1
|
Khanra S, Singh S, Singh TG. Mechanistic exploration of ubiquitination-mediated pathways in cerebral ischemic injury. Mol Biol Rep 2024; 52:22. [PMID: 39607439 DOI: 10.1007/s11033-024-10123-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
The ubiquitin-proteasome system (UPS) plays a pivotal role in regulating protein homeostasis and cellular processes, including protein degradation, trafficking, DNA repair, and cell signaling. During cerebral ischemia, ischemic conditions profoundly disrupt UPS activity, leading to proteasomal dysfunction and the accumulation of abnormal proteins. This imbalance contributes to neuronal injury and cell death observed in ischemic stroke. The UPS is intricately linked to various signaling pathways crucial for neuronal survival, inflammation, and cellular stress response, such as NF-κB, TRIM, TRIP, JAK-STAT, PI3K/Akt, and ERK1/2. Alterations in the ubiquitination process can significantly impact the activation and regulation of these pathways, exacerbating ischemic brain injury. Therapeutic approaches targeting the UPS in cerebral ischemia aim to rebalance protein levels, reduce proteotoxic stress, and mitigate neuronal injury. Strategies include proteasome inhibition, targeting specific ubiquitin ligases and deubiquitinating enzymes, and modulating ubiquitination-mediated regulation of key signaling pathways implicated in ischemia-induced pathophysiology. Therefore, the present review discusses the molecular mechanisms underlying UPS dysfunction in ischemic stroke is crucial for developing effective therapeutic interventions. Modulating ubiquitination-mediated pathways through therapeutic interventions targeting specific UPS components holds significant promise for mitigating ischemic brain injury and promoting neuroprotection and functional recovery in patients with cerebral ischemia.
Collapse
Affiliation(s)
- Supriya Khanra
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| |
Collapse
|
2
|
Lee DH, Song J. Impaired olfactory system in metabolic imbalance-related neuropathology. Life Sci 2024; 355:122967. [PMID: 39142504 DOI: 10.1016/j.lfs.2024.122967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/25/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
Olfactory dysfunction, influenced by factors such as aging and environmental stress, is linked to various neurological disorders. The olfactory bulb's connections to brain areas like the hypothalamus, piriform cortex, entorhinal cortex, and limbic system make olfactory dysfunction a contributor to a range of neuropathological conditions. Recent research has underscored that olfactory deficits are prevalent in individuals with both metabolic syndrome and dementia. These systemic metabolic alterations correlate with olfactory impairments, potentially affecting brain regions associated with the olfactory bulb. In cases of metabolic syndrome, phenomena such as insulin resistance and disrupted glucose metabolism may result in compromised olfactory function, leading to multiple neurological issues. This review synthesizes key findings on the interplay between metabolic-induced olfactory dysfunction and neuropathology. It emphasizes the critical role of olfactory assessment in diagnosing and managing neurological diseases related to metabolic syndrome.
Collapse
Affiliation(s)
- Dong Hoon Lee
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School & Hwasun Hospital, Hwasun 58128, Republic of Korea.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Republic of Korea.
| |
Collapse
|
3
|
Lin W, Zhao XY, Cheng JW, Li LT, Jiang Q, Zhang YX, Han F. Signaling pathways in brain ischemia: Mechanisms and therapeutic implications. Pharmacol Ther 2023; 251:108541. [PMID: 37783348 DOI: 10.1016/j.pharmthera.2023.108541] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
Ischemic stroke occurs when the arteries supplying blood to the brain are narrowed or blocked, inducing damage to brain tissue due to a lack of blood supply. One effective way to reduce brain damage and alleviate symptoms is to reopen blocked blood vessels in a timely manner and reduce neuronal damage. To achieve this, researchers have focused on identifying key cellular signaling pathways that can be targeted with drugs. These pathways include oxidative/nitrosative stress, excitatory amino acids and their receptors, inflammatory signaling molecules, metabolic pathways, ion channels, and other molecular events involved in stroke pathology. However, evidence suggests that solely focusing on protecting neurons may not yield satisfactory clinical results. Instead, researchers should consider the multifactorial and complex mechanisms underlying stroke pathology, including the interactions between different components of the neurovascular unit. Such an approach is more representative of the actual pathological process observed in clinical settings. This review summarizes recent research on the multiple molecular mechanisms and drug targets in ischemic stroke, as well as recent advances in novel therapeutic strategies. Finally, we discuss the challenges and future prospects of new strategies based on the biological characteristics of stroke.
Collapse
Affiliation(s)
- Wen Lin
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiang-Yu Zhao
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jia-Wen Cheng
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Li-Tao Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang 050051, Hebei, China
| | - Quan Jiang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Yi-Xuan Zhang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, China.
| | - Feng Han
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, China; Institute of Brain Science, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
4
|
Mahemuti Y, Kadeer K, Su R, Abula A, Aili Y, Maimaiti A, Abulaiti S, Maimaitituerxun M, Miao T, Jiang S, Axier A, Aisha M, Wang Y, Cheng X. TSPO exacerbates acute cerebral ischemia/reperfusion injury by inducing autophagy dysfunction. Exp Neurol 2023; 369:114542. [PMID: 37717810 DOI: 10.1016/j.expneurol.2023.114542] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/19/2023]
Abstract
Autophagy is considered a double-edged sword, with a role in the regulation of the pathophysiological processes of the central nervous system (CNS) after cerebral ischemia-reperfusion injury (CIRI). The 18-kDa translocator protein (TSPO) is a highly conserved protein, with its expression level in the nervous system closely associated with the regulation of pathophysiological processes. In addition, the ligand of TSPO reduces neuroinflammation in brain diseases, but the potential role of TSPO in CIRI is largely undiscovered. On this basis, we investigated whether TSPO regulates neuroinflammatory response by affecting autophagy in microglia. In our study, increased expression of TSPO was detected in rat brain tissues with transient middle cerebral artery occlusion (tMCAO) and in BV2 microglial cells exposed to oxygen-glucose deprivation or reoxygenation (OGD/R) treatment, respectively. In addition, we confirmed that autophagy was over-activated during CIRI by increased expression of autophagy activation related proteins with Beclin-1 and LC3B, while the expression of p62 was decreased. The degradation process of autophagy was inhibited, while the expression levels of LAMP-1 and Cathepsin-D were significantly reduced. Results of confocal laser microscopy and transmission electron microscopy (TEM) indicated that autophagy flux was disordered. In contrast, inhibition of TSPO prevented autophagy over-activation both in vivo and in vitro. Interestingly, suppression of TSPO alleviated nerve cell damage by reducing reactive oxygen species (ROS) and pro-inflammatory factors, including TNF-α and IL-6 in microglia cells. In summary, these results indicated that TSPO might affect CIRI by mediating autophagy dysfunction and thus might serve as a potential target for ischemic stroke treatment.
Collapse
Affiliation(s)
- Yusufu Mahemuti
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China; School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, PR China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, Zhejiang, PR China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, Zhejiang, PR China
| | - Kaheerman Kadeer
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Riqing Su
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Abudureheman Abula
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Yirizhati Aili
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Aierpati Maimaiti
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Subinuer Abulaiti
- Department of Epidemiology and Biostatistics, Institute of Public Health, Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | | | - Tong Miao
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Shihao Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Aximujiang Axier
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Maimaitili Aisha
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Yongxin Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Xiaojiang Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China.
| |
Collapse
|
5
|
Wang X, Tian X, Shen H, Zhang X, Xie L, Chen M. Moderate Hyperkalemia Regulates Autophagy to Reduce Cerebral Ischemia-Reperfusion Injury in a CA/CPR Rat Model. Brain Sci 2023; 13:1285. [PMID: 37759886 PMCID: PMC10526941 DOI: 10.3390/brainsci13091285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/26/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Cerebral ischemia-reperfusion injury (CIRI) can cause irreversible brain damage and autophagy has been implicated in the pathophysiology. Increasing serum potassium (K+) levels reduces CIRI, but the relationship between its protective mechanism and autophagy is unclear. In this study, we aimed to find the optimal degree of raising serum (K+) and to investigate the relationship between high (K+) and autophagy and the underlying mechanisms in a cardiac arrest/cardiopulmonary resuscitation (CA/CPR) rat model. METHODS Sprague Dawley (SD) rats were divided into four groups: S group, N group, P group, and Q group. The rats S group and N group were administered saline. The rats P group and Q group were administered 640 mg/kg of potassium chloride (KCl) continuously pumped at 4 mL/h (21.3 mg/(kg·min) and divided according to the electrocardiogram (ECG) changes during the administration of KCl. After 24-h of resuscitation, neural damage was assessed by measuring neurological deficit score (NDS), oxidative stress markers, and pathological staining of the cerebral cortex. The level of autophagy and the expression of mTOR-ULK1-Beclin1 pathway-related proteins were evaluated using transmission electron microscopy (TEM), immunostaining, and western blotting. RESULTS Our results revealed that high (K+) improved NDS and decreased the oxidative stress markers. The autophagosomes, autolysosomes, and lysosomes were decreased following treatment KCl. Furthermore, the levels of micro-tubule-associated protein 1 light chain 3 (LC3) Ⅱ/Ⅰ, Unc-51-like kinase 1 (ULK1), and Beclin1 were decreased, whereas mTOR expression was increased in the cortex. CONCLUSION The results demonstrated that moderate hyperkalemia could alleviate autophagy after CIRI via regulating the mTOR-ULK1-Beclin1 pathway.
