1
|
Han Z, Song Y, Qin C, Zhou H, Han D, Yan S, Ni H. S-Nitrosylation of Dexras1 Controls Post-Stroke Recovery via Regulation of Neuronal Excitability and Dendritic Remodeling. CNS Neurosci Ther 2025; 31:e70199. [PMID: 39749632 PMCID: PMC11696243 DOI: 10.1111/cns.70199] [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: 10/09/2024] [Revised: 11/29/2024] [Accepted: 12/15/2024] [Indexed: 01/04/2025] Open
Abstract
AIMS Stroke is a major public health concern leading to high rates of death and disability worldwide, unfortunately with no effective treatment available for stroke recovery during the repair phase. METHODS Photothrombotic stroke was induced in mice. Adeno-associated viruses (AAV) were microinjected into the peri-infarct cortex immediately after photothrombotic stroke. Grid-walking task and cylinder task were used to assess motor function. Western blotting, Golgi staining, and electrophysiology recordings were performed to uncover the mechanisms. RESULTS The ternary complex of neuronal nitric oxide synthase (nNOS), carboxy-terminal PDZ ligand of nNOS (CAPON) and dexamethasone-induced ras protein 1 (Dexras1) is structurally beneficial for S-nitrosylation of Dexras1 (SNO-Dexras1). In our previous study, uncoupling nNOS-CAPON interaction by Tat-CAPON-12C promoted functional recovery after stroke. Here, we show that ischemia elevated the levels of nNOS-Dexras1 complex and SNO-Dexras1 in the peri-infarct cortex in the days 4-10 after stroke induction, and as excepted, Tat-CAPON-12C, a peptide disrupting nNOS-CAPON interaction, significantly reversed these changes. The above information implies that repressed SNO-Dexras1 may mediate functional-promoting effects of Tat-CAPON-12C and SNO-Dexras1 could be the vital molecular substrate for post-stroke functional recovery in the repair phage. Inhibiting the ischemia-induced SNO-Dexras1 by AAV vector-mediated knockdown of Dexras1 or over-expression of dominant negative Dexras1 (Dexras1-C11S) produced sustained recovery of motor function from stroke. In contrast, up-regulation of SNO-Dexras1 by over-expressing Dexras1 worsened stroke outcome. Using electrophysiology recordings, we also observed that silence of Dexras1 in the peri-infarct cortex increased the spike number and the miniature excitatory postsynaptic currents (mEPSCs) frequency, suggesting enhancement of neuronal excitability. In addition, silence of Dexras1 increased dendritic complexity in cultured neuron and more importantly enhanced dendritic spine density in the peri-infarct cortex, implying dendritic remodeling. CONCLUSION Thus, inhibition of SNO-Dexras1 positively regulates post-stroke functional recovery via enhanced neuronal excitability and dendritic remodeling. Our results identify that SNO-Dexras1 may serve as a novel target for promoting motor functional restoration from stroke in the delayed phase, shedding light on stroke treatment.
Collapse
Affiliation(s)
- Zhou Han
- Department of Pharmacy, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
- Nanjing Medical Center for Clinical PharmacyNanjingJiangsuChina
| | - Yixuan Song
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive MedicineShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Cheng Qin
- School of Life Sciences and Chemical EngineeringJiangsu Second Normal UniversityNanjingChina
| | - Haihui Zhou
- Department of Pharmacy, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
- Nanjing Medical Center for Clinical PharmacyNanjingJiangsuChina
| | - Dan Han
- Department of Pharmacy, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
- Nanjing Medical Center for Clinical PharmacyNanjingJiangsuChina
| | - Simin Yan
- Department of Pharmacy, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
- Nanjing Medical Center for Clinical PharmacyNanjingJiangsuChina
| | - Huanyu Ni
- Department of Pharmacy, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
- Nanjing Medical Center for Clinical PharmacyNanjingJiangsuChina
| |
Collapse
|
2
|
Fronza MG, Ferreira BF, Pavan-Silva I, Guimarães FS, Lisboa SF. "NO" Time in Fear Response: Possible Implication of Nitric-Oxide-Related Mechanisms in PTSD. Molecules 2023; 29:89. [PMID: 38202672 PMCID: PMC10779493 DOI: 10.3390/molecules29010089] [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: 10/31/2023] [Revised: 12/05/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Post-traumatic stress disorder (PTSD) is a psychiatric condition characterized by persistent fear responses and altered neurotransmitter functioning due to traumatic experiences. Stress predominantly affects glutamate, a neurotransmitter crucial for synaptic plasticity and memory formation. Activation of the N-Methyl-D-Aspartate glutamate receptors (NMDAR) can trigger the formation of a complex comprising postsynaptic density protein-95 (PSD95), the neuronal nitric oxide synthase (nNOS), and its adaptor protein (NOS1AP). This complex is pivotal in activating nNOS and nitric oxide (NO) production, which, in turn, activates downstream pathways that modulate neuronal signaling, including synaptic plasticity/transmission, inflammation, and cell death. The involvement of nNOS and NOS1AP in the susceptibility of PTSD and its comorbidities has been widely shown. Therefore, understanding the interplay between stress, fear, and NO is essential for comprehending the maintenance and progression of PTSD, since NO is involved in fear acquisition and extinction processes. Moreover, NO induces post-translational modifications (PTMs), including S-nitrosylation and nitration, which alter protein function and structure for intracellular signaling. Although evidence suggests that NO influences synaptic plasticity and memory processing, the specific role of PTMs in the pathophysiology of PTSD remains unclear. This review highlights pathways modulated by NO that could be relevant to stress and PTSD.
Collapse
Affiliation(s)
- Mariana G. Fronza
- Pharmacology Departament, Ribeirão Preto Medical School, University of São Paulo, São Paulo 14049-900, Brazil; (M.G.F.); (B.F.F.); (I.P.-S.)
| | - Bruna F. Ferreira
- Pharmacology Departament, Ribeirão Preto Medical School, University of São Paulo, São Paulo 14049-900, Brazil; (M.G.F.); (B.F.F.); (I.P.-S.)
| | - Isabela Pavan-Silva
- Pharmacology Departament, Ribeirão Preto Medical School, University of São Paulo, São Paulo 14049-900, Brazil; (M.G.F.); (B.F.F.); (I.P.-S.)
| | - Francisco S. Guimarães
- Pharmacology Departament, Ribeirão Preto Medical School, University of São Paulo, São Paulo 14049-900, Brazil; (M.G.F.); (B.F.F.); (I.P.-S.)
| | - Sabrina F. Lisboa
- Pharmacology Departament, Ribeirão Preto Medical School, University of São Paulo, São Paulo 14049-900, Brazil; (M.G.F.); (B.F.F.); (I.P.-S.)
- Biomolecular Sciences Department, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo 14040-903, Brazil
| |
Collapse
|
3
|
Wu H, Li D, Zhang T, Zhao G. Novel Mechanisms of Perioperative Neurocognitive Disorders: Ferroptosis and Pyroptosis. Neurochem Res 2023; 48:2969-2982. [PMID: 37289349 DOI: 10.1007/s11064-023-03963-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/29/2023] [Accepted: 06/01/2023] [Indexed: 06/09/2023]
Abstract
Perioperative neurocognitive disorders (PNDs) are some of the most common postoperative complications among the elderly and susceptible individuals, which significantly worsens the clinical outcome of patients. However, the prevention and treatment strategies of PNDs are difficult to determine and implement since the pathogenesis of PNDs is not well understood. The development of living organisms is associated with active and organized cell death, which is essential for maintaining the homeostasis of life. Ferroptosis is a programmed cell death (different from apoptosis and necrosis) mainly caused by an imbalance in the generation and degradation of intracellular lipid peroxides due to iron overload. Pyroptosis is an inflammatory cell death characterized by the creation of membrane holes mediated by the gasdermin (GSDM) family, followed by cell lysis and the release of pro-inflammatory cytokines. Ferroptosis and pyroptosis are involved in the pathogenesis of various central nervous system (CNS) diseases. Furthermore, ferroptosis and pyroptosis are closely associated with the occurrence and development of PNDs. This review summarizes the main regulatory mechanisms of ferroptosis and pyroptosis and the latest related to PNDs. Based on the available evidence, potential intervention strategies that can alleviate PNDs by inhibiting ferroptosis and pyroptosis have also been provided.
Collapse
Affiliation(s)
- Hang Wu
- Department of Anaesthesiology, China-Japan Union Hospital of Jilin University, 126 Sendai Street, Changchun, Jilin, China
| | - Dongmei Li
- Department of Anaesthesiology, China-Japan Union Hospital of Jilin University, 126 Sendai Street, Changchun, Jilin, China
| | - Te Zhang
- Department of Anaesthesiology, China-Japan Union Hospital of Jilin University, 126 Sendai Street, Changchun, Jilin, China
| | - Guoqing Zhao
- Department of Anaesthesiology, China-Japan Union Hospital of Jilin University, 126 Sendai Street, Changchun, Jilin, China.
- Jilin University, 2699 Forward Avenue, Changchun, Jilin, China.
| |
Collapse
|
4
|
Cheng HP, Feng DD, Li XH, Gao LH, Qiu YJ, Liang XY, Zhou Y, Huang P, Shao M, Zhang YN, Chang YF, Fu JF, Huang YH, Liu W, Tang SY, Li C, Luo ZQ. NMDA receptor activation induces damage of alveolar type II cells and lung fibrogenesis through ferroptosis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119535. [PMID: 37451346 DOI: 10.1016/j.bbamcr.2023.119535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
Ferroptosis, a newly discovered type of regulated cell death, has been implicated in numerous human diseases. Idiopathic pulmonary fibrosis (IPF) is a progressive and ultimately fatal interstitial lung disease with poor prognosis and limited treatment options. Emerging evidence has linked ferroptosis and glutamate-determined cell fate which is considered a new light on the etiology of pulmonary fibrosis. Here, we observed that N-methyl d-aspartate receptor (NMDAR) activation promoted cell damage and iron deposition in MLE-12 cells in a dose-, time-, and receptor-dependent manner. This mediated substantial Ca2+ influx, upregulated the expression levels of nNOS and IRP1, and affected intracellular iron homeostasis by regulating the expression of iron transport-related proteins (i.e., TFR1, DMT1, and FPN). Excessive iron load promoted the continuous accumulation of total intracellular and mitochondrial reactive oxygen species, which ultimately led to ferroptosis. NMDAR inhibition reduced lung injury and pulmonary fibrosis in bleomycin-induced mice. Bleomycin stimulation upregulated the expression of NMDAR1, nNOS, and IRP1 in mouse lung tissues, which ultimately led to iron deposition via regulation of the expression of various iron metabolism-related genes. NMDAR activation initiated the pulmonary fibrosis process by inducing iron deposition in lung tissues and ferroptosis of alveolar type II cells. Our data suggest that NMDAR activation regulates the expression of iron metabolism-related genes by promoting calcium influx, increasing nNOS and IRP1 expression, and increasing iron deposition by affecting cellular iron homeostasis, ultimately leading to mitochondrial damage, mitochondrial dysfunction, and ferroptosis. NMDAR activation-induced ferroptosis of alveolar type II cells might be a key event to the initiation of pulmonary fibrosis.
