the mRNA and protein expression of tumor necrosis factor alpha, interleukin (IL)-1β, IL-6, monocyte chemoattractant protein-1, and nitric oxide in the hippocampus. The results of this study suggest that lanthanum can impair learning and memory in mice, possibly by over-activating the microglia.
Collapse
Affiliation(s)
- Licheng Yan
- Department of Toxicology, School of Public Health, China Medical University, No.77 Puhe road, Shenyang North New Area, Shenyang, 110122, Liaoning province, People's Republic of China
- Department of Toxicology, School of Public Health, North China University of Science and Technology, No.21 Bohai road, Caofeidian New Area, Tangshan, 063210, Hebei province, People's Republic of China
| | - Jinghua Yang
- Department of Toxicology, School of Public Health, China Medical University, No.77 Puhe road, Shenyang North New Area, Shenyang, 110122, Liaoning province, People's Republic of China
| | - Miao Yu
- Department of Toxicology, School of Public Health, China Medical University, No.77 Puhe road, Shenyang North New Area, Shenyang, 110122, Liaoning province, People's Republic of China
| | - Wenchang Sun
- Department of Toxicology, School of Public Health, China Medical University, No.77 Puhe road, Shenyang North New Area, Shenyang, 110122, Liaoning province, People's Republic of China
| | - Yarao Han
- Department of Toxicology, School of Public Health, China Medical University, No.77 Puhe road, Shenyang North New Area, Shenyang, 110122, Liaoning province, People's Republic of China
| | - Xiaobo Lu
- Department of Toxicology, School of Public Health, China Medical University, No.77 Puhe road, Shenyang North New Area, Shenyang, 110122, Liaoning province, People's Republic of China
| | - Cuihong Jin
- Department of Toxicology, School of Public Health, China Medical University, No.77 Puhe road, Shenyang North New Area, Shenyang, 110122, Liaoning province, People's Republic of China
| | - Shengwen Wu
- Department of Toxicology, School of Public Health, China Medical University, No.77 Puhe road, Shenyang North New Area, Shenyang, 110122, Liaoning province, People's Republic of China
| | - Yuan Cai
- Department of Toxicology, School of Public Health, China Medical University, No.77 Puhe road, Shenyang North New Area, Shenyang, 110122, Liaoning province, People's Republic of China.
| |
Collapse
|
21
|
Liu J, Xu Y, Liao G, Tu H, Huang Y, Peng T, Chen X, Huang Z, Zhang Y, Meng X, Zou F. The role of ambra1 in Pb-induced developmental neurotoxicity in zebrafish. Biochem Biophys Res Commun 2022; 594:139-145. [PMID: 35085890 DOI: 10.1016/j.bbrc.2021.12.084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 12/21/2022]
Abstract
Lead is a highly toxic metal that displays developmental neurotoxicity. Ambra1 plays a crucial role in embryonic neural development. At present, the role of Ambra1 in lead-induced developmental neurotoxicity remains unknown. In this study, we investigated the mechanism of Ambra1 concerning its role in lead-induced neurotoxicity. Zebrafish (Danio rerio) embryos were exposed to 0.1, 1, or 10 μM Pb until 5 days post-fertilization, and their locomotor activity was significantly impaired by the 10 μM treatment. Meanwhile, Pb reduced the expression of ambra1a and ambra1b in the brain at 48 and 72 h post-fertilization. Overexpression of ambra1a or ambra1b reversed Pb-induced alterations in locomotor activity, and decreased the apoptotic cell numbers in the brains of Pb-treated zebrafish. Our data reveal a novel protective role of Ambra1 against Pb-induced neural damage in the developing zebrafish.
Collapse
Affiliation(s)
- Jiaxian Liu
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Yongjie Xu
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Gengze Liao
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongwei Tu
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying Huang
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Tao Peng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaohui Chen
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Guangdong Higher Education Institutes, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhibin Huang
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Guangdong Higher Education Institutes, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yiyue Zhang
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Guangdong Higher Education Institutes, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaojing Meng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| | - Fei Zou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
22
|
孙 芳, 韦 伟. [Effect of GnRH agonist combined with menstrual blood-derived stem cell transplantation on ovarian function in mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:1850-1856. [PMID: 35012918 PMCID: PMC8752426 DOI: 10.12122/j.issn.1673-4254.2021.12.14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To investigate the effect of combined treatment with gonadotropin-releasing hormone (GnRH) agonist and menstrual blood-derived stem cell (MenSC) transplantation on ovarian function in mice. METHODS We tested the effect of GnRH agonist and MenSC transplantation, either alone or in combination, in a mouse model of premature ovarian failure (POF) induced by daily intraperitoneal injection of 120 mg/kg cyclophosphamide (CTX) for 15 days. Venirelin was administered at the daily dose of 0.1 mg via intravenous infusion, starting at 14 days before modeling; MenSC suspension (2× 106 cells) was injected into the ovarium 1 day after modeling. HE staining was used to observe ovarian pathologies of the mice. ELISA was used to detect serum levels of LH, FSH, AMH and E2. The expression levels of ki-67, Bcl-2, BAX, caspase 9, caspase 3, their cleavages, P-PI3K, PI3K, P-Akt and Akt proteins in ovarian granulosa cells were determined with Western blotting. RESULTS The mouse models of POF showed obvious ovarian fibrosis with increased atresia follicles and had significantly elevated serum levels of LH and FSH (P < 0.01), decreased serum levels of AMH and E2 (P < 0.01), significantly increased ovarian expression levels of caspase 9, caspase 3, and their cleavages (P < 0.01), and decreased expression levels of Ki-67, Bcl-2, P-PI3K and P-Akt (P < 0.01). The combined treatment with GnRH agonist and MenSC transplantation obviously inhibited the production of ovarian atresia follicles, lowered serum levels of LH and FSH (P < 0.01), and increased the levels of AMH and E2 (P < 0.05 or 0.01) in the mouse models. The combined treatment also inhibited the expressions of BAX and cleaved caspase 9 and caspase 3 proteins (P < 0.05 or 0.01) and enhanced the expressions of Ki-67, Bcl-2, P-PI3K, and p-Akt proteins (P < 0.05 or 0.01) in ovarian granulococcus cells. CONCLUSION In mouse models of POF, GnRH agonist combined with MenSC transplantation can reduce the generation of ovarian atresia and improve ovarian reserve by up-regulating PI3K-Akt signaling pathway to achieve ovarian repair and protection.