Collapse
Affiliation(s)
- Xiaoqin Wang
- The Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China; (X.W.); (X.T.); (H.S.)
| | - Xinyue Tian
- The Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China; (X.W.); (X.T.); (H.S.)
| | - Haiying Shen
- The Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China; (X.W.); (X.T.); (H.S.)
| | - Xiaohua Zhang
- The Department of Physiology, Guangxi Medical University, Nanning 530021, China; (X.Z.); (L.X.)
| | - Lu Xie
- The Department of Physiology, Guangxi Medical University, Nanning 530021, China; (X.Z.); (L.X.)
| | - Menghua Chen
- The Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China; (X.W.); (X.T.); (H.S.)
| |
Collapse
|
6
|
Novel Therapeutic Strategies for Ischemic Stroke: Recent Insights into Autophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3450207. [PMID: 35720192 PMCID: PMC9200548 DOI: 10.1155/2022/3450207] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 04/24/2022] [Accepted: 05/11/2022] [Indexed: 11/18/2022]
Abstract
Stroke is one of the leading causes of death and disability worldwide. Autophagy is a conserved cellular catabolic pathway that maintains cellular homeostasis by removal of damaged proteins and organelles, which is critical for the maintenance of energy and function homeostasis of cells. Accumulating evidence demonstrates that autophagy plays important roles in pathophysiological mechanisms under ischemic stroke. Previous investigations show that autophagy serves as a “double-edged sword” in ischemic stroke as it can either promote the survival of neuronal cells or induce cell death in special conditions. Following ischemic stroke, autophagy is activated or inhibited in several cell types in brain, including neurons, astrocytes, and microglia, as well as microvascular endothelial cells, which involves in inflammatory activation, modulation of microglial phenotypes, and blood-brain barrier permeability. However, the exact mechanisms of underlying the role of autophagy in ischemic stroke are not fully understood. This review focuses on the recent advances regarding potential molecular mechanisms of autophagy in different cell types. The focus is also on discussing the “double-edged sword” effect of autophagy in ischemic stroke and its possible underlying mechanisms. In addition, potential therapeutic strategies for ischemic stroke targeting autophagy are also reviewed.
Collapse
|
7
|
Huang R, Zhang C, Wang X, Hu H. PPARγ in Ischemia-Reperfusion Injury: Overview of the Biology and Therapy. Front Pharmacol 2021; 12:600618. [PMID: 33995008 PMCID: PMC8117354 DOI: 10.3389/fphar.2021.600618] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Ischemia-reperfusion injury (IRI) is a complex pathophysiological process that is often characterized as a blood circulation disorder caused due to various factors (such as traumatic shock, surgery, organ transplantation, burn, and thrombus). Severe metabolic dysregulation and tissue structure destruction are observed upon restoration of blood flow to the ischemic tissue. Theoretically, IRI can occur in various tissues and organs, including the kidney, liver, myocardium, and brain, among others. The advances made in research regarding restoring tissue perfusion in ischemic areas have been inadequate with regard to decreasing the mortality and infarct size associated with IRI. Hence, the clinical treatment of patients with severe IRI remains a thorny issue. Peroxisome proliferator-activated receptor γ (PPARγ) is a member of a superfamily of nuclear transcription factors activated by agonists and is a promising therapeutic target for ameliorating IRI. Therefore, this review focuses on the role of PPARγ in IRI. The protective effects of PPARγ, such as attenuating oxidative stress, inhibiting inflammatory responses, and antagonizing apoptosis, are described, envisaging certain therapeutic perspectives.
Collapse
Affiliation(s)
- Ruizhen Huang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chiyu Zhang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xing Wang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Honglin Hu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
8
|
Almeida MR, Silva AR, Elias I, Fernandes C, Machado R, Galego O, Santo GC. SQSTM1 gene as a potential genetic modifier of CADASIL phenotype. J Neurol 2020; 268:1453-1460. [PMID: 33216222 DOI: 10.1007/s00415-020-10308-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/05/2020] [Accepted: 11/08/2020] [Indexed: 10/23/2022]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most common inherited cerebral small vessel disease and is caused by mutations in the NOTCH3 gene. Interestingly, CADASIL patients present a large phenotypic variability even harboring the same pathogenic variant. We describe two CADASIL siblings with a particularly aggressive clinical phenotype characterized by early-onset stroke, gait disturbances and/or dementia, severe emotional dysregulation, and dysexecutive syndrome together with a severe white matter burden on MRI. The genetic analysis revealed the co-occurrence of NOTCH3 (p.Gly420Cys) and SQSTM1 (p.Ser275Phefs*17) pathogenic variants which might worsen the aggressiveness of disease progression in both siblings. Interestingly, to the best of our knowledge, mutations in SQSTM1 gene have never been described in CADASIL patients before. Curiously, both Notch3 and p62 encoded proteins have a key role in the autophagy-lysosomal pathway which is impaired in CADASIL patients. Thus, the contribution of SQSTM1 gene to the clinical heterogeneity of CADASIL patients, in particular for those who develop cognitive impairment or dementia at an early age, is certainly overlooked. Therefore, we advocate expanding the genetic analysis to other genes associated with the phenotype spectrum of CADASIL patients using NGS-customized gene panel.
Collapse
Affiliation(s)
- Maria Rosário Almeida
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marques de Pombal, 3004-517, Coimbra, Portugal.
| | - Ana Rita Silva
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marques de Pombal, 3004-517, Coimbra, Portugal
| | - Inês Elias
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marques de Pombal, 3004-517, Coimbra, Portugal
| | | | - Rita Machado
- Neurology Department, Coimbra University Hospital, Coimbra, Portugal
| | - Orlando Galego
- Neuroradiology Department, Coimbra University Hospital, Coimbra, Portugal
| | - Gustavo Cordeiro Santo
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marques de Pombal, 3004-517, Coimbra, Portugal.,Neurology Department, Coimbra University Hospital, Coimbra, Portugal
| |
Collapse
|
9
|
He GQ, Chen Y, Liao HJ, Xu WM, Zhang W, He GL. Associations between Huwe1 and autophagy in rat cerebral neuron oxygen‑glucose deprivation and reperfusion injury. Mol Med Rep 2020; 22:5083-5094. [PMID: 33173969 PMCID: PMC7646962 DOI: 10.3892/mmr.2020.11611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 07/15/2020] [Indexed: 12/22/2022] Open
Abstract
Autophagy and the ubiquitin proteasome system (UPS) are two major protein degradation pathways involved in brain ischemia. Autophagy can compensate for UPS impairment-induced cellular dysfunction. HECT, UBA and WWE domain containing E3 ubiquitin protein ligase 1 (Huwe1), an E3 ubiquitin ligase, serves critical roles in nervous system plasticity, regeneration and disease. However, the role of Huwe1 in autophagy in brain ischemia/reperfusion (I/R) injury remains unknown. The aim of the present study was to investigate the crosstalk between autophagy and the UPS in brain ischemia. The present study established an oxygen-glucose deprivation and reperfusion (OGD/R) model in rat primary cortex neurons in vitro. Lentiviral interference was used to silence the expression of Huwe1. An autophagy promoter (rapamycin), an autophagy inhibitor (wortmannin) and a JNK pathway inhibitor (SP600125) were also used in the current study. Cellular autophagy-related proteins, including Beclin-1, autophagy related (ATG) 7, ATG5, ATG3 and microtubule associated protein 1 light chain 3 α, and apoptosis-related proteins, such as P53, cleaved caspase 3, Bax and Bcl2, were detected via western blotting and immunocytochemistry. Neuronal apoptosis was evaluated using a TUNEL assay. The results demonstrated that silencing Huwe1 increased the expression levels of autophagy-related proteins at 24 h after OGD/R. Treatment with a JNK inhibitor or cotreatment with Huwe1 shRNA significantly increased autophagy. Rapamycin increased apoptosis under OGD/R conditions. However, treatment with Huwe1 shRNA decreased the number of TUNEL-positive cells at 24 h after OGD/R. Cotreatment with Huwe1 shRNA and wortmannin alleviated neuronal apoptosis under OGD/R conditions compared with cotreatment with DMSO. Collectively, the present results suggested that silencing Huwe1 was accompanied by a compensatory induction of autophagy under OGD/R conditions. Furthermore, the JNK pathway may be a key mediator of the interaction between Huwe1 and autophagy in response to UPS impairment.
Collapse
Affiliation(s)
- Guo-Qian He
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Yan Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Hui-Juan Liao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Wen-Ming Xu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Wei Zhang
- Department of Medical Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Cancer Hospital Affiliated to School of Medicine, Chengdu, Sichuan 610041, P.R. China
| | - Guo-Lin He
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
10
|
Remote Ischemic Postconditioning Inhibits Hippocampal Neuronal Apoptosis and Mitophagy After Cardiopulmonary Resuscitation in Rats. Shock 2020; 55:74-82. [PMID: 32590695 DOI: 10.1097/shk.0000000000001596] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Studies have shown that remote ischemic post-conditioning can improve brain damage caused by ischemia and hypoxia. However, the specific mechanism underlying this phenomenon is still unclear. The purpose of this study was to investigate the effects of remote ischemic post-conditioning on neuronal apoptosis and mitophagy after cardiopulmonary resuscitation (CPR) in rats. METHODS Male Sprague-Dawley rats were used to establish an asphyxia cardiac arrest model by clamping the tracheal duct. First, the expression levels of P53, Cytochrome c (Cytc), and Parkin in the cytoplasm and mitochondria were observed at 3, 6, 24, and 72 h after the restoration of spontaneous circulation (ROSC). Then neurological deficit scores, hippocampal neuron apoptosis, mitochondrial P53 and Parkin, cytoplasmic Cytc, and neuron ultrastructure were evaluated 24 h after ROSC. RESULTS P53 and Parkin can translocate from the cytoplasm to the mitochondria, promoting the translocation of cytoplasmic Cytc to mitochondria after CPR, reaching a peak at 24 h after the ROSC. The P53 inhibitor Pifithrin-μ reduced apoptosis induced by P53 mitochondrial translocation. Apoptosis was induced after cardiac arrest and attenuated by remote ischemic postconditioning via inhibiting P53 mitochondrial translocation and the release of Cytc to the cytoplasm. In addition, remote ischemic postconditioning could inhibit Parkin-mediated mitophagy. CONCLUSION Taken together, our results show that remote ischemic post-conditioning improves neural function after CPR by inhibiting P53 mitochondrial translocation-induced apoptosis and Parkin-mediated mitophagy.