Collapse
Affiliation(s)
- Hai-Peng Cheng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dan-Dan Feng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiao-Hong Li
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li-Hua Gao
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yu-Jia Qiu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xing-Yue Liang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yan Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Pu Huang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Min Shao
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yun-Na Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yan-Fen Chang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jia-Feng Fu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yan-Hong Huang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Wei Liu
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Si-Yuan Tang
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Chen Li
- Department of Physiology, Changzhi Medical College, Changzhi, Shanxi, China.
| | - Zi-Qiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, Hunan, China.
| |
Collapse
|
5
|
Zeng T, Li J, Xie L, Dong Z, Chen Q, Huang S, Xie S, Lai Y, Li J, Yan W, Wang Y, Xie Z, Hu C, Zhang J, Kuang S, Song Y, Gao L, Lv Z. Nrf2 regulates iron-dependent hippocampal synapses and functional connectivity damage in depression. J Neuroinflammation 2023; 20:212. [PMID: 37735410 PMCID: PMC10512501 DOI: 10.1186/s12974-023-02875-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 08/16/2023] [Indexed: 09/23/2023] Open
Abstract
Neuronal iron overload contributes to synaptic damage and neuropsychiatric disorders. However, the molecular mechanisms underlying iron deposition in depression remain largely unexplored. Our study aims to investigate how nuclear factor-erythroid 2 (NF-E2)-related factor 2 (Nrf2) ameliorates hippocampal synaptic dysfunction and reduces brain functional connectivity (FC) associated with excessive iron in depression. We treated mice with chronic unpredictable mild stress (CUMS) with the iron chelator deferoxamine mesylate (DFOM) and a high-iron diet (2.5% carbonyl iron) to examine the role of iron overload in synaptic plasticity. The involvement of Nrf2 in iron metabolism and brain function was assessed using molecular biological techniques and in vivo resting-state functional magnetic resonance imaging (rs-fMRI) through genetic deletion or pharmacologic activation of Nrf2. The results demonstrated a significant correlation between elevated serum iron levels and impaired hippocampal functional connectivity (FC), which contributed to the development of depression-induced CUMS. Iron overload plays a crucial role in CUMS-induced depression and synaptic dysfunction, as evidenced by the therapeutic effects of a high-iron diet and DFOM. The observed iron overload in this study was associated with decreased Nrf2 levels and increased expression of transferrin receptors (TfR). Notably, inhibition of iron accumulation effectively attenuated CUMS-induced synaptic damage mediated by downregulation of brain-derived neurotrophic factor (BDNF). Nrf2-/- mice exhibited compromised FC within the limbic system and the basal ganglia, particularly in the hippocampus, and inhibition of iron accumulation effectively attenuated CUMS-induced synaptic damage mediated by downregulation of brain-derived neurotrophic factor (BDNF). Activation of Nrf2 restored iron homeostasis and reversed vulnerability to depression. Mechanistically, we further identified that Nrf2 deletion promoted iron overload via upregulation of TfR and downregulation of ferritin light chain (FtL), leading to BDNF-mediated synapse damage in the hippocampus. Therefore, our findings unveil a novel role for Nrf2 in regulating iron homeostasis while providing mechanistic insights into poststress susceptibility to depression. Targeting Nrf2-mediated iron metabolism may offer promising strategies for developing more effective antidepressant therapies.
Collapse
Affiliation(s)
- Ting Zeng
- School of Traditional Chinese Medicine, Southern Medical University (SMU), Sha Tai Nan Road No. 1063, Guangzhou, 510515, Guangdong, China
- Department of Brain Diseases, The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Junjie Li
- School of Traditional Chinese Medicine, Southern Medical University (SMU), Sha Tai Nan Road No. 1063, Guangzhou, 510515, Guangdong, China
| | - Lingpeng Xie
- Department of Hepatology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China
| | - Zhaoyang Dong
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Qing Chen
- School of Traditional Chinese Medicine, Southern Medical University (SMU), Sha Tai Nan Road No. 1063, Guangzhou, 510515, Guangdong, China
| | - Sha Huang
- School of Traditional Chinese Medicine, Southern Medical University (SMU), Sha Tai Nan Road No. 1063, Guangzhou, 510515, Guangdong, China
| | - Shuwen Xie
- School of Traditional Chinese Medicine, Southern Medical University (SMU), Sha Tai Nan Road No. 1063, Guangzhou, 510515, Guangdong, China
| | - Yuqi Lai
- School of Traditional Chinese Medicine, Southern Medical University (SMU), Sha Tai Nan Road No. 1063, Guangzhou, 510515, Guangdong, China
| | - Jun Li
- School of Traditional Chinese Medicine, Southern Medical University (SMU), Sha Tai Nan Road No. 1063, Guangzhou, 510515, Guangdong, China
| | - Weixin Yan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - YuHua Wang
- School of Traditional Chinese Medicine, Southern Medical University (SMU), Sha Tai Nan Road No. 1063, Guangzhou, 510515, Guangdong, China
| | - Zeping Xie
- School of Traditional Chinese Medicine, Southern Medical University (SMU), Sha Tai Nan Road No. 1063, Guangzhou, 510515, Guangdong, China
| | - Changlei Hu
- School of Traditional Chinese Medicine, Southern Medical University (SMU), Sha Tai Nan Road No. 1063, Guangzhou, 510515, Guangdong, China
| | - Jiayi Zhang
- School of Traditional Chinese Medicine, Southern Medical University (SMU), Sha Tai Nan Road No. 1063, Guangzhou, 510515, Guangdong, China
| | - Shanshan Kuang
- School of Traditional Chinese Medicine, Southern Medical University (SMU), Sha Tai Nan Road No. 1063, Guangzhou, 510515, Guangdong, China
| | - Yuhong Song
- Department of Traditional Chinese Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China.
| | - Lei Gao
- School of Traditional Chinese Medicine, Southern Medical University (SMU), Sha Tai Nan Road No. 1063, Guangzhou, 510515, Guangdong, China.
| | - Zhiping Lv
- School of Traditional Chinese Medicine, Southern Medical University (SMU), Sha Tai Nan Road No. 1063, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
6
|
Yang L, Fu Q, Yang L, Zhang Y. HIF-1α/MMP-9 promotes spinal cord central sensitization in rats with bone cancer pain. Eur J Pharmacol 2023; 954:175858. [PMID: 37356787 DOI: 10.1016/j.ejphar.2023.175858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/27/2023]
Abstract
Bone cancer pain (BCP) is one of the most prevalent and serious symptoms of patients with cancer. Currently, the medical interventions used for the treatment of BCP do not act with optimal safety and efficacy. In this study, we appraised whether the hypoxia-inducible factor 1α (HIF-1α)/metalloproteinase-9 (MMP9) axis activates the PI3K/AKT pathway, resulting in elevated spinal cord central sensitization and aggravated BCP. BCP rats were established by tibial injection of Walker 256 cells, followed by different interventions in rats using HIF-1ɑ inhibitor LW6 or antibody treatments. After treatment with LW6 or antibody against HIF-1α, central sensitization in the spinal cord tissues of rats was inhibited, and pain perception in rats was reduced. Moreover, the activation of glial cells in the spinal cord tissues was ameliorated. The expression of MMP9 was remarkably suppressed in spinal cord tissues after inhibition of HIF-1ɑ activity, and the activity of the PI3K/AKT signaling pathway was inhibited. Further activation of MMP9 expression suppressed the alleviating effect of HIF-1ɑ inhibitor LW6 or antibody on pain perception in rats inoculated with tumors. Taken together, our studies suggest a HIF-1α/MMP9-mediated activation of PI3K/AKT in the spinal cord tissues, resulting in increased pain perception in a rat model with BCP.
Collapse
Affiliation(s)
- Liyu Yang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110003, Liaoning, PR China
| | - Qin Fu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110003, Liaoning, PR China
| | - Liqing Yang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110003, Liaoning, PR China
| | - Yiqi Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110003, Liaoning, PR China.
| |
Collapse
|
7
|
Irollo E, Nash B, Luchetta J, Brandimarti R, Meucci O. The Endolysosomal Transporter DMT1 is Required for Morphine Regulation of Neuronal Ferritin Heavy Chain. J Neuroimmune Pharmacol 2023; 18:495-508. [PMID: 37661197 PMCID: PMC10577102 DOI: 10.1007/s11481-023-10082-x] [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: 01/20/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023]
Abstract
NeuroHIV and other neurologic disorders present with altered iron metabolism in central nervous system neurons. Many people with HIV also use opioids, which can worsen neuroHIV symptoms by further dysregulating neuronal iron metabolism. Our previous work demonstrated that the μ-opioid agonist morphine causes neuronal endolysosomes to release their iron stores, and neurons respond by upregulating ferritin heavy chain (FHC), an iron storage protein associated with cognitive impairment in neuroHIV. Here, we investigated if this process required divalent metal transporter 1 (DMT1), a well-known iron transporter expressed on endolysosomes. We first optimized conditions to detect DMT1 isoforms (DMT1 1B ± iron responsive element) using fluorescently labeled rat DMT1 constructs expressed in HEK-293 cells. We also expressed these constructs in primary rat cortical neurons to compare their expression and subcellular distribution with endogenous DMT1 isoforms. We found endogenous DMT1 isoforms in the cytoplasm that colocalized with lysosomal-associated protein 1 (LAMP1), a marker of endolysosomes. Next, we blocked endogenous DMT1 isoforms using ebselen, a potent pharmacological inhibitor of DMT1 iron transport. Ebselen pre-treatment blocked morphine's ability to upregulate FHC protein, suggesting this pathway requires DMT1 iron transport from endolysosomes. This was further validated using viral-mediated genetic silencing of DMT1±IRE in cortical neurons, which also blocked FHC upregulation in the presence of morphine. Overall, our work demonstrates that the μ-opioid agonist morphine utilizes the endolysosomal iron transporter DMT1 to modulate neuronal cellular iron metabolism, upregulate FHC protein, and contribute to cognitive decline in neuroHIV. Morphine requires DMT1 to upregulate neuronal FHC. Cortical neurons treated with morphine release their endolysosomal iron stores to the cytoplasm and upregulate FHC, an iron storage protein associated with dendritic spine deficits and cognitive impairment in neuroHIV. This pathway requires the endolysosomal iron transporter DMT1, as pharmacological and genetic inhibitors of the transporter completely block morphine's ability to upregulate FHC. Created with BioRender.com .
Collapse
Affiliation(s)
- Elena Irollo
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, 19102, USA
| | - Bradley Nash
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, 19102, USA
| | - Jared Luchetta
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, 19102, USA
| | - Renato Brandimarti
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, 19102, USA
- Department of Pharmacy and Biotechnology, University of Bologna, Via Marsala, 49, Bologna, BO, 40126, Italy
| | - Olimpia Meucci
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, 19102, USA.
- Department of Microbiology & Immunology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, 19102, USA.
- Center for Neuroimmunology & CNS Therapeutics, Institute for Molecular Medicine & Infectious Disease, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
8
|
Lotan A, Luza S, Opazo CM, Ayton S, Lane DJR, Mancuso S, Pereira A, Sundram S, Weickert CS, Bousman C, Pantelis C, Everall IP, Bush AI. Perturbed iron biology in the prefrontal cortex of people with schizophrenia. Mol Psychiatry 2023; 28:2058-2070. [PMID: 36750734 PMCID: PMC10575779 DOI: 10.1038/s41380-023-01979-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/10/2023] [Accepted: 01/20/2023] [Indexed: 02/09/2023]
Abstract
Despite loss of grey matter volume and emergence of distinct cognitive deficits in young adults diagnosed with schizophrenia, current treatments for schizophrenia do not target disruptions in late maturational reshaping of the prefrontal cortex. Iron, the most abundant transition metal in the brain, is essential to brain development and function, but in excess, it can impair major neurotransmission systems and lead to lipid peroxidation, neuroinflammation and accelerated aging. However, analysis of cortical iron biology in schizophrenia has not been reported in modern literature. Using a combination of inductively coupled plasma-mass spectrometry and western blots, we quantified iron and its major-storage protein, ferritin, in post-mortem prefrontal cortex specimens obtained from three independent, well-characterised brain tissue resources. Compared to matched controls (n = 85), among schizophrenia cases (n = 86) we found elevated tissue iron, unlikely to be confounded by demographic and lifestyle variables, by duration, dose and type of antipsychotic medications used or by copper and zinc levels. We further observed a loss of physiologic age-dependent iron accumulation among people with schizophrenia, in that the iron level among cases was already high in young adulthood. Ferritin, which stores iron in a redox-inactive form, was paradoxically decreased in individuals with the disorder. Such iron-ferritin uncoupling could alter free, chemically reactive, tissue iron in key reasoning and planning areas of the young-adult schizophrenia cortex. Using a prediction model based on iron and ferritin, our data provide a pathophysiologic link between perturbed cortical iron biology and schizophrenia and indicate that achievement of optimal cortical iron homeostasis could offer a new therapeutic target.
Collapse
Affiliation(s)
- Amit Lotan
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Psychiatry and the Biological Psychiatry Laboratory, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Sandra Luza
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
| | - Carlos M Opazo
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia.
| | - Scott Ayton
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Darius J R Lane
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Serafino Mancuso
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
| | - Avril Pereira
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
| | - Suresh Sundram
- Department of Psychiatry, School of Clinical Sciences, Monash University, Melbourne, VIC, Australia
- Mental Health Program, Monash Health, Melbourne, VIC, Australia
| | - Cynthia Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Chad Bousman
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Departments of Medical Genetics, Psychiatry, Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada
- The Cooperative Research Centre (CRC) for Mental Health, Melbourne, VIC, Australia
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
- North Western Mental Health, Melbourne, VIC, Australia
| | - Ian P Everall
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
- North Western Mental Health, Melbourne, VIC, Australia
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.
- The Cooperative Research Centre (CRC) for Mental Health, Melbourne, VIC, Australia.
| |
Collapse
|
9
|
Wen J, Yang F, Fang CX, Chen HL, Yang L. Sulforaphane triggers iron overload-mediated ferroptosis in gastric carcinoma cells by activating the PI3K/IRP2/DMT1 pathway. Hum Exp Toxicol 2023; 42:9603271231177295. [PMID: 37201195 DOI: 10.1177/09603271231177295] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
OBJECTIVE Increasing evidence indicates that prolonged exposure to sulforaphane (SFN) can improve malignancies. However, the role of iron in SFN-triggered death in gastric carcinoma cells and the underlying molecular mechanisms remain unclear. Thus, the current study explored the effects of SFN on iron overload-mediated ferroptosis and the PI3K/IRP2/DMT1 pathway in gastric carcinoma cells. METHODS We utilized the MGC-803 cell line to assess whether SFN affected iron metabolism and whether this effect contributed to cell death. Pharmacological inhibition of iron metabolism also was performed to determine the molecular mechanism underlying SFN-triggered iron overload and the disturbance in iron metabolism. RESULTS Our data revealed that SFN treatment altered iron homeostasis and led to iron overload in vitro. Interestingly, SFN-stimulated cell death resulted from ferroptosis, a recently identified iron-dependent form of regulated cell death. Furthermore, an iron chelator, deferiprone, ameliorated the SFN-triggered mitochondrial dysfunction and reduced the iron overload. In addition, we found that the SFN-triggered iron overload was regulated by the PI3K/IRP2/DMT1 signaling pathway. CONCLUSION We discovered that disturbance in iron metabolism might be involved in the SFN-triggered cell death in gastric carcinoma cells. Blockade of the PI3K/IRP2/DMT1 axis could provide a feedback effect on SFN-induced ferroptosis to protect tumor cells from growth.
Collapse
Affiliation(s)
- Jing Wen
- Department of Oncology, Minda Hospital of Hubei Minzu University, Enshi, P.R.China
| | - Fan Yang
- Department of General Surgery II, Minda Hospital of Hubei Minzu University, Enshi, P.R.China
| | - Cheng-Xiang Fang
- Department of Oncology, Minda Hospital of Hubei Minzu University, Enshi, P.R.China
| | - Hong-Liu Chen
- Department of General Surgery II, Minda Hospital of Hubei Minzu University, Enshi, P.R.China
| | - Li Yang
- Department of Oncology, Minda Hospital of Hubei Minzu University, Enshi, P.R.China
| |
Collapse
|
10
|
Durmaz CD, Karabulut HG, Saka MC, Sucularlı C, Gümüş Akay G, Atbaşoğlu C, Ilgın Ruhi H. Genetic Analysis of RASD1 as a Candidate Gene for Schizophrenia. Balkan Med J 2022; 39:422-428. [PMID: 36305088 PMCID: PMC9667215 DOI: 10.4274/balkanmedj.galenos.2022.2022-5-90] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/29/2022] [Indexed: 12/01/2022] Open
Abstract
Background RASD1 encodes Dexamethasone-induced Ras-related protein 1 (Dexras1), a protein with a critical role in signal transduction in neurons. There is a strong suspicion that dysfunction of Dexras1 might contribute to the pathogenesis of neuropsychiatric diseases. Related to its functions in intracellular signaling pathways, Dexras1 has a potential role in the etiology of schizophrenia because of its close interaction with NOS1, NOS1AP, and NMDAR, which have previously been associated with schizophrenia. Aims To investigate the association of RASD1 variants with schizophrenia in a selected cohort from Turkey. Study Design A case-control study. Methods We performed targeted sequencing for the two exons, single intron, and untranslated regions of RASD1 gene in 200 individuals with schizophrenia and 100 healthy controls of Turkish origin. Results Two rare variants, RASD1 (NM_016084.5): c.722A>T and c*31G>A were identified in individuals with schizophrenia but not in any controls. The c.722A>T was found in a single individual with schizophrenia and is a missense heterozygous variant in the second exon of RASD1, which is extremely rare in GnomAD. The other variant, c*31G>A, which was found in another individual from this schizophrenia cohort, has not been reported previously. Seven previously identified common single nucleotide polymorphisms were also detected; however, they were not significantly associated with schizophrenia in this study cohort. Conclusion Our findings suggest that rare variants of RASD1 might be contributing to the etiopathogenesis of schizophrenia. Further studies are needed to elucidate the underlying mechanism of this association.
Collapse
Affiliation(s)
- Ceren Damla Durmaz
- Department of Medical Genetics, School of Medicine, Ankara University, Ankara, Turkey
- Department of Medical Genetics, School of Medicine, Hacettepe University, Ankara, Turkey
| | | | - Meram Can Saka
- Department of Psychiatry, School of Medicine, Ankara University, Ankara, Turkey
| | - Ceren Sucularlı
- Department of Bioinformatics, Institute of Health Sciences, Hacettepe University, Ankara, Turkey
| | - Güvem Gümüş Akay
- Department of Physiology, School of Medicine, Ankara University, Ankara, Turkey
- Ankara University Brain Research Center (AUBAUM), Ankara University, Ankara, Turkey
- Neuroscience and Neurotechnology Center of Excellence (NÖROM), Ankara, Turkey
| | - Cem Atbaşoğlu
- Ankara University, School of Medicine Retired Faculty Member, Professor of Psychiatry, Ankara, Turkey
| | - Hatice Ilgın Ruhi
- Department of Medical Genetics, School of Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
11
|
Zeng X, Li J, Yang F, Xia R. The effect of narcotics on ferroptosis-related molecular mechanisms and signalling pathways. Front Pharmacol 2022; 13:1020447. [PMID: 36313359 PMCID: PMC9606818 DOI: 10.3389/fphar.2022.1020447] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/30/2022] [Indexed: 11/26/2022] Open
Abstract
Ferroptosis is a novel programmed cell death form characterized by iron-mediated reactive oxygen species-induced lipid peroxidation and subsequent cell damage that is distinct from apoptosis, necroptosis, pyroptosis, and autophagy. Most studies on ferroptosis are based on its function and mechanism, but there have been relatively few studies on the effects of drugs, especially anaesthetics, on ferroptosis. Therefore, we summarized the recent literature on the effects of anaesthetics on ferroptosis to understand the underlying mechanism. In particular, we focused on the targets of various anaesthetics in different mechanisms of ferroptosis and the effects of ferroptosis induction or inhibition by narcotics on various diseases. The aims of this review are to provide a relatively reasonable drug regimen for clinicians, to explore potential ferroptosis protection drugs and targets, to reduce perioperative complications and to improve the postoperative performance of patients, especially those who are critically ill.
Collapse
Affiliation(s)
- Xiaoqin Zeng
- Department of Anaesthesiology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Jingda Li
- College of Life Sciences, Yangtze University, Jingzhou, Hubei, China
| | - Fuyuan Yang
- School of Basic Medicine, Yangtze University Health Science Center, Jingzhou, Hubei, China
- *Correspondence: Fuyuan Yang, ; Rui Xia,
| | - Rui Xia
- Department of Anaesthesiology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
- *Correspondence: Fuyuan Yang, ; Rui Xia,
| |
Collapse
|
12
|
Ferroptosis is involved in regulating perioperative neurocognitive disorders: emerging perspectives. J Neuroinflammation 2022; 19:219. [PMID: 36068571 PMCID: PMC9450301 DOI: 10.1186/s12974-022-02570-3] [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: 06/09/2022] [Accepted: 08/09/2022] [Indexed: 11/10/2022] Open
Abstract
Since the twenty-first century, the development of technological advances in anesthesia and surgery has brought benefits to human health. However, the adverse neurological effects of perioperative-related factors (e.g., surgical trauma, anesthesia, etc.) as stressors cannot be ignored as well. The nervous system appears to be more "fragile" and vulnerable to damage in developing and aging individuals. Ferroptosis is a novel form of programmed cell death proposed in 2012. In recent years, the regulation of ferroptosis to treat cancer, immune system disorders, and neurodegenerative diseases have seen an unprecedented surge of interest. The association of ferroptosis with perioperative neurocognitive disorders has also received much attention. Cognitive impairment can not only affect the individual's quality of life, but also impose a burden on the family and society. Therefore, the search for effective preventive and therapeutic methods to alleviate cognitive impairment caused by perioperative-related factors is a challenge that needs to be urgently addressed. In our review, we first briefly describe the connection between iron accumulation in neurons and impairment of brain function during development and aging. It is followed by a review of the pathways of ferroptosis, mainly including iron metabolism, amino acid metabolism, and lipid metabolism pathway. Furthermore, we analyze the connection between ferroptosis and perioperative-related factors. The surgery itself, general anesthetic drugs, and many other relevant factors in the perioperative period may affect neuronal iron homeostasis. Finally, we summarize the experimental evidence for ameliorating developmental and degenerative neurotoxicity by modulating ferroptosis. The suppression of ferroptosis seems to provide the possibility to prevent and improve perioperative neurocognitive impairment.
Collapse
|
13
|
Grochowska KM, Andres‐Alonso M, Karpova A, Kreutz MR. The needs of a synapse—How local organelles serve synaptic proteostasis. EMBO J 2022; 41:e110057. [PMID: 35285533 PMCID: PMC8982616 DOI: 10.15252/embj.2021110057] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/24/2021] [Accepted: 02/10/2022] [Indexed: 12/12/2022] Open
Abstract
Synaptic function crucially relies on the constant supply and removal of neuronal membranes. The morphological complexity of neurons poses a significant challenge for neuronal protein transport since the machineries for protein synthesis and degradation are mainly localized in the cell soma. In response to this unique challenge, local micro‐secretory systems have evolved that are adapted to the requirements of neuronal membrane protein proteostasis. However, our knowledge of how neuronal proteins are synthesized, trafficked to membranes, and eventually replaced and degraded remains scarce. Here, we review recent insights into membrane trafficking at synaptic sites and into the contribution of local organelles and micro‐secretory pathways to synaptic function. We describe the role of endoplasmic reticulum specializations in neurons, Golgi‐related organelles, and protein complexes like retromer in the synthesis and trafficking of synaptic transmembrane proteins. We discuss the contribution of autophagy and of proteasome‐mediated and endo‐lysosomal degradation to presynaptic proteostasis and synaptic function, as well as nondegradative roles of autophagosomes and lysosomes in signaling and synapse remodeling. We conclude that the complexity of neuronal cyto‐architecture necessitates long‐distance protein transport that combines degradation with signaling functions.
Collapse
Affiliation(s)
- Katarzyna M Grochowska
- Leibniz Group “Dendritic Organelles and Synaptic Function” Center for Molecular Neurobiology ZMNH University Medical Center Hamburg‐Eppendorf Hamburg Germany
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
| | - Maria Andres‐Alonso
- Leibniz Group “Dendritic Organelles and Synaptic Function” Center for Molecular Neurobiology ZMNH University Medical Center Hamburg‐Eppendorf Hamburg Germany
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
| | - Anna Karpova
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
- Center for Behavioral Brain Sciences Otto von Guericke University Magdeburg Germany
| | - Michael R Kreutz
- Leibniz Group “Dendritic Organelles and Synaptic Function” Center for Molecular Neurobiology ZMNH University Medical Center Hamburg‐Eppendorf Hamburg Germany
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
- Center for Behavioral Brain Sciences Otto von Guericke University Magdeburg Germany
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg Germany
| |
Collapse
|
14
|
Zimmer TS, David B, Broekaart DWM, Schidlowski M, Ruffolo G, Korotkov A, van der Wel NN, van Rijen PC, Mühlebner A, van Hecke W, Baayen JC, Idema S, François L, van Eyll J, Dedeurwaerdere S, Kessels HW, Surges R, Rüber T, Gorter JA, Mills JD, van Vliet EA, Aronica E. Seizure-mediated iron accumulation and dysregulated iron metabolism after status epilepticus and in temporal lobe epilepsy. Acta Neuropathol 2021; 142:729-759. [PMID: 34292399 PMCID: PMC8423709 DOI: 10.1007/s00401-021-02348-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/02/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022]
Abstract
Neuronal dysfunction due to iron accumulation in conjunction with reactive oxygen species (ROS) could represent an important, yet underappreciated, component of the epileptogenic process. However, to date, alterations in iron metabolism in the epileptogenic brain have not been addressed in detail. Iron-related neuropathology and antioxidant metabolic processes were investigated in resected brain tissue from patients with temporal lobe epilepsy and hippocampal sclerosis (TLE-HS), post-mortem brain tissue from patients who died after status epilepticus (SE) as well as brain tissue from the electrically induced SE rat model of TLE. Magnetic susceptibility of the presumed seizure-onset zone from three patients with focal epilepsy was compared during and after seizure activity. Finally, the cellular effects of iron overload were studied in vitro using an acute mouse hippocampal slice preparation and cultured human fetal astrocytes. While iron-accumulating neurons had a pyknotic morphology, astrocytes appeared to acquire iron-sequestrating capacity as indicated by prominent ferritin expression and iron retention in the hippocampus of patients with SE or TLE. Interictal to postictal comparison revealed increased magnetic susceptibility in the seizure-onset zone of epilepsy patients. Post-SE rats had consistently higher hippocampal iron levels during the acute and chronic phase (when spontaneous recurrent seizures are evident). In vitro, in acute slices that were exposed to iron, neurons readily took up iron, which was exacerbated by induced epileptiform activity. Human astrocyte cultures challenged with iron and ROS increased their antioxidant and iron-binding capacity, but simultaneously developed a pro-inflammatory phenotype upon chronic exposure. These data suggest that seizure-mediated, chronic neuronal iron uptake might play a role in neuronal dysfunction/loss in TLE-HS. On the other hand, astrocytes sequester iron, specifically in chronic epilepsy. This function might transform astrocytes into a highly resistant, pro-inflammatory phenotype potentially contributing to pro-epileptogenic inflammatory processes.
Collapse
Affiliation(s)
- Till S Zimmer
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Bastian David
- Department of Epileptology, University Hospital Bonn, Bonn, Germany
| | - Diede W M Broekaart
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Martin Schidlowski
- Department of Epileptology, University Hospital Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Gabriele Ruffolo
- Laboratory affiliated to Istituto Pasteur Italia, Department of Physiology and Pharmacology, University of Rome Sapienza, Rome, Italy
| | - Anatoly Korotkov
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Nicole N van der Wel
- Department Cell Biology and Histology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department Electron Microscopy Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Peter C van Rijen
- Department of Neurosurgery, Brain Centre, Rudolf Magnus Institute for Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Angelika Mühlebner
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wim van Hecke
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Johannes C Baayen
- Department of Neurosurgery, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Sander Idema
- Department of Neurosurgery, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Liesbeth François
- Neurosciences Therapeutic Area, UCB Pharma, Braine-l'Alleud, Belgium
| | - Jonathan van Eyll
- Neurosciences Therapeutic Area, UCB Pharma, Braine-l'Alleud, Belgium
| | | | - Helmut W Kessels
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Rainer Surges
- Department of Epileptology, University Hospital Bonn, Bonn, Germany
| | - Theodor Rüber
- Department of Epileptology, University Hospital Bonn, Bonn, Germany
| | - Jan A Gorter
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - James D Mills
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Department of Clinical and Experimental Epilepsy, UCL, London, UK
- Chalfont Centre for Epilepsy, Chalfont St Peter, UK
| | - Erwin A van Vliet
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands.
| |
Collapse
|
15
|
Li Y, Lu YX, Chi HL, Xiao T, Chen YM, Fu LY, Zibrila AI, Qi J, Li HB, Su Q, Gao HL, Zhang Y, Shi XL, Yu XJ, Kang YM. Chronic Blockade of NMDAR Subunit 2A in the Hypothalamic Paraventricular Nucleus Alleviates Hypertension Through Suppression of MEK/ERK/CREB Pathway. Am J Hypertens 2021; 34:840-850. [PMID: 33856436 DOI: 10.1093/ajh/hpab047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 02/05/2021] [Accepted: 04/14/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND N-Methyl-d-aspartate receptor (NMDAR) in the hypothalamic paraventricular nucleus (PVN) plays critical roles in regulating sympathetic outflow. Studies showed that acute application of the antagonists of NMDAR or its subunits would reduce sympathetic nerve discharges. However, little is known about the effect of long-term management of NMDAR in hypertensive animals. METHODS PEAQX, the specific antagonist of NMDAR subunit 2A (GluN2A) was injected into both sides of the PVN of two-kidney, one-clip (2K1C) renal hypertensive rats and control (normotensive rats) for 3 weeks. RESULTS Three weeks of PEAQX infusion significantly reduced the blood pressure of the 2K1C rats. It managed to resume the balance between excitatory and inhibitory neural transmitters, reduce the level of proinflammatory cytokines and reactive oxygen species in the PVN, and reduce the level of norepinephrine in plasma of the 2K1C rats. PEAQX administration also largely reduced the transcription and translation levels of GluN2A and changed the expression levels of NMDAR subunits 1 and 2B (GluN1 and GluN2B). In addition, NMDAR was known to function through activating the extracellular regulated protein kinases (ERK) or phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) pathways. In our study, we found that in the PVN of 2K1C rats treated with PEAQX, the phosphorylation levels of mitogen-activated protein kinase kinase (MEK), ERK1/2, and cAMP-response element-binding protein (CREB) significantly reduced, while the phosphorylation level of PI3K did not change significantly. CONCLUSIONS Chronic blockade of GluN2A alleviates hypertension through suppression of MEK/ERK/CREB pathway.
Collapse
Affiliation(s)
- Ying Li
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi’an, China
| | - Yu-Xin Lu
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi’an, China
| | - Hong-Li Chi
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi’an, China
| | - Tong Xiao
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi’an, China
| | - Yan-Mei Chen
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi’an, China
| | - Li-Yan Fu
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi’an, China
| | - Abdoulaye Issotina Zibrila
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi’an, China
| | - Jie Qi
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi’an, China
| | - Hong-Bao Li
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi’an, China
| | - Qing Su
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi’an, China
| | - Hong-Li Gao
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi’an, China
| | - Yan Zhang
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi’an, China
| | - Xiao-Lian Shi
- Department of Pharmacology, Xi’an Jiaotong University School of Basic Medical Sciences, Xi’an, China
| | - Xiao-Jing Yu
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi’an, China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi’an, China
| |
Collapse
|
16
|
Thomas GEC, Zarkali A, Ryten M, Shmueli K, Gil-Martinez AL, Leyland LA, McColgan P, Acosta-Cabronero J, Lees AJ, Weil RS. Regional brain iron and gene expression provide insights into neurodegeneration in Parkinson's disease. Brain 2021; 144:1787-1798. [PMID: 33704443 PMCID: PMC8320305 DOI: 10.1093/brain/awab084] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/20/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022] Open
Abstract
The mechanisms responsible for the selective vulnerability of specific neuronal populations in Parkinson's disease are poorly understood. Oxidative stress secondary to brain iron accumulation is one postulated mechanism. We measured iron deposition in 180 cortical regions of 96 patients with Parkinson's disease and 35 control subjects using quantitative susceptibility mapping. We estimated the expression of 15 745 genes in the same regions using transcriptomic data from the Allen Human Brain Atlas. Using partial least squares regression, we then identified the profile of gene transcription in the healthy brain that underlies increased cortical iron in patients with Parkinson's disease relative to controls. Applying gene ontological tools, we investigated the biological processes and cell types associated with this transcriptomic profile and identified the sets of genes with spatial expression profiles in control brains that correlated significantly with the spatial pattern of cortical iron deposition in Parkinson's disease. Gene ontological analyses revealed that these genes were enriched for biological processes relating to heavy metal detoxification, synaptic function and nervous system development and were predominantly expressed in astrocytes and glutamatergic neurons. Furthermore, we demonstrated that the genes differentially expressed in Parkinson's disease are associated with the pattern of cortical expression identified in this study. Our findings provide mechanistic insights into regional selective vulnerabilities in Parkinson's disease, particularly the processes involving iron accumulation.
Collapse
Affiliation(s)
| | | | - Mina Ryten
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1B 5EH, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL, London, WC1N 1EH, UK
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, UCL, London, WC1N 1EH, UK
| | - Karin Shmueli
- Department of Medical Physics and Biomedical Engineering, Malet Place Engineering Building, UCL, London, WC1E 6BT, UK
| | - Ana Luisa Gil-Martinez
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1B 5EH, UK
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, UCL, London, WC1N 1EH, UK
| | | | - Peter McColgan
- Huntington’s Disease Centre, UCL Institute of Neurology, London, WC1B 5EH, UK
| | | | - Andrew J Lees
- Reta Lila Weston Institute of Neurological Studies, London, WC1N 1PJ, UK
| | - Rimona S Weil
- Dementia Research Centre, UCL, London, WC1N 3AR, UK
- Wellcome Centre for Human Neuroimaging, UCL, London, WC1N 3AR, UK
- Movement Disorders Consortium, UCL, London, WC1N 3BG, UK
| |
Collapse
|
17
|
Kutchukian C, Vivas O, Casas M, Jones JG, Tiscione SA, Simó S, Ory DS, Dixon RE, Dickson EJ. NPC1 regulates the distribution of phosphatidylinositol 4-kinases at Golgi and lysosomal membranes. EMBO J 2021; 40:e105990. [PMID: 34019311 PMCID: PMC8246069 DOI: 10.15252/embj.2020105990] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 03/11/2021] [Accepted: 03/19/2021] [Indexed: 12/15/2022] Open
Abstract
Cholesterol and phosphoinositides (PI) are two critically important lipids that are found in cellular membranes and dysregulated in many disorders. Therefore, uncovering molecular pathways connecting these essential lipids may offer new therapeutic insights. We report that loss of function of lysosomal Niemann-Pick Type C1 (NPC1) cholesterol transporter, which leads to neurodegenerative NPC disease, initiates a signaling cascade that alters the cholesterol/phosphatidylinositol 4-phosphate (PtdIns4P) countertransport cycle between Golgi-endoplasmic reticulum (ER), as well as lysosome-ER membrane contact sites (MCS). Central to these disruptions is increased recruitment of phosphatidylinositol 4-kinases-PI4KIIα and PI4KIIIβ-which boosts PtdIns4P metabolism at Golgi and lysosomal membranes. Aberrantly increased PtdIns4P levels elevate constitutive anterograde secretion from the Golgi complex, and mTORC1 recruitment to lysosomes. NPC1 disease mutations phenocopy the transporter loss of function and can be rescued by inhibition or knockdown of either key phosphoinositide enzymes or their recruiting partners. In summary, we show that the lysosomal NPC1 cholesterol transporter tunes the molecular content of Golgi and lysosome MCS to regulate intracellular trafficking and growth signaling in health and disease.
Collapse
Affiliation(s)
- Candice Kutchukian
- Department of Physiology and Membrane BiologyUniversity of CaliforniaDavisCAUSA
| | - Oscar Vivas
- Department of Physiology and Membrane BiologyUniversity of CaliforniaDavisCAUSA
- Present address:
Department of Physiology and BiophysicsUniversity of WashingtonSeattleWAUSA
| | - Maria Casas
- Department of Physiology and Membrane BiologyUniversity of CaliforniaDavisCAUSA
| | - Julia G Jones
- Department of Physiology and Membrane BiologyUniversity of CaliforniaDavisCAUSA
| | - Scott A Tiscione
- Department of Physiology and Membrane BiologyUniversity of CaliforniaDavisCAUSA
| | - Sergi Simó
- Department of Cell Biology & Human AnatomyUniversity of CaliforniaDavisCAUSA
| | - Daniel S Ory
- Department of Internal MedicineWashington University School of MedicineSt. LouisMOUSA
| | - Rose E Dixon
- Department of Physiology and Membrane BiologyUniversity of CaliforniaDavisCAUSA
| | - Eamonn J Dickson
- Department of Physiology and Membrane BiologyUniversity of CaliforniaDavisCAUSA
| |
Collapse
|
18
|
Nash B, Irollo E, Brandimarti R, Meucci O. Opioid Modulation of Neuronal Iron and Potential Contributions to NeuroHIV. Methods Mol Biol 2021; 2201:139-162. [PMID: 32975796 DOI: 10.1007/978-1-0716-0884-5_13] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Opioid use has substantially increased over recent years and remains a major driver of new HIV infections worldwide. Clinical studies indicate that opioids may exacerbate the symptoms of HIV-associated neurocognitive disorders (HAND), but the mechanisms underlying opioid-induced cognitive decline remain obscure. We recently reported that the μ-opioid agonist morphine increased neuronal iron levels and levels of ferritin proteins that store iron, suggesting that opioids modulate neuronal iron homeostasis. Additionally, increased iron and ferritin heavy chain protein were necessary for morphine's ability to reduce the density of thin and mushroom dendritic spines in cortical neurons, which are considered critical mediators of learning and memory, respectively. As altered iron homeostasis has been reported in HAND and related neurocognitive disorders like Alzheimer's, Parkinson's, and Huntington's disease, understanding how opioids regulate neuronal iron metabolism may help identify novel drug targets in HAND with potential relevance to these other neurocognitive disorders. Here, we review the known mechanisms of opioid-mediated regulation of neuronal iron and corresponding cellular responses and discuss the implications of these findings for patients with HAND. Furthermore, we discuss a new molecular approach that can be used to understand if opioid modulation of iron affects the expression and processing of amyloid precursor protein and the contributions of this pathway to HAND.
Collapse
Affiliation(s)
- Bradley Nash
- Department of Pharmacology & Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Elena Irollo
- Department of Pharmacology & Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Renato Brandimarti
- Department of Pharmacology & Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Olimpia Meucci
- Department of Pharmacology & Physiology, Drexel University College of Medicine, Philadelphia, PA, USA.
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA, USA.
- Center for Neuroimmunology and CNS Therapeutics, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Irollo E, Luchetta J, Ho C, Nash B, Meucci O. Mechanisms of neuronal dysfunction in HIV-associated neurocognitive disorders. Cell Mol Life Sci 2021; 78:4283-4303. [PMID: 33585975 PMCID: PMC8164580 DOI: 10.1007/s00018-021-03785-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/14/2021] [Accepted: 01/29/2021] [Indexed: 12/13/2022]
Abstract
HIV-associated neurocognitive disorder (HAND) is characterized by cognitive and behavioral deficits in people living with HIV. HAND is still common in patients that take antiretroviral therapies, although they tend to present with less severe symptoms. The continued prevalence of HAND in treated patients is a major therapeutic challenge, as even minor cognitive impairment decreases patient’s quality of life. Therefore, modern HAND research aims to broaden our understanding of the mechanisms that drive cognitive impairment in people with HIV and identify promising molecular pathways and targets that could be exploited therapeutically. Recent studies suggest that HAND in treated patients is at least partially induced by subtle synaptodendritic damage and disruption of neuronal networks in brain areas that mediate learning, memory, and executive functions. Although the causes of subtle neuronal dysfunction are varied, reversing synaptodendritic damage in animal models restores cognitive function and thus highlights a promising therapeutic approach. In this review, we examine evidence of synaptodendritic damage and disrupted neuronal connectivity in HAND from clinical neuroimaging and neuropathology studies and discuss studies in HAND models that define structural and functional impairment of neurotransmission. Then, we report molecular pathways, mechanisms, and comorbidities involved in this neuronal dysfunction, discuss new approaches to reverse neuronal damage, and highlight current gaps in knowledge. Continued research on the manifestation and mechanisms of synaptic injury and network dysfunction in HAND patients and experimental models will be critical if we are to develop safe and effective therapies that reverse subtle neuropathology and cognitive impairment.
Collapse
Affiliation(s)
- Elena Irollo
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Jared Luchetta
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Chunta Ho
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Bradley Nash
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA. .,Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA. .,Center for Neuroimmunology and CNS Therapeutics, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
20
|
Yang C, Xia Z, Li T, Chen Y, Zhao M, Sun Y, Ma J, Wu Y, Wang X, Wang P, Wang H. Antioxidant Effect of Propofol in Gliomas and Its Association With Divalent Metal Transporter 1. Front Oncol 2020; 10:590931. [PMID: 33330075 PMCID: PMC7732593 DOI: 10.3389/fonc.2020.590931] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/26/2020] [Indexed: 01/31/2023] Open
Abstract
Background Oxidative stress enhances tumor invasion and metastasis in brain cancer. The activation of divalent metal transporter 1 (DMT1), which is regulated by glutamate receptors, can result in the increase of oxidative stress and risk of cancer development. Propofol, an anesthetic with antioxidant capacity, has been shown to decrease oxidative stress in several different types of cancer. However, the underlying mechanism remains unclear. Therefore, the present study aimed to elucidate the mechanism underlying the suppression of oxidative stress in glioma cells by propofol. It was hypothesized that propofol may inhibit oxidative stress in gliomas via suppressing Ca2+-permeable α-amino-3-hydroxyl-5-methylisoxazole-4-propionic acid (AMPA) receptor (CPAR)-DMT1 signaling. Methods Male Wistar rats with C6 gliomas, which were established by intracranial injection of C6 glioma cells, were either treated with propofol or not for 6 h before being sacrificed. The levels of AMPA receptor subunit GluR2 and DMT1 protein expression were assessed using western blotting. The association between CPARs and DMT1 was confirmed in vitro using the AMPA receptor activator (R, S)-AMPA. Glutathione and reactive oxygen species assay kits were used to evaluate tumor oxidative stress. The effect of propofol on glioma proliferation was evaluated by determining tumor weight, cell cycles and a growth curve. Results Propofol infusion at either 20 or 40 mg/kg-1/h-1 increased GluR2 levels and downregulated DMT1 expression as well as glutathione content markedly in the periphery compared with that in the glioma core. The in vitro results revealed that (R, S)-AMPA increased DMT1 expression and reactive oxygen species levels, which were partly reversed by propofol treatment. Conclusion Propofol regulated DMT1 expression by modulating CPARs, resulting in the inhibition of tumor oxidative stress and glioma growth. The present study provides evidence for optimizing the selection of anesthetic drugs in perioperative management and prognosis of patients with glioma.
Collapse
Affiliation(s)
- Chenyi Yang
- Department of Anesthesiology, The Third Central Hospital of Tianjin, Nankai University Affinity the Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Zhengyuan Xia
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| | - Tang Li
- Department of Anesthesiology, The Third Central Hospital of Tianjin, Nankai University Affinity the Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Yimeng Chen
- Department of Anesthesiology, The Third Central Hospital of Tianjin, Nankai University Affinity the Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Mingshu Zhao
- Department of Anesthesiology, The Third Central Hospital of Tianjin, Nankai University Affinity the Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Yi Sun
- Department of Anesthesiology, The Third Central Hospital of Tianjin, Nankai University Affinity the Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Ji Ma
- Department of Anesthesiology, The Third Central Hospital of Tianjin, Nankai University Affinity the Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Yi Wu
- Department of Anesthesiology, The Third Central Hospital of Tianjin, Nankai University Affinity the Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Xinyue Wang
- Department of Anesthesiology, The Third Central Hospital of Tianjin, Nankai University Affinity the Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Peng Wang
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Haiyun Wang
- Department of Anesthesiology, The Third Central Hospital of Tianjin, Nankai University Affinity the Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| |
Collapse
|
21
|
Chen JL, Lu JH, Xie CS, Shen YJ, Wang JW, Ye XY, Zhang MB, Jia GL, Tao YX, Li J, Cao H. Caveolin-1 in spinal cord modulates type-2 diabetic neuropathic pain through the Rac1/NOX2/NR2B signaling pathway. Am J Transl Res 2020; 12:1714-1727. [PMID: 32509171 PMCID: PMC7269978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 04/25/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVE The present study determines whether Cav-1 modulates the initiation, development and maintenance of type-2 DNP via the Rac1/NOX2-NR2B signaling pathway. METHODS After regular feeding for three days, these rats were randomly divided into two groups: control group with normal-diet (maintenance feed) (n=8); type-2 DM group (n=8). In the type-2 DM group, the rats were fed with a high-fat and high-sugar diet, and received a single intraperitoneal streptozotocin (STZ) injection (35 mg/kg). At two weeks after STZ injection, these diabetic neuropathic pain (DNP) rats were treated with daidzein (0.4 mg/kg/day) and N-tert-Butyl-α-phenylnitrone (PBN, 100 mg/kg/day) for 14 days. After the type-2 DNP model was successfully established, the rats were assigned into four groups: DNP group, DNP+Da group (DNP rats with Cav-1 specific inhibitor daidzein), DNP+PBN group (DNP rats treated with ROS scavenger PBN), and SC group (solvent control group). Then, the mechanical and thermal hyperalgesia were assayed to evaluate the function of the caveolin 1-Recombinant Human Ras-Related C1/nicotinamide adenosine diphosphate oxidase 2-NR2B gene (Cav-1-Rac1/NOX2-NR2B) signaling pathway. In the mechanism study, the protein expression levels of p-Caveolin-1, Rac1, NOX2, p-NR2B and t-NR2B, the production of ROS, and the distribution of Cav-1 and NOX2 in the spinal cord were observed. RESULTS The present study revealed that p-Cav-1 was persistently upregulated and activated in the spinal cord microglia in type-2 DNP rats. The use of the pharmacological inhibitor of Cav-1 and a ROS scavenger resulted to a significantly relieved mechanical allodynia and thermal hyperalgesia. In addition, it was demonstrated that Cav-1 promoted ROS generation via the activation of Rac1-dependent NADPH oxidase (NOX). CONCLUSION The present data suggests that Cav-1 in the spinal cord modulates type-2 DNP via regulating the Rac1/NOX2-NR2B pathway.
Collapse
Affiliation(s)
- Jia-Li Chen
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityZhejiang 325035, China
| | - Jia-Hui Lu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityZhejiang 325035, China
| | - Ci-Shan Xie
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityZhejiang 325035, China
| | - Yu-Jing Shen
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityZhejiang 325035, China
| | - Jun-Wu Wang
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityZhejiang 325035, China
| | - Xiu-Ying Ye
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityZhejiang 325035, China
| | - Mao-Biao Zhang
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityZhejiang 325035, China
| | - Gai-Li Jia
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityZhejiang 325035, China
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New JerseyNewark, New Jersey 07103, USA
| | - Jun Li
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityZhejiang 325035, China
| | - Hong Cao
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityZhejiang 325035, China
| |
Collapse
|
22
|
Wu J, Yang JJ, Cao Y, Li H, Zhao H, Yang S, Li K. Iron overload contributes to general anaesthesia-induced neurotoxicity and cognitive deficits. J Neuroinflammation 2020; 17:110. [PMID: 32276637 PMCID: PMC7149901 DOI: 10.1186/s12974-020-01777-6] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/17/2020] [Indexed: 11/30/2022] Open
Abstract
Background Increasing evidence suggests that multiple or long-time exposure to general anaesthesia (GA) could be detrimental to cognitive development in young subjects and might also contribute to accelerated neurodegeneration in the elderly. Iron is essential for normal neuronal function, and excess iron in the brain is implicated in several neurodegenerative diseases. However, the role of iron in GA-induced neurotoxicity and cognitive deficits remains elusive. Methods We used the primary hippocampal neurons and rodents including young rats and aged mice to examine whether GA impacted iron metabolism and whether the impact contributed to neuronal outcomes. In addition, a pharmacological suppression of iron metabolism was performed to explore the molecular mechanism underlying GA-mediated iron overload in the brain. Results Our results demonstrated that GA, induced by intravenous ketamine or inhalational sevoflurane, disturbed iron homeostasis and caused iron overload in both in vitro hippocampal neuron culture and in vivo hippocampus. Interestingly, ketamine- or sevoflurane-induced cognitive deficits, very likely, resulted from a novel iron-dependent regulated cell death, ferroptosis. Notably, iron chelator deferiprone attenuated the GA-induced mitochondrial dysfunction, ferroptosis, and further cognitive deficits. Moreover, we found that GA-induced iron overload was activated by NMDAR-RASD1 signalling via DMT1 action in the brain. Conclusion We conclude that disturbed iron metabolism may be involved in the pathogenesis of GA-induced neurotoxicity and cognitive deficits. Our study provides new vision for consideration in GA-associated neurological disorders.
Collapse
Affiliation(s)
- Jing Wu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing, 210093, China
| | - Jian-Jun Yang
- Department of Anesthesiology, The first Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Yan Cao
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing, 210093, China
| | - Huihui Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing, 210093, China
| | - Hongting Zhao
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing, 210093, China
| | - Shuofei Yang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China.
| | - Kuanyu Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing, 210093, China.
| |
Collapse
|
23
|
De La Fuente-Ortega E, Plaza-Briceño W, Vargas-Robert S, Haeger P. Prenatal Ethanol Exposure Misregulates Genes Involved in Iron Homeostasis Promoting a Maladaptation of Iron Dependent Hippocampal Synaptic Transmission and Plasticity. Front Pharmacol 2019; 10:1312. [PMID: 31787896 PMCID: PMC6855190 DOI: 10.3389/fphar.2019.01312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/15/2019] [Indexed: 01/05/2023] Open
Abstract
Prenatal ethanol exposure (PAE) induces behavioral maladptations in offspring, including a deficit in memory formation which is part of the umbrella sign of fetal alcohol spectrum disorder. Clinical and preclinical studies have shown that iron depletion exacerbates cognitive problems in offspring exposed to ethanol in utero and that PAE promotes dysregulation in brain iron homeostasis. However, the mechanisms underlying brain iron dysregulation and neuronal activity defects in adolescent offspring of PAE are unclear and poorly understand. Here, we used a PAE rat model to analyze messenger RNA (mRNA) and protein expression of iron homeostasis genes such as transferrin receptor (TfR), divalent metal transporter (DMT1), ferroportin (FPN1), and ferritin (FT) in brain areas associated with memory formation such as the prefrontal cortex (PFC), ventral tegmental area, and hippocampus. Interestingly, we found that 21 day old PAE rats have higher mRNA expression of DMT1 in the PFC, and TfR in the hippocampus, compared to control animals. In contrast FPN has lower mRNA expression in the PFC, and FT and FPN1 have lower expression in the hippocampus. In agreement with these results, we found a 1.5–2 fold increase of TfR and DMT1 protein levels both in the hippocampus and the PFC. Additionally, using an electrophysiological approach, we found that in hippocampal slices from PAE rats, iron treatment decreased long-term potentiation (LTP), but not AMPAR basal transmission (AMPAR fEPSP). In contrast, in control slices Fe-NTA did not affect LTP but decreased significantly the AMPAR fEPSP. Meanwhile, iron chelation with deferiprone decreased AMPAR transmission in PAE and control slices and decreased LTP only in controls slices. These results suggest that PAE affects iron homeostasis of specific brain areas—PFC and hippocampus—which could be involved in maladaptive cognition observed in this animal model.
Collapse
Affiliation(s)
- Erwin De La Fuente-Ortega
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Wladimir Plaza-Briceño
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Sofía Vargas-Robert
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Paola Haeger
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| |
Collapse
|
24
|
Iron Pathophysiology in Alzheimer’s Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1173:67-104. [DOI: 10.1007/978-981-13-9589-5_5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
25
|
Nash B, Tarn K, Irollo E, Luchetta J, Festa L, Halcrow P, Datta G, Geiger JD, Meucci O. Morphine-Induced Modulation of Endolysosomal Iron Mediates Upregulation of Ferritin Heavy Chain in Cortical Neurons. eNeuro 2019; 6:ENEURO.0237-19.2019. [PMID: 31300544 PMCID: PMC6675873 DOI: 10.1523/eneuro.0237-19.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 07/01/2019] [Indexed: 01/01/2023] Open
Abstract
HIV-associated neurocognitive disorders (HAND) remain prevalent and are aggravated by µ-opioid use. We have previously shown that morphine and other µ-opioids may contribute to HAND by inhibiting the homeostatic and neuroprotective chemokine receptor CXCR4 in cortical neurons, and this novel mechanism depends on upregulation of the protein ferritin heavy chain (FHC). Here, we examined the cellular events and potential mechanisms involved in morphine-mediated FHC upregulation using rat cortical neurons of either sex in vitro and in vivo. Morphine dose dependently increased FHC protein levels in primary neurons through µ-opioid receptor (µOR) and Gαi-protein signaling. Cytoplasmic FHC levels were significantly elevated, but nuclear FHC levels and FHC gene expression were unchanged. Morphine-treated rats also displayed increased FHC levels in layer 2/3 neurons of the prefrontal cortex. Importantly, both in vitro and in vivo FHC upregulation was accompanied by loss of mature dendritic spines, which was also dependent on µOR and Gαi-protein signaling. Moreover, morphine upregulated ferritin light chain (FLC), a component of the ferritin iron storage complex, suggesting that morphine altered neuronal iron metabolism. Indeed, prior to FHC upregulation, morphine increased cytoplasmic labile iron levels as a function of decreased endolysosomal iron. In line with this, chelation of endolysosomal iron (but not extracellular iron) blocked morphine-induced FHC upregulation and dendritic spine reduction, whereas iron overloading mimicked the effect of morphine on FHC and dendritic spines. Overall, these data demonstrate that iron mediates morphine-induced FHC upregulation and consequent dendritic spine deficits and implicate endolysosomal iron efflux to the cytoplasm in these effects.
Collapse
Affiliation(s)
- Bradley Nash
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Kevin Tarn
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Elena Irollo
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Jared Luchetta
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Lindsay Festa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Peter Halcrow
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203
| | - Gaurav Datta
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203
| | - Jonathan D Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19102
| |
Collapse
|
26
|
Ingrassia R, Garavaglia B, Memo M. DMT1 Expression and Iron Levels at the Crossroads Between Aging and Neurodegeneration. Front Neurosci 2019; 13:575. [PMID: 31231185 PMCID: PMC6560079 DOI: 10.3389/fnins.2019.00575] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 05/20/2019] [Indexed: 12/15/2022] Open
Abstract
Iron homeostasis is an essential prerequisite for metabolic and neurological functions throughout the healthy human life, with a dynamic interplay between intracellular and systemic iron metabolism. The development of different neurodegenerative diseases is associated with alterations of the intracellular transport of iron and heavy metals, principally mediated by Divalent Metal Transporter 1 (DMT1), responsible for Non-Transferrin Bound Iron transport (NTBI). In addition, DMT1 regulation and its compartmentalization in specific brain regions play important roles during aging. This review highlights the contribution of DMT1 to the physiological exchange and distribution of body iron and heavy metals during aging and neurodegenerative diseases. DMT1 also mediates the crosstalk between central nervous system and peripheral tissues, by systemic diffusion through the Blood Brain Barrier (BBB), with the involvement of peripheral iron homeostasis in association with inflammation. In conclusion, a survey about the role of DMT1 and iron will illustrate the complex panel of interrelationship with aging, neurodegeneration and neuroinflammation.
Collapse
Affiliation(s)
- Rosaria Ingrassia
- Section of Biotechnologies, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Barbara Garavaglia
- Medical Genetics and Neurogenetics Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Maurizio Memo
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
27
|
Spiers JG, Chen HJC, Bourgognon JM, Steinert JR. Dysregulation of stress systems and nitric oxide signaling underlies neuronal dysfunction in Alzheimer's disease. Free Radic Biol Med 2019; 134:468-483. [PMID: 30716433 DOI: 10.1016/j.freeradbiomed.2019.01.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/19/2018] [Accepted: 01/21/2019] [Indexed: 12/12/2022]
Abstract
Stress is a multimodal response involving the coordination of numerous body systems in order to maximize the chance of survival. However, long term activation of the stress response results in neuronal oxidative stress via reactive oxygen and nitrogen species generation, contributing to the development of depression. Stress-induced depression shares a high comorbidity with other neurological conditions including Alzheimer's disease (AD) and dementia, often appearing as one of the earliest observable symptoms in these diseases. Furthermore, stress and/or depression appear to exacerbate cognitive impairment in the context of AD associated with dysfunctional catecholaminergic signaling. Given there are a number of homologous pathways involved in the pathophysiology of depression and AD, this article will highlight the mechanisms by which stress-induced perturbations in oxidative stress, and particularly NO signaling, contribute to neurodegeneration.
Collapse
Affiliation(s)
- Jereme G Spiers
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, 3083, Australia.
| | - Hsiao-Jou Cortina Chen
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, 4072, Australia
| | | | - Joern R Steinert
- Department of Neuroscience, Psychology and Behavior, University of Leicester, Leicester, LE1 9HN, United Kingdom.
| |
Collapse
|
28
|
Neely CLC, Lippi SLP, Lanzirotti A, Flinn JM. Localization of Free and Bound Metal Species through X-Ray Synchrotron Fluorescence Microscopy in the Rodent Brain and Their Relation to Behavior. Brain Sci 2019; 9:E74. [PMID: 30925761 PMCID: PMC6523809 DOI: 10.3390/brainsci9040074] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 03/23/2019] [Accepted: 03/26/2019] [Indexed: 12/27/2022] Open
Abstract
Biometals in the brain, such as zinc, copper, and iron, are often discussed in cases of neurological disorders; however, these metals also have important regulatory functions and mediate cell signaling and plasticity. With the use of synchrotron X-ray fluorescence, our lab localized total, both bound and free, levels of zinc, copper, and iron in a cross section of one hemisphere of a rat brain, which also showed differing metal distributions in different regions within the hippocampus, the site in the brain known to be crucial for certain types of memory. This review discusses the several roles of these metals in brain regions with an emphasis on hippocampal cell signaling, based on spatial mapping obtained from X-ray fluorescence microscopy. We also discuss the localization of these metals and emphasize different cell types and receptors in regions with metal accumulation, as well as the potential relationship between this physiology and behavior.
Collapse
Affiliation(s)
- Caroline L C Neely
- Department of Psychology, George Mason University, 4400 University Drive, Fairfax, VA 22030, USA.
| | - Stephen L P Lippi
- Department of Psychology & Sociology, Angelo State University, 2601 W. Avenue N, ASU Station #10907, San Angelo, TX 76909, USA.
| | - Antonio Lanzirotti
- Center for Advanced Radiation Sources, University of Chicago, 9700 South Cass Avenue, Argonne, IL 60439, USA.
| | - Jane M Flinn
- Department of Psychology, George Mason University, 4400 University Drive, Fairfax, VA 22030, USA.
| |
Collapse
|
29
|
DeGregorio-Rocasolano N, Martí-Sistac O, Gasull T. Deciphering the Iron Side of Stroke: Neurodegeneration at the Crossroads Between Iron Dyshomeostasis, Excitotoxicity, and Ferroptosis. Front Neurosci 2019; 13:85. [PMID: 30837827 PMCID: PMC6389709 DOI: 10.3389/fnins.2019.00085] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 01/25/2019] [Indexed: 12/21/2022] Open
Abstract
In general, iron represents a double-edged sword in metabolism in most tissues, especially in the brain. Although the high metabolic demands of brain cells require iron as a redox-active metal for ATP-producing enzymes, the brain is highly vulnerable to the devastating consequences of excessive iron-induced oxidative stress and, as recently found, to ferroptosis as well. The blood-brain barrier (BBB) protects the brain from fluctuations in systemic iron. Under pathological conditions, especially in acute brain pathologies such as stroke, the BBB is disrupted, and iron pools from the blood gain sudden access to the brain parenchyma, which is crucial in mediating stroke-induced neurodegeneration. Each brain cell type reacts with changes in their expression of proteins involved in iron uptake, efflux, storage, and mobilization to preserve its internal iron homeostasis, with specific organelles such as mitochondria showing specialized responses. However, during ischemia, neurons are challenged with excess extracellular glutamate in the presence of high levels of extracellular iron; this causes glutamate receptor overactivation that boosts neuronal iron uptake and a subsequent overproduction of membrane peroxides. This glutamate-driven neuronal death can be attenuated by iron-chelating compounds or free radical scavenger molecules. Moreover, vascular wall rupture in hemorrhagic stroke results in the accumulation and lysis of iron-rich red blood cells at the brain parenchyma and the subsequent presence of hemoglobin and heme iron at the extracellular milieu, thereby contributing to iron-induced lipid peroxidation and cell death. This review summarizes recent progresses made in understanding the ferroptosis component underlying both ischemic and hemorrhagic stroke subtypes.
Collapse
Affiliation(s)
- Núria DeGregorio-Rocasolano
- Cellular and Molecular Neurobiology Research Group, Department of Neurosciences, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | - Octavi Martí-Sistac
- Cellular and Molecular Neurobiology Research Group, Department of Neurosciences, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Teresa Gasull
- Cellular and Molecular Neurobiology Research Group, Department of Neurosciences, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| |
Collapse
|
30
|
Abstract
The key molecular events that provoke Parkinson's disease (PD) are not fully understood. Iron deposit was found in the substantia nigra pars compacta (SNpc) of PD patients and animal models, where dopaminergic neurons degeneration occurred selectively. The mechanisms involved in disturbed iron metabolism remain unknown, however, considerable evidence indicates that iron transporters dysregulation, activation of L-type voltage-gated calcium channel (LTCC) and ATP-sensitive potassium (KATP) channels, as well as N-methyl-D-aspartate (NMDA) receptors (NMDARs) contribute to this process. There is emerging evidence on the structural links and functional modulations between iron and α-synuclein, and the key player in PD which aggregates in Lewy bodies. Iron is believed to modulate α-synuclein synthesis, post-translational modification, and aggregation. Furthermore, glia, especially activated astroglia and microglia, are involved in iron deposit in PD. Glial contributions were largely dependent on the factors they released, e.g., neurotrophic factors, pro-inflammatory factors, lactoferrin, and those undetermined. Therefore, iron chelation using iron chelators, the extracts from many natural foods with iron chelating properties, may be an effective therapy for prevention and treatment of the disease.
Collapse
|
31
|
Association of metals with the risk and clinical characteristics of Parkinson's disease. Parkinsonism Relat Disord 2018; 55:117-121. [DOI: 10.1016/j.parkreldis.2018.05.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/15/2018] [Accepted: 05/27/2018] [Indexed: 01/12/2023]
|
32
|
Dexras1 is a homeostatic regulator of exercise-dependent proliferation and cell survival in the hippocampal neurogenic niche. Sci Rep 2018; 8:5294. [PMID: 29593295 PMCID: PMC5871767 DOI: 10.1038/s41598-018-23673-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 03/19/2018] [Indexed: 02/07/2023] Open
Abstract
Adult hippocampal neurogenesis is highly responsive to exercise, which promotes the proliferation of neural progenitor cells and the integration of newborn granule neurons in the dentate gyrus. Here we show that genetic ablation of the small GTPase, Dexras1, suppresses exercise-induced proliferation of neural progenitors, alters survival of mitotic and post-mitotic cells in a stage-specific manner, and increases the number of mature newborn granule neurons. Dexras1 is required for exercise-triggered recruitment of quiescent neural progenitors into the cell cycle. Pharmacological inhibition of NMDA receptors enhances SGZ cell proliferation in wild-type but not dexras1-deficient mice, suggesting that NMDA receptor-mediated signaling is dependent on Dexras1. At the molecular level, the absence of Dexras1 abolishes exercise-dependent activation of ERK/MAPK and CREB, and inhibits the upregulation of NMDA receptor subunit NR2A, bdnf, trkB and vegf-a expression in the dentate gyrus. Our study reveals Dexras1 as an important stage-specific regulator of exercise-induced neurogenesis in the adult hippocampus by enhancing pro-mitogenic signaling to neural progenitor cells and modulating cell survival.
Collapse
|
33
|
Xu H, Jiang H, Xie J. New Insights into the Crosstalk between NMDARs and Iron: Implications for Understanding Pathology of Neurological Diseases. Front Mol Neurosci 2017; 10:71. [PMID: 28360837 PMCID: PMC5352910 DOI: 10.3389/fnmol.2017.00071] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 03/01/2017] [Indexed: 12/11/2022] Open
Abstract
Both iron dyshomeostasis and N-methyl-D-aspartate receptors (NMDARs)-mediated neurotoxicity have been shown to have an important role in neurological diseases such as Parkinson’s disease (PD) and Alzheimer’s disease (AD). Evidence proved that activation of NMDARs could promote iron overload and iron-induced neurotoxicity by enhancing iron importer divalent metal transporter 1 (DMT1)-mediated iron uptake and iron releasing from lysosome. Also, iron overload could regulate NMDARs-mediated synaptic transmission. This indicates that there might be a possible relationship between iron and activation of NMDARs in neurological diseases. Understanding this interaction between iron and activation of NMDARs may provide new therapeutic avenues for a more targeted neurotherapeutic strategy for these diseases. Therefore, in this review article, we will describe the dysfunction of iron metabolism and NMDARs in neurological diseases including PD and AD, and summarize the new insight into the mechanisms underlying the interaction between iron and activation of NMDARs.
Collapse
Affiliation(s)
- Huamin Xu
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao UniversityQingdao, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao UniversityQingdao, China
| | - Hong Jiang
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao UniversityQingdao, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao UniversityQingdao, China
| | - Junxia Xie
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao UniversityQingdao, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao UniversityQingdao, China
| |
Collapse
|
34
|
Jafri AJA, Arfuzir NNN, Lambuk L, Iezhitsa I, Agarwal R, Agarwal P, Razali N, Krasilnikova A, Kharitonova M, Demidov V, Serebryansky E, Skalny A, Spasov A, Yusof APM, Ismail NM. Protective effect of magnesium acetyltaurate against NMDA-induced retinal damage involves restoration of minerals and trace elements homeostasis. J Trace Elem Med Biol 2017; 39:147-154. [PMID: 27908408 DOI: 10.1016/j.jtemb.2016.09.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/05/2016] [Accepted: 09/14/2016] [Indexed: 02/08/2023]
Abstract
Glutamate-mediated excitotoxicity involving N-methyl-d-aspartate (NMDA) receptors has been recognized as a final common outcome in pathological conditions involving death of retinal ganglion cells (RGCs). Overstimulation of NMDA receptors results in influx of calcium (Ca) and sodium (Na) ions and efflux of potassium (K). NMDA receptors are blocked by magnesium (Mg). Such changes due to NMDA overstimulation are also associated with not only the altered levels of minerals but also that of trace elements and redox status. Both the decreased and elevated levels of trace elements such as iron (Fe), zinc (Zn), copper (Cu) affect NMDA receptor excitability and redox status. Manganese (Mn), and selenium (Se) are also part of antioxidant defense mechanisms in retina. Additionally endogenous substances such as taurine also affect NMDA receptor activity and retinal redox status. Therefore, the aim of this study was to evaluate the effect of Mg acetyltaurate (MgAT) on the retinal mineral and trace element concentration, oxidative stress, retinal morphology and retinal cell apoptosis in rats after-NMDA exposure. One group of Sprague Dawley rats received intravitreal injection of vehicle while 4 other groups similarly received NMDA (160nmolL-1). Among the NMDA injected groups, 3 groups also received MgAT (320nmolL-1) as pre-treatment, co-treatment or post-treatment. Seven days after intravitreal injection, rats were sacrificed, eyes were enucleated and retinae were isolated for estimation of mineral (Ca, Na, K, Mg) and trace element (Mn, Cu, Fe, Se, Zn) concentration using Inductively Coupled Plasma (DRC ICP-MS) techniques (NexION 300D), retinal oxidative stress using Elisa, retinal morphology using H&E staining and retinal cell apoptosis using terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL). Intravitreal NMDA injection resulted in increased concentration of Ca (4.6 times, p<0.0001), Mg (1.5 times, p<0.01), Na (3 times, p<0.0001) and K (2.3 times, p<0.0001) compared to vehicle injected group. This was accompanied with significant increase of Ca/Mg and Na/K ratios, 3 and 1.27 times respectively, compared to control group. The trace elements such as Cu, Fe and Zn also showed a significant increase amounting to 3.3 (p<0.001), 2.3 (p<0.0001) and 3 (p<0.0001) times respectively compared to control group. Se was increased by 60% (p<0.005). Pre-treatment with MgAT abolished effect of NMDA on minerals and trace elements more effectively than co- and post-treatment. Similar observations were made for retinal oxidative stress, retinal morphology and retinal cell apoptosis. In conclusion, current study demonstrated the protective effect of MgAT against NMDA-induced oxidative stress and retinal cell apoptosis. This effect of MgAT was associated with restoration of retinal concentrations of minerals and trace elements. Further studies are warranted to explore the precise molecular targets of MgAT. Nevertheless, MgAT seems a potential candidate in the management of diseases involving NMDA-induced excitotoxicity.
Collapse
Affiliation(s)
- Azliana Jusnida Ahmad Jafri
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Selangor, Malaysia
| | - Natasha Najwa Nor Arfuzir
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Selangor, Malaysia
| | - Lidawani Lambuk
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Selangor, Malaysia
| | - Igor Iezhitsa
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Selangor, Malaysia; Volgograd State Medical University, Research Institute of Pharmacology, Volgograd, Russia.
| | - Renu Agarwal
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Selangor, Malaysia
| | - Puneet Agarwal
- International Medical University, IMU Clinical School, Seremban, Malaysia
| | - Norhafiza Razali
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Selangor, Malaysia
| | - Anna Krasilnikova
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Selangor, Malaysia
| | - Maria Kharitonova
- Volgograd State Medical University, Research Institute of Pharmacology, Volgograd, Russia; Institute of Pharmacy, Department of Pharmacology and Toxicology, University of Innsbruck, Center for Chemistry and Biomedicine, Innrain 80 - 82/III, A-6020, Innsbruck, Austria
| | - Vasily Demidov
- Russian Society of Trace Elements in Medicine, 46 Zemlyanoy Val str., Moscow, 105064, Russia
| | - Evgeny Serebryansky
- Russian Society of Trace Elements in Medicine, 46 Zemlyanoy Val str., Moscow, 105064, Russia
| | - Anatoly Skalny
- Russian Society of Trace Elements in Medicine, 46 Zemlyanoy Val str., Moscow, 105064, Russia; Peoples' Friendship University of Russia, Moscow, Russia; All-Russian Research Institute of Medicinal and Aromatic Plants, Moscow, Russia
| | - Alexander Spasov
- Volgograd State Medical University, Research Institute of Pharmacology, Volgograd, Russia
| | - Ahmad Pauzi Md Yusof
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Selangor, Malaysia
| | - Nafeeza Mohd Ismail
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Selangor, Malaysia
| |
Collapse
|