1
|
Shang N, Li X, Zhang L, Wang S, He C, Zhang L, Niu Q, Zheng X. Zinc as a Mediator Through the ROCK1 Pathway of Cognitive Impairment in Aluminum-Exposed Workers: A Clinical and Animal Study. Biol Trace Elem Res 2024; 202:5413-5428. [PMID: 38407795 DOI: 10.1007/s12011-024-04119-2] [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: 01/16/2024] [Accepted: 02/19/2024] [Indexed: 02/27/2024]
Abstract
Aluminum (Al) exposure was implicated in neurodegenerative diseases and cognitive impairment, yet the involvement of zinc (Zn) and its mechanism in Al-induced mild cognitive impairment (MCI) remains poorly understood. The objective is to explore the role of Zn in Al-induced cognitive impairment and its potential mechanisms. Montreal cognitive assessment (MoCA) test scores and serum Al, Zn from Al industry workers were collected. A mediation analysis was performed to evaluate the role of serum Zn among serum Al and MoCA test scores. Subsequently, an Al-exposure study was conducted on a rat model categorized into control, low-, medium-, and high-dose groups. After a Morris Water Maze test and detection of Al, Zn content in the hippocampus, integrated transcriptomic and proteomic analyses between the control group and the high-dose group were performed to identify the differentially expressed genes (DEPs), proteins (DEPs), and pathways. To corroborate these findings, quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting (WB) were selected to identify the gene and protein results. Zn overall mediates the relationship between serum Al and cognitive function (mediation effect 17.82%, effect value = - 0.0351). In the Al-exposed rat model, 734 DEGs, 18 miRNAs, 35 lncRNAs, 64 circRNAs, and 113 DEPs were identified between the high-dose group and the control group. Among them, ROCK1, DMD, and other four DEPs were identified as related to zinc finger proteins (ZNF). Co-enrichment analyses of the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) linked these changes to the RHOA/ROCK1 signaling axis. ZNF-related proteins Rock1, DMD, and DHX57 in the high-dose group were downregulated (p = 0.006, 0.003, 0.04), and the expression of Myl9, Rhoa, miR431, and miR182 was also downregulated (p = 0.003, 0.032, 0.032, and 0.046). These findings also show correlations between Al, Zn levels in the hippocampus, water maze performance, and expressions of Myl9, Rhoa, miR431, miR182, DMD, ROCK1, and DHX57, with both negative and positive associations. Based on the results, we determined that Zn was involved in Al-induced MCI in Al workers and Al-exposed rat models. Al exposure and interaction with Zn could trigger the downregulation of ZNF of ROCK1, DMD, and DHX57. miR431, miR182 regulate RHOA/ROCK1 was one of the Zn-involved pathways in Al-induced cognitive impairment.
Collapse
Affiliation(s)
- Nan Shang
- Department of Pharmacy, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Xianlin Li
- School of Pharmacy, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Lan Zhang
- School of Public Health, Capital Medical University, Beijing, 100069, China
| | - ShanShan Wang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Section of Occupational Medicine, Department of Special Medicine, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Chanting He
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Ling Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xiaojun Zheng
- Department of Pharmacy, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, China.
| |
Collapse
|
2
|
Rajabi P, Noori AS, Sargolzaei J. Autism spectrum disorder and various mechanisms behind it. Pharmacol Biochem Behav 2024; 245:173887. [PMID: 39378931 DOI: 10.1016/j.pbb.2024.173887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024]
Abstract
Autism Spectrum Disorder (ASD) is a complex and heterogeneous neurodevelopmental condition characterized by a range of social, communicative, and behavioral challenges. This comprehensive review delves into key aspects of ASD. Clinical Overview and genetic features provide a foundational understanding of ASD, highlighting the clinical presentation and genetic underpinnings that contribute to its complexity. We explore the intricate neurobiological mechanisms at play in ASD, including structural and functional differences that may underlie the condition's hallmark traits. Emerging research has shed light on the role of the immune system and neuroinflammation in ASD. This section investigates the potential links between immunological factors and ASD, offering insights into the condition's pathophysiology. We examine how atypical functional connectivity and alterations in neurotransmitter systems may contribute to the unique cognitive and behavioral features of ASD. In the pursuit of effective interventions, this section reviews current therapeutic strategies, ranging from behavioral and educational interventions to pharmacological approaches, providing a glimpse into the diverse and evolving landscape of ASD treatment. This holistic exploration of mechanisms in ASD aims to contribute to our evolving understanding of the condition and to guide the development of more targeted and personalized interventions for individuals living with ASD.
Collapse
Affiliation(s)
- Parisa Rajabi
- Department of Psychiatry, Arak University of Medical Sciences, Arak, Iran
| | - Ali Sabbah Noori
- Department of Biology, Faculty of Science, Arak University, Arak, Iran
| | - Javad Sargolzaei
- Department of Biology, Faculty of Science, Arak University, Arak, Iran.
| |
Collapse
|
3
|
Fang H, Li J, Zhang L, Li B, Song J, Lu X, Niu Q, Wang L. LncRNA 51A: A promising diagnostic biomarker for assessing cognitive decline in occupationally exposed aluminum workers. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 111:104548. [PMID: 39222898 DOI: 10.1016/j.etap.2024.104548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 08/03/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE To assess the diagnostic utility of lncRNA 51 A in detecting cognitive decline among aluminum-exposed workers occupationally. METHODS 921 male workers from an aluminum manufacturing facility underwent cognitive assessments, measurement of plasma aluminum levels and quantification of lncRNA 51 A levels. Receiver Operating Characteristic (ROC) curves were constructed to assess the diagnostic potential of lncRNA 51 A. Bayesian network model was utilized to predict the likelihood of cognitive decline among the study population. RESULTS Significant differences in lncRNA 51 A levels, plasma aluminum concentration and MMSE scores were observed between cognitive normal and decline groups. The lncRNA 51 A expression was negatively correlated with MMSE scores. The area under the curve (AUC) was 0.894, with 89.3 % sensitivity and 73.9 % specificity. The Bayesian network model indicated varying probabilities of cognitive decline based on lncRNA 51 A expression levels. CONCLUSION Plasma lncRNA 51 A shows potential as an excellent biomarker for cognitive decline diagnosis in aluminum-exposed workers.
Collapse
Affiliation(s)
- Hailun Fang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Juan Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Lei Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Baichun Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Jing Song
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Shanxi Key Laboratory of Environmental Health Impairment and Prevention, Shanxi Medical University, Taiyuan, China; NHC Key Laboratory of Pneumoconiosis, Shanxi Medical University, Taiyuan, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention,Shanxi Medical University, Taiyuan, China
| | - Xiaoting Lu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Shanxi Key Laboratory of Environmental Health Impairment and Prevention, Shanxi Medical University, Taiyuan, China; NHC Key Laboratory of Pneumoconiosis, Shanxi Medical University, Taiyuan, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention,Shanxi Medical University, Taiyuan, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Shanxi Key Laboratory of Environmental Health Impairment and Prevention, Shanxi Medical University, Taiyuan, China; NHC Key Laboratory of Pneumoconiosis, Shanxi Medical University, Taiyuan, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention,Shanxi Medical University, Taiyuan, China
| | - Linping Wang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Shanxi Key Laboratory of Environmental Health Impairment and Prevention, Shanxi Medical University, Taiyuan, China; NHC Key Laboratory of Pneumoconiosis, Shanxi Medical University, Taiyuan, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention,Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
4
|
Boopathi S, Mendonca E, Gandhi A, Rady A, Darwish NM, Arokiyaraj S, Kumar TTA, Pachaiappan R, Guru A, Arockiaraj J. Exploring the Combined Effect of Exercise and Apigenin on Aluminium-Induced Neurotoxicity in Zebrafish. Mol Neurobiol 2024; 61:5320-5336. [PMID: 38191695 DOI: 10.1007/s12035-024-03913-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 12/30/2023] [Indexed: 01/10/2024]
Abstract
Aluminium (AL) is a strong environmental neurotoxin linked to neurodegenerative disorders. Widespread industrial use leads to its presence in water systems, causing bioaccumulation in organisms. This, in turn, results in the bioaccumulation of AL in various organisms. Several studies have highlighted the benefits of enhanced physical activity in combating neurodegenerative diseases. Meanwhile widespread presence of apigenin in aquatic environment has been largely overlooked, in terms of its potential to counter AL-induced neurotoxicity. The combined impact of exercise and apigenin in mitigating the effects of AL-induced neurotoxicity in aquatic animals remains unexplored. Hence, the objective of this study is to determine whether the combined treatment of exercise and apigenin can effectively alleviate the chronic neurotoxicity induced by AL. Zebrafish that were exposed to AL showed behaviours resembling anxiety, increased aggression, unusual swimming pattern, and memory impairment, which are typical features observed in Alzheimer's disease (AD)-like syndrome. Combined treatment of exercise and apigenin protects zebrafish from AL-induced neurotoxicity, which was measured by improvements in memory, reduced anxiety and aggression, and increased levels of antioxidant enzymes and acetylcholinesterase (AChE) activity. Furthermore, AL exposure is associated with increased expression of genes related to neuroinflammation and AD. However, synergistic effect of exercise and apigenin counteract this effect in AL-treated zebrafish. These findings suggest that AL is involved in neurodegenerative diseases in fish, which could affect the integrity of aquatic ecosystem. Hence, there is a strong correlation between enhanced physical activity, apigenin, and the well-being of the ecosystem.
Collapse
Affiliation(s)
- Seenivasan Boopathi
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, Chengalpattu District, 603203, India
| | - Edrea Mendonca
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, Chengalpattu District, 603203, India
| | - Akash Gandhi
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, Chengalpattu District, 603203, India
| | - Ahmed Rady
- Department of Zoology, College of Science, King Saud University, P. O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Noura M Darwish
- Biochemistry Department, Faculty of Science Ain Shams University, Abbasaya, P.O. Box, Cairo, 11566, Egypt
| | - Selvaraj Arokiyaraj
- Department of Food Science & Biotechnology, Sejong University, Seoul, 05006, Korea
| | | | - Raman Pachaiappan
- Department of Biotechnology, Faculty of Engineering and Technology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, Chengalpattu District, 603203, India
| | - Ajay Guru
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, 600 077, India.