Collapse
|
11
|
Neuroprotection by Phytoestrogens in the Model of Deprivation and Resupply of Oxygen and Glucose In Vitro: The Contribution of Autophagy and Related Signaling Mechanisms. Antioxidants (Basel) 2020; 9:antiox9060545. [PMID: 32580379 PMCID: PMC7346137 DOI: 10.3390/antiox9060545] [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: 04/27/2020] [Revised: 06/13/2020] [Accepted: 06/18/2020] [Indexed: 12/19/2022] Open
Abstract
Phytoestrogens can have a neuroprotective effect towards ischemia-reperfusion-induced neuronal damage. However, their mechanism of action has not been well described. In this work, we investigate the type of neuronal cell death induced by oxygen and glucose deprivation (OGD) and resupply (OGDR) and pinpoint some of the signaling mechanisms whereby the neuroprotective effects of phytoestrogens occur in these conditions. First, we found that autophagy initiation affords neuronal protection upon neuronal damage induced by OGD and OGDR. The mammalian target of rapamycin/ribosomal S6 kinase (mTOR/S6K) pathway is blocked in these conditions, and we provide evidence that this is mediated by modulation of both the 5′ AMP-activated protein kinase (AMPK) and phosphatidylinositol-3-kinase/protein kinase B (PI3K/AKT) pathways. These are dampened up or down, respectively, under OGDR-induced neuronal damage. In contrast, the MAPK-Erk kinase/extracellular signal-regulated kinase (MEK/ERK) pathway is increased under these conditions. Regarding the pathways affected by phytoestrogens, we show that their protective properties require autophagy initiation, but at later stages, they decrease mitogen-activated protein kinase (MAPK) and AMPK activation and increase mTOR/S6K activation. Collectively, our results put forward a novel mode of action where phytoestrogens play a dual role in the regulation of autophagy by acting as autophagy initiation enhancers when autophagy is a neuroprotective and pro-survival mechanism, and as autophagy initiation inhibitors when autophagy is a pro-death mechanism. Finally, our results support the therapeutic potential of phytoestrogens in brain ischemia by modulating autophagy.
Collapse
|
12
|
Cell Death Pathways in Ischemic Stroke and Targeted Pharmacotherapy. Transl Stroke Res 2020; 11:1185-1202. [PMID: 32219729 DOI: 10.1007/s12975-020-00806-z] [Citation(s) in RCA: 201] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 03/04/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023]
Abstract
Ischemic stroke is one of the significant causes of morbidity and mortality, affecting millions of people across the globe. Cell injury in the infarct region is an inevitable consequence of focal cerebral ischemia. Subsequent reperfusion exacerbates the harmful effect and increases the infarct volume. These cellular injuries follow either a regulated pathway involving tightly structured signaling cascades and molecularly defined effector mechanisms or a non-regulated pathway, also known as accidental cell death, where the process is biologically uncontrolled. Classical cell death pathways are long established and well reported in several articles that majorly define apoptotic cell death. A recent focus on cell death study also considers investigation on non-classical pathways that are tightly regulated, may or may not involve caspases, but non-apoptotic. Pathological cell death is a cardinal feature of different neurodegenerative diseases. Although ischemia cannot be classified as a neurodegenerative disease, it is a cerebrovascular event where the infarct region exhibits aberrant cell death. Over the past few decades, several therapeutic options have been implicated for ischemic stroke. However, their use has been hampered owing to the number of limitations that they possess. Ischemic penumbral neurons undergo apoptosis and become dysfunctional; however, they are salvageable. Thus, understanding the role of different cell death pathways is crucial to aid in the modern treatment of protecting apoptotic neurons.
Collapse
|
13
|
Yang M, Sun W, Xiao L, He M, Gu Y, Yang T, Chen J, Liang X. Mesenchymal Stromal Cells Suppress Hippocampal Neuron Autophagy Stress Induced by Hypoxic-Ischemic Brain Damage: The Possible Role of Endogenous IL-6 Secretion. Neural Plast 2020; 2020:8822579. [PMID: 32908484 PMCID: PMC7474748 DOI: 10.1155/2020/8822579] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/17/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Increasing evidence has revealed that mesenchymal stromal cell (MSC) transplantation alleviates hypoxic-ischemic brain damage (HIBD) induced neurological impairments via immunomodulating astrocyte antiapoptosis effects. However, it remains unclear whether MSCs regulate neuron autophagy following HIBD. RESULTS In the present study, MSC transplantation effectively ameliorated learning-memory function and suppressed stress-induced hippocampal neuron autophagy in HIBD rats. Moreover, the suppressive effects of MSCs on autophagy were significantly weakened following endogenous IL-6 silencing in MSCs. Suppressing IL-6 expression also significantly increased p-AMPK protein expression and decreased p-mTOR protein expression in injured hippocampal neurons. CONCLUSION Endogenous IL-6 in MSCs may reduce autophagy in hippocampal neurons partly through the AMPK/mTOR pathway.
Collapse
Affiliation(s)
- Miao Yang
- 1Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- 2Chongqing Key Laboratory of Child Nutrition and Health, Chongqing 400014, China
- 3Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- 4China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Wuqing Sun
- 2Chongqing Key Laboratory of Child Nutrition and Health, Chongqing 400014, China
- 3Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- 4China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
- 5Information Technological Service Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Lu Xiao
- 1Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- 2Chongqing Key Laboratory of Child Nutrition and Health, Chongqing 400014, China
- 3Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- 4China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Mulan He
- 1Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- 2Chongqing Key Laboratory of Child Nutrition and Health, Chongqing 400014, China
- 3Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- 4China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Yan Gu
- 1Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- 2Chongqing Key Laboratory of Child Nutrition and Health, Chongqing 400014, China
- 3Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- 4China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Ting Yang
- 1Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- 2Chongqing Key Laboratory of Child Nutrition and Health, Chongqing 400014, China
- 3Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- 4China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Jie Chen
- 1Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- 2Chongqing Key Laboratory of Child Nutrition and Health, Chongqing 400014, China
- 3Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- 4China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Xiaohua Liang
- 1Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- 2Chongqing Key Laboratory of Child Nutrition and Health, Chongqing 400014, China
- 3Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- 4China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| |
Collapse
|
14
|
Mo Z, Tang C, Li H, Lei J, Zhu L, Kou L, Li H, Luo S, Li C, Chen W, Zhang L. Eicosapentaenoic acid prevents inflammation induced by acute cerebral infarction through inhibition of NLRP3 inflammasome activation. Life Sci 2019; 242:117133. [PMID: 31830477 DOI: 10.1016/j.lfs.2019.117133] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/27/2019] [Accepted: 11/29/2019] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Acute cerebral infarction (ACI) is the most common type of acute cerebrovascular diseases resulting in high rate of death and disability. Numerous evidences show that inflammation is the leading cause of ischemic brain injury, thus anti-inflammatory therapy is an attractive candidate for ischemic brain damage. Eicosapentaenoic acid (EPA) exerts anti-inflammatory activity in lots of human inflammatory diseases, whereas its effect in ACI is left to elucidate. METHOD Nlpr3-/- mice, Gpr40-/-; Gpr120-/- mice and mice with right middle cerebral artery occlusion (MCAO) were used to detect NLR family pyrin domain containing 3 (NLRP3) inflammasome activation by Western Blot and the release of proinflammatory cytokines by ELISA. To estimate the acute ischemic condition in vitro, oxygen-glucose deprivation (OGD) was induced in BV2 microglia cells. Transfection of the shRNA targeting GPR40 and GPR120 mRNA into BV2 cells was also assessed. Apoptosis in ischemic cerebral tissues and BV2 cells was detected by terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) assay and flow cytometry. RESULT Here we show that EPA suppresses ACI-induced inflammatory responses through blocking NLRP3 inflammasome activation. In addition, EPA inhibits NLRP3 inflammasome activation through G protein-coupled receptor 40 (GPR40) and GPR120. Importantly, EPA ameliorates ACI-induced apoptosis. CONCLUSION EPA exerts beneficial effect on ACI-induced inflammation through blocking NLRP3 inflammasome activation by GPR40 and GPR120. Our findings suggest the potential clinical use of EPA in ACI.
Collapse
Affiliation(s)
- Zhihuai Mo
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Chaogang Tang
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China; Department of Neurology, Maoming People's Hospital, 525000, Guangdong, China
| | - Huiqing Li
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Junjie Lei
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Lingjuan Zhu
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Li Kou
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Hao Li
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China; Department of Neurology, Maoming People's Hospital, 525000, Guangdong, China
| | - Shijian Luo
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Chunyi Li
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Wenli Chen
- Department of Pharmacology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China.
| | - Lei Zhang
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China.
| |
Collapse
|
15
|
Wu C, Yan X, Liao Y, Liao L, Huang S, Zuo Q, Zhou L, Gao L, Wang Y, Lin J, Li S, Wang K, Ge X, Song H, Yang R, Lu F. Increased perihematomal neuron autophagy and plasma thrombin-antithrombin levels in patients with intracerebral hemorrhage: An observational study. Medicine (Baltimore) 2019; 98:e17130. [PMID: 31574813 PMCID: PMC6775380 DOI: 10.1097/md.0000000000017130] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Animal studies have demonstrated that autophagy was involved in neuronal damage after intracerebral hemorrhage (ICH). Several studies showed thrombin-antithrombin (TAT) plasma levels were elevated in patients with ICH. In this study, we aimed to evaluate if autophagy occurred in patients with ICH; and the relationship between the severity of brain injury and plasma TAT levels.A novel tissue harvesting device was used during hematoma removal surgery to collect loose fragments of tissue surrounding the affected brain area in 27 ICH patients with hematoma volumes of >30 mL in the basal ganglia. Control tissues were obtained from patients who underwent surgery for arteriovenous malformation (n = 25). Transmission electron microscopy (TEM) and immunohistochemistry for autophagy-related proteins were used to evaluate the ultrastructural and morphologic cellular characteristics; and the extent of autophagy in the recovered tissue specimens. Stroke severity was assessed by using the Glasgow Coma Scale (GCS) and the National Institutes of Health Stroke Scale (NIHSS). An enzyme-linked immunosorbent assay (ELISA) was used to measure plasma TAT levels.Transmission electron microscopy showed autophagosomes and autolysosomes exist in neurons surrounding the hematoma, but not in the control tissues. The number of cells containing autophagic vacuoles correlated with the severity of brain injury. Immunohistochemistry showed strong LC3, beclin 1, and cathepsin D staining in ICH tissue specimens. Plasma TAT levels correlated positively with autophagic cells and ICH severity (P < .01).Autophagy was induced in perihematomal neurons after ICH. Autophagy and plasma TAT levels correlated positively with severity of brain injury. These results suggest that autophagy and increased plasma TAT levels may contribute to the secondary damage in ICH patients.
Collapse
Affiliation(s)
- Chenghan Wu
- Department of Neurology, Second Affiliated Clinical College of Fujian University of Traditional Chinese Medicine
| | - Xiaohua Yan
- Department of Neurosurgery and TCM, Fujian Provincial Hospital
| | - Yuansheng Liao
- Department of Neurology, Second Affiliated Clinical College of Fujian University of Traditional Chinese Medicine
| | - Lianming Liao
- Central Laboratory, Union Hospital of Fujian Medical University
| | - Shengyue Huang
- Department of Neurosurgery and TCM, Fujian Provincial Hospital
| | - Quanting Zuo
- Department of Neurology, Second Affiliated Clinical College of Fujian University of Traditional Chinese Medicine
| | - Linying Zhou
- Laboratory of Electron Microscopy, Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Lili Gao
- Department of Neurology, Second Affiliated Clinical College of Fujian University of Traditional Chinese Medicine
| | - Yinzhou Wang
- Department of Neurosurgery and TCM, Fujian Provincial Hospital
| | - Jushan Lin
- Department of Neurology, Second Affiliated Clinical College of Fujian University of Traditional Chinese Medicine
| | - Shiju Li
- Department of Neurology, Second Affiliated Clinical College of Fujian University of Traditional Chinese Medicine
| | - Kaiyu Wang
- Department of Neurosurgery and TCM, Fujian Provincial Hospital
| | - Xiuming Ge
- Department of Neurology, Second Affiliated Clinical College of Fujian University of Traditional Chinese Medicine
| | - Hailong Song
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO
| | - Ruiling Yang
- Department of Neurology, Second Affiliated Clinical College of Fujian University of Traditional Chinese Medicine
| | - Feng Lu
- Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
16
|
Hase Y, Chen A, Bates LL, Craggs LJL, Yamamoto Y, Gemmell E, Oakley AE, Korolchuk VI, Kalaria RN. Severe white matter astrocytopathy in CADASIL. Brain Pathol 2019; 28:832-843. [PMID: 29757481 DOI: 10.1111/bpa.12621] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/08/2018] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is characterized by strategic white matter (WM) hyperintensities on MRI. Pathological features include WM degeneration, arteriolosclerosis, lacunar infarcts, and the deposition of granular osmiophilic material. Based on the hypothesis that the gliovascular unit is compromised, we assessed the nature of astrocyte damage in the deep WM of CADASIL subjects. METHODS We evaluated post-mortem brains from CADASIL, cerebral small vessel disease, similar age cognitively normal and older control subjects. Standard immunohistochemical, immunofluorescent, and unbiased stereological methods were used to evaluate the distribution of astrocytes, microvessels, and autophagy markers in five different brain regions. RESULTS Compared to the controls, the deep WM of CADASIL subjects overall showed increased numbers of glial fibrillary acidic protein (GFAP)-positive clasmatodendritic astrocytes (P=0.037) and a decrease in the percentage of normal appearing astrocytes (P=0.025). In accord with confluent WM hyperintensities, the anterior temporal pole contained abundant clasmatodendritic astrocytes with displaced aquaporin 4 immunoreactivity. Remarkably, we also found strong evidence for the immunolocalization of autophagy markers including microtubule-associated protein 1, light chain 3 (LC3), and sequestosome 1/p62 and Caspase-3 in GFAP-positive clasmatodendritic cells, particularly within perivascular regions of the deep WM. LC3 was co-localized in more than 90% of the GFAP-positive clasmatodendrocytes. CONCLUSIONS Our novel findings show astrocytes undergo autophagy-like cell death in CADASIL, with the anterior temporal pole being highly vulnerable. We propose astrocytes transform from normal appearing type A to hypertrophic type B and eventually to clasmatodendritic type C cells. These observations also suggest the gliovascular unit of the deep WM is severely impaired in CADASIL.
Collapse
Affiliation(s)
- Yoshiki Hase
- Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne, UK
| | - Aiqing Chen
- Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne, UK
| | - Letitia L Bates
- Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne, UK
| | - Lucinda J L Craggs
- Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne, UK
| | - Yumi Yamamoto
- Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne, UK
| | - Elizabeth Gemmell
- Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne, UK
| | - Arthur E Oakley
- Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne, UK
| | - Viktor I Korolchuk
- Institute for Cell and Molecular Biosciences, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne, UK.,Institute for Ageing, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne, UK
| | - Raj N Kalaria
- Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne, UK.,Institute for Ageing, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne, UK
| |
Collapse
|
17
|
Hou K, Xu D, Li F, Chen S, Li Y. The progress of neuronal autophagy in cerebral ischemia stroke: Mechanisms, roles and research methods. J Neurol Sci 2019; 400:72-82. [PMID: 30904689 DOI: 10.1016/j.jns.2019.03.015] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 02/25/2019] [Accepted: 03/15/2019] [Indexed: 12/26/2022]
Abstract
There is increasing evidence indicating that autophagy may be a new target in the treatment of ischemic stroke. Moderate autophagy can clear damaged organelles, thereby protecting cells against various injuries. However, long-term excessive autophagy brings redundant degradation of cell contents, leading to cell death and eventually serious damage to tissues and organs. A number of different animal models of ischemic brain injury shows that autophagy is activated and involved in the regulation of neuronal death during ischemic brain injury. This article summarizes the role of autophagy, its underlying regulators and mechanisms in ischemic neuronal injury. We briefly introduce the relationship between apoptosis and autophagy and give a summary of research methods and modulators of autophagy.
Collapse
Affiliation(s)
- Kai Hou
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Dan Xu
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Fengyang Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Shijie Chen
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Yunman Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
18
|
Fang Y, Chen S, Reis C, Zhang J. The Role of Autophagy in Subarachnoid Hemorrhage: An Update. Curr Neuropharmacol 2018; 16:1255-1266. [PMID: 28382869 PMCID: PMC6251055 DOI: 10.2174/1570159x15666170406142631] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/16/2017] [Accepted: 04/05/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Autophagy is an extensive self-degradation process for the disposition of cytosolic aggregated or misfolded proteins and defective organelles which executes the functions of pro-survival and pro-death to maintain cellular homeostasis. The pathway plays essential roles in several neurological disorders. Subarachnoid Hemorrhage (SAH) is a devastating subtype of hemorrhagic stroke with high risk of neurological deficit and high mortality. Early brain injury (EBI) plays a role in the poor clinical course and outcome after SAH. Recent studies have paid attention on the role of the autophagy pathway in the development of EBI after SAH. We aim to update the multifaceted roles of autophagy pathway in the pathogenesis of SAH, especially in the phase of EBI. METHODS We reviewed early researches related to autophagy and SAH. The following three aspects of contents will be mainly discussed: the process of the autophagy pathway, the role of the autophagy in SAH and the interaction between organelle dysfunction and autophagy pathway after SAH. RESULTS Accumulating evidence shows an increased autophagy reaction in response to early stages of SAH. However, others suggest inadequate or excessive autophagy activation can result in cell injury and death. In addition to autophagy, apoptosis and necrosis can occur in neurons simultaneously after SAH, leading to mixed features of cell death morphologies. And it is also known that there is extensive crosstalk between autophagy and apoptosis pathway. Subcellular organelles of neural cells generally participate in the formation and functional parts of autophagy process. CONCLUSION Autophagy plays an important role in the SAH-induced brain injury. A better understanding of the interrelationship among autophagy, apoptosis, and necrosis might provide us better therapeutic targets for the treatment of SAH.
Collapse
Affiliation(s)
- Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Sheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Cesar Reis
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, United States.,Department of Preventive Medicine, Loma Linda University Medical Center, Loma Linda, CA, United States
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China.,Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
19
|
Lonati E, Sala G, Tresoldi V, Coco S, Salerno D, Milani C, Losurdo M, Farina F, Botto L, Ferrarese C, Palestini P, Bulbarelli A. Ischemic Conditions Affect Rerouting of Tau Protein Levels: Evidences for Alteration in Tau Processing and Secretion in Hippocampal Neurons. J Mol Neurosci 2018; 66:604-616. [DOI: 10.1007/s12031-018-1199-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 10/17/2018] [Indexed: 11/30/2022]
|
20
|
Chimeh U, Zimmerman MA, Gilyazova N, Li PA. B355252, A Novel Small Molecule, Confers Neuroprotection Against Cobalt Chloride Toxicity In Mouse Hippocampal Cells Through Altering Mitochondrial Dynamics And Limiting Autophagy Induction. Int J Med Sci 2018; 15:1384-1396. [PMID: 30275767 PMCID: PMC6158673 DOI: 10.7150/ijms.24702] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/12/2018] [Indexed: 12/31/2022] Open
Abstract
Cerebral hypoxia as often occurs in cases of stroke, hemorrhage, or other traumatic brain injuries, is one of the leading causes of death worldwide and a main driver of disabilities in the elderly. Using a chemical mimetic of hypoxia, cobalt chloride (CoCl2), we tested the ability of a novel small molecule, 4-chloro-N-(naphthalen-1-ylmethyl)-5-(3-(piperazin-1-yl)phenoxy)thiophene-2-sulfonamide (B355252), to alleviate CoCl2-induced damage in mouse hippocampal HT22 cells. A dose-dependent decrease in cell viability was observed during CoCl2 treatment along with increases in mitochondrial membrane potential and generation of reactive oxygen species (ROS). B355252 conferred protection against these changes. We further found that mitochondrial dynamics, the balance between mitochondrial fusion and fission, were perturbed by CoCl2 treatment. Mitochondrial fusion, which was assessed by measuring the expression of proteins optic atrophy protein 1 (OPA1) and mitofusin 2 (Mfn2), declined due to CoCl2 exposure, but B355252 addition was able to elevate Mfn2 expression while OPA1 expression was unchanged. Mitochondrial fission, measured by phosphorylated dynamin-related protein 1 (p-DRP1) and fission protein 1 (FIS1) expression, also decreased following CoCl2 exposure, and was stabilized by B355252 addition. Finally, autophagy was assessed by measuring the conversion of cytosolic microtubule-associated protein 1A/1B-light chain three-I (LC3-I) to autophagosome-bound microtubule-associated protein 1A/1B-light chain three-II (LC3-II) and was found to be increased by CoCl2. B355252 addition significantly reduced autophagy induction. Taken together, our results indicate B355252 has therapeutic potential to reduce the damaging effects caused by CoCl2 and should be further evaluated for applications in cerebral ischemia therapy.
Collapse
Affiliation(s)
| | | | | | - P. Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, Durham, NC USA
| |
Collapse
|
21
|
Mohammad Alizadeh E, Mahdavi M, Jenani Fard F, Chamani S, Farajdokht F, Karimi P. Metformin protects PC12 cells against oxygen-glucose deprivation/reperfusion injury. Toxicol Mech Methods 2018; 28:622-629. [DOI: 10.1080/15376516.2018.1486495] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
| | - Majid Mahdavi
- Department of Biology, University of Tabriz, Tabriz, Iran
| | | | | | - Fereshteh Farajdokht
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pouran Karimi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
22
|
Inhibition of excessive autophagy and mitophagy mediates neuroprotective effects of URB597 against chronic cerebral hypoperfusion. Cell Death Dis 2018; 9:733. [PMID: 29955058 PMCID: PMC6023888 DOI: 10.1038/s41419-018-0755-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/11/2018] [Accepted: 05/24/2018] [Indexed: 12/13/2022]
Abstract
URB597 (URB) has therapeutic potential for treating chronic cerebral hypoperfusion (CCH)-induced neuronal death. The present study investigated the protective effects of URB on autopahgy and mitophagy in a CCH model as well as the underlying mechanisms. The ultrastructural changes were examined by electron microscopy. The mitochondrial membrane potential was assessed by immunofluorescence. The expressions of autophagy-related proteins (beclin-1, p62, and LC3), lysosome-related proteins (CTSD and LAMP1), and mitophagy-related proteins (BNIP3, cyt C and parkin) were evaluated by western blotting, and the interaction of beclin-1 and Bcl-2 were determined by immunoprecipitation. CCH significantly decreased the protein expression of p62, CTSD, and LAMP1 and increased the protein expression of beclin-1, parkin, and BNIP3, the LC3-II to LC3-I ratio, and the release of cyt C from mitochondria to cytoplasm. Furthermore, CCH induced the accumulation of ubiquitinated proteins in PSDs. However, URB significantly reversed these results. Besides, URB significantly inhibited the beclin-1 from beclin-1/Bcl-2 complex to whole-cell lysates. The above results indicate that URB could inhibit impaired autophagy degradation and the disruption of beclin-1/Bcl-2 complex and subsequently cut off BNIP3-cyt C- and parkin-required mitophagy, finally preventing the abnormal excessive autophagy and mitophagy. These findings provide new insights that URB is a promising agent for therapeutic management of CCH.
Collapse
|
23
|
Yang G, Wang N, Seto SW, Chang D, Liang H. Hydroxysafflor yellow a protects brain microvascular endothelial cells against oxygen glucose deprivation/reoxygenation injury: Involvement of inhibiting autophagy via class I PI3K/Akt/mTOR signaling pathway. Brain Res Bull 2018; 140:243-257. [PMID: 29775658 DOI: 10.1016/j.brainresbull.2018.05.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/27/2018] [Accepted: 05/11/2018] [Indexed: 12/31/2022]
Abstract
The present study aimed to test whether Hydroxysafflor yellow A (HSYA) protects the brain microvascular endothelial cells (BMECs) injury induced by oxygen glucose deprivation/reoxygenation (OGD/R) via the PI3K/Akt/mTOR autophagy signaling pathway. Primary rat BMECs were cultured and identified by the expression of factor VIII-related antigen before being exposed to OGD/R to imitate ischemia/reperfusion (I/R) damage in vitro. The protective effect of HSYA was evaluated by assessing (1) cellular morphologic and ultrastructural changes; (2) cell viability and cytotoxicity; (3) transendothelial electrical resistance (TEER) of monolayer BMECs; (4) cell apoptosis; (5) fluorescence intensity of LC3B; (6) LC3 mRNA expression; (7) protein expressions of LC3, Beclin-1, Zonula occludens-1 (ZO-1), phospho-Akt (p-Akt), Akt, phospho-mTOR (p-mTOR) and mTOR. It was found that HSYA (20, 40, and 80 μM) and 3-MA effectively reversed the cellular morphological and ultrastructural changes, increased cell survival, normalized the permeability of BMECs, and suppressed apoptosis induced by OGD/R (2 h OGD followed by 24 h reoxygenation). Concurrently, HSYA and 3-MA also inhibited OGD/R-induced autophagy evidenced by the decreased number of autophagosomes and down-regulated levels of LC3 and Beclin-1 proteins and mRNAs. HSYA (80 μM), in combination with 3-MA showed a synergistic effect. Mechanistic studies revealed that HSYA (80 μM) markedly increased the levels of p-Akt and p-mTOR proteins. Blockade of PI3K activity by ZSTK474 abolished its anti-autophagic and pro-survival effect and lowered both Akt and mTOR phosphorylation levels. Taken together, these results suggest that HSYA protects BMECs against OGD/R-induced injury by inhibiting autophagy via the Class I PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Guang Yang
- Anhui University of Chinese Medicine, Hefei 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China.
| | - Ning Wang
- Anhui University of Chinese Medicine, Hefei 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; National Institute of Complementary Medicine, Western Sydney University, Penrith, NSW 2751, Australia.
| | - Sai Wang Seto
- National Institute of Complementary Medicine, Western Sydney University, Penrith, NSW 2751, Australia
| | - Dennis Chang
- National Institute of Complementary Medicine, Western Sydney University, Penrith, NSW 2751, Australia
| | - Huangzheng Liang
- School of Medical, Western Sydney University, Penrith, NSW 2751, Australia
| |
Collapse
|
24
|
PD149163 induces hypothermia to protect against brain injury in acute cerebral ischemic rats. J Pharmacol Sci 2017; 135:105-113. [PMID: 29113791 DOI: 10.1016/j.jphs.2017.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 08/24/2017] [Accepted: 09/15/2017] [Indexed: 12/19/2022] Open
Abstract
Therapeutic hypothermia is a promising strategy for acute cerebral ischemia via physical or pharmacological methods. In this study, we pharmacologically induced hypothermia on Sprague Dawley rats by intraperitoneally injecting PD149163. We found that mild hypothermia was induced by PD149163 treatment without local cerebral blood flow (LCBF) alteration. To evaluate the neuroprotective effects of PD149163, TTC staining, HE staining and Nissl's staining were performed in our study. We found that PD149163 could prevent neuronal damage, and inhibit proliferation and activation of glial cells induced by ischemia. Simultaneously, we observed PD149163 ameliorated apoptosis characterized by down-regulated caspase-3 and Bax, but elevated Bcl-2. Moreover, PD149163 dramatically reduced JNK and AMPK/mTOR signaling pathway activation, and thereby inhibited autophagy by increased P62 expression, decreased the ratio of LC3-Ⅱ to LC3-Ⅰ and the expression of Beclin. Taken together, the present findings reveal the therapeutic effects of PD149163-induced hypothermia in brain ischemia, and provide a new strategy for stroke treatment.
Collapse
|
25
|
Zhu Y, Shui M, Liu X, Hu W, Wang Y. Increased autophagic degradation contributes to the neuroprotection of hydrogen sulfide against cerebral ischemia/reperfusion injury. Metab Brain Dis 2017; 32:1449-1458. [PMID: 28421304 DOI: 10.1007/s11011-017-0014-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 04/06/2017] [Indexed: 12/17/2022]
Abstract
Hydrogen sulfide (H2S), an endogenous gaseous signal molecule, exhibits protective effect against ischemic injury. However, its underlying mechanism is not fully understood. We have recently reported that exogenous H2S decreases the accumulation of autophagic vacuoles in mouse brain with ischemia/reperfusion (I/R) injury. To further investigate whether this H2S-induced reduction of autophagic vacuoles is caused by the decreased autophagosome synthesis and/or the increased autophagic degradation inautophagic flux, we performed in vitro and in vivo studies using SH-SY5Y cells for the oxygen and glucose deprivation/reoxygenation (OGD/R) and mice for the cerebral I/R, respectively. NaHS (a donor of H2S) treatment significantly increased cell viability and reduced cerebral infarct volume. NaHS treatment reduced the OGD/R-induced elevation in LC3-II (an autophagic marker), which was completely reversed by co-treatment with an autophagic flux inhibitor bafilomycin A1 (BafA1). However, H2S did not affect the OGD/R-induced increase of the ULK1 self-association and decrease of the ATG13 phosphorylation, which are the critical steps for the initiation of autophagosome formation. Cerebral I/R injury caused an increase in LC3-II, a decrease in p62 and the accumulation of autophagosomes in the cortex and the hippocampus, which were inhibited by NaHS treatment. This H2S-induced decline of LC3-II in ischemic brain was reversed by BafA1. Moreover, BafA1 treatment abolished the protection of H2S on the cerebral infarction. Collectively, the neuroprotection of exogenous H2S against ischemia/hypoxia and reperfusion/reoxygenation injury is mediated by the enhancement of autophagic degradation.
Collapse
Affiliation(s)
- Yuanjun Zhu
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Mengyang Shui
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiaoyan Liu
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Wenhui Hu
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA.
| | - Yinye Wang
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
26
|
Bao L, Li RH, Li M, Jin MF, Li G, Han X, Yang YY, Sun B, Xu LX, Feng X. Autophagy-regulated AMPAR subunit upregulation in in vitro oxygen glucose deprivation/reoxygenation-induced hippocampal injury. Brain Res 2017; 1668:65-71. [PMID: 28549968 DOI: 10.1016/j.brainres.2017.05.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/17/2017] [Accepted: 05/18/2017] [Indexed: 01/11/2023]
Abstract
Autophagy has been implicated to mediate experimental cerebral ischemia/reperfusion-induced neuronal death; the underlying molecular mechanisms, though, are poorly understood. In this study, we investigated the role of autophagy in regulating the expression of AMPAR subunits (GluR1, GluR2, and GluR3) in oxygen glucose deprivation/reperfusion (OGD/R)-mediated injury of hippocampal neurons. Our results showed that, OGD/R-induced hippocampal neuron injury was accompanied by accumulation of autophagosomes and autolysosomes in cytoplasm alongside a dramatic increase in expression of autophagy-related genes, LC3 and Beclin 1 and increased intracellular Ca2+ levels. Pre-treatment with autophagy inhibitor 3-methyladenine (3-MA) significantly reduced this effect. Moreover, the OGD/R-induced upregulation of mRNA and protein expressions of GluR1, GluR2, and GluR3 were also effectively reversed in cells pretreated with 3-MA. Our findings indicate that OGD/R induced the expression of GluRs by activating autophagy in in vitro cultured hippocampal neurons, which could be effectively reversed by the administration of 3-MA.
Collapse
Affiliation(s)
- Li Bao
- Department of Neonatology, People's Hospital of Yixing City, Jiangsu, China
| | - Rong-Hu Li
- Department of Neonatology, Children's Hospital of Jinan City, Jinan, China
| | - Mei Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Mei-Fang Jin
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Gang Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Xing Han
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| | - Yuan-Yuan Yang
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| | - Bin Sun
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| | - Li-Xiao Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China.
| | - Xing Feng
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
27
|
Li H, Zhang Q, Yang X, Wang L. PPAR-γ agonist rosiglitazone reduces autophagy and promotes functional recovery in experimental traumaticspinal cord injury. Neurosci Lett 2017; 650:89-96. [PMID: 28433567 DOI: 10.1016/j.neulet.2017.02.075] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 02/27/2017] [Accepted: 02/28/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Secondary damage is often more important in determining the functional outcome and provides a practical target for therapeutic intervention. Rosiglitazone (ROSG) is a potent PPAR-γ agonist and has been shown to induce neuroprotection in animal models of spinal cord injury (SCI). However, it is still unclear whether this PPAR-γ agonist can mediate neuronal autophagy after SCI. METHODS SCI was induced by application of vascular clips (force of 24g) to the dura via a four-level T5-T8 laminectomy. The role of the PPAR-γ agonist ROSG on neuronal autophagy induced by SCI was investigated. RESULTS The expression of autophagy-related proteins, including microtubule-associated protein 1 light chain 3 type II (LC3-II), beclin-1, and cathepsin D, increased significantly after SCI. ROSG downregulated autophagy-related protein expression and improved the locomotor function after SCI. GW9662 (a PPAR-γ inhibitor) significantly antagonized the effect of ROSG and abolished the protective effect on SCI. CONCLUSIONS Our results clearly demonstrated that the administration of ROSG after SCI reduced autophagy and promoted functional recovery after SCI in rats.
Collapse
Affiliation(s)
- Hongpeng Li
- Yuncheng Central Hospital, Shanxi 044000, PR China; Department of Emergency and Critical Care Medicine, Zhoupu Hospital affiliated with Shanghai University of Medicine and Health Sciences, Shanghai 201318, PR China.
| | - Qin Zhang
- Yuncheng Central Hospital, Shanxi 044000, PR China.
| | - Xiaohui Yang
- Yuncheng Central Hospital, Shanxi 044000, PR China
| | - Liping Wang
- Second Hospital of Lanzhou University, Gansu 730000, PR China
| |
Collapse
|
28
|
Li H, Wu J, Shen H, Yao X, Liu C, Pianta S, Han J, Borlongan CV, Chen G. Autophagy in hemorrhagic stroke: Mechanisms and clinical implications. Prog Neurobiol 2017; 163-164:79-97. [PMID: 28414101 DOI: 10.1016/j.pneurobio.2017.04.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/13/2017] [Accepted: 04/08/2017] [Indexed: 02/07/2023]
Abstract
Accumulating evidence advances the critical role of autophagy in brain pathology after stroke. Investigations employing autophagy induction or inhibition using pharmacological tools or autophagy-related gene knockout mice have recently revealed the biological significance of intact and functional autophagy in stroke. Most of the reported cases attest to a pro-survival role for autophagy in stroke, by facilitating removal of damaged proteins and organelles, which can be recycled for energy generation and cellular defenses. However, these observations are difficult to reconcile with equally compelling evidence demonstrating stroke-induced upregulation of brain cell death index that parallels enhanced autophagy. This begs the question of whether drug-induced autophagy during stroke culminates in improved or worsened pathological outcomes. A corollary fascinating hypothesis, but presents as a tricky conundrum, involves the effects of autophagy on cell death and inflammation, which are two main culprits in the disease progression of stroke-induced brain injury. Evidence has extended the roles of autophagy in inflammation via cytokine regulation in an unconventional secretion manner or by targeting inflammasomes for degradation. Moreover, in the recently concluded Vancouver Autophagy Symposium (VAS) held in 2014, the potential of selective autophagy for clinical treatment has been recognized. The role of autophagy in ischemic stroke has been reviewed previously in detail. Here, we evaluate the strength of laboratory and clinical evidence by providing a comprehensive summary of the literature on autophagy, and thereafter we offer our perspectives on exploiting autophagy as a drug target for cerebral ischemia, especially in hemorrhagic stroke.
Collapse
Affiliation(s)
- Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University,188 Shizi Street, Suzhou 215006, China
| | - Jiang Wu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University,188 Shizi Street, Suzhou 215006, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University,188 Shizi Street, Suzhou 215006, China
| | - Xiyang Yao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University,188 Shizi Street, Suzhou 215006, China
| | - Chenglin Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University,188 Shizi Street, Suzhou 215006, China
| | - S Pianta
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery & Brain Repair, University of South Florida Morsani College of Medicine,12901 Bruce B Downs Blvd Tampa, FL 33612 USA
| | - J Han
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery & Brain Repair, University of South Florida Morsani College of Medicine,12901 Bruce B Downs Blvd Tampa, FL 33612 USA
| | - C V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery & Brain Repair, University of South Florida Morsani College of Medicine,12901 Bruce B Downs Blvd Tampa, FL 33612 USA
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University,188 Shizi Street, Suzhou 215006, China.
| |
Collapse
|
29
|
Chao MW, Yang CH, Lin PT, Yang YH, Chuang YC, Chung MC, Tseng CY. Exposure to PM 2.5 causes genetic changes in fetal rat cerebral cortex and hippocampus. ENVIRONMENTAL TOXICOLOGY 2017; 32:1412-1425. [PMID: 27539004 DOI: 10.1002/tox.22335] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 07/29/2016] [Accepted: 07/30/2016] [Indexed: 05/06/2023]
Abstract
PM2.5 travels along the respiratory tract and enters systemic blood circulation. Studies have shown that PM2.5 increases the incidence of various diseases not only in adults but also in newborn infants. It causes chronic inflammation in pregnant women and retards fetal development. In this study, pregnant rats were exposed to PM2.5 for extended periods of time and it was found that PM2.5 exposure increased immune cells in mother rats. In addition, cytokines and free radicals rapidly accumulated in the amniotic fluid and indirectly affected the fetuses. The authors collected cerebral cortex and hippocampus samples at E18 and analyzed changes of miRNA levels. Expression levels of cortical miR-6315, miR-3588, and miR-466b-5p were upregulated, and positively correlated with the genes Pkn2 (astrocyte migration), Gorab (neuritogenesis), and Mobp (allergic encephalomyelitis). In contrast, PM2.5 decreased expression of miR-338-5p and let-7e-5p, both related to mental development. Further, PM2.5 exposure increased miR-3560 and let-7b-5p in the hippocampus, two proteins that regulate genes Oxct1 and Lin28b that control ketogenesis and glycosylation, and neural cell differentiation, respectively. miR-99b-5p, miR-92b-5p, and miR-99a-5p were decreased, leading to reduced expression of Kbtbd8 and Adam11 which reduced cell mitosis, migration, and differentiation, and inhibited learning abilities and motor coordination of the fetus. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 1412-1425, 2017.
Collapse
Affiliation(s)
- Ming-Wei Chao
- Department of Bioscience Technology College of Science, Chung Yuan Christian University, Zhongli district, Taoyaun, 320, Taiwan
- Center for Nanotechnology, College of Science, Chung Yuan Christian University, Zhongli district, Taoyaun, 320, Taiwan
| | - Chin-Hua Yang
- Department of Diagnostic Radiology, Taoyuan General Hospital, Taoyaun, 310, Taiwan
- Departmewnt of Biomedical Engineering and Environmental Science, National Tsing Hua University, East District, Hsinchu 300, Taiwan
| | - Po-Ting Lin
- Department of Mechanical Engineering, College of Engineering, Chung Yuan Christian University, Zhongli district, Taoyaun, 320, Taiwan
| | - Yu-Hsiu Yang
- Department of Biomedical Engineering College of Engineering, Chung Yuan Christian University, Zhongli district, Taoyaun, 320, Taiwan
| | - Yu-Chen Chuang
- Department of Biomedical Engineering College of Engineering, Chung Yuan Christian University, Zhongli district, Taoyaun, 320, Taiwan
| | - Meng-Chi Chung
- Department of Bioscience Technology College of Science, Chung Yuan Christian University, Zhongli district, Taoyaun, 320, Taiwan
| | - Chia-Yi Tseng
- Center for Nanotechnology, College of Science, Chung Yuan Christian University, Zhongli district, Taoyaun, 320, Taiwan
- Department of Biomedical Engineering College of Engineering, Chung Yuan Christian University, Zhongli district, Taoyaun, 320, Taiwan
| |
Collapse
|
30
|
Huang XP, Ding H, Yang XQ, Li JX, Tang B, Liu XD, Tang YH, Deng CQ. Synergism and mechanism of Astragaloside IV combined with Ginsenoside Rg1 against autophagic injury of PC12 cells induced by oxygen glucose deprivation/reoxygenation. Biomed Pharmacother 2017; 89:124-134. [PMID: 28219050 DOI: 10.1016/j.biopha.2017.02.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 02/03/2017] [Accepted: 02/07/2017] [Indexed: 01/30/2023] Open
Abstract
The aim of this study was to explore the effect by which the combination of Astragaloside IV (AST IV) and Ginsenoside Rg1 (Rg1) resisted autophagic injury in PC12 cells induced by oxygen glucose deprivation/reoxygenation (OGD/R). We studied the nature of the interaction between AST IV and Rg1 that inhibited autophagy through the Isobologram method, and investigated the synergistic mechanism via the PI3K I/Akt/mTOR and PI3K III/Becline-1/Bcl-2 signaling pathways. Our results showed that, based on the 50% inhibiting concentration (IC50), AST IV combined with Rg1 at a 1:1 ratio resulted in a synergistic effect, whereas the combination of the two had an antagonistic effect on autophagy at ratios of 1:2 and 2:1. Meanwhile, AST IV and Rg1 alone increased cell survival and decreased lactate dehydrogenase (LDH) leakage induced by OGD/R, reduced autophagosomes and the LC3 II positive patch, down-regulated the LC3 II/LC3 I ratio and up-regulated the p62 protein; the 1:1 combination enhanced these effects. Mechanistic study showed that Rg1 and the 1:1 combination increased the phosphorylation of PI3K I, Akt and mTOR; the effects of the combination were greater than those of the drugs alone. AST IV and the 1:1 combination suppressed the expression of PI3K III and Becline-1, and the combination elevated Bcl-2 protein expression; the effects of the combination were better than those of the drugs alone. These results suggest that after 2 h-OGD followed by reoxygenation for 24h, PC12 cells suffer excessive autophagy and damage, which are blocked by AST IV or Rg1; moreover, the combination of AST IV and Rg1 at a 1:1 ratio of their IC50 concentrations has a synergistic inhibition on autophagic injury. The synergistic mechanism may be associated with the PI3K I/Akt/mTOR and PI3K III/Becline-1/Bcl-2 signaling pathways.
Collapse
Affiliation(s)
- Xiao-Ping Huang
- Molecular Pathology Laboratory, Hunan Provincial Key Laboratory for Prevention and Treatment of Integrated Traditional Chinese and Western Medicine on Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Huang Ding
- Molecular Pathology Laboratory, Hunan Provincial Key Laboratory for Prevention and Treatment of Integrated Traditional Chinese and Western Medicine on Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Xiao-Qian Yang
- Molecular Pathology Laboratory, Hunan Provincial Key Laboratory for Prevention and Treatment of Integrated Traditional Chinese and Western Medicine on Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Jing-Xian Li
- Molecular Pathology Laboratory, Hunan Provincial Key Laboratory for Prevention and Treatment of Integrated Traditional Chinese and Western Medicine on Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Biao Tang
- Hunan Education Department's Key Laboratory of Cell Biology and Molecular Technology, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Xiao-Dan Liu
- Hunan Education Department's Key Laboratory of Cell Biology and Molecular Technology, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Ying-Hong Tang
- Hunan Education Department's Key Laboratory of Cell Biology and Molecular Technology, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Chang-Qing Deng
- Molecular Pathology Laboratory, Hunan Provincial Key Laboratory for Prevention and Treatment of Integrated Traditional Chinese and Western Medicine on Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China.
| |
Collapse
|
31
|
Jing YH, Qi CC, Yuan L, Liu XW, Gao LP, Yin J. Adult neural stem cell dysfunction in the subventricular zone of the lateral ventricle leads to diabetic olfactory defects. Neural Regen Res 2017; 12:1111-1118. [PMID: 28852393 PMCID: PMC5558490 DOI: 10.4103/1673-5374.211190] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Sensitive smell discrimination is based on structural plasticity of the olfactory bulb, which depends on migration and integration of newborn neurons from the subventricular zone. In this study, we examined the relationship between neural stem cell status in the subventricular zone and olfactory function in rats with diabetes mellitus. Streptozotocin was injected through the femoral vein to induce type 1 diabetes mellitus in Sprague-Dawley rats. Two months after injection, olfactory sensitivity was decreased in diabetic rats. Meanwhile, the number of BrdU-positive and BrdU+/DCX+ double-labeled cells was lower in the subventricular zone of diabetic rats compared with age-matched normal rats. Western blot results revealed downregulated expression of insulin receptor β, phosphorylated glycogen synthase kinase 3β, and β-catenin in the subventricular zone of diabetic rats. Altogether, these results indicate that diabetes mellitus causes insulin deficiency, which negatively regulates glycogen synthase kinase 3β and enhances β-catenin degradation, with these changes inhibiting neural stem cell proliferation. Further, these signaling pathways affect proliferation and differentiation of neural stem cells in the subventricular zone. Dysfunction of subventricular zone neural stem cells causes a decline in olfactory bulb structural plasticity and impairs olfactory sensitivity in diabetic rats.
Collapse
Affiliation(s)
- Yu-Hong Jing
- Institute of Anatomy and Histology & Embryology and Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, China.,Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, Gansu Province, China
| | - Chu-Chu Qi
- Institute of Anatomy and Histology & Embryology and Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Li Yuan
- Institute of Anatomy and Histology & Embryology and Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Xiang-Wen Liu
- Institute of Anatomy and Histology & Embryology and Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Li-Ping Gao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Jie Yin
- Institute of Anatomy and Histology & Embryology and Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| |
Collapse
|
32
|
Shu S, Li CM, You YL, Qian XL, Zhou S, Ling CQ. Electroacupuncture Ameliorates Cerebral Ischemia-Reperfusion Injury by Regulation of Autophagy and Apoptosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2016; 2016:7297425. [PMID: 27800003 PMCID: PMC5075311 DOI: 10.1155/2016/7297425] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 07/20/2016] [Indexed: 01/24/2023]
Abstract
Background. The therapeutic mechanisms of cerebral ischemia treatment by acupuncture are yet not well addressed. Objective. We investigated the effects of electroacupuncture (EA) at GV26 observing the expression of autophagy-related proteins Beclin-1 and LC3B and proportion of apoptotic cells and Bcl-2 positive cells in MCAO/R model rats. Methods. Sprague-Dawley (SD) male rats were randomly assigned to 7 groups: model groups (M6h, M24h, and M72h), EA treatment groups (T6h, T24h, and T72h), and sham operation group (S). Neurological deficit and cerebral infarction volume were measured to assess the improvement effect, while the expression of Beclin-1 and LC3B and proportion of Tunel-positive and Bcl-2 positive cells were examined to explore EA effect on autophagy and apoptosis. Results. EA significantly decreased neurological deficit scores and the volume of cerebral infarction. Beclin-1 was significantly decreased in T24h, while LC3B-II/LC3B-I ratio markedly reduced in 6th hour. EA groups markedly reduced the number of Tunel positive cells, especially in T24h. Meanwhile, the number of Bcl-2 positive cells obviously increased after EA treatment, especially in T6h and T24h. Conclusions. The alleviation of inadequate autophagy and apoptosis may be a key mechanism involved in the reflex regulation of EA at GV26 to treat cerebral ischemia.
Collapse
Affiliation(s)
- Shi Shu
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, No. 168, Changhai Road, Yangpu District, Shanghai 200433, China
| | - Chun-Ming Li
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, No. 168, Changhai Road, Yangpu District, Shanghai 200433, China
| | - Yan-Li You
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, No. 168, Changhai Road, Yangpu District, Shanghai 200433, China
| | - Xiao-Lu Qian
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, No. 168, Changhai Road, Yangpu District, Shanghai 200433, China
| | - Shuang Zhou
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, No. 168, Changhai Road, Yangpu District, Shanghai 200433, China
| | - Chang-quan Ling
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, No. 168, Changhai Road, Yangpu District, Shanghai 200433, China
| |
Collapse
|
33
|
Liu P, Yang X, Hei C, Meli Y, Niu J, Sun T, Li PA. Rapamycin Reduced Ischemic Brain Damage in Diabetic Animals Is Associated with Suppressions of mTOR and ERK1/2 Signaling. Int J Biol Sci 2016; 12:1032-40. [PMID: 27489506 PMCID: PMC4971741 DOI: 10.7150/ijbs.15624] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/28/2016] [Indexed: 12/17/2022] Open
Abstract
The objectives of the present study are to investigate the activation of mTOR and ERK1/2 signaling after cerebral ischemia in diabetic rats and to examine the neuroprotective effects of rapamycin. Ten minutes transient global cerebral ischemia was induced in straptozotocin-induced diabetic hyperglycemic rats and non-diabetic, euglycemic rats. Brain samples were harvested after 16 h of reperfusion. Rapamycin or vehicle was injected 1 month prior to the induction of ischemia. The results showed that diabetes increased ischemic neuronal cell death and associated with elevations of p-P70S6K and Ras/ERK1/2 and suppression of p-AMPKα. Rapamycin ameliorated diabetes-enhanced ischemic brain damage and suppressed phosphorylation of P70S6K and ERK1/2. It is concluded that diabetes activates mTOR and ERK1/2 signaling pathways in rats subjected to transient cerebral ischemia and inhibition of mTOR by rapamycin reduces ischemic brain damage and suppresses the mTOR and ERK1/2 signaling in diabetic settings.
Collapse
Affiliation(s)
- Ping Liu
- 1. Department of Endocrinology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
- 2. Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
| | - Xiao Yang
- 2. Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
- 3. Neuroscience Center, General Hospital of Ningcia Medical University, and Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Yinchuan 750004, China
| | - Changchun Hei
- 2. Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
- 4. Department of Human Anatomy, Histology and Embryology, Ningxia Medical University, Yinchuan 75004, China
| | - Yvonne Meli
- 2. Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
| | - Jianguo Niu
- 4. Department of Human Anatomy, Histology and Embryology, Ningxia Medical University, Yinchuan 75004, China
| | - Tao Sun
- 4. Department of Human Anatomy, Histology and Embryology, Ningxia Medical University, Yinchuan 75004, China
| | - P. Andy Li
- 2. Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
| |
Collapse
|
34
|
Vavilis T, Delivanoglou N, Aggelidou E, Stamoula E, Mellidis K, Kaidoglou A, Cheva A, Pourzitaki C, Chatzimeletiou K, Lazou A, Albani M, Kritis A. Oxygen-Glucose Deprivation (OGD) Modulates the Unfolded Protein Response (UPR) and Inflicts Autophagy in a PC12 Hypoxia Cell Line Model. Cell Mol Neurobiol 2016; 36:701-12. [PMID: 26239244 PMCID: PMC11482405 DOI: 10.1007/s10571-015-0250-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 07/29/2015] [Indexed: 11/29/2022]
Abstract
Hypoxia is the lack of sufficient oxygenation of tissue, imposing severe stress upon cells. It is a major feature of many pathological conditions such as stroke, traumatic brain injury, cerebral hemorrhage, perinatal asphyxia and can lead to cell death due to energy depletion and increased free radical generation. The present study investigates the effect of hypoxia on the unfolded protein response of the cell (UPR), utilizing a 16-h oxygen-glucose deprivation protocol (OGD) in a PC12 cell line model. Expression of glucose-regulated protein 78 (GRP78) and glucose-regulated protein 94 (GRP94), key players of the UPR, was studied along with the expression of glucose-regulated protein 75 (GRP75), heat shock cognate 70 (HSC70), and glyceraldehyde 3-phosphate dehydrogenase, all with respect to the cell death mechanism(s). Cells subjected to OGD displayed upregulation of GRP78 and GRP94 and concurrent downregulation of GRP75. These findings were accompanied with minimal apoptotic cell death and induction of autophagy. The above observation warrants further investigation to elucidate whether autophagy acts as a pro-survival mechanism that upon severe and prolonged hypoxia acts as a concerted cell response leading to cell death. In our OGD model, hypoxia modulates UPR and induces autophagy.
Collapse
Affiliation(s)
- Theofanis Vavilis
- Laboratory of Physiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Nikoleta Delivanoglou
- Laboratory of Physiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Eleni Aggelidou
- Laboratory of Physiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Eleni Stamoula
- Laboratory of Physiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Kyriakos Mellidis
- Laboratory of Physiology, School of Biology, Faculty of Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Aikaterini Kaidoglou
- Laboratory of Histology, Embryology and Anthropology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Angeliki Cheva
- Department of Pathology, General Hospital of Thessaloniki "G. Papanikolaou", Thessaloniki, Greece
| | - Chryssa Pourzitaki
- Laboratory of Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Katerina Chatzimeletiou
- Unit of Human Reproduction, 1st Department of Obstetrics and Gynaecology, Aristotle University Medical School, Papageorgiou General Hospital, Thessaloniki, Greece
| | - Antigone Lazou
- Laboratory of Physiology, School of Biology, Faculty of Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria Albani
- Laboratory of Physiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Aristeidis Kritis
- Laboratory of Physiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece.
| |
Collapse
|
35
|
Salidroside protects cortical neurons against glutamate-induced cytotoxicity by inhibiting autophagy. Mol Cell Biochem 2016; 419:53-64. [PMID: 27357827 DOI: 10.1007/s11010-016-2749-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 06/18/2016] [Indexed: 12/20/2022]
Abstract
Recent evidence suggests that glutamate-induced cytotoxicity contributes to autophagic neuron death and is partially mediated by increased oxidative stress. Salidroside has been demonstrated to have neuroprotective effects in glutamate-induced neuronal damage. The precise mechanism of its regulatory role in neuronal autophagy is, however, poorly understood. This study aimed to probe the effects and mechanisms of salidroside in glutamate-induced autophagy activation in cultured rat cortical neurons. Cell viability assay, Western blotting, coimmunoprecipitation, and small interfering RNA were performed to analyze autophagy activities during glutamate-evoked oxidative injury. We found that salidroside protected neonatal neurons from glutamate-induced apoptotic cell death. Salidroside significantly attenuated the LC3-II/LC3-I ratio and expression of Beclin-1, but increased (SQSTM1)/p62 expression under glutamate exposure. Pretreatment with 3-methyladenine (3-MA), an autophagy inhibitor, decreased LC3-II/LC3-I ratio, attenuated glutamate-induced cell injury, and mimicked some of the protective effects of salidroside against glutamate-induced cell injury. Molecular analysis demonstrated that salidroside inhibited cortical neuron autophagy in response to glutamate exposure through p53 signaling by increasing the accumulation of cytoplasmic p53. Salidroside inhibited the glutamate-induced dissociation of the Bcl-2-Beclin-1 complex with minor affects on the PI3K/Akt/mTOR signaling pathways. These data demonstrate that the inhibition of autophagy could be responsible for the neuroprotective effects of salidroside on glutamate-induced neuronal injury.
Collapse
|
36
|
He G, Xu W, Tong L, Li S, Su S, Tan X, Li C. Gadd45b prevents autophagy and apoptosis against rat cerebral neuron oxygen-glucose deprivation/reperfusion injury. Apoptosis 2016; 21:390-403. [DOI: 10.1007/s10495-016-1213-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
37
|
Pérez-Rodríguez D, Anuncibay-Soto B, Llorente IL, Pérez-García CC, Fernández-López A. Hippocampus and cerebral cortex present a different autophagic response after oxygen and glucose deprivation in an ex vivo rat brain slice model. Neuropathol Appl Neurobiol 2016; 41:e68-79. [PMID: 24861158 DOI: 10.1111/nan.12152] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 05/21/2014] [Indexed: 01/06/2023]
Abstract
AIMS To evaluate the neuroprotective role of autophagy in the cerebral cortex and hippocampus using an ex vivo animal model of stroke in brain slices. METHODS Brain slices were maintained for 30 min in oxygen and glucose deprivation (OGD) followed by 3 h in normoxic conditions to simulate the reperfusion that follows ischaemia in vivo (RL, reperfusion-like). Phagophore formation (Beclin 1 and LC3B) as well as autophagy flux (p62/SQSTM1, Atg5, Atg7 and polyubiquitin) markers were quantified by Western blot and/or qPCR. The release of lactate dehydrogenase (LDH) and glutamate in the medium was used as a measure of the mortality in the absence and in the presence of the autophagy inhibitor 3-methyladenine. RESULTS Striking differences in the autophagy markers were observed between the hippocampus and cerebral cortex in normoxic conditions. OGD/RL induced increases both in the phagophore formation and in the autophagy flux in the first three hours in the cerebral cortex that were not observed in the hippocampus. The blocking of autophagy increased the OGD/RL-induced mortality, increased the glutamate release in both the cerebral cortex and hippocampus and abolished the OGD-induced decrease in the polyubiquitinated proteins in the cerebral cortex. CONCLUSIONS We conclude that OGD induces a rapid autophagic response in the cerebral cortex that plays a neuroprotective role. Polyubiquitination levels and control of the glutamate release appear to be involved in the neuroprotective role of autophagy.
Collapse
Affiliation(s)
- Diego Pérez-Rodríguez
- Área de Biología Celular, Instituto de Biomedicina, Universidad de León, Leon, Spain
| | - Berta Anuncibay-Soto
- Área de Biología Celular, Instituto de Biomedicina, Universidad de León, Leon, Spain
| | - Irene L Llorente
- Área de Biología Celular, Instituto de Biomedicina, Universidad de León, Leon, Spain
| | - Carlos C Pérez-García
- Área de Medicina y Cirugía Veterinaria, Instituto de Biomedicina, Universidad de León, Leon, Spain
| | | |
Collapse
|
38
|
Liu W, Shang G, Yang S, Huang J, Xue X, Lin Y, Zheng Y, Wang X, Wang L, Lin R, Tao J, Chen L. Electroacupuncture protects against ischemic stroke by reducing autophagosome formation and inhibiting autophagy through the mTORC1-ULK1 complex-Beclin1 pathway. Int J Mol Med 2015; 37:309-18. [PMID: 26647915 PMCID: PMC4716798 DOI: 10.3892/ijmm.2015.2425] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 11/12/2015] [Indexed: 11/06/2022] Open
Abstract
In a previous study by our group, we demonstrated that electroacupuncture (EA) activates the class I phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. There is considerable evidence that the downