1
|
Shi S, Sun Y, Zan G, Zhao M. The interaction between central and peripheral immune systems in methamphetamine use disorder: current status and future directions. J Neuroinflammation 2025; 22:40. [PMID: 39955589 PMCID: PMC11829452 DOI: 10.1186/s12974-025-03372-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/07/2025] [Indexed: 02/17/2025] Open
Abstract
Methamphetamine (METH) use disorder (MUD) is characterized by compulsive drug-seeking behavior and substantial neurotoxicity, posing a considerable burden on individuals and society. Traditionally perceived as a localized central nervous system disorder, recent preclinical and clinical studies have elucidated that MUD is a multifaceted disorder influenced by various biological systems, particularly the immune system. Emerging evidence suggests that both central and peripheral immune responses play a crucial role in the initiation and persistence of MUD. Conceptualizing it as a systemic immune process prompts significant inquiries regarding the mechanisms of communication between peripheral and central compartments. Also, whether this intercommunication could serve as diagnostic biomarkers or therapeutic targets. This review begins by offering an overview of mechanistic studies pertaining to the neuroimmune and peripheral immune systems. Finally, future directions are suggested through the integration of innovative technologies and multidimensional data to promote the translation of basic mechanistic research into clinical diagnostic and therapeutic interventions.
Collapse
Affiliation(s)
- Sai Shi
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiwen Sun
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guiying Zan
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Min Zhao
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China.
- CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Sciences, Shanghai, China.
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wan Ping Road, Shanghai, 200030, China.
| |
Collapse
|
2
|
Ding J, Sun B, Gao Y, Zheng J, Liu C, Huang J, Jia N, Pei X, Jiang X, Hu S, Xia B, Meng Y, Dai Z, Qi X, Wang J. Evidence for chromium crosses blood brain barrier from the hypothalamus in chromium mice model. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 273:116179. [PMID: 38460200 DOI: 10.1016/j.ecoenv.2024.116179] [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: 12/26/2023] [Revised: 02/27/2024] [Accepted: 03/03/2024] [Indexed: 03/11/2024]
Abstract
It has been shown that exposure to hexavalent Chromium, Cr (Ⅵ), via nasal cavity can have neurotoxicological effects and induces behavioral impairment due to the fact that blood brain barrier (BBB) does not cover olfactory bulb. But whether Cr (Ⅵ) can cross the BBB and have a toxicological effects in central nervous system (CNS) remains unclear. Therefore, we investigated the effects of Cr (Ⅵ) on mice treated with different concentrations and exposure time (14 days and 28 days) of Cr (Ⅵ) via intraperitoneal injection. Results revealed that Cr accumulated in hypothalamus (HY) in a timely dependent manner. Much more severer neuropathologies was observed in the group of mice exposed to Cr (Ⅵ) for 28 days than that for 14 days. Gliosis, neuronal morphological abnormalities, synaptic degeneration, BBB disruption and neuronal number loss were observed in HY. In terms of mechanism, the Nrf2 related antioxidant stress signaling dysfunction and activated NF-κB related inflammatory pathway were observed in HY of Cr (Ⅵ) intoxication mice. And these neuropathologies and signaling defects appeared in a timely dependent manner. Taking together, we proved that Cr (Ⅵ) can enter HY due to weaker BBB in HY and HY is the most vulnerable CNS region to Cr (Ⅵ) exposure. The concentration of Cr in HY increased along with time. The accumulated Cr in HY can cause BBB disruption, neuronal morphological abnormalities, synaptic degeneration and gliosis through Nrf2 and NF-κB signaling pathway. This finding improves our understanding of the neurological dysfunctions observed in individuals who have occupational exposure to Cr (Ⅵ), and provided potential therapeutic targets to treat neurotoxicological pathologies induced by Cr (Ⅵ).