Collapse
Affiliation(s)
- 芳 孙
- 绍兴第二医院医共体总院药剂科,浙江 绍兴 312000Department of Pharmacy, General Hospital of Shaoxing Second Hospital, Shaoxing 312000, China
| | - 伟 韦
- 浙江大学医学院附属邵逸夫医院妇产科,浙江 杭州 310000Department of Obstetrics and Gynecology, Run Run Shaw Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou 310000, China
| |
Collapse
|
23
|
Wu L, Li S, Pang S, Zhang B, Wang J, He B, Lv L, Wang W, Zhao N, Zhang Y. Effects of lead exposure on the activation of microglia in mice fed with high-fat diets. ENVIRONMENTAL TOXICOLOGY 2021; 36:1923-1931. [PMID: 34156151 DOI: 10.1002/tox.23312] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/05/2021] [Accepted: 06/13/2021] [Indexed: 06/13/2023]
Abstract
Lead (Pb) exposure can cause central nervous system (CNS) damage. The process of Pb neurotoxicity is accompanied by the microglia activation. In addition, microglia activation was observed under the intervention of high-fat diets (HFD). This study was designed to investigate the effect of Pb on the cognitive function of mice with HFD, with focus on the microglia activation in brain. Male C57BL/6J mice, 8 weeks of age, were randomly divided into control, HFD, Pb, and HFD + Pb groups. The results showed that HFD following Pb exposure could exacerbate the learning and memory impairment in mice. Pb exposure could promote microglia activation and increase the expression of M1 microglia marker and decrease the expression of M2 microglia marker in the hippocampus of mice with HFD. Our finding suggested that Pb exposure may aggravate CNS damage by promoting M1 polarization and inhibiting M2 polarization of hippocampal microglia in HFD mice.
Collapse
Affiliation(s)
- Lei Wu
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| | - Shuang Li
- Experiment Animal Center, North China University of Science and Technology, Tangshan, Hebei, China
| | - Shulan Pang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| | - Bo Zhang
- JiTang college of North China University of Science and Technology, Tangshan, Hebei, China
| | - Jierui Wang
- Rheumatology Department, Kailuan General Hospital, Tangshan, Hebei, China
| | - Bin He
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| | - Linyi Lv
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| | - Weixuan Wang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| | - Na Zhao
- Experiment Animal Center, North China University of Science and Technology, Tangshan, Hebei, China
| | - Yanshu Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
- Experiment Animal Center, North China University of Science and Technology, Tangshan, Hebei, China
| |
Collapse
|
24
|
Gadde R, Betharia S. N,N'bis-(2-mercaptoethyl) isophthalamide (NBMI) exerts neuroprotection against lead-induced toxicity in U-87 MG cells. Arch Toxicol 2021; 95:2643-2657. [PMID: 34165617 DOI: 10.1007/s00204-021-03103-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/15/2021] [Indexed: 11/30/2022]
Abstract
N,N'-bis(2-mercaptoethyl)isophthalamide (NBMI) is a novel lipophilic heavy metal chelator and thiol redox antioxidant. This study was designed to investigate the neuroprotective activity of NBMI in U-87 MG cells exposed to lead acetate (PbAc). Cells were pretreated with NBMI for 24 h prior to a 48 h exposure to PbAc. Cell death (55%, p < 0.0001) and reduction of intracellular GSH levels (0.70-fold, p < 0.005) induced by 250 µM Pb were successfully attenuated by NBMI pretreatment at concentrations as low as 10 µM. A similar pretreatment with the FDA-approved Pb chelator dimercaptosuccinic acid (DMSA) proved ineffective, indicating a superior PKPD profile for NBMI. Pretreatment with NBMI successfully counteracted Pb-induced neuroinflammation by reducing IL-1β (0.59-fold, p < 0.05) and GFAP expression levels. NBMI alone was also found to significantly increase ferroportin expression (1.97-fold, p < 0.05) thereby enhancing cellular ability to efflux heavy metals. While no response was observed on the apoptotic pathway, this study demonstrated for the first time that necrotic cell death induced by Pb in U-87 MG cells is successfully attenuated by NBMI. Collectively these data demonstrate NBMI to be a promising neuroprotective compound in the realm of Pb poisoning.
Collapse
Affiliation(s)
- Rajitha Gadde
- Department of Pharmaceutical Sciences, School of Pharmacy, MCPHS University, 179 Longwood Avenue, Boston, MA, 02115, USA.
| | - Swati Betharia
- Department of Pharmaceutical Sciences, School of Pharmacy, MCPHS University, 179 Longwood Avenue, Boston, MA, 02115, USA
| |
Collapse
|
25
|
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
|
26
|
Signal transduction associated with lead-induced neurological disorders: A review. Food Chem Toxicol 2021; 150:112063. [PMID: 33596455 DOI: 10.1016/j.fct.2021.112063] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/28/2022]
Abstract
Lead is a heavy metal pollutant that is widely present in the environment. It affects every organ system, yet the nervous system appears to be the most sensitive and primary target. Although many countries have made significant strides in controlling Pb pollution, Pb poisoning continuous to be a major public health concern. Exposure to Pb causes neurotoxicity that ranges from neurodevelopmental disorders to severe neurodegenerative lesions, leading to impairments in learning, memory, and cognitive function. Studies on the mechanisms of Pb-induced nervous system injury have convincingly shown that this metal can affect a plethora of cellular pathways affecting on cell survival, altering calcium dyshomeostasis, and inducing apoptosis, inflammation, energy metabolism disorders, oxidative stress, autophagy and glial stress. This review summarizes recent knowledge on multiple signaling pathways associated with Pb-induced neurological disorders in vivo and in vitro.