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, Chengalpattu District, 603203, India.
| |
Collapse
|
5
|
Gao R, Ali T, Liu Z, Li A, He K, Yang C, Feng J, Li S. NMDAR (2C) deletion in astrocytes relieved LPS-induced neuroinflammation and depression. Int Immunopharmacol 2024; 132:111964. [PMID: 38603856 DOI: 10.1016/j.intimp.2024.111964] [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: 02/09/2024] [Revised: 03/16/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024]
Abstract
The link between neuroinflammation and depression is a subject of growing interest in neuroscience and psychiatry; meanwhile, the precise mechanisms are still being unrevealed. However, glial cell activation, together with cytokine level elevation, suggests a connection between neuroinflammation and the development or exacerbation of depression. Glial cells (astrocytes) communicate with neurons via their extracellular neurotransmitter receptors, including glutamate receptors NMDARs. However, these receptor roles are controversial and enigmatic in neurological disorders, including depression. Therefore, we hypothesized whether NMDAR subnit NR2C deletion in the astrocytes exhibited anti-depressive effects concurrent with neuroinflammation prevention. To assess, we prepared astrocytic-NR2C knockout mice (G-2C: GFAPCre+Grin2Cflox/flox), followed by LPS administration, behavior tests, and biochemical analysis. Stimulatingly, astrocytic-NR2C knockout mice (G-2C) did not display depressive-like behaviors, neuroinflammation, and synaptic deficits upon LPS treatment. PI3K was impaired upon LPS administration in control mice (Grin2Cflox/flox); however, they were intact in the hippocampus of LPS-treated G-2C mice. Further, PI3K activation (via PTEN inhibition by BPV) restored neuroinflammation and depressive-like behavior, accompanied by altered synaptic protein and spine numbers in G-2C mice in the presence of LPS. In addition, NF-κB and JNK inhibitor (BAY, SP600125) treatments reversed the effects of BPV. Moreover, these results were further validated with an NR2C antagonist DQP-1105. Collectively, these observations support the astrocytic-NR2C contribution to LPS-induced neuroinflammation, depression, and synaptic deficits.
Collapse
Affiliation(s)
- Ruyan Gao
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, PR China, 518055.
| | - Tahir Ali
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, PR China, 518055; Institute of Chemical Biology, Shenzhen Bay Laboratory Shenzhen 518132 China.
| | - Zizhen Liu
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, PR China, 518055.
| | - Axiang Li
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, People's Republic of China.
| | - Kaiwu He
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, PR China, 518055.
| | - Canyu Yang
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, People's Republic of China.
| | - Jinxing Feng
- Department of Neonatology, Shenzhen Children's Hospital, Shenzhen, China.
| | - Shupeng Li
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, PR China, 518055; Institute of Chemical Biology, Shenzhen Bay Laboratory Shenzhen 518132 China; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
6
|
Farid HA, Sayed RH, El-Shamarka MES, Abdel-Salam OME, El Sayed NS. PI3K/AKT signaling activation by roflumilast ameliorates rotenone-induced Parkinson's disease in rats. Inflammopharmacology 2024; 32:1421-1437. [PMID: 37541971 PMCID: PMC11006765 DOI: 10.1007/s10787-023-01305-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 07/20/2023] [Indexed: 08/06/2023]
Abstract
Parkinson's disease (PD) is the second most common progressive age-related neurodegenerative disorder. Paramount evidence shed light on the role of PI3K/AKT signaling activation in the treatment of neurodegenerative disorders. PI3K/AKT signaling can be activated via cAMP-dependent pathways achieved by phosphodiesterase 4 (PDE4) inhibition. Roflumilast is a well-known PDE4 inhibitor that is currently used in the treatment of chronic obstructive pulmonary disease. Furthermore, roflumilast has been proposed as a favorable candidate for the treatment of neurological disorders. The current study aimed to unravel the neuroprotective role of roflumilast in the rotenone model of PD in rats. Ninety male rats were allocated into six groups as follows: control, rotenone (1.5 mg/kg/48 h, s.c.), L-dopa (22.5 mg/kg, p.o), and roflumilast (0.2, 0.4 or 0.8 mg/kg, p.o). All treatments were administrated for 21 days 1 h after rotenone injection. Rats treated with roflumilast showed an improvement in motor activity and coordination as well as preservation of dopaminergic neurons in the striatum. Moreover, roflumilast increased cAMP level and activated the PI3K/AKT axis via stimulation of CREB/BDNF/TrkB and SIRT1/PTP1B/IGF1 signaling cascades. Roflumilast also caused an upsurge in mTOR and Nrf2, halted GSK-3β and NF-ĸB, and suppressed FoxO1 and caspase-3. Our study revealed that roflumilast exerted neuroprotective effects in rotenone-induced neurotoxicity in rats. These neuroprotective effects were mediated via the crosstalk between CREB/BDNF/TrkB and SIRT1/PTP1B/IGF1 signaling pathways which activates PI3K/AKT trajectory. Therefore, PDE4 inhibition is likely to offer a reliable persuasive avenue in curing PD via PI3K/AKT signaling activation.
Collapse
Affiliation(s)
- Heba A Farid
- Department of Narcotics, Ergogenic Aids and Poisons, National Research Centre, Cairo, Egypt
| | - Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo, 11562, Egypt.
| | | | - Omar M E Abdel-Salam
- Department of Narcotics, Ergogenic Aids and Poisons, National Research Centre, Cairo, Egypt
| | - Nesrine S El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo, 11562, Egypt
| |
Collapse
|
7
|
Zhang Z, Li X, Ma L, Wang S, Zhang J, Zhou Y, Guo X, Niu Q. LNC000152 Mediates Aluminum-Induced Proliferation of Reactive Astrocytes. ACS OMEGA 2024; 9:11958-11968. [PMID: 38496998 PMCID: PMC10938322 DOI: 10.1021/acsomega.3c09702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/29/2023] [Accepted: 01/05/2024] [Indexed: 03/19/2024]
Abstract
Aluminum is a metal element with significant neurotoxicity, and there is a substantial correlation between aluminum exposure and cognitive dysfunction. Glial fibrillary acidic protein (GFAP) is widely used as a marker of reactive astrocyte proliferation in response to pathological injury of the central nervous system. Studies of various neurodegenerative diseases have confirmed that the expression changes in GFAP are associated with nerve injury. We investigated the role of LNC000152 in the aluminum-induced reactive proliferation of astrocytes. By establishing two aluminum-exposed cell models of rat primary astrocytes and CTX-TNA2 cell lines, we examined the expression of LNC000152 and GFAP and detected cell proliferation with EdU and cell cycle changes with flow cytometry. The role of aluminum in promoting glial cell proliferation was verified; the expression levels of LNC000152 and GFAP increased with the concentration of aluminum exposure. Intervention of LNC000152 expression by siRNA technology revealed that LNC000152 affected glial cell responsive proliferation by influencing GFAP expression. These results suggest that LNC000152 plays a role in the reactive proliferation of astrocytes induced by aluminum.
Collapse
Affiliation(s)
- Zhuoran Zhang
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Xiaoyan Li
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Limin Ma
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Shanshan Wang
- Section
of Occupational Medicine, Department of Special Medicine, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Jingsi Zhang
- Section
of Occupational Medicine, Department of Special Medicine, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Yue Zhou
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Xin Guo
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Qiao Niu
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| |
Collapse
|
8
|
Jiang YY, Wei RY, Tang K, Wang Z, Tan NH. Ginsenoside Rg1 promotes neurite growth of retinal ganglion cells through cAMP/PKA/CREB pathways. J Ginseng Res 2024; 48:163-170. [PMID: 38465221 PMCID: PMC10920000 DOI: 10.1016/j.jgr.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 05/02/2022] [Indexed: 11/20/2022] Open
Abstract
Background Mechanisms of synaptic plasticity in retinal ganglion cells (RGCs) are complex and the current knowledge cannot explain. Growth and regeneration of dendrites together with synaptic formation are the most important parameters for evaluating the cellular protective effects of various molecules. The effect of ginsenoside Rg1 (Rg1) on the growth of retinal ganglion cell processes has been poorly understood. Therefore, we investigated the effect of ginsenoside Rg1 on the neurite growth of RGCs. Methods Expression of proteins and mRNA were detected by Western blot and qPCR. cAMP levels were determined by ELISA. In vivo effects of Rg1 on RGCs were evaluated by hematoxylin and eosin, and immunohistochemistry staining. Results This study found that Rg1 promoted the growth and synaptic plasticity of RGCs neurite by activating the cAMP/PKA/CREB pathways. Meanwhile, Rg1 upregulated the expression of GAP43, Rac1 and PAX6, which are closely related to the growth of neurons. Meantime, H89, an antagonist of PKA, could block this effect of Rg1. In addition, we preliminarily explored the effect of Rg1 on enhancing the glycolysis of RGCs, which could be one of the mechanisms for its neuroprotective effects. Conclusion Rg1 promoted neurite growth of RGCs through cAMP/PKA/CREB pathways. This study may lay a foundation for its clinical use of optic nerve diseases in the future.