Collapse
Affiliation(s)
- Jiuyang Ding
- Key Laboratory of Human Brain bank for Functions and Diseases of Department of Education of Guizhou Province, Guizhou Medical University, Guiyang 550025, China; Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang 550004, China; School of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Baofei Sun
- Key Laboratory of Human Brain bank for Functions and Diseases of Department of Education of Guizhou Province, Guizhou Medical University, Guiyang 550025, China
| | - Yingdong Gao
- Department of Reproductive Medicine, Taian Maternity and Child Health Hospital, Taian 271000, China
| | - Juan Zheng
- Department of Reproductive Medicine, Taian Maternity and Child Health Hospital, Taian 271000, China
| | - Changyou Liu
- Department of Pediatrics, Taian Maternity and Child Health Hospital, Taian 271000, China
| | - Jian Huang
- School of Forensic Medicine, Kunming Medical University, Kunming 650500, China
| | - Nannan Jia
- Neonatal Screening Center, Taian Maternity and Child Health Hospital, Taian, China
| | - Xianglin Pei
- School of Materials and Architectural Engineering, Guizhou Normal University, Guiyang 550025, China
| | - Xueyu Jiang
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Shanshan Hu
- Good Clinical Practice Center, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
| | - Bing Xia
- School of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Yunle Meng
- Institute of Forensic Science, Ministry of Public Security, Beijing 100038, China
| | - Zhuihui Dai
- State Key Laboratory of Ore Deposit Geochemistry, Institute of Geochemistry, Chinese Academy of Sciences, Guiyang 550081, China.
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang 550004, China.
| | - Jiawen Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China.
| |
Collapse
|
3
|
Ding J, Wu J, Hou X, Yang L, Gao Y, Zheng J, Jia N, He Z, Zhang H, Wang C, Qi X, Huang J, Pei X, Wang J. α-synuclein-lack expression rescues methamphetamine-induced mossy fiber degeneration in dorsal hippocampal CA3. Neurotoxicology 2024; 101:36-45. [PMID: 38311184 DOI: 10.1016/j.neuro.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 01/20/2024] [Accepted: 01/31/2024] [Indexed: 02/09/2024]
Abstract
Methamphetamine (METH) - induced cognitive impairments may be related to synaptic degeneration at mossy fiber terminals, critical for spatial memory formation in hippocampal circuits. We have previously found METH-induced neurodegeneration in the striatum by increasing the α-synuclein (α-SYN) level. However, whether and how the METH-induced mossy fiber degeneration is also blamed for the abnormal accumulation of α-SYN remains to be elucidated. Chronic METH exposure decreased mossy fiber density but upregulatedα-SYN and phosphorylated TAU (TAU-pSer396) in hippocampal CA3, associated with glial cell overactivation, axonal neuropathies, and memory impairment. Notably, the knockout of the α-SYN gene significantly alleviated the METH-induced mossy fiber degeneration and memory impairment. Meanwhile, the TAU-pSer396 accumulation and glial activation were ameliorated by α-SYN knockout. Our findings suggest an essential role of α-SYN in mediating METH-induced mossy fiber degeneration, providing promising therapeutic and prophylactic targets for METH-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Jiuyang Ding
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China; Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Jun Wu
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Xiaotao Hou
- Guangzhou KingMed Center for Clinical Laboratory Co., Ltd., Guangzhou, China; Guangdong Provincial Key Laboratory of Genetic Disease Diagnostic, Guangzhou, China
| | - Li Yang
- Department of Reproductive Medicine, Taian Maternity and Child Health Hospital, Taian, China
| | - Yingdong Gao
- Department of Reproductive Medicine, Taian Maternity and Child Health Hospital, Taian, China
| | - Juan Zheng
- Department of Reproductive Medicine, Taian Maternity and Child Health Hospital, Taian, China
| | - Nannan Jia
- Neonatal Screening Center, Taian Maternity and Child Health Hospital, Taian, China
| | - Zheng He
- Neonatal Screening Center, Taian Maternity and Child Health Hospital, Taian, China
| | - Hui Zhang
- Department of Reproductive Medicine, Taian Maternity and Child Health Hospital, Taian, China
| | - Chengfei Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Jiang Huang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Xianglin Pei
- School of Materials and Architectural Engineering, Guizhou Normal University, Guiyang China.