Collapse
|
27
|
Clark I, Vissel B. Broader Insights into Understanding Tumor Necrosis Factor and Neurodegenerative Disease Pathogenesis Infer New Therapeutic Approaches. J Alzheimers Dis 2021; 79:931-948. [PMID: 33459706 PMCID: PMC7990436 DOI: 10.3233/jad-201186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2020] [Indexed: 12/12/2022]
Abstract
Proinflammatory cytokines such as tumor necrosis factor (TNF), with its now appreciated key roles in neurophysiology as well as neuropathophysiology, are sufficiently well-documented to be useful tools for enquiry into the natural history of neurodegenerative diseases. We review the broader literature on TNF to rationalize why abruptly-acquired neurodegenerative states do not exhibit the remorseless clinical progression seen in those states with gradual onsets. We propose that the three typically non-worsening neurodegenerative syndromes, post-stroke, post-traumatic brain injury (TBI), and post cardiac arrest, usually become and remain static because of excess cerebral TNF induced by the initial dramatic peak keeping microglia chronically activated through an autocrine loop of microglial activation through excess cerebral TNF. The existence of this autocrine loop rationalizes post-damage repair with perispinal etanercept and proposes a treatment for cerebral aspects of COVID-19 chronicity. Another insufficiently considered aspect of cerebral proinflammatory cytokines is the fitness of the endogenous cerebral anti-TNF system provided by norepinephrine (NE), generated and distributed throughout the brain from the locus coeruleus (LC). We propose that an intact LC, and therefore an intact NE-mediated endogenous anti-cerebral TNF system, plus the DAMP (damage or danger-associated molecular pattern) input having diminished, is what allows post-stroke, post-TBI, and post cardiac arrest patients a strong long-term survival advantage over Alzheimer's disease and Parkinson's disease sufferers. In contrast, Alzheimer's disease and Parkinson's disease patients remorselessly worsen, being handicapped by sustained, accumulating, DAMP and PAMP (pathogen-associated molecular patterns) input, as well as loss of the LC-origin, NE-mediated, endogenous anti-cerebral TNF system. Adrenergic receptor agonists may counter this.
Collapse
Affiliation(s)
- I.A. Clark
- Research School of Biology, Australian National University, Canberra, Australia
| | - B. Vissel
- Centre for Neuroscience and Regenerative Medicine, Faculty of Science, University of Technology, Sydney, Australia
- St. Vincent’s Centre for Applied Medical Research, Sydney, Australia
| |
Collapse
|
28
|
Sabogal-Guáqueta AM, Marmolejo-Garza A, de Pádua VP, Eggen B, Boddeke E, Dolga AM. Microglia alterations in neurodegenerative diseases and their modeling with human induced pluripotent stem cell and other platforms. Prog Neurobiol 2020; 190:101805. [PMID: 32335273 DOI: 10.1016/j.pneurobio.2020.101805] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/16/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022]
Abstract
Microglia are the main innate immune cells of the central nervous system (CNS). Unlike neurons and glial cells, which derive from ectoderm, microglia migrate early during embryo development from the yolk-sac, a mesodermal-derived structure. Microglia regulate synaptic pruning during development and induce or modulate inflammation during aging and chronic diseases. Microglia are sensitive to brain injuries and threats, altering their phenotype and function to adopt a so-called immune-activated state in response to any perceived threat to the CNS integrity. Here, we present a short overview on the role of microglia in human neurodegenerative diseases and provide an update on the current model systems to study microglia, including cell lines, iPSC-derived microglia with an emphasis in their transcriptomic profile and integration into 3D brain organoids. We present various strategies to model and study their role in neurodegeneration providing a relevant platform for the development of novel and more effective therapies.
Collapse
Affiliation(s)
- Angélica María Sabogal-Guáqueta
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, Behavioral and Cognitive Neurosciences (BCN), University of Groningen, Groningen, The Netherlands; Department of Biomedical Sciences of Cells & Systems, section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Neuroscience Group of Antioquia, Cellular and Molecular Neurobiology Area-School of Medicine, SIU, University of Antioquia, Medellín, Colombia
| | - Alejandro Marmolejo-Garza
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, Behavioral and Cognitive Neurosciences (BCN), University of Groningen, Groningen, The Netherlands; Department of Biomedical Sciences of Cells & Systems, section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vítor Passos de Pádua
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, Behavioral and Cognitive Neurosciences (BCN), University of Groningen, Groningen, The Netherlands; Neurology Department, Medical School, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Bart Eggen
- Department of Biomedical Sciences of Cells & Systems, section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Erik Boddeke
- Department of Biomedical Sciences of Cells & Systems, section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Amalia M Dolga
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, Behavioral and Cognitive Neurosciences (BCN), University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
29
|
Malik A, Ashraf MAB, Khan MW, Zahid A, Shafique H, Waquar S, Gan SH, Ashraf M. Implication of Physiological and Biochemical Variables of Prognostic Importance in Lead Exposed Subjects. ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2020; 78:329-336. [PMID: 31620805 DOI: 10.1007/s00244-019-00673-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 10/09/2019] [Indexed: 06/10/2023]
Abstract
The use of leaded gasoline adversely affects cardiovascular, nervous, and immune systems. Study projects to rule out different variables of prognostic importance in lead-exposed subjects. A total of 317 traffic wardens with 5 years of outdoor experience and Hb levels < 10 µg/dl, and 100 traffic wardens with indoor duties were substituted in two groups. Levels of vitamins, cytokines, lead, iron, minerals, oxidative stress, and lipid peroxidation were estimated with help of their standard ELISA and spectrophotometric methods respectively. The present study show increased levels of lead in subjects (29.8 ± 3.8 vs. 1.5 ± 0.2 µg/dl) that may be involved in increasing oxidative stress, i.e., levels of malondialdehyde (MDA), 4-hydroxynonenal (4-HNE), and isoprostanes were increased in subjects (4.6 ± 0.5, 4.3 ± 0.6 and 37.2 ± 5.1). Moreover, levels of antioxidants, i.e., superoxide dismutase (SOD), glutathione (GSH), and catalase (CAT), were decreased. It also exhibits reduced levels of different enzymes in anemic traffic wardens. Current study concludes that wardens exposed to environmental lead are more susceptible to develop cardiovascular and neurological disorders. It shows that toxicity of lead maybe responsible for redox imbalance and production of proinflammatory cytokines. Thus, early detection of these biomarkers may help to reduce lead toxicity and it also may help to control the dilemma of uncontrolled environmental pollution by implicating strict actions against substandard gasoline.
Collapse
Affiliation(s)
- Arif Malik
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | | | | | - Ayesha Zahid
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Hassan Shafique
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Sulayman Waquar
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Siew Hua Gan
- School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Muhammad Ashraf
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan.
| |
Collapse
|
30
|
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: 39] [Impact Index Per Article: 7.8] [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
|
31
|
Flores-Montoya MG, Bill CA, Vines CM, Sobin C. Early chronic low-level lead exposure reduced C-C chemokine receptor 7 in hippocampal microglia. Toxicol Lett 2019; 314:106-116. [PMID: 31306743 PMCID: PMC7815484 DOI: 10.1016/j.toxlet.2019.07.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 06/11/2019] [Accepted: 07/06/2019] [Indexed: 01/10/2023]
Abstract
Chronic low-level lead exposure alters cognitive function in young children however the mechanisms mediating these deficits in the brain are not known. Previous studies in our laboratory showed that early lead exposure reduced the number of microglial cells in hippocampus/dentate gyrus of C57BL/6 J mice. In the current study, C-C chemokine receptor 7 (CCR7) and major histocompatibility complex II (MHC II) were examined to investigate whether these neuroimmune factors which are known to trigger cell migration and antigen presentation, were altered by early chronic lead exposure. Thirty-six C57BL/6 J male mice were exposed to 0 ppm (controls, n = 12), 30 ppm (low-dose, n = 12), or 430 ppm (higher-dose, n = 12) of lead acetate via dams' milk from postnatal day (PND) 0 to 28. Flow cytometry was used to quantify cell types and cell surface expression of MHC II and CCR7 in hippocampal and whole brain microglia. Non-parametric independent samples median tests were used to test for statistically significant differences between groups. As compared to controls, CCR7 in hippocampal microglia was decreased in the low-dose group, measured as geometric mean fluorescence intensity (GMFI); in the higher-dose group CCR7+MHC II- hippocampal microglia were decreased. Further analyses revealed that the higher-dose group had decreased percentage of CCR7+MHC II- hippocampal macrophages as compared to controls but increased MHC II levels in CCR7+MHC II+ hippocampal macrophages as compared to controls. It was also noted that lead exposure disrupted the balance of MHC II and/or CCR7 in lead exposed animals. Reduced CCR7 in hippocampal microglia might alter the neuroimmune environment in hippocampi of lead exposed animals. Additional studies are needed to test this possibility.
Collapse
Affiliation(s)
- Mayra Gisel Flores-Montoya
- Dept of Psychology, The University of Texas at El Paso, El Paso, TX, USA; Dept of Psychology, Carleton College, Northfield, MN, USA.
| | - Colin A Bill
- Dept of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - Charlotte M Vines
- Dept of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - Christina Sobin
- Dept of Public Health Sciences, The University of Texas at El Paso, El Paso, TX, USA; Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| |
Collapse
|
32
|
Jin M, Ji X, Zhang B, Sheng W, Wang R, Liu K. Synergistic effects of Pb and repeated heat pulse on developmental neurotoxicity in zebrafish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 172:460-470. [PMID: 30738228 DOI: 10.1016/j.ecoenv.2019.01.104] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/27/2019] [Accepted: 01/29/2019] [Indexed: 06/09/2023]
Abstract
Pollutant discharges to the aquatic environment often contain multiple environmental stressors, affecting aquatic organisms. To mimic the discharges from nuclear and industry facilities, the combined effects of two independent types of stressors, heavy metal Pb and repeated heat pulse were addressed in this study. We investigated the developmental toxicity of combined treatment, especially its toxic effects on zebrafish neurodevelopment. The normal embryos at 4 hpf were exposed to 0.2 mM of Pb dissolved in the bathing medium with different temperatures (30, 32, and 34 °C) and then maintained in an incubator at 28 °C. After performing above treatment once every 24 h for 6 days, we found that combined treatment significantly affected neural development, including loss of dopaminergic (DA) neurons and brain vasculature, disruption of locomotor activity and neurodevelopmental genes expression in a temperature-dependent manner as compared to the Pb alone exposure group, indicating that repeated heat pulse enhances these negative impacts induced by Pb. In contrast, no apparent toxicity was observed in repeated heat pulse alone groups, suggesting that Pb treatment reduces thermal tolerance in zebrafish, which emphasized the importance to evaluate synergistic effects of Pb and repeated heat pulse. Moreover, repeated heat pulse aggravated Pb-induced apoptosis in the zebrafish brain. Further study of the underlying mechanism suggested that Caspase 3 regulated apoptosis was involved in this process. Taken together, our findings shed light on the full understanding of toxic effects of discharges from industrial applications on living organisms and its environmental impact.
Collapse
Affiliation(s)
- Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Jinan 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Jinan 250103, Shandong Province, PR China.
| | - Xiuna Ji
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Jinan 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Jinan 250103, Shandong Province, PR China
| | - Baoyue Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Jinan 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Jinan 250103, Shandong Province, PR China
| | - Wenlong Sheng
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Jinan 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Jinan 250103, Shandong Province, PR China
| | - Rongchun Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Jinan 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Jinan 250103, Shandong Province, PR China
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Jinan 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Jinan 250103, Shandong Province, PR China.
| |
Collapse
|
33
|
Rahman A, Al-Qenaie S, Rao MS, Khan KM, Guillemin GJ. Memantine Is Protective against Cytotoxicity Caused by Lead and Quinolinic Acid in Cultured Rat Embryonic Hippocampal Cells. Chem Res Toxicol 2019; 32:1134-1143. [PMID: 30950269 DOI: 10.1021/acs.chemrestox.8b00421] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Quinolinic acid (QA) is an excitotoxic metabolite of the kynurenine pathway of tryptophan metabolism produced in response to inflammation and oxidative stress. Lead (Pb) causes oxidative stress and thus may produce neurotoxicity by increasing QA production. We investigated the in vitro cytotoxic effects of Pb and QA and the protective effects of the NMDA receptor antagonist memantine. Primary cultures of embryonic hippocampal cells from Wistar rats were treated with different concentrations of Pb, QA, and Pb + QA with and without memantine. Cell viability was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). Apoptosis was analyzed by flow cytometry after Annexin-V/propidium iodide staining. The numbers of immunostained neurons (with β3-Tubulin; Tuj1) and astrocytes (with glial fibrillary acidic protein) were counted. Pb at 20 μg/dL (0.97 μM) and QA at 500 nM concentrations showed significant cytotoxic effects, as evidenced by decreased cell viability, increased apoptosis, and a decrease in the number of both astrocytes and neurons. The combination of Pb and QA showed significant synergistic apoptotic effects at lower doses. Memantine (500 nM) was largely protective against the cytotoxic effects of both Pb and QA, suggesting that Pb's and QA's cytotoxicity involves NMDA receptor activation. Whereas the neuroprotection by memantine from QA-induced toxicity has been previously reported, this is the first study reporting the protection by memantine against Pb-induced cytotoxicity in cultured hippocampal cells. Protection by memantine against these neurotoxicants in vivo needs to be investigated.