Collapse
Affiliation(s)
| | | | - Kai Tang
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhen Wang
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ning-hua Tan
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
9
|
Li H, Liu Y, Wang X, Xu C, Zhang X, Zhang J, Lin L, Niu Q. miR-128-3p is involved in aluminum-induced cognitive impairment by regulating the Sirt1-Keap1/Nrf2 pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 271:115966. [PMID: 38219620 DOI: 10.1016/j.ecoenv.2024.115966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/05/2024] [Accepted: 01/07/2024] [Indexed: 01/16/2024]
Abstract
Aluminum (Al) is a common neurotoxicant in the environment, but the molecular mechanism of its toxic effects is still unclear. Studies have shown that aluminum exposure causes an increase in neuronal apoptosis. The aim of this study was to investigate the mechanism and signaling pathway of neuronal apoptosis induced by aluminum exposure. The rat model was established by intraperitoneal injection of maltol aluminum for 90 days. The results showed that the escape latency of the three groups exposed to maltol aluminum was higher than that of the control group on the 3rd, 4th and 5th days of the positioning cruise experiment (P < 0.05). On the 6th day of the space exploration experiment, compared with the control group(6.00 ± 0.71,15.33 ± 1.08) and the low-dose group(5.08 ± 1.69,13.67 ± 1.09), the number of times that the high-dose group crossed the platform(2.25 ± 0.76) and the platform quadrant(7.58 ± 1.43) was significantly reduced (P < 0.01). The relative expression levels of Sirt1 and Nrf2 in hippocampal tissues of all groups decreased gradually with increasing maltol aluminum exposure dose the relative expression levels of Sirt1 and Nrf2 in high-dose group (0.261 ± 0.094,0.325 ± 0.108) were significantly lower than those in control group (1.018 ± 0.222,1.009 ± 0.156)(P < 0.05). The relative expression level of Keap1 increased gradually with increasing maltol aluminum exposure dose (P < 0.05). The relative expression level of miR-128-3p in the high-dose group(1.520 ± 0.280) was significantly higher than that in the control group(1.000 ± 0.420) (P < 0.05). The content of GSH-Px in the hippocampus of rats decreased with increasing dose. ROS levels gradually increased. We speculated that subchronic aluminum exposure may lead to the activation of miR-128-3p in rat hippocampus of rats, thereby inhibiting the Sirt1-Keap1/Nrf2 pathway so that the Sirt1-Keap1/Nrf2 pathway could not be activated to exert antioxidant capacity, resulting in an imbalance in the antioxidant system of rats and the apoptosis of neurons, which caused reduced cognitive impairment in rats.
Collapse
Affiliation(s)
- Huan Li
- Department of Occupational Health, School of Public Health, Jining Medical University, Jining, China; Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Yan Liu
- Department of Occupational Health, School of Public Health, Jining Medical University, Jining, China; Department of Occupational Health, School of Public Health, Binzhou Medical University, Binzhou, China
| | - Xiangmeng Wang
- Department of Osteoarthrosis, Jining Second People's Hospital, Jining, China
| | - Chaoqun Xu
- Department of Occupational Health, School of Public Health, Jining Medical University, Jining, China
| | - Xiaoyu Zhang
- Department of Occupational Health, School of Public Health, Jining Medical University, Jining, China
| | - Jing Zhang
- Department of Occupational Health, School of Public Health, Jining Medical University, Jining, China; Department of Occupational Health, School of Public Health, Binzhou Medical University, Binzhou, China
| | - Li Lin
- Department of Occupational Health, School of Public Health, Jining Medical University, Jining, China; Department of Occupational Health, School of Public Health, Binzhou Medical University, Binzhou, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Department of Occupational Health, School of Public Health, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
10
|
Wang Y, Yu Q, Liu S, Liu C, Ju Y, Song Q, Cheng D. Aluminum-maltol induced oxidative stress and reduced AMPK activity via BCK-related energy supply failure in C6 cell. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 270:115831. [PMID: 38101974 DOI: 10.1016/j.ecoenv.2023.115831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/17/2023]
Abstract
Aluminum (Al) exposure significantly interferes with the energy supply in astrocytes, which may be a potential mechanism of Al-induced neurotoxicity. This study was designed to explore the mechanisms of Al-induced energy supply impairment in rat C6 astroglioma cell line. Aluminum-maltolate (Al(mal)3) (0.1 mM, 24 h) exposure significantly decreased brain-type creatine kinase (BCK) co-localization with the endoplasmic reticulum (ER) and resulted in mitochondrial dysfunctions, accompanied by a decrease in AMPK phosphorylation. The results of molecular docking showed that Al(mal)3 increased BCK's hydrophobicity and hindered the localization movement of BCK between subcells·H2O2 co-administration was found to exacerbate mitochondrial dysfunction, Ca2+ dyshomeostasis, and apoptosis. After treated with Al(mal)3, additional oxidative stress contributed to BCK activity inhibition but did not promote a further decrease in AMPK phosphorylation. The activation of p-AMPK by its agonist can partially restore mitochondrial function, BCK activity, and ER-localized-BCK levels in Al(mal)3-treated astrocytes. In summary, Al exposure resulted in a sustained depletion of the mitochondrial and antioxidant systems, which was associated with reduced p-AMPK activity and decreased ER-localized-BCK levels in astrocytes. This study provides a theoretical basis for exploring the mechanisms of neurotoxicity induced by Al exposure.
Collapse
Affiliation(s)
- Yingjie Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Qianqian Yu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Sijia Liu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Chunxu Liu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Yaojun Ju
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Qi Song
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Dai Cheng
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China.
| |
Collapse
|
11
|
Koyya P, Manthari RK, Pandrangi SL. Brain-Derived Neurotrophic Factor - The Protective Agent Against Neurological Disorders. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:353-366. [PMID: 37287291 PMCID: PMC11348470 DOI: 10.2174/1871527322666230607110617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/19/2023] [Accepted: 05/29/2023] [Indexed: 06/09/2023]
Abstract
The burden of neurological illnesses on global health is significant. Our perception of the molecular and biological mechanisms underlying intellectual processing and behavior has significantly advanced over the last few decades, laying the groundwork for potential therapies for various neurodegenerative diseases. A growing body of literature reveals that most neurodegenerative diseases could be due to the gradual failure of neurons in the brain's neocortex, hippocampus, and various subcortical areas. Research on various experimental models has uncovered several gene components to understand the pathogenesis of neurodegenerative disorders. One among them is the brain-derived neurotrophic factor (BDNF), which performs several vital functions, enhancing synaptic plasticity and assisting in the emergence of long-term thoughts. The pathophysiology of some neurodegenerative diseases, including Alzheimer's, Parkinson's, Schizophrenia, and Huntington's, has been linked to BDNF. According to numerous research, high levels of BDNF are connected to a lower risk of developing a neurodegenerative disease. As a result, we want to concentrate on BDNF in this article and outline its protective role against neurological disorders.
Collapse
Affiliation(s)
- Prathyusha Koyya
- Department of Biotechnology, GITAM School of Science, Gandhi Institute of Technology and Management (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| | - Ram Kumar Manthari
- Department of Biotechnology, GITAM School of Science, Gandhi Institute of Technology and Management (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| | - Santhi Latha Pandrangi
- Department of Biochemistry and Bioinformatics, GITAM School of Science, Gandhi Institute of Technology and Management (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| |
Collapse
|
12
|
Shehata SA, Kolieb E, Ali DA, Maher SA, Korayem HE, Ibrahim MA, Nafie MS, Ameen SH. Selenium alleviates modafinil-induced neurobehavioral toxicity in rat via PI3K/Akt/mTOR/GSK3B signaling pathway and suppression of oxidative stress and apoptosis: in vivo and in silico study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:458-480. [PMID: 38015391 DOI: 10.1007/s11356-023-31093-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023]
Abstract
Nonmedical use of modafinil (MOD) led to increased rates of overdose toxicity, road accidents, addiction, withdrawal, suicide, and mental illnesses. The current study aims to determine the probable MOD brain toxicity and elucidate the possible role of selenium (Se) in ameliorating the neurotoxicity in rat models. Fifty-four male Albino rats were randomly assigned into nine groups. The groups were G1 (control negative), G2 (Se0.1), G3 (Se0.2), G4 (MOD300), G5 (MOD600), G6 (Se0.1 + MOD300), G7 (Se0.2 + MOD300), G8 (Se0.1 + MOD600), and G9 (Se0.2 + MOD600). After finishing the experiment, blood and brain tissue were harvested for biochemical and histological investigation. Neurobehavior parameters were assessed. Tissue neurotransmitter levels and oxidative stress markers were assessed. Gene expression of PI3K/Akt/mTOR-GSK3B, orexin, and orexin receptor2 was measured by qRT-PCR. Histological and immunohistochemistry assessments, as well as molecular docking, were carried out. MOD-induced neurobehavioral toxicity exhibited by behavioral and cognitive function impairments, which are associated with decreased antioxidant activities, increased MDA levels, and decreases in neurotransmitter levels. Brain levels of mRNA expression of PI3K, Akt, and mTOR were decreased, while GS3K, orexin, and orexin receptors were significantly elevated. These disturbances were confirmed by histopathological brain changes with increased silver and Bax immunostaining and decreased crystal violet levels. MOD induced neurotoxic effects in a dose-dependent manner. Compared with the MOD groups, SE coadministration significantly attenuates MOD-induced toxic changes. Docking study shows the protective role of Se as an apoptosis inhibitor and inflammation inhibitor. In conclusion, Se could be used as a biologically effective antioxidant compound to protect from MOD neurobehavioral toxicity in Wistar rats by reversing behavioral alterations, inflammation, apoptosis, and oxidative injury.
Collapse
Affiliation(s)
- Shaimaa A Shehata
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Eman Kolieb
- Physiology Department, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Dina A Ali
- Clinical Pharmacology Department, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
- Center of Excellence in Molecular & Cellular Medicine, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Shymaa Ahmed Maher
- Center of Excellence in Molecular & Cellular Medicine, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Horeya Erfan Korayem
- Histology and Cell Biology Department, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Mahrous A Ibrahim
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt.
- Forensic Medicine and Clinical Toxicology, College of Medicine, Jouf University, 72341, Aljouf, Saudi Arabia.
| | - Mohamed S Nafie
- Department of Chemistry, College of Sciences, University of Sharjah, P. O. Box 27272, Sharjah, United Arab Emirates
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia, 41522, Egypt
| | - Shimaa H Ameen
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Zagazig University, Alsharqia, Egypt
| |
Collapse
|
13
|
Zhang H, Zhu Y, Zhu G, Yang S. Clinical Efficacy and Mechanistic Insights of Anshen Dingzhi Prescription on Breast Cancer-Related PTSD Through Network Pharmacology and Molecular Docking. Integr Cancer Ther 2024; 23:15347354241285435. [PMID: 39344020 PMCID: PMC11450869 DOI: 10.1177/15347354241285435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 09/18/2024] [Accepted: 09/05/2024] [Indexed: 10/01/2024] Open
Abstract
Anshen Dingzhi prescription (ADP) is a classic prescription of traditional Chinese medicine, which has been used in the treatment of neuropsychiatric diseases. However, its treatment of breast cancer-related post-traumatic stress disorder (BC-PTSD) lacks clinical research evidence and its mechanism is not clear. The present study investigated the efficacy and action mechanism of ADP against BC-PTSD. The results of the clinical trial showed that after 4 weeks of treatment, both groups showed reduced post-traumatic stress disorder checklist-civilian version (PCL-C), Pittsburgh sleep quality index (PSQI), self-rating depression scale (SDS) and self-rating anxiety scale (SAS) scores, and increased functional assessment of cancer therapy-breast (FACT-B) scores. The serum cortisol (CORT), tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1β) levels were decreased and brain-derived neurotrophic factor (BDNF) level were increased, and the improvement of serum TNF-α, IL-1β, and BDNF in treatment group was better than that of the control group. The overall treatment efficacy in the treatment group (43.90%) was superior to that in the control group (23.81%), and the overall incidence of adverse effects was lower than that in the control group. The results of network analysis and molecular docking showed that ADP blood components could act on IL1B, TNF, and BDNF. ADP contributes to the treatment of BC-PTSD symptoms, with a mechanism possibly related to its regulatory effect on TNF-α, IL-1β, and BDNF levels.Trial registration: Chinese Clinical Trial Registry, http://www.chictr.org.cn,ChiCTR2300077801.
Collapse
Affiliation(s)
- Hao Zhang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- Anhui University of Chinese Medicine, Hefei, China
| | - Yongfu Zhu
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Guoqi Zhu
- Anhui University of Chinese Medicine, Hefei, China
| | - Shaojie Yang
- The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
14
|
Xu W, Sun X, Jiang H, Wang X, Wang B, Niu Q, Meng H, Du J, Yang G, Liu B, Zhang H, Tan Y. Diffusion Kurtosis Imaging in Evaluating the Mild Cognitive Impairment of Occupational Aluminum Workers. Acad Radiol 2023; 30:2225-2233. [PMID: 36690563 DOI: 10.1016/j.acra.2022.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 12/02/2022] [Accepted: 12/02/2022] [Indexed: 01/23/2023]
Abstract
RATIONALE AND OBJECTIVES To investigate whether diffusion kurtosis imaging (DKI) can distinguish mild cognitive impairment (MCI) from normal controls (NC) in aluminum (Al)-exposed workers, and to explore the association of DKI with cognitive performance and plasma Al concentration. MATERIALS AND METHODS 28 patients with MCI and 25 NC at Al factory were enrolled in this study. All subjects underwent conventional MRI and DKI scans. The mean kurtosis (MK), axial kurtosis (Ka), radial kurtosis (Kr), mean diffusivity (MD) and fractional anisotropy (FA) parameters of the hippocampus, substantia nigra, red nucleus, thalamus, anterior cingulate gyrus, genu and crus of the corpus callosum, frontal, parietal and temporal lobe were measured. To compare the parameters between the two groups, the Mann-Whitney rank sum test was used. The correlation of parameter values with cognitive performance and plasma Al concentration was analyzed using Spearman correlation analysis. The receiver operating characteristic (ROC) curve and the Z-scores were used to evaluate the diagnostic efficacy of each parameter. RESULTS Compared with the NC group, the MK, Ka, Kr, and FA values in the MCI group were significantly decreased, and the MD values were significantly increased (p<0.05). For the diagnosis of MCI, MK in the right hippocampus showed the largest AUC (0.924). The MK, Kr, MD and FA values were correlated with the Montreal Cognitive Assessment (MoCA) scores, and MK values in the right hippocampus showed the greatest correlation with MoCA scores (r=0.744, p <0.001). Plasma Al in the MCI group was higher than that in the NC group, although there was no significant difference in plasma Al between the two groups (p=0.057). There was no correlation between DKI parameters and plasma Al. CONCLUSION The DKI method might be a sensitive imaging biomarker to discriminate MCI from NC, and could preliminarily assess the severity of cognitive impairment in Al-exposed workers. MK in the right hippocampus appeared to be the best independent predictor. The mechanism of cognitive decline is an important content of aluminum exposure research. This study indicates that the DKI technique could provide valuable information for the diagnosis of MCI.
Collapse
Affiliation(s)
- Wenji Xu
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Xiangru Sun
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Haoru Jiang
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Xiaochun Wang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.; Shanxi Key Laboratory of Intelligent Imaging and Nanomedicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.; Intelligent Imaging Big Data and Functional Nano-imaging Engineering Research Center of Shanxi Province, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Bin Wang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.; Shanxi Key Laboratory of Intelligent Imaging and Nanomedicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.; Intelligent Imaging Big Data and Functional Nano-imaging Engineering Research Center of Shanxi Province, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Qiao Niu
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Huaxing Meng
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Jiangfeng Du
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.; Shanxi Key Laboratory of Intelligent Imaging and Nanomedicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.; Intelligent Imaging Big Data and Functional Nano-imaging Engineering Research Center of Shanxi Province, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Guoqiang Yang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.; Shanxi Key Laboratory of Intelligent Imaging and Nanomedicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.; Intelligent Imaging Big Data and Functional Nano-imaging Engineering Research Center of Shanxi Province, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Bo Liu
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.; Shanxi Key Laboratory of Intelligent Imaging and Nanomedicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.; Intelligent Imaging Big Data and Functional Nano-imaging Engineering Research Center of Shanxi Province, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Hui Zhang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.; Shanxi Key Laboratory of Intelligent Imaging and Nanomedicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.; Intelligent Imaging Big Data and Functional Nano-imaging Engineering Research Center of Shanxi Province, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Yan Tan
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.; Shanxi Key Laboratory of Intelligent Imaging and Nanomedicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.; Intelligent Imaging Big Data and Functional Nano-imaging Engineering Research Center of Shanxi Province, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China..
| |
Collapse
|
15
|
Aleksandrova Y, Neganova M. Deciphering the Mysterious Relationship between the Cross-Pathogenetic Mechanisms of Neurodegenerative and Oncological Diseases. Int J Mol Sci 2023; 24:14766. [PMID: 37834214 PMCID: PMC10573395 DOI: 10.3390/ijms241914766] [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: 08/10/2023] [Revised: 09/22/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
The relationship between oncological pathologies and neurodegenerative disorders is extremely complex and is a topic of concern among a growing number of researchers around the world. In recent years, convincing scientific evidence has accumulated that indicates the contribution of a number of etiological factors and pathophysiological processes to the pathogenesis of these two fundamentally different diseases, thus demonstrating an intriguing relationship between oncology and neurodegeneration. In this review, we establish the general links between three intersecting aspects of oncological pathologies and neurodegenerative disorders, i.e., oxidative stress, epigenetic dysregulation, and metabolic dysfunction, examining each process in detail to establish an unusual epidemiological relationship. We also focus on reviewing the current trends in the research and the clinical application of the most promising chemical structures and therapeutic platforms that have a modulating effect on the above processes. Thus, our comprehensive analysis of the set of molecular determinants that have obvious cross-functional pathways in the pathogenesis of oncological and neurodegenerative diseases can help in the creation of advanced diagnostic tools and in the development of innovative pharmacological strategies.
Collapse
Affiliation(s)
- Yulia Aleksandrova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Margarita Neganova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 420088 Kazan, Russia
| |
Collapse
|
16
|
Li Q, Feng Y, Wang R, Liu R, Ba Y, Huang H. Recent insights into autophagy and metals/nanoparticles exposure. Toxicol Res 2023; 39:355-372. [PMID: 37398566 PMCID: PMC10313637 DOI: 10.1007/s43188-023-00184-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 03/08/2023] [Accepted: 04/04/2023] [Indexed: 07/04/2023] Open
Abstract
Some anthropogenic pollutants, such as heavy metals and nanoparticles (NPs), are widely distributed and a major threat to environmental safety and public health. In particular, lead (Pb), cadmium (Cd), chromium (Cr), arsenic (As), and mercury (Hg) have systemic toxicity even at extremely low concentrations, so they are listed as priority metals in relation to their significant public health burden. Aluminum (Al) is also toxic to multiple organs and is linked to Alzheimer's disease. As the utilization of many metal nanoparticles (MNPs) gradually gain traction in industrial and medical applications, they are increasingly being investigated to address potential toxicity by impairing certain biological barriers. The dominant toxic mechanism of these metals and MNPs is the induction of oxidative stress, which subsequently triggers lipid peroxidation, protein modification, and DNA damage. Notably, a growing body of research has revealed the linkage between dysregulated autophagy and some diseases, including neurodegenerative diseases and cancers. Among them, some metals or metal mixtures can act as environmental stimuli and disturb basal autophagic activity, which has an underlying adverse health effect. Some studies also revealed that specific autophagy inhibitors or activators could modify the abnormal autophagic flux attributed to continuous exposure to metals. In this review, we have gathered recent data about the contribution of the autophagy/mitophagy mediated toxic effects and focused on the involvement of some key regulatory factors of autophagic signaling during exposure to selected metals, metal mixtures, as well as MNPs in the real world. Besides this, we summarized the potential significance of interactions between autophagy and excessive reactive oxygen species (ROS)-mediated oxidative damage in the regulation of cell survival response to metals/NPs. A critical view is given on the application of autophagy activators/inhibitors to modulate the systematic toxicity of various metals/MNPs.
Collapse
Affiliation(s)
- Qiong Li
- Department of Environmental Health and Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Yajing Feng
- Department of Environmental Health and Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Ruike Wang
- Department of Environmental Health and Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Rundong Liu
- Department of Environmental Health and Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Yue Ba
- Department of Environmental Health and Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Hui Huang
- Department of Environmental Health and Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| |
Collapse
|
17
|
Miao J, Ma H, Yang Y, Liao Y, Lin C, Zheng J, Yu M, Lan J. Microglia in Alzheimer's disease: pathogenesis, mechanisms, and therapeutic potentials. Front Aging Neurosci 2023; 15:1201982. [PMID: 37396657 PMCID: PMC10309009 DOI: 10.3389/fnagi.2023.1201982] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/30/2023] [Indexed: 07/04/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by protein aggregation in the brain. Recent studies have revealed the critical role of microglia in AD pathogenesis. This review provides a comprehensive summary of the current understanding of microglial involvement in AD, focusing on genetic determinants, phenotypic state, phagocytic capacity, neuroinflammatory response, and impact on synaptic plasticity and neuronal regulation. Furthermore, recent developments in drug discovery targeting microglia in AD are reviewed, highlighting potential avenues for therapeutic intervention. This review emphasizes the essential role of microglia in AD and provides insights into potential treatments.
Collapse
Affiliation(s)
- Jifei Miao
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Shenzhen, China
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Haixia Ma
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yang Yang
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yuanpin Liao
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Cui Lin
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Juanxia Zheng
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Muli Yu
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Jiao Lan
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Shenzhen, China
| |
Collapse
|
18
|
Li H, Zhang Q, Xue X, Zhang J, Wang S, Zhang J, Lin L, Niu Q. Lnc001209 Participates in aluminium-induced apoptosis of PC12 cells by regulating PI3K-AKT-mTOR signalling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 259:115062. [PMID: 37229874 DOI: 10.1016/j.ecoenv.2023.115062] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/07/2023] [Accepted: 05/21/2023] [Indexed: 05/27/2023]
Abstract
Aluminium (Al) is a common environmental neurotoxin, but the molecular mechanism underlying its toxic effects remains unclear. Many studies have shown that aluminium exposure leads to increased neuronal apoptosis. This study aimed to investigate the mechanisms and signalling pathways involved in aluminium exposure-induced neuronal apoptosis. The results showed a decrease in the number of PC12 cells and changes in cell morphology in the aluminium maltol exposure group. The viability of PC12 cells decreased gradually with increasing of exposure doses, and the apoptosis rate increased. The expression of Lnc001209 decreased gradually with an increase in the aluminium exposure dose. After transfection of Lnc001209 siRNA in aluminium-exposed PC12 cells, the protein expression levels of p-Akt Ser473, p-Akt Thr308, p-P85 Tyr467, p-mTOR Ser2448 and CD36 were increased. RNA pull-down MS showed that Lnc001209 interacts with the CD36 protein. Expression of the CD36 protein was increased in PC12 cells exposed to aluminium. The results of the CD36 intervention experiment showed that the protein expression levels of p-Akt Ser473, p-Akt Thr308, p-P85 Tyr467, and p-mTOR Ser2448 likely increased after CD36 overexpression. In addition, the phosphorylation level of AKT had the most significant increase. The enhancement of p-Akt activity promotes neuronal apoptosis.
Collapse
Affiliation(s)
- Huan Li
- Department of Occupational Health, School of Public Health, Jining Medical University, Jining 272067, Shandong, China; Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Qinli Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Xingli Xue
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Jingsi Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Shanshan Wang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Jing Zhang
- Department of Occupational Health, School of Public Health, Jining Medical University, Jining 272067, Shandong, China
| | - Li Lin
- Department of Occupational Health, School of Public Health, Jining Medical University, Jining 272067, Shandong, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Department of Occupational Health, School of Public Health, Xuzhou Medical University, Xuzhou 221000, Jiangsu, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| |
Collapse
|
19
|
Samman WA, Selim SM, El Fayoumi HM, El-Sayed NM, Mehanna ET, Hazem RM. Dapagliflozin Ameliorates Cognitive Impairment in Aluminum-Chloride-Induced Alzheimer's Disease via Modulation of AMPK/mTOR, Oxidative Stress and Glucose Metabolism. Pharmaceuticals (Basel) 2023; 16:ph16050753. [PMID: 37242536 DOI: 10.3390/ph16050753] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/02/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurological illness characterized by memory loss and cognitive deterioration. Dapagliflozin was suggested to attenuate the memory impairment associated with AD; however, its mechanisms were not fully elucidated. This study aims to examine the possible mechanisms of the neuroprotective effects of dapagliflozin against aluminum chloride (AlCl3)-induced AD. Rats were distributed into four groups: group 1 received saline, group 2 received AlCl3 (70 mg/kg) daily for 9 weeks, and groups 3 and 4 were administered AlCl3 (70 mg/kg) daily for 5 weeks. Dapagliflozin (1 mg/kg) and dapagliflozin (5 mg/kg) were then given daily with AlCl3 for another 4 weeks. Two behavioral experiments were performed: the Morris Water Maze (MWM) and the Y-maze spontaneous alternation (Y-maze) task. Histopathological alterations in the brain, as well as changes in acetylcholinesterase (AChE) and amyloid β (Aβ) peptide activities and oxidative stress (OS) markers, were all evaluated. A western blot analysis was used for the detection of phosphorylated 5' AMP-activated protein kinase (p-AMPK), phosphorylated mammalian target of Rapamycin (p-mTOR) and heme oxygenase-1 (HO-1). Tissue samples were collected for the isolation of glucose transporters (GLUTs) and glycolytic enzymes using PCR analysis, and brain glucose levels were also measured. The current data demonstrate that dapagliflozin represents a possible approach to combat AlCl3-induced AD in rats through inhibiting oxidative stress, enhancing glucose metabolism and activating AMPK signaling.
Collapse
Affiliation(s)
- Waad A Samman
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Medina 30078, Saudi Arabia
| | - Salma M Selim
- Department of Pharmacology and Toxicology, Faculty of Dentistry, Sinai University, Kantara, Ismailia 41636, Egypt
| | - Hassan M El Fayoumi
- Department of Pharmacology and Toxicology, Faculty of Dentistry, Sinai University, Kantara, Ismailia 41636, Egypt
| | - Norhan M El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Eman T Mehanna
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Reem M Hazem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
20
|
Shi J, Liu D, Jin Q, Chen X, Zhang R, Shi T, Zhu S, Zhang Y, Zong X, Wang C, Li L. Whole-Transcriptome Analysis of Repeated Low-Level Sarin-Exposed Rat Hippocampus and Identification of Cerna Networks to Investigate the Mechanism of Sarin-Induced Cognitive Impairment. BIOLOGY 2023; 12:biology12040627. [PMID: 37106826 PMCID: PMC10136365 DOI: 10.3390/biology12040627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023]
Abstract
Sarin is a potent organophosphorus nerve agent that causes cognitive dysfunction, but its underlying molecular mechanisms are poorly understood. In this study, a rat model of repeated low-level sarin exposure was established using the subcutaneous injection of 0.4 × LD50 for 21 consecutive days. Sarin-exposed rats showed persistent learning and memory impairment and reduced hippocampal dendritic spine density. A whole-transcriptome analysis was applied to study the mechanism of sarin-induced cognitive impairment, and a total of 1035 differentially expressed mRNA (DEmRNA), including 44 DEmiRNA, 305 DElncRNA, and 412 DEcircRNA, were found in the hippocampus of sarin-treated rats. According to Gene Ontology (GO) annotation, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment, and Protein-Protein Interaction (PPI) analysis, these DERNAs were mainly involved in neuronal synaptic plasticity and were related to the pathogenesis of neurodegenerative diseases. The circRNA/lncRNA-miRNA-mRNA ceRNA network was constructed, in which Circ_Fmn1, miR-741-3p, miR-764-3p, miR-871-3p, KIF1A, PTPN11, SYN1, and MT-CO3 formed one circuit, and Circ_Cacna1c, miR-10b-5p, miR-18a-5p, CACNA1C, PRKCD, and RASGRP1 constituted another circuit. The balance between the two circuits was crucial for maintaining synaptic plasticity and may be the regulatory mechanism by which sarin causes cognitive impairment. Our study reveals the ceRNA regulation mechanism of sarin exposure for the first time and provides new insights into the molecular mechanisms of other organophosphorus toxicants.
Collapse
Affiliation(s)
- Jingjing Shi
- State Key Laboratory of NBC Protection for Civilians, Beijing 102205, China
| | - Dongxin Liu
- State Key Laboratory of NBC Protection for Civilians, Beijing 102205, China
| | - Qian Jin
- State Key Laboratory of NBC Protection for Civilians, Beijing 102205, China
| | - Xuejun Chen
- State Key Laboratory of NBC Protection for Civilians, Beijing 102205, China
| | - Ruihua Zhang
- State Key Laboratory of NBC Protection for Civilians, Beijing 102205, China
| | - Tong Shi
- State Key Laboratory of NBC Protection for Civilians, Beijing 102205, China
| | - Siqing Zhu
- State Key Laboratory of NBC Protection for Civilians, Beijing 102205, China
| | - Yi Zhang
- State Key Laboratory of NBC Protection for Civilians, Beijing 102205, China
| | - Xingxing Zong
- State Key Laboratory of NBC Protection for Civilians, Beijing 102205, China
| | - Chen Wang
- State Key Laboratory of NBC Protection for Civilians, Beijing 102205, China
| | - Liqin Li
- State Key Laboratory of NBC Protection for Civilians, Beijing 102205, China
| |
Collapse
|
21
|
Shang N, Zhang L, Gao Q, Li W, Wang S, Gao X, Chen J, Zhang L, Niu Q, Zhang Q. Simultaneous effects of aluminum exposure on the homeostasis of essential metal content in rat brain and perturbation of gut microbiota. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 254:114707. [PMID: 36893695 DOI: 10.1016/j.ecoenv.2023.114707] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/21/2023] [Accepted: 02/26/2023] [Indexed: 06/18/2023]
Abstract
The theory of the brain-gut axis has confirmed that gut microbiota and metabolites are involved in the progression of neurodegenerative diseases through multiple pathways. However, few studies have highlighted the role of gut microbiota in cognitive impairment induced by aluminum (Al) exposure and its correlations with the homeostasis of essential metal content in the brain. To explore the relationship between alterations in the content of essential metals in the brain and relative abundance changes in gut microbiota induced by Al exposure, the Al, zinc (Zn), copper (Cu), iron (Fe), chromium (Cr), manganese (Mn), and cobalt (Co) content level in the hippocampus, olfactory bulb, and midbrain tissue were measured by inductively coupled plasma mass spectrometry (ICP-MS) methods after Al maltolate was intraperitoneally injected every other day for exposed groups. Then the unsupervised principal coordinates analysis (PCoA) and linear discriminant analysis effect size (LEfSe) were used to analyze the relative abundance of the gut microbiota community and the structure of the gut microbiome. Finally, the correlations between gut microbiota composition and essential metal content in the different exposure groups were explored by using the Pearson correlation coefficient method. Based on the results, we indicated that the content of Al in the hippocampus, olfactory bulb, and midbrain tissue was increased and then decreased with the increasing exposure duration, with peaks occurring between 14 and 30 days. Concomitantly, Al-exposure decreased the Zn, Fe, and Mn levels in these tissues. 16 S rRNA gene sequencing results indicated that significant differences in the intestinal microbial community structure at the phylum, family, and genus levels were found in the Day 90 exposed group compared with the Day 7 exposed group. Ten enriched species in the exposed group were identified as markers at the three levels. Furthermore, ten bacteria at the genus level were identified to have a significantly strong correlation (r = 0.70-0.90) with Fe, Zn, Mn, and Co.