| | - Jiawen Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China.
| |
Collapse
|
4
|
Hu S, Huang X, Huang J, Qian Y, Tian Y, Xiao Y, Qi X, Zhou X, Yang Z, Chen Z. Iron chelation prevents nigrostriatal neurodegeneration in a chronic methamphetamine mice model. Neurotoxicology 2023; 99:24-33. [PMID: 37717738 DOI: 10.1016/j.neuro.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/31/2023] [Accepted: 09/12/2023] [Indexed: 09/19/2023]
Abstract
Methamphetamine (METH) has been established to selectively target and impair dopaminergic neurons through multiple pathways. Ferroptosis is a unique form of non-apoptotic cell death driven by cellular iron accumulation-induced lipid peroxidation. Nonetheless, it remains unclear whether METH can induce ferroptosis. In the present study, we sought to assess alterations in iron levels after chronic METH exposure and reveal the modulatory role of iron on METH-induced pathologies. Importantly, we demonstrated that METH increased iron deposition in the nigrostriatal system, including the substantia nigra (SN) and caudate putamen (CPu). Moreover, decreases in GPx4 levels, increases in lipid peroxidation products, and pathological alterations were observed in the nigrostriatal system as a consequence of chronic METH exposure. The iron chelator deferiprone not only alleviated nigrostriatal iron deposition, dopaminergic cell death, and lipid peroxidation, but alsoattenuated the decreases in GPx4 induced by METH. These findings suggest an alleviation of ferroptosis in dopaminergic neurons. In addition, we found that the ferroptosis inhibitor liproxstatin-1 attenuated METH-induced dopaminergic degeneration in the nigrostriatal system. Our findings corroborated that METH might induce dopaminergic neurodegeneration through iron-dependent ferroptosis. Interestingly, reducing iron levels or inhibiting ferroptosis may alleviate METH-induced dopaminergic neurodegeneration.
Collapse
Affiliation(s)
- Shanshan Hu
- Good Clinical Practice Center, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Xiaorong Huang
- Good Clinical Practice Center, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Jian Huang
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China; NHC Key Laboratory of Drug Addiction Medicine,Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming 650500, China
| | - Ying Qian
- Department of Pharmacy, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Yingbiao Tian
- Department of Pharmacy, The Second Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
| | - Ye Xiao
- Good Clinical Practice Center, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang 550004, China
| | - Xiaoxian Zhou
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
| | - Zhusheng Yang
- School of Materials and Architectural Engineering, Guizhou Normal University, Guiyang 550025, China.
| | - Zehui Chen
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China.
| |
Collapse
|
5
|
Loftis JM, Ramani S, Firsick EJ, Hudson R, Le-Cook A, Murnane KS, Vandenbark A, Shirley RL. Immunotherapeutic treatment of inflammation in mice exposed to methamphetamine. Front Psychiatry 2023; 14:1259041. [PMID: 38025429 PMCID: PMC10666795 DOI: 10.3389/fpsyt.2023.1259041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/15/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Currently, there are no FDA-approved medications to treat methamphetamine addiction, including the inflammatory, neurotoxic, and adverse neuropsychiatric effects. We have shown that partial (p)MHC class II constructs (i.e., Recombinant T-cell receptor Ligand - RTL1000), comprised of the extracellular α1 and β1 domains of MHC class II molecules linked covalently to myelin oligodendrocyte glycoprotein (MOG)-35-55 peptide, can address the neuroimmune effects of methamphetamine addiction through its ability to bind to and down-regulate CD74 expression, block macrophage migration inhibitory factor (MIF) signaling, and reduce levels of pro-inflammatory chemokine ligand 2 (CCL2). The present study evaluated the effects of our third-generation pMHC II construct, DRmQ, on cognitive function and concentration of inflammatory cytokines in the frontal cortex, a region critical for cognitive functions such as memory, impulse control, and problem solving. Methods Female and male C57BL/6J mice were exposed to methamphetamine (or saline) via subcutaneous (s.c.) injections administered four times per day every other day for 14 days. Following methamphetamine exposure, mice received immunotherapy (DRmQ or ibudilast) or vehicle s.c. injections daily for five days. Cognitive function was assessed using the novel object recognition test (NORT). To evaluate the effects of immunotherapy on inflammation in the frontal cortex, multiplex immunoassays were conducted. ANOVA was used to compare exploration times on the NORT and immune factor concentrations. Results Post hoc analysis revealed increased novel object exploration time in MA-DRmQ treated mice, as compared to MA-VEH treated mice (non-significant trend). One-way ANOVA detected a significant difference across the groups in the concentration of macrophage inflammatory protein-2 (MIP-2) (p = 0.03). Post hoc tests indicated that mice treated with methamphetamine and DRmQ or ibudilast had significantly lower levels of MIP-2 in frontal cortex, as compared to mice treated with methamphetamine and vehicle (p > 0.05). Discussion By specifically targeting CD74, our DRQ constructs can block the signaling of MIF, inhibiting the downstream signaling and pro-inflammatory effects that contribute to and perpetuate methamphetamine addiction.
Collapse
Affiliation(s)
- Jennifer M. Loftis
- Research and Development Service, Veterans Affairs Portland Health Care System, Portland, OR, United States
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
- Methamphetamine Research Center, Oregon Health & Science University, Portland, OR, United States
| | - Sankrith Ramani
- Research and Development Service, Veterans Affairs Portland Health Care System, Portland, OR, United States
| | - Evan J. Firsick
- Research and Development Service, Veterans Affairs Portland Health Care System, Portland, OR, United States
| | - Rebekah Hudson
- Research and Development Service, Veterans Affairs Portland Health Care System, Portland, OR, United States
| | - Anh Le-Cook
- Research and Development Service, Veterans Affairs Portland Health Care System, Portland, OR, United States
| | - Kevin S. Murnane
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, United States
- Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, United States
- Department of Psychiatry and Behavioral Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, United States
| | - Arthur Vandenbark
- Research and Development Service, Veterans Affairs Portland Health Care System, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, United States
| | | |
Collapse
|
6
|
Wan L, He W, Wang YY, Xu Y, Lu Q, Zhang MN, Wang QH, Dun S, Liu LY, Shi XY, Wang J, Hu LY, Zhang B, Yang G, Zou LP. Vigabatrin-associated brain abnormalities on MRI in tuberous sclerosis complex patients with infantile spasms: are they preventable? Ther Adv Neurol Disord 2022; 15:17562864221138148. [PMID: 36601084 PMCID: PMC9806385 DOI: 10.1177/17562864221138148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 10/22/2022] [Indexed: 12/28/2022] Open
Abstract
Background Vigabatrin (VGB) is currently the most widely prescribed first-line medication for individuals with infantile spasms (IS) and especially for those with tuberous sclerosis complex (TSC), with demonstrated efficacy. Meanwhile, its adverse events, such as vigabatrin-associated brain abnormalities on magnetic resonance imaging (MRI; VABAM), have also been widely reported. Objectives The objectives of this study were to observe the occurrences of VABAM in patients with IS caused by TSC (IST) and further explore the associated risk factors. Methods Children with IS receiving VGB were recruited from our institution; clinical, imaging, and medication data were collected. Cerebral MRI was reviewed to determine the occurrence of VABAM. Group comparisons (IS caused by TSC and other etiologies) were performed; subgroup analyses on IST were also performed. Next, a retrospective cohort study of children taking VGB was conducted to explore risk/protective factors associated with VABAM. Results The study enrolled 172 children with IS who received VGB. VABAM was observed in 38 patients (22.1%) with a peak dosage of 103.5 ± 26.7 mg/kg/day. Subsequent analysis found the incidence of VABAM was significantly lower in the 80 patients with IST than in the 92 patients with IS caused by other etiologies (10% versus 32.6%, p-value < 0.001). In subgroup analyses within the IST cohort, VABAM was significantly lower in children who received concomitant rapamycin therapy. Univariate and multivariate logistic regression analysis of the 172 IS children showed that treatment with rapamycin was the independent factor associated with a lower risk of VABAM; similar results were observed in the survival analysis. Conclusion The incidence of VABAM was significantly lower in IST patients. Further research is needed to examine the mechanisms that underlie this phenomenon and to determine if treatment with rapamycin may reduce the risk of VABAM.
Collapse
Affiliation(s)
| | | | | | - Yong Xu
- Department of Pediatrics, PLA General Hospital,
Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China
| | - Qian Lu
- Department of Pediatrics, PLA General
Hospital, Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China
| | - Meng-Na Zhang
- Department of Pediatrics, PLA General
Hospital, Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China
| | - Qiu-Hong Wang
- Department of Pediatrics, PLA General
Hospital, Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China
| | - Shuo Dun
- Department of Pediatrics, PLA General
Hospital, Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China
| | - Li-Ying Liu
- Department of Pediatrics, PLA General
Hospital, Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China
| | - Xiu-Yu Shi
- Department of Pediatrics, PLA General
Hospital, Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China,The Second School of Clinical Medicine,
Southern Medical University, Guangzhou, China
| | - Jing Wang
- Department of Pediatrics, PLA General
Hospital, Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China
| | - Lin-Yan Hu
- Department of Pediatrics, PLA General
Hospital, Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China
| | - Bo Zhang
- Department of Neurology and ICCTR
Biostatistics and Research Design Center, Boston Children’s Hospital,
Harvard Medical School, Boston, MA, USA
| | - Guang Yang
- Department of Pediatrics, PLA General
Hospital, Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China,Medical School of Chinese People’s Liberation
Army, Beijing, China,The Second School of Clinical Medicine,
Southern Medical University, Guangzhou, China
| | | |
Collapse
|
7
|
Ding J, Huang J, Tang X, Shen L, Hu S, He J, Liu T, Yu Z, Liu Y, Wang Q, Wang J, Zhao N, Qi X, Huang J. Low and high dose methamphetamine differentially regulate synaptic structural plasticity in cortex and hippocampus. Front Cell Neurosci 2022; 16:1003617. [PMID: 36406748 PMCID: PMC9666390 DOI: 10.3389/fncel.2022.1003617] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/19/2022] [Indexed: 03/24/2024] Open
Abstract
Psychostimulants, such as methamphetamine (METH) can induce structural remodeling of synapses by remodeling presynaptic and postsynaptic morphology. Escalating or long-lasting high dose METH accounts for neurodegeneration by targeting multiple neurotransmitters. However, the effects of low dose METH on synaptic structure and the modulation mechanism remain elusive. This study aims to assess the effects of low dose (2 mg/kg) and high dose (10 mg/kg) of METH on synaptic structure alternation in hippocampus and prefrontal cortex (PFC) and to reveal the underlying mechanism involved in the process. Low dose METH promoted spine formation, synaptic number increase, post-synaptic density length elongation, and memory function. High dose of METH induced synaptic degeneration, neuronal number loss and memory impairment. Moreover, high dose, but not low dose, of METH caused gliosis in PFC and hippocampus. Mechanism-wise, low dose METH inactivated ras-related C3 botulinum toxin substrate 1 (Rac1) and activated cell division control protein 42 homolog (Cdc42); whereas high dose METH inactivated Cdc42 and activated Rac1. We provided evidence that low and high doses of METH differentially regulate synaptic plasticity in cortex and hippocampus.
Collapse
Affiliation(s)
- Jiuyang Ding
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Jian Huang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Xiang Tang
- Department of Children Rehabilitation, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Lingyi Shen
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Shanshan Hu
- Good Clinical Practice Center, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiaojiao He
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Ting Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, China
| | - Zhixing Yu
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Yubo Liu
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Qiyan Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Jiawen Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Na Zhao
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Jiang Huang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
| |
Collapse
|
8
|
Huang J, Ding J, Wang X, Gu C, He Y, Li Y, Fan H, Xie Q, Qi X, Wang Z, Qiu P. Transfer of neuron-derived α-synuclein to astrocytes induces neuroinflammation and blood-brain barrier damage after methamphetamine exposure: Involving the regulation of nuclear receptor-associated protein 1. Brain Behav Immun 2022; 106:247-261. [PMID: 36089218 DOI: 10.1016/j.bbi.2022.09.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 09/01/2022] [Accepted: 09/04/2022] [Indexed: 10/31/2022] Open
Abstract
The α-synuclein (α-syn) is involved in methamphetamine (METH)-induced neurotoxicity. Neurons can transfer excessive α-syn to neighboring neurons and glial cells. The effects of α-syn aggregation in astrocytes after METH exposure on the blood-brain barrier (BBB) remains unclear. Our previous study demonstrated that nuclear receptor-related protein 1 (Nurr1), a member of the nuclear receptor family widely expressed in the brain, was involved in the process of METH-induced α-syn accumulated in astrocytes to activate neuroinflammation. The role Nurr1 plays in astrocyte-mediated neuroinflammation, which results in BBB injury induced by METH, remains uncertain. This study found that METH up-regulated α-syn expression in neurons extended to astrocytes, thereby eliciting astrocyte activation, increasing and decreasing IL-1β, IL-6, TNF-α, and GDNF levels by down-regulating Nurr1 expression, and ultimately damaging the BBB. Specifically, the permeability of BBB to Evans blue and sodium fluorescein (NaF) increased; IgG deposits in the brain parenchyma increased; the Claudin5, Occludin, and PDGFRβ levels decreased. Several ultrastructural pathological changes occurred in the BBB, such as abnormal cerebral microvascular diameter, astrocyte end-foot swelling, decreased pericyte coverage, and loss of tight junctions. However, knockout or inhibition of α-syn or astrocyte-specific overexpression of Nurr1 partially alleviated these symptoms and BBB injury. Moreover, the in vitro experiments confirmed that METH increased α-syn level in the primary cultured neurons, which could be further transferred to primary cultured astrocytes, resulting in decreased Nurr1 levels. The decreased Nurr1 levels mediated the increase of IL-1β, IL-6, and TNF-α, and the decrease of GDNF, thereby changing the permeability to NaF, transendothelial electrical resistance, and Claudin5 and Occludin levels of primary cultured brain microvascular endothelial cells. Based on our findings, we proposed a new mechanism to elucidate METH-induced BBB injury and presented α-syn and Nurr1 as promising drug intervention targets to reduce BBB injury and resulting neurotoxicity in METH abusers.
Collapse
Affiliation(s)
- Jian Huang
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Jiuyang Ding
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China; Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Xiaohan Wang
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Cihang Gu
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Yitong He
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Yanning Li
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Haoliang Fan
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Qiqian Xie
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Zhuo Wang
- School of Medicine, South China University of Technology, Guangzhou, China; Department of Infertility and Sexual Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Pingming Qiu
- School of Forensic Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Icariside II Attenuates Methamphetamine-Induced Neurotoxicity and Behavioral Impairments via Activating the Keap1-Nrf2 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8400876. [PMID: 35387263 PMCID: PMC8979738 DOI: 10.1155/2022/8400876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/26/2021] [Accepted: 03/11/2022] [Indexed: 12/15/2022]
Abstract
Chronic and long-term methamphetamine (METH) abuse is bound to cause damages to multiple organs and systems, especially the central nervous system (CNS). Icariside II (ICS), a type of flavonoid and one of the main active ingredients of the traditional Chinese medicine Epimedium, exhibits a variety of biological and pharmacological properties such as anti-inflammatory, antioxidant, and anticancer activities. However, whether ICS could protect against METH-induced neurotoxicity remains unknown. Based on a chronic METH abuse mouse model, we detected the neurotoxicity after METH exposure and determined the intervention effect of ICS and the potential mechanism of action. Here, we found that METH could trigger neurotoxicity, which was characterized by loss of dopaminergic neurons, depletion of dopamine (DA), activation of glial cells, upregulation of α-synuclein (α-syn), abnormal dendritic spine plasticity, and dysfunction of motor coordination and balance. ICS treatment, however, alleviated the above-mentioned neurotoxicity elicited by METH. Our data also indicated that when ICS combated METH-induced neurotoxicity, it was accompanied by partial correction of the abnormal Kelch 2 like ECH2 associated protein 1 (Keap1)-nuclear factor erythroid-2-related factor 2 (Nrf2) pathway and oxidative stress response. In the presence of ML385, an inhibitor of Nrf2, ICS failed to activate the Nrf2-related protein expression and reduce the oxidative stress response. More importantly, ICS could not attenuate METH-induced dopaminergic neurotoxicity and behavioral damage when the Nrf2 was inhibited, suggesting that the neuroprotective effect of ICS on METH-induced neurotoxicity was dependent on activating the Keap1-Nrf2 pathway. Although further research is needed to dig deeper into the actual molecular targets of ICS, it is undeniable that the current results imply the potential value of ICS to reduce the neurotoxicity of METH abusers.
Collapse
|
10
|
Occurrence of Total and Proteinase K-Resistant Alpha-Synuclein in Glioblastoma Cells Depends on mTOR Activity. Cancers (Basel) 2022; 14:cancers14061382. [PMID: 35326535 PMCID: PMC8946689 DOI: 10.3390/cancers14061382] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/01/2022] [Accepted: 03/07/2022] [Indexed: 01/18/2023] Open
Abstract
Simple Summary The accumulation of alpha-synuclein (α-syn) is considered a pathological hallmark of the neurodegenerative disorders known as synucleinopathies. The clearance of α-syn depends on autophagy activity, which is inhibited by the mechanistic target of rapamycin (mTOR). Thus, it is likely that α-syn accumulation may occur whenever mTOR is overactive and autophagy is suppressed. In fact, the lack of effective autophagy increases the amount of α-syn and may produce protein aggregation. Therefore, in the present study, we questioned whether cells from glioblastoma multiforme (GBM), a lethal brain neoplasm, wherein mTOR is upregulated and autophagy is suppressed, may overexpress α-syn. In fact, a large quantity of α-syn is measured in GBM cells compared with astrocytes, which includes proteinase K-resistant α-syn. Rapamycin, while inhibiting mTOR activity, significantly reduces the amount of α-syn and allocates α-syn within autophagy-like vacuoles. Abstract Alpha-synuclein (α-syn) is a protein considered to be detrimental in a number of degenerative disorders (synucleinopathies) of which α-syn aggregates are considered a pathological hallmark. The clearance of α-syn strongly depends on autophagy, which can be stimulated by inhibiting the mechanistic target of rapamycin (mTOR). Thus, the overexpression of mTOR and severe autophagy suppression may produce α-syn accumulation, including the proteinase K-resistant protein isoform. Glioblastoma multiforme (GBM) is a lethal brain tumor that features mTOR overexpression and severe autophagy inhibition. Cell pathology in GBM is reminiscent of a fast, progressive degenerative disorder. Therefore, the present work questions whether, as is analogous to neurons during degenerative disorders, an overexpression of α-syn occurs within GBM cells. A high amount of α-syn was documented in GBM cells via real-time PCR (RT-PCR), Western blotting, immunohistochemistry, immuno-fluorescence, and ultrastructural stoichiometry, compared with the amount of β- and γ-synucleins and compared with the amount of α-syn counted within astrocytes. The present study indicates that (i) α-syn is overexpressed in GBM cells, (ii) α-syn expression includes a proteinase-K resistant isoform, (iii) α-syn is dispersed from autophagy-like vacuoles to the cytosol, (iv) α-syn overexpression and cytosol dispersion are mitigated by rapamycin, and (v) the α-syn-related GBM-like phenotype is mitigated by silencing the SNCA gene.
Collapse
|