Collapse
Affiliation(s)
- Abdur Rahman
- Department of Food Science and Nutrition, College of Life Sciences , Kuwait University , 13060 Kuwait City , Kuwait
| | - Sara Al-Qenaie
- Department of Food Science and Nutrition, College of Life Sciences , Kuwait University , 13060 Kuwait City , Kuwait.,Kuwait Oil Company Hospital , 61008 Ahmadi , Kuwait
| | - Muddanna S Rao
- Department of Anatomy, Faculty of Medicine , Kuwait University , 13060 Kuwait City , Kuwait
| | - Khalid M Khan
- Department of Anatomy, Faculty of Medicine , Kuwait University , 13060 Kuwait City , Kuwait
| | - Gilles J Guillemin
- Neuroinflammation Group, Faculty of Medicine and Health Sciences , Macquarie University , Macquarie Park , New South Wales 2109 , Australia
| |
Collapse
|
34
|
Huat TJ, Camats-Perna J, Newcombe EA, Valmas N, Kitazawa M, Medeiros R. Metal Toxicity Links to Alzheimer's Disease and Neuroinflammation. J Mol Biol 2019; 431:1843-1868. [PMID: 30664867 DOI: 10.1016/j.jmb.2019.01.018] [Citation(s) in RCA: 271] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/10/2019] [Accepted: 01/11/2019] [Indexed: 12/11/2022]
Abstract
As the median age of the population increases, the number of individuals with Alzheimer's disease (AD) and the associated socio-economic burden are predicted to worsen. While aging and inherent genetic predisposition play major roles in the onset of AD, lifestyle, physical fitness, medical condition, and social environment have emerged as relevant disease modifiers. These environmental risk factors can play a key role in accelerating or decelerating disease onset and progression. Among known environmental risk factors, chronic exposure to various metals has become more common among the public as the aggressive pace of anthropogenic activities releases excess amount of metals into the environment. As a result, we are exposed not only to essential metals, such as iron, copper, zinc and manganese, but also to toxic metals including lead, aluminum, and cadmium, which perturb metal homeostasis at the cellular and organismal levels. Herein, we review how these metals affect brain physiology and immunity, as well as their roles in the accumulation of toxic AD proteinaceous species (i.e., β-amyloid and tau). We also discuss studies that validate the disruption of immune-related pathways as an important mechanism of toxicity by which metals can contribute to AD. Our goal is to increase the awareness of metals as players in the onset and progression of AD.
Collapse
Affiliation(s)
- Tee Jong Huat
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia; Centre for Stem Cell Ageing and Regenerative Engineering, The University of Queensland, Brisbane, Australia.
| | - Judith Camats-Perna
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Estella A Newcombe
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Nicholas Valmas
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Masashi Kitazawa
- Center for Occupational and Environmental Health, Department of Medicine, University of California, Irvine, CA, USA
| | - Rodrigo Medeiros
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
35
|
Yan L, Yang J, Yu M, Lu Y, Huang L, Wang J, Lu X, Jin C, Wu S, Cai Y. Lanthanum chloride induces neuron damage by activating the nuclear factor-kappa B signaling pathway in activated microglia. Metallomics 2019; 11:1277-1287. [DOI: 10.1039/c9mt00108e] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Lanthanum is a rare earth element which can have adverse effects on the central nervous system (CNS).
Collapse
|
36
|
Caspases orchestrate microglia instrumental functions. Prog Neurobiol 2018; 171:50-71. [DOI: 10.1016/j.pneurobio.2018.09.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 09/21/2018] [Accepted: 09/29/2018] [Indexed: 12/16/2022]
|
37
|
Peng J, Zhou F, Wang Y, Xu Y, Zhang H, Zou F, Meng X. Differential response to lead toxicity in rat primary microglia and astrocytes. Toxicol Appl Pharmacol 2018; 363:64-71. [PMID: 30476502 DOI: 10.1016/j.taap.2018.11.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 11/10/2018] [Accepted: 11/22/2018] [Indexed: 01/25/2023]
Abstract
Lead (Pb) is one of the most widely studied occupational and environmental toxins. Chronic exposure to Pb affects neural function in the central nervous system (CNS). Glial cells in the CNS, such as microglia and astrocytes, respond differently to Pb-induced toxicity. However, the underlying mechanism has not yet been identified. We measured the cell viability and intracellular Pb uptake in rat primary microglia and astrocytes using the CCK-8 assay and inductively coupled plasma mass spectrometry, and found that Pb decreased microglial viability at lower dosages than in astrocytes, while Pb uptake was greater in astrocytes. Pb-induced oxidative stress in microglia results in increased production of reactive oxygen species, down-regulation of glutathione, and enhanced Nrf2 protein expression, while there was no obvious change in astrocytes. The role of Nrf2 in Pb-induced oxidative stress has also been confirmed in primary microglia with the use of Nrf2 small interfering RNA and an Nrf2 agonist. These data indicate that primary microglia were more sensitive to Pb exposure than astrocytes, which is associated with an obvious oxidative stress response and up-regulation of Nrf2 might be involved in this process.
Collapse
Affiliation(s)
- Jiawen Peng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Fan Zhou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuhao Wang
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Yongjie Xu
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongnan Zhang
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Fei Zou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| | - Xiaojing Meng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
38
|
Bjørklund G, Skalny AV, Rahman MM, Dadar M, Yassa HA, Aaseth J, Chirumbolo S, Skalnaya MG, Tinkov AA. Toxic metal(loid)-based pollutants and their possible role in autism spectrum disorder. ENVIRONMENTAL RESEARCH 2018; 166:234-250. [PMID: 29902778 DOI: 10.1016/j.envres.2018.05.020] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/18/2018] [Accepted: 05/18/2018] [Indexed: 06/08/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social interaction, verbal and non-verbal communication, and stereotypic behaviors. Many studies support a significant relationship between many different environmental factors in ASD etiology. These factors include increased daily exposure to various toxic metal-based environmental pollutants, which represent a cause for concern in public health. This article reviews the most relevant toxic metals, commonly found, environmental pollutants, i.e., lead (Pb), mercury (Hg), aluminum (Al), and the metalloid arsenic (As). Additionally, it discusses how pollutants can be a possible pathogenetic cause of ASD through various mechanisms including neuroinflammation in different regions of the brain, fundamentally occurring through elevation of the proinflammatory profile of cytokines and aberrant expression of nuclear factor kappa B (NF-κB). Due to the worldwide increase in toxic environmental pollution, studies on the role of pollutants in neurodevelopmental disorders, including direct effects on the developing brain and the subjects' genetic susceptibility and polymorphism, are of utmost importance to achieve the best therapeutic approach and preventive strategies.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway.
| | - Anatoly V Skalny
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia; All-Russian Research Institute of Medicinal and Aromatic Plants, Moscow, Russia
| | - Md Mostafizur Rahman
- Department of Environmental Sciences, Jahangirnagar University, Dhaka, Bangladesh; Graduate School of Environmental Science, Hokkaido University, Japan
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Heba A Yassa
- Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Jan Aaseth
- Faculty of Health and Social Sciences, Inland Norway University of Applied Sciences, Elverum, Norway; Department of Research, Innlandet Hospital Trust, Brumunddal, Norway
| | - Salvatore Chirumbolo
- Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | | | - Alexey A Tinkov
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia
| |
Collapse
|
39
|
Rahman A, Rao MS, Khan KM. Intraventricular infusion of quinolinic acid impairs spatial learning and memory in young rats: a novel mechanism of lead-induced neurotoxicity. J Neuroinflammation 2018; 15:263. [PMID: 30217162 PMCID: PMC6137743 DOI: 10.1186/s12974-018-1306-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/04/2018] [Indexed: 12/20/2022] Open
Abstract
Background Lead (Pb), a heavy metal, and quinolinic acid (QA), a metabolite of the kynurenine pathway of tryptophan metabolism, are known neurotoxicants. Both Pb and QA impair spatial learning and memory. Pb activates astrocytes and microglia, which in turn induce the synthesis of QA. We hypothesized increased QA production in response to Pb exposure as a novel mechanism of Pb-neurotoxicity. Methods Two experimental paradigms were used. In experiment one, Wistar rat pups were exposed to Pb via their dams’ drinking water from postnatal day 1 to 21. Control group was given regular water. In the second protocol, QA (9 mM) or normal saline (as Vehicle Control) was infused into right lateral ventricle of 21-day old rats for 7 days using osmotic pumps. Learning and memory were assessed by Morris water maze test on postnatal day 30 or 45 in both Pb- and QA-exposed rats. QA levels in the Pb exposed rats were measured in blood by ELISA and in the brain by immunohistochemistry on postnatal days 45 and 60. Expression of various molecules involved in learning and memory was analyzed by Western blot. Means of control and experimental groups were compared with two-way repeated measure ANOVA (learning) and t test (all other variables). Results Pb exposure increased QA level in the blood (by ~ 58%) and increased (p < 0.05) the number of QA-immunoreactive cells in the cortex, and CA1, CA3 and dentate gyrus regions of the hippocampus, compared to control rats. In separate experiments, QA infusion impaired learning and short-term memory similar to Pb. PSD-95, PP1, and PP2A were decreased (p < 0.05) in the QA-infused rats, whereas tau phosphorylation was increased, compared to vehicle infused rats. Conclusion Putting together the results of the two experimental paradigms, we propose that increased QA production in response to Pb exposure is a novel mechanism of Pb-induced neurotoxicity.
Collapse
Affiliation(s)
- Abdur Rahman
- Department of Food Science and Nutrition, College of Life Sciences, Kuwait University, Kuwait City, Kuwait.
| | - Muddanna S Rao
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Khalid M Khan
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
40
|
Clark IA, Vissel B. Therapeutic implications of how TNF links apolipoprotein E, phosphorylated tau, α-synuclein, amyloid-β and insulin resistance in neurodegenerative diseases. Br J Pharmacol 2018; 175:3859-3875. [PMID: 30097997 PMCID: PMC6151331 DOI: 10.1111/bph.14471] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/26/2018] [Accepted: 07/23/2018] [Indexed: 12/24/2022] Open
Abstract
While cytokines such as TNF have long been recognized as essential to normal cerebral physiology, the implications of their chronic excessive production within the brain are now also increasingly appreciated. Syndromes as diverse as malaria and lead poisoning, as well as non‐infectious neurodegenerative diseases, illustrate this. These cytokines also orchestrate changes in tau, α‐synuclein, amyloid‐β levels and degree of insulin resistance in most neurodegenerative states. New data on the effects of salbutamol, an indirect anti‐TNF agent, on α‐synuclein and Parkinson's disease, APOE4 and tau add considerably to the rationale of the anti‐TNF approach to understanding, and treating, these diseases. Therapeutic advances being tested, and arguably useful for a number of the neurodegenerative diseases, include a reduction of excess cerebral TNF, whether directly, with a specific anti‐TNF biological agent such as etanercept via Batson's plexus, or indirectly via surgically implanting stem cells. Inhaled salbutamol also warrants investigating further across the neurodegenerative disease spectrum. It is now timely to integrate this range of new information across the neurodegenerative disease spectrum, rather than keep seeing it through the lens of individual disease states.
Collapse
Affiliation(s)
- I A Clark
- Research School of Biology, Australian National University, Canberra, Australia
| | - B Vissel
- Centre for Neuroscience and Regenerative Medicine, Faculty of Science, University of Technology, Sydney, NSW, Australia.,St. Vincent's Centre for Applied Medical Research, Sydney, NSW, Australia
| |
Collapse
|
41
|
Willis LM, Park H, Watson MWL, Majonis D, Watson JL, Nitz M. Tellurium-based mass cytometry barcode for live and fixed cells. Cytometry A 2018; 93:685-694. [DOI: 10.1002/cyto.a.23495] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/15/2018] [Accepted: 04/26/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Lisa M. Willis
- Department of Chemistry; University of Toronto; Toronto Ontario M5S 3H6 Canada
| | - Hanuel Park
- Department of Chemistry; University of Toronto; Toronto Ontario M5S 3H6 Canada
| | - Michael W. L. Watson
- Fluidigm Corporation, 7000 Shoreline Court; South San Francisco California 94080
| | - Daniel Majonis
- Fluidigm Canada Inc.; 1380 Rodick Road, Markham Ontario L3R 4G5 Canada
| | - Jessica L. Watson
- Fluidigm Canada Inc.; 1380 Rodick Road, Markham Ontario L3R 4G5 Canada
| | - Mark Nitz
- Department of Chemistry; University of Toronto; Toronto Ontario M5S 3H6 Canada
| |
Collapse
|
42
|
Chen S, Fang Y, Xu S, Reis C, Zhang J. Mammalian Sterile20-like Kinases: Signalings and Roles in Central Nervous System. Aging Dis 2018; 9:537-552. [PMID: 29896440 PMCID: PMC5988607 DOI: 10.14336/ad.2017.0702] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/02/2017] [Indexed: 12/25/2022] Open
Abstract
Mammalian Sterile20-like (MST) kinases are located upstream in the mitogen-activated protein kinase pathway, and play an important role in cell proliferation, differentiation, renewal, polarization and migration. Generally, five MST kinases exist in mammalian signal transduction pathways, including MST1, MST2, MST3, MST4 and YSK1. The central nervous system (CNS) is a sophisticated entity that takes charge of information reception, integration and response. Recently, accumulating evidence proposes that MST kinases are critical in the development of disease in different systems involving the CNS. In this review, we summarized the signal transduction pathways and interacting proteins of MST kinases. The potential biological function of each MST kinase and the commonly reported MST-related diseases in the neural system are also reviewed. Further investigation of MST kinases and their interaction with CNS diseases would provide the medical community with new therapeutic targets for human diseases.
Collapse
Affiliation(s)
- Sheng Chen
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuanjian Fang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shenbin Xu
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Cesar Reis
- 2Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, USA.,3Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianmin Zhang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,4Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
43
|
Developmental toxicant exposure in a mouse model of Alzheimer’s disease induces differential sex-associated microglial activation and increased susceptibility to amyloid accumulation. J Dev Orig Health Dis 2017; 8:493-501. [DOI: 10.1017/s2040174417000277] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
As the resident macrophage of the central nervous system, microglia are thought to contribute to Alzheimer’s disease (AD) pathology through lack of neuroprotection. The role of immune dysfunction in AD may be due to disruption of regulatory signals for the activation of microglia that may occur early in development. We hypothesized that early toxicant exposure would systematically activate microglia, possibly reversing the pathological severity of AD. Offspring of a triple transgenic murine model for AD (3×TgAD) were exposed to a model neurotoxicant, lead acetate, from postnatal days (PND) 5–10. Our results indicated that female mice exposed to Pb had a greater and earlier incidence of amyloid burden within the hippocampus, coinciding with decreased markers of microglial activation at PND 50. Pb-exposed males had increased microglial activation at PND 50, as evidence by CD11b expression and microglial abundance, with no significant increase in amyloid burden at that time. There was greater amyloid burden at PND 90 and 180 in both male and female mice exposed to Pb compared with control. Together, these data suggest that activated microglia are neuroprotective against amyloid accumulation early in AD pathology, and that early exposure to Pb could increase susceptibility to later-life neurodegeneration. Likewise, females may be more susceptible to early-life microglial damage, and, subsequently, AD. Further investigation into the sex biased mechanisms by which microglial activation is altered by an early-life immune insult will provide critical insight into the temporal susceptibility of the developing neuroimmune system and its potential role in AD etiopathology.
Collapse
|
44
|
Yang Y, Wang L, Wu Y, Su D, Wang N, Wang J, Shi C, Lv L, Zhang S. Tanshinol suppresses inflammatory factors in a rat model of vascular dementia and protects LPS-treated neurons via the MST1-FOXO3 signaling pathway. Brain Res 2016; 1646:304-314. [DOI: 10.1016/j.brainres.2016.06.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 06/07/2016] [Accepted: 06/11/2016] [Indexed: 01/07/2023]
|
45
|
Zheng S, Song H, Gao H, Liu C, Zhang Z, Fu J. The Antagonistic Effect of Selenium on Lead-Induced Inflammatory Factors and Heat Shock Protein mRNA Level in Chicken Cartilage Tissue. Biol Trace Elem Res 2016; 173:177-84. [PMID: 26831653 DOI: 10.1007/s12011-016-0630-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 01/20/2016] [Indexed: 10/22/2022]
Abstract
Selenium (Se) is recognized as a necessary trace mineral in animal diets, including those of birds. Lead (Pb) is a toxic heavy metal and can damage organs in humans and animals. Complex antagonistic interactions between Se and heavy metals have been reported in previous studies. However, little is known regarding the effects of Se on Pb-induced toxicity and the expression of inflammatory factors and heat shock proteins (HSPs) in the cartilage of chickens. In this present study, we fed chickens either with Se or Pb or both Se and Pb supplement and later analyzed the mRNA expressions of inflammatory factors (inducible nitric oxide synthase (iNOS), nuclear factor-kappa B (NF-κB), tumor necrosis factor-α (TNF-α), and cyclooxygenase-2 (COX-2)) and HSPs (Hsp27, Hsp40, Hsp60, Hsp70, and Hsp90). The results showed that Se and Pb influenced the expression of inflammatory factors and HSP genes in the chicken cartilage tissues. Additionally, we also found that antagonistic interaction existed between Se and Pb supplementation. Our findings suggested that Se could exert a antagonistic effect on Pb in chicken cartilage tissues.
Collapse
Affiliation(s)
- Shufang Zheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Huanyu Song
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Han Gao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Chunpeng Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Ziwei Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| | - Jing Fu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
46
|
Sun GX, Chen Y, Liu CP, Li S, Fu J. Effect of Selenium Against Lead-Induced Damage on the Gene Expression of Heat Shock Proteins and Inflammatory Cytokines in Peripheral Blood Lymphocytes of Chickens. Biol Trace Elem Res 2016; 172:474-480. [PMID: 26728796 DOI: 10.1007/s12011-015-0602-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 12/17/2015] [Indexed: 11/26/2022]
Abstract
The possible beneficial role of selenium (Se) in heat shock proteins (HSPs) and inflammation damage induced by lead (Pb) in chickens is unclear. Therefore, the aim of this study was to investigate the effect of Se against Pb on the messenger RNA (mRNA) expression levels of HSPs (HSP 27, 40, 60, 70, and 90); heme oxygenase-1 (HO-1); and the inflammatory cytokines nuclear factor kappa B (NF-κB), tumor necrosis factor alpha (TNF-α), cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS) in the peripheral blood lymphocytes of chickens. A total of 360 1-day-old broiler chickens were randomly allocated into four groups (n = 90/group). The control group was fed a basic diet containing 0.2 mg/kg Se and 0.5 mg/kg Pb; the Se supplementation group (+Se group) was fed a Se-adequate (sodium selenite) diet containing 1 mg/kg Se and 0.5 mg/kg Pb; the Pb-supplemented group (+Pb group) was fed a Pb acetate diet containing 0.2 mg/kg Se and 350 mg/kg Pb; and the Se and Pb compound group (Se + Pb group) was fed a diet containing 1 mg/kg Se and 350 mg/kg Pb. The blood was collected and examined for the mRNA levels of HSP and inflammatory cytokine genes at 30 and 60 days old. The results showed that Pb poisoning induced the mRNA expression of HSPs and inflammatory cytokines in the peripheral blood lymphocytes of chickens. In addition, Se alleviated the Pb-induced increase in HSP and inflammatory cytokine mRNA levels in chicken peripheral blood lymphocytes. In conclusion, Se can antagonize the toxic effects of Pb on chickens and protect the chickens' peripheral blood lymphocytes in normal physiological function.
Collapse
Affiliation(s)
- G X Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Y Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - C P Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - S Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| | - J Fu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
47
|
Dobrakowski M, Kasperczyk A, Pawlas N, Birkner E, Hudziec E, Chwalińska E, Kasperczyk S. Association between subchronic and chronic lead exposure and levels of antioxidants and chemokines. Int Arch Occup Environ Health 2016; 89:1077-85. [PMID: 27294390 PMCID: PMC5005401 DOI: 10.1007/s00420-016-1144-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 06/06/2016] [Indexed: 11/22/2022]
Abstract
Purpose This study aimed to compare the influence of lead on the non-enzymatic antioxidant defenses and the levels of chemokines in workers subchronically and chronically exposed to lead. Methods The study population was divided into three groups. The first group consisted of male workers subchronically exposed to lead for 40 ± 3.2 days, while the second group included male workers chronically exposed to lead. The third group was a control group. Results The levels of uric acid and bilirubin were significantly higher after a subchronic exposure to lead compared to the baseline by 22 and 35 %, respectively. Similarly, the values of total antioxidant capacity (TAC), total oxidant status (TOS), and oxidative stress index (OSI) increased by 15, 50, and 33 %, respectively. At the same time, the levels of thiol groups and albumin decreased by 5 and 8 %, respectively. Additionally, the levels of interleukin-8 (IL-8) and macrophage inflammatory protein-1β (MIP-1β) were significantly higher after a subchronic exposure to lead compared to the baseline by 34 and 20 %, respectively. Moreover, IL-8 level was significantly higher by 40 % in the group of workers chronically exposed to lead than in the control group, while the level of interferon gamma-induced protein-10 (IP-10) was significantly lower by 28 %. Conclusions Similar to chronic lead exposure, subchronic exposure to lead is associated with elevated blood levels of uric acid and bilirubin in humans. This probably results in increased TAC value despite thiol depletion. However, the compensatory activation of non-enzymatic antioxidant defenses seems to be insufficient to protect against lead-induced oxidative stress, which may be additively enhanced by the pro-inflammatory action of chemokines, especially IL-8.
Collapse
Affiliation(s)
- Michał Dobrakowski
- Department of Biochemistry, School of Medicine with the Division of Dentistry, Medical University of Silesia, ul. Jordana 19, 41-808, Zabrze, Poland.
| | - Aleksandra Kasperczyk
- Department of Biochemistry, School of Medicine with the Division of Dentistry, Medical University of Silesia, ul. Jordana 19, 41-808, Zabrze, Poland
| | - Natalia Pawlas
- Institute of Occupational Medicine and Environmental Health in Sosnowiec, ul. Kościelna 13, 41-200, Sosnowiec, Poland
| | - Ewa Birkner
- Department of Biochemistry, School of Medicine with the Division of Dentistry, Medical University of Silesia, ul. Jordana 19, 41-808, Zabrze, Poland
| | - Edyta Hudziec
- Department of Biochemistry, School of Medicine with the Division of Dentistry, Medical University of Silesia, ul. Jordana 19, 41-808, Zabrze, Poland
| | - Ewa Chwalińska
- Department of Biochemistry, School of Medicine with the Division of Dentistry, Medical University of Silesia, ul. Jordana 19, 41-808, Zabrze, Poland
| | - Sławomir Kasperczyk
- Department of Biochemistry, School of Medicine with the Division of Dentistry, Medical University of Silesia, ul. Jordana 19, 41-808, Zabrze, Poland
| |
Collapse
|
48
|
Wang H, Li S, Teng X. The Antagonistic Effect of Selenium on Lead-Induced Inflammatory Factors and Heat Shock Proteins mRNA Expression in Chicken Livers. Biol Trace Elem Res 2016; 171:437-444. [PMID: 26470710 DOI: 10.1007/s12011-015-0532-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/06/2015] [Indexed: 01/15/2023]
Abstract
The aim of this study was to investigate the effect of lead (Pb) poisoning on nitric oxide (NO) content, inducible nitric oxide synthase (iNOS) activity, the messenger RNA (mRNA) levels of inflammatory factors (nuclear factor-kappa B (NF-κB), tumor necrosis factor-α (TNF-α), cyclooxygenase-2 (COX-2), prostaglandin E synthases (PTGEs), and iNOS), heat shock proteins (HSPs) (HSP27, HSP40, HSP60, HSP70, and HSP90), and the antagonistic effect of selenium (Se) on Pb in chicken livers. One hundred eighty 7-day-old male chickens were randomly divided into four groups and were fed commercial diet and drinking water, Na2SeO3-added commercial diet and drinking water, commercial diet and (CH3OO)2Pb-added drinking water, and Na2SeO3-added commercial diet and (CH3OO)2Pb-added drinking water, respectively, for 30, 60, and 90 days. Then, NO content, iNOS activity, and the mRNA levels of NF-κB, TNF-α, COX-2, PTGEs, iNOS, HSP27, HSP40, HSP60, HSP70, and HSP90 were examined in chicken livers. The results showed that Pb poisoning induced NO content, iNOS activity, and mRNA expression of inflammation factors and HSPs in chicken livers. In addition, Se alleviated Pb-induced increase of inflammation factor and HSP expression in chicken livers.
Collapse
Affiliation(s)
- Hao Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| | - Xiaohua Teng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| |
Collapse
|
49
|
Hussein SA, Hassanein MRR, Amin A, Hussein AHM. Alpha-Lipoic Acid Protects Rat Kidney Against Oxidative Stress-Mediated DNA Damage and Apoptosis Induced by Lead. ACTA ACUST UNITED AC 2015. [DOI: 10.3923/ajbmb.2016.1.14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
50
|
Meuser MV, Quattrociocchi DG, Da Costa LM, Ferreira GB, Carneiro JWDM. Computational study of the interaction between the [Pb(H2O)3]2+ cation and ligands containing oxygen, nitrogen and sulfur donor atoms. Polyhedron 2015. [DOI: 10.1016/j.poly.2015.09.059] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|