Collapse
Affiliation(s)
- Nan Shang
- Department of Pharmacy, First Hospital of Shanxi Medical University, Taiyuan Shanxi 030001, China.
| | - Lan Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan Shanxi 030001, China
| | - Qi Gao
- School of Pharmacy, Shanxi Medical University, Taiyuan Shanxi 030001, China
| | - Weipeng Li
- School of Pharmacy, Shanxi Medical University, Taiyuan Shanxi 030001, China
| | - Shanshan Wang
- Section of Occupational Medicine, Department of Special Medicine, Shanxi Medical University, Taiyuan Shanxi 030001, China
| | - Xiaocheng Gao
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan Shanxi 030001, China
| | - Jin Chen
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan Shanxi 030001, China
| | - Ling Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan Shanxi 030001, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan Shanxi 030001, China
| | - Qinli Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan Shanxi 030001, China
| |
Collapse
|
22
|
Khezri MR, Ghasemnejad-Berenji M. The Role of Caspases in Alzheimer's Disease: Pathophysiology Implications and Pharmacologic Modulation. J Alzheimers Dis 2023; 91:71-90. [PMID: 36442198 DOI: 10.3233/jad-220873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder worldwide. Although the main cause of the onset and development of AD is not known yet, neuronal death due to pathologic changes such as amyloid-β (Aβ) deposition, tau aggregation, neuroinflammation, oxidative stress, and calcium dyshomeostasis are considered to be the main cause. At the present, there is no cure for this insidious disorder. However, accurate identification of molecular changes in AD can help provide new therapeutic goals. Caspases are a group of proteases which are known because of their role in cellular apoptosis. In addition, different caspases are involved in other cellular responses to the environment, such as induction of inflammation. Emerging evidence suggest that these proteases play a central role in AD pathophysiology due to their role in the processing of amyloid-β protein precursor, tau cleavage, and neuroinflammation. Therefore, it seems that targeting caspases may be a suitable therapeutic option to slow the progression of AD. This review focuses on the role of caspases in AD pathophysiology and introduce results from studies targeted caspases in different models of AD.
Collapse
Affiliation(s)
| | - Morteza Ghasemnejad-Berenji
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran.,Research Center for Experimental and Applied Pharmaceutical Sciences, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
23
|
Bittencourt LO, Damasceno-Silva RD, Aragão WAB, Eiró-Quirino L, Oliveira ACA, Fernandes RM, Freire MAM, Cartágenes SC, Dionizio A, Buzalaf MAR, Cassoli JS, Cirovic A, Cirovic A, Maia CDSF, Lima RR. Global Proteomic Profile of Aluminum-Induced Hippocampal Impairments in Rats: Are Low Doses of Aluminum Really Safe? Int J Mol Sci 2022; 23:ijms232012523. [PMID: 36293377 PMCID: PMC9603961 DOI: 10.3390/ijms232012523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/12/2022] [Accepted: 09/29/2022] [Indexed: 11/21/2022] Open
Abstract
Hippocampus is the brain area where aluminum (Al) accumulates in abundance and is widely associated with learning and memory. In the present study, we evaluate behavioral, tissue, and proteomic changes in the hippocampus of Wistar rats caused by exposure to doses that mimic human consumption of aluminum chloride (AlCl3) in urban areas. For this, male Wistar rats were divided into two groups: Control (distilled water) and AlCl3 (8.3 mg/kg/day), both groups were exposed orally for 60 days. After the Al exposure protocol, cognitive functions were assessed by the Water maze test, followed by a collection for analysis of the global proteomic profile of the hippocampus by mass spectrometry. Aside from proteomic analysis, we performed a histological analysis of the hippocampus, to the determination of cell body density by cresyl violet staining in Cornu Ammonis fields (CA) 1 and 3, and hilus regions. Our results indicated that exposure to low doses of aluminum chloride triggered a decreased cognitive performance in learning and memory, being associated with the deregulation of proteins expression, mainly those related to the regulation of the cytoskeleton, cellular metabolism, mitochondrial activity, redox regulation, nervous system regulation, and synaptic signaling, reduced cell body density in CA1, CA3, and hilus.
Collapse
Affiliation(s)
- Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Rakhel Dayanne Damasceno-Silva
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Walessa Alana Bragança Aragão
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Luciana Eiró-Quirino
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Ana Carolina Alves Oliveira
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Rafael Monteiro Fernandes
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Marco Aurelio M. Freire
- Graduate Program in Health and Society, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoro 59610210, Brazil
| | - Sabrina Carvalho Cartágenes
- Laboratory of Pharmacology of Inflammation and Behavior, Institute of Health Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Aline Dionizio
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru 05508060, Brazil
| | | | - Juliana Silva Cassoli
- Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Ana Cirovic
- Faculty of Medicine, Institute of Anatomy, University of Belgrade, 11000 Belgrade, Serbia
| | - Aleksandar Cirovic
- Faculty of Medicine, Institute of Anatomy, University of Belgrade, 11000 Belgrade, Serbia
| | - Cristiane do Socorro Ferraz Maia
- Laboratory of Pharmacology of Inflammation and Behavior, Institute of Health Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
- Correspondence:
| |
Collapse
|
24
|
Activation of PI3k/Akt/mTOR Signaling Induces Deposition of p-tau to Promote Aluminum Neurotoxicity. Neurotox Res 2022; 40:1516-1525. [PMID: 36066748 DOI: 10.1007/s12640-022-00573-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/16/2022] [Accepted: 08/29/2022] [Indexed: 10/14/2022]
Abstract
Aluminum neurotoxicity impairs learning and memory ability, but the molecular mechanism has not been elucidated. The aim of this study was to examine the role of phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling in regulating the expression of synaptic plasticity-related proteins (PRPs) and p-tau deposition to explore the mechanism underlying aluminum-induced neurotoxicity. We constructed a sub-chronic aluminum-exposed Sprague Dawley (SD) rat model to assess aluminum neurotoxicity in vivo. The learning and memory abilities of rats were examined using the Morris water maze test. We also assessed the effect of aluminum in vitro using rat pheochromocytoma (PC12) cells. To explore the role of PI3K/Akt/mTOR signaling in aluminum neurotoxicity, we used the PI3K inhibitor wortmannin and the mTOR inhibitor rapamycin in aluminum-treated PC12 cells. Protein expression was examined by western blotting. Aluminum disrupted the learning and memory abilities of SD rats. Mechanistically, aluminum reduced the levels of the synaptic PRPs (cAMP-response element binding protein (CREB), glutamate receptor 1 (GluR1), glutamate receptor 2 (GluR2), and postsynaptic density protein 95 (PSD-95), and it increased p-tau deposition in the hippocampus of SD rats. We observed similar results in aluminum-treated PC12 cells. Further, PI3K/Akt/mTOR signaling was abnormally activated in aluminum-treated PC12 cells, and treatment with rapamycin reversed the decrease in synaptic PRPs levels and the increase in p-tau deposition. In conclusion, the activation of PI3K/Akt/mTOR signaling reduces the levels of synaptic PRPs and increases p-tau deposition induced by aluminum. Therefore, the PI3K/Akt/mTOR pathway participates in the mechanism of aluminum neurotoxicity.
Collapse
|
25
|
王 菲, 王 路, 熊 玥, 邓 静, 吕 明, 汤 博, 张 潇, 李 英. [Mechanism of valproic acid-induced dendritic spine and synaptic impairment in the prefrontal cortex for causing core autistic symptoms in mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:101-107. [PMID: 35249876 PMCID: PMC8901407 DOI: 10.12122/j.issn.1673-4254.2022.01.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To investigate the mechanism of valproic acid (VPA) -induced impairment of the dendritic spines and synapses in the prefrontal cortex (PFC) for causing core symptoms of autism spectrum disorder (ASD) in mice. METHODS Female C57 mice were subjected to injections of saline or VPA on gestational days 10 and 12, and the male offspring mice in the two groups were used as the normal control group and ASD model group (n=10), respectively. Another 20 male mice with fetal exposure to VPA were randomized into two groups for stereotactic injection of DMSO or Wortmannin into the PFC (n=10). Open field test, juvenile play test and 3-chamber test were used to evaluate autistic behaviors of the mice. The density of dendrite spines in the PFC was observed with Golgi staining. Western blotting and immunofluorescence staining were used to detect the expressions of p-PI3K, PI3K, p-AKT, AKT, p-mTOR, mTOR and the synaptic proteins PSD95, p-Syn, and Syn in the PFC of the mice. RESULTS Compared with the normal control mice, the mice with fetal exposure to VPA exhibited obvious autism-like behaviors with significantly decreased density of total, mushroom and stubby dendritic spines (P < 0.05) and increased filopodia dendritic spines (P < 0.05) in the PFC. The VPA-exposed mice also showed significantly increased expressions of p-PI3K/PI3K, p-AKT/AKT, and p-mTOR/mTOR (P < 0.01) and lowered expressions of PSD95 and p-Syn/Syn in the PFC (P < 0.05 or 0.001). Wortmannin injection into the PFC obviously improved the ASD-like phenotype and dendritic spine development, down-regulated PI3K/Akt/mTOR signaling pathway and up-regulated the synaptic proteins in VPA-exposed mice. CONCLUSION In male mice with fetal exposure to VPA, excessive activation of PI3K/Akt/mTOR signaling pathway and decreased expressions of the synaptic proteins PSD95 and p-Syn cause dendritic spine damage and synaptic development disturbance in the PFC, which eventually leads to ASD-like phenotype.
Collapse
Affiliation(s)
- 菲菲 王
- 重庆医科大学基础医学院生理学教研室,重庆 400016Department of Physiology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - 路义 王
- 重庆医科大学基础医学院生理学教研室,重庆 400016Department of Physiology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - 玥 熊
- 重庆医科大学基础医学院生理学教研室,重庆 400016Department of Physiology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - 静 邓
- 重庆医科大学基础医学院生理学教研室,重庆 400016Department of Physiology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - 明其 吕
- 重庆医科大学实验教学管理中心,重庆 400016Experimental Teaching Management Center, Chongqing Medical University, Chongqing 400016, China
| | - 博诣 汤
- 重庆医科大学临床医学院,重庆 400016Clinical Medical College, Chongqing Medical University, Chongqing 400016, China
| | - 潇月 张
- 重庆医科大学临床医学院,重庆 400016Clinical Medical College, Chongqing Medical University, Chongqing 400016, China
| | - 英博 李
- 重庆医科大学基础医学院生理学教研室,重庆 400016Department of Physiology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
26
|
Necrostatin-1 Relieves Learning and Memory Deficits in a Zebrafish Model of Alzheimer's Disease Induced by Aluminum. Neurotox Res 2022; 40:198-214. [PMID: 34982355 DOI: 10.1007/s12640-021-00463-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 12/21/2022]
Abstract
Aluminum (Al) is considered one of the environmental risk factors for Alzheimer's disease (AD). The present study aims to establish a zebrafish AD model induced by Al and explore if necrostation-1 (Nec-1), a specific inhibitor of necroptosis, is effective in relieving learning and memory deficits in the zebrafish AD models. We treated adult zebrafish with aluminum trichloride at various doses for 1 month, followed by a T-maze test to evaluate learning and memory performance. Al concentration, levels of acetylcholine (Ach), and AD-related protein and gene expression in the brain tissue were evaluated in the zebrafish AD models. Our results demonstrated that in the brain tissue of Al-treated zebrafish, Al accumulated, Ach levels decreased, and AD-related genes and proteins increased. As a result, the learning and memory performance of Al-treated zebrafish was impaired. This suggested that a zebrafish AD model was established. To test the effect of Nec-1 on the zebrafish AD model, we added Nec-1 into the culture medium of the Al-treated adult zebrafish. The results demonstrated that Nec-1 could relive the learning and memory deficits, enhance Ach levels and the numbers of neural cells, and impact necroptosis-related gene expression. We concluded that Nec-1 could reverse Al-induced learning and memory impairment and had potential theoretical value in the zebrafish AD model.
Collapse
|
27
|
Zhao X, He C, Wang S, Lei Y, Niu Q. The association between blood lymphocyte NMDAR, group I mGluRs and cognitive function changes in occupationally aluminum-exposed workers and verification in rats. J Trace Elem Med Biol 2022; 69:126875. [PMID: 34673477 DOI: 10.1016/j.jtemb.2021.126875] [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: 04/30/2021] [Revised: 08/27/2021] [Accepted: 10/12/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Many studies have shown that occupational aluminum (Al) exposure could affect the cognitive functions of workers and cause mild cognitive impairment (MCI). Glutamate receptors (GluRs) play an important role in learning and memory functions. METHODS 352 workers in a large Al production enterprise were investigated in this research. MMSE, CDT, DST, VFT, FOM were used to evaluate the cognitive functions of workers. Plasma Al levels as exposure indices were measured by Graphite Furnace Atomic Absorption Method (GFAAS). The expression of GluRs was measured by ELISA. Cognitive function comprehensive scores were obtained through factor analysis. Then a rat model of chronic AlCl3 exposure was established. The detection method of Al levels and protein expression were the same as mentioned-above. RESULTS Compared with the Q1 group, the DST, VFT, and comprehensive cognitive function scores of the Q4 group were lower(P < 0.05). For every 1μg/L increase in plasma Al concentration, the risk of cognitive impairment increases 1.051 times (95 %CI:1.031,1.072). Both NMDAR1 and NMDAR2A protein expression level of Q1 group were higher than those of Q2, Q3, Q4 group (all P < 0.05). The mediating effect ratio of NMDAR1 between plasma Al levels and cognitive function comprehensive scores was a1*b1/c=11.30 %, and the mediating effect ratio of NMDAR2A was |a2*b2/c|=21.77 %. Compared with control group, the escape latency of rats in the high Al dose group was longer day by day (P < 0.05). With the increase of Al dose, the relative expression of NMDAR1, NMDAR2A, NMDAR2B, GluR1 and mGluR5 in cerebral cortex and lymphocytes of rats were decreased (P < 0.05). The result of correlation analysis on NMDAR1 protein expression between brain cortex and lymphocyte showed that the correlation coefficient is r = 0.646(P < 0.05). CONCLUSION Taking together the results from both Al exposed workers and animal, there is a certain correlation between NMDAR1 protein contents of brain cortex and peripheral lymphocytes. We propose that lymphocyte NMDAR1 could be considered as a peripheral potential marker of cognitive impairment for further observation.
Collapse
Affiliation(s)
- Xiaoyan Zhao
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Chanting He
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan, Shanxi, 030001, China; Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, Taiyuan, Shanxi, 030001, China; Department of Anatomy, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Shanshan Wang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Yang Lei
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan, Shanxi, 030001, China; Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, Taiyuan, Shanxi, 030001, China.
| |
Collapse
|
28
|
Pan B, Lu X, Han X, Huan J, Gao D, Cui S, Ju X, Zhang Y, Xu S, Song J, Wang L, Zhang H, Niu Q. Mechanism by Which Aluminum Regulates the Abnormal Phosphorylation of the Tau Protein in Different Cell Lines. ACS OMEGA 2021; 6:31782-31796. [PMID: 34870001 PMCID: PMC8637959 DOI: 10.1021/acsomega.1c04434] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/02/2021] [Indexed: 06/04/2023]
Abstract
Aluminum (Al) is an environmental neurotoxin to which humans are extensively exposed; however, the molecular mechanism of aluminum toxicity is unclear. Several studies have indicated that exposure to aluminum can cause abnormal phosphorylation of the tau protein. The purpose of this study was to investigate respectively the special molecular mechanism of abnormal regulation on synthesis and degradation of the tau protein induced by AlCl3 in cells of different species. The results of tau protein showed that the sites of abnormal tau phosphorylation induced by AlCl3 are Thr231, Ser262, and Ser396 in N2a cells. Meanwhile, the expressions of Thr181, Thr231, and Ser262 increased abnormally in SH-SY5Y cells. The result of the study showed that PP2A expression was high in N2a cells, while GSK-3β and PP2A in SH-SY5Y cells were involved in the synthesis process of abnormal tau phosphorylation induced by AlCl3. In N2a cells, the ubiquitin-proteasome pathway (UPP) mainly regulated tau phosphorylation at Ser262 and Ser396. Meanwhile, in SH-SY5Y cells, the UPP mainly regulated tau phosphorylation at Thr231 and Ser396. In summary, the UPP is involved in the degradation of Tau that is abnormally phosphorylated induced by AlCl3, but this process is site-specific and differs in cells of different species.
Collapse
Affiliation(s)
- Baolong Pan
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
- Sixth
Hospital of Shanxi Medical University (General Hospital of Tisco), Taiyuan 030001, China
| | - Xiaoting Lu
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Xiao Han
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Jiaping Huan
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Dan Gao
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Shuangjie Cui
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Xiaofen Ju
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Yunwei Zhang
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Shimeng Xu
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Jing Song
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Linping Wang
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Huifang Zhang
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Qiao Niu
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
29
|
Han Q, Jiang J, Yuan Y, Tang B, Zhang J. Bicaudal-C protein, a potential antidepressant target. Neuroreport 2021; 32:1293-1298. [PMID: 34554934 DOI: 10.1097/wnr.0000000000001729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Bicaudal-C protein is a highly conserved RNA binding protein, which contains K homology domains and sterile alpha motif domain. Genome-wide association study identified that Bicaudal-C protein was associated with depression. The expression of Bicaudal-C increased in depression patients, also increased expression of Bicaudal-C induces the behavior of depression. The decrease of synaptic plasticity plays a part in depression. Bicaudal-C protein reduces the synaptic plasticity of neurons via TrkB/mTOR/AMPA/pGluA1 pathways, Wnt pathway, or influencing some proteins related to synaptic plasticity. The decreased expression of Bicaudal-C plays an important role in the action of several antidepressants, such as ketamine, biperiden, and scopolamine. Therefore, Bicaudal-C protein may be a potential antidepressant target. Clarifying the relationship between Bicaudal-C protein and depression may help us to find new antidepressants. This review focuses on the research advances of the relationship between Bicaudal-C protein and depression.
Collapse
Affiliation(s)
- Qinghua Han
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | | | | | | | | |
Collapse
|
30
|
Cornell J, Salinas S, Huang HY, Zhou M. Microglia regulation of synaptic plasticity and learning and memory. Neural Regen Res 2021; 17:705-716. [PMID: 34472455 PMCID: PMC8530121 DOI: 10.4103/1673-5374.322423] [Citation(s) in RCA: 179] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Microglia are the resident macrophages of the central nervous system. Microglia possess varied morphologies and functions. Under normal physiological conditions, microglia mainly exist in a resting state and constantly monitor their microenvironment and survey neuronal and synaptic activity. Through the C1q, C3 and CR3 “Eat Me” and CD47 and SIRPα “Don’t Eat Me” complement pathways, as well as other pathways such as CX3CR1 signaling, resting microglia regulate synaptic pruning, a process crucial for the promotion of synapse formation and the regulation of neuronal activity and synaptic plasticity. By mediating synaptic pruning, resting microglia play an important role in the regulation of experience-dependent plasticity in the barrel cortex and visual cortex after whisker removal or monocular deprivation, and also in the regulation of learning and memory, including the modulation of memory strength, forgetfulness, and memory quality. As a response to brain injury, infection or neuroinflammation, microglia become activated and increase in number. Activated microglia change to an amoeboid shape, migrate to sites of inflammation and secrete proteins such as cytokines, chemokines and reactive oxygen species. These molecules released by microglia can lead to synaptic plasticity and learning and memory deficits associated with aging, Alzheimer’s disease, traumatic brain injury, HIV-associated neurocognitive disorder, and other neurological or mental disorders such as autism, depression and post-traumatic stress disorder. With a focus mainly on recently published literature, here we reviewed the studies investigating the role of resting microglia in synaptic plasticity and learning and memory, as well as how activated microglia modulate disease-related plasticity and learning and memory deficits. By summarizing the function of microglia in these processes, we aim to provide an overview of microglia regulation of synaptic plasticity and learning and memory, and to discuss the possibility of microglia manipulation as a therapeutic to ameliorate cognitive deficits associated with aging, Alzheimer’s disease, traumatic brain injury, HIV-associated neurocognitive disorder, and mental disorders.
Collapse
Affiliation(s)
- Jessica Cornell
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Shelbi Salinas
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Hou-Yuan Huang
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Miou Zhou
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
31
|
Zhang H, Cai X, Xiang C, Han Y, Niu Q. miR-29a and the PTEN-GSK3β axis are involved in aluminum-induced damage to primary hippocampal neuronal networks. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 224:112701. [PMID: 34461321 DOI: 10.1016/j.ecoenv.2021.112701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/17/2021] [Accepted: 08/23/2021] [Indexed: 06/13/2023]
Abstract
We previously reported that aluminum (Al) can cause a range of neurotoxic injuries including progressive irreversible synaptic structural damage and synaptic dysfunction, and eventually neuronal deaths. Mechanism of Al-induced electrophysiological and neuronal connectivity changes in neurons may indicate damage to the neuronal network. Here, mouse primary hippocampal neurons were cultured on micro-electrode array (MEA)- and high-content analysis (HCA)-related plates, showing that Al exposure significantly inhibited hippocampal neuronal electrical spike activity and neurite outgrowth characterized by a reduction in neurite branching and a decrease in the average total neurite length in relation to both Al dose and time of incubation. In recent years, miR-29a/ phosphatase and tensin homolog (PTEN) have been found to play pivotal roles in the morphogenesis of neurons, it has been confirmed in vitro and in vivo that the PTEN-Glycogen synthase kinase-3β (GSK-3β) axis regulates neurite outgrowth. The present study demonstrated that increases in Al exposure and dose gradually reduce miR-29a expression. Up-regulation of miR-29a in the hippocampal neurons by lentivirus transfection reversed the decrease in electrical spike activity and the reduction in both neurite branching and length induced by Al. Moreover, miR-29a suppressed the expression of PTEN and increased the level of phosphorylated Protein Kinase B (p-AKT) and p-GSK-3β which were inhibited by the Al treatment. This suggests that miR-29a is critically involved in the functional and structural neuronal damage induced by Al and is a potential target for Al neurotoxicity. Moreover, the reduction of neurite length and branching induced by Al exposure was regulated by miR-29a and its target neuronal PTEN-GSK3β signaling pathway, which also represents a possible mechanism of Al-induced the inhibition of the electrical activity. Collectively, Al-induced damage to the neuronal network occurred through miR-29a-mediated alterations of the PTEN-GSK3β signaling pathway.
Collapse
Affiliation(s)
- Huifang Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China.
| | - Xiaoya Cai
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China
| | - Changxin Xiang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China
| | - Yingchao Han
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China.
| |
Collapse
|
32
|
Molecular mechanisms of aluminum neurotoxicity: Update on adverse effects and therapeutic strategies. ADVANCES IN NEUROTOXICOLOGY 2021; 5:1-34. [PMID: 34263089 DOI: 10.1016/bs.ant.2020.12.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
33
|
Huang S, Yuan H, Li W, Liu X, Zhang X, Xiang D, Luo S. Polygonatum sibiricum Polysaccharides Protect against MPP-Induced Neurotoxicity via the Akt/mTOR and Nrf2 Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8843899. [PMID: 33520086 PMCID: PMC7817274 DOI: 10.1155/2021/8843899] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/15/2020] [Accepted: 12/31/2020] [Indexed: 12/20/2022]
Abstract
Polygonatum sibiricum, a well-known life-prolonging tonic in Chinese medicine, has been widely used for nourishing nerves in the orient, but the underlying molecular mechanisms remain unclear. In this study, we found that P. sibiricum polysaccharides (PSP) ameliorated 1-methyl-4-phenyl-1,2.3,6-tetrahydropyridine- (MPTP-) induced locomotor activity deficiency and dopaminergic neuronal loss in an in vivo Parkinson's disease (PD) mouse model. Additionally, PSP pretreatment inhibited N-methyl-4-phenylpyridine (MPP+) induced the production of reactive oxygen species, increasing the ratio of reduced glutathione/oxidized glutathione. In vitro experiments showed that PSP promoted the proliferation of N2a cells in a dose-dependent manner, while exhibiting effects against oxidative stress and neuronal apoptosis elicited by MPP+. These effects were found to be associated with the activation of Akt/mTOR-mediated p70S6K and 4E-BP1 signaling pathways, as well as nuclear factor erythroid 2-related factor 2- (Nrf2-) mediated NAD(P)H quinone oxidoreductase 1 (NQO1), heme oxygenase-1 (HO-1), glutamate-cysteine ligase catalytic subunit (Gclc), and glutamate-cysteine ligase modulatory subunit (Gclm), resulting in antiapoptotic and antioxidative effects. Meanwhile, PSP exhibited no chronic toxicity in C57BJ/6 mice. Together, our results suggest that PSP can serve as a promising therapeutic candidate with neuroprotective properties in preventing PD.
Collapse
Affiliation(s)
- Si Huang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
| | - Haiyan Yuan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
| | - Xinyi Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
| | - Xiaojie Zhang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Daxiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
| | - Shilin Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
| |
Collapse
|
34
|
Pan B, Li Y, Zhang J, Zhou Y, Li L, Xue X, Li H, Niu Q. Role of mGluR 1 in synaptic plasticity impairment induced by maltol aluminium in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 78:103406. [PMID: 32438325 DOI: 10.1016/j.etap.2020.103406] [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: 01/11/2020] [Revised: 04/27/2020] [Accepted: 04/30/2020] [Indexed: 06/11/2023]
Abstract
The main symptoms of Alzheimer's disease (AD) is the loss of learning and memory ability, of which biological basis is synaptic plasticity. Aluminium has been found to cause changes in synaptic plasticity, but its molecular mechanism was unclear. In this study, Sprague-Dawley rats were injected with aluminium maltol (Al(mal)3) through the lateral ventricle to establish an AD-like model. Y-maze, electrophysiological measurements, Golgi staining, scanning electron microscopy, quantitative real-time polymerase chain reaction, and western blot techniques were used to investigate regulation of the metabolic glutamate receptor 1 (mGluR1) in synaptic plasticity impairment induced by Al(mal)3. The results showed that Al(mal)3 inhibited the induction and maintenance of long-term potentiation in the hippocampal CA1 region. During this process, the expression of mGluR1 was up-regulated and it inhibited the expression and phosphorylation of the N-methyl-D-aspartic acid receptors (NMDARs). This mainly affected NMDAR1 and NMDAR2B but did not affect protein kinase C expression.
Collapse
Affiliation(s)
- Baolong Pan
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; General Hospital of Tisco, Sixth Hospital of Shanxi Medical University, Taiyuan 030001,PR China
| | - Yaqin Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China
| | - Jingsi Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China
| | - Yue Zhou
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China
| | - Liang Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China
| | - Xingli Xue
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China
| | - Huan Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, China.
| |
Collapse
|
35
|
Shang N, Zhang P, Wang S, Chen J, Fan R, Chen J, Huang T, Wang Y, Duncan J, Zhang L, Niu Q, Zhang Q. Aluminum-Induced Cognitive Impairment and PI3K/Akt/mTOR Signaling Pathway Involvement in Occupational Aluminum Workers. Neurotox Res 2020; 38:344-358. [PMID: 32506341 DOI: 10.1007/s12640-020-00230-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/20/2020] [Accepted: 05/25/2020] [Indexed: 01/17/2023]
Abstract
Epidemiological studies indicate that long-term occupational exposure to aluminum (Al) causes neurotoxicity and cognitive impairment. While the molecular underpinnings associated with workers' cognitive impairment is unclear, one mechanism may involve Al-induced PI3K/Akt/mTOR activation and neuronal cell death, which impairs learning and memory in rats. Here, we sought to determine whether PI3K/Akt/mTOR is also associated with cognitive impairment in Al-exposed occupational workers. Cognitive function was screened by Mini-Mental State Examination (MMSE) and Clock-Drawing Test (CDT), and serum Al and PI3K/Akt/mTOR-associated gene expression was quantified. A negative correlation between serum Al and scores of MMSE and CDT was found, which might relate with downregulation of PI3K/Akt/mTOR. To determine the role of the PI3K/Akt/mTOR pathway cognitive function, we treated zebrafish with Al and observed a profound impairment in learning and memory. Increased brain Al levels was associated with decreased expression of PI3K/Akt/mTOR in Al-exposed zebrafish. Finally, rapamycin, an mTOR inhibitor, was added to isolate the role of mTOR specifically in the Al exposed zebrafish. The results suggested that Al induces learning and memory deficits by downregulating PI3K, Akt, and mTOR1 expression and inducing neuronal cell death like rapamycin group. This study indicates that aluminum exposure can cause cognitive impairment through PI3K/Akt/mTOR pathway, with mTOR activity being a critical player involved in this mechanism. Future studies are necessary to further characterize the role of PI3K/Akt/mTOR1 signaling in Al-induced neurocognitive decline among Al occupational workers. These findings draw attention to Al risk exposure among occupational workers and the need to implement novel safety and protective measures to mitigate neurocognitive health risks in the Al industrial workspace.
Collapse
Affiliation(s)
- Nan Shang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Department of Pharmacy, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Ping Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Shuo Wang
- Department of Physical and Chemical, Beijing Chaoyang District Center for Disease Control and Prevention, Beijing, 100021, China
| | - Jianping Chen
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Rong Fan
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jin Chen
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Tao Huang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yanhong Wang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jeremy Duncan
- Department of Physiology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Ling Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Qinli Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China. .,Department of Pathology, University of Mississippi Medical Center, Jackson, MS, 39216, USA.
| |
Collapse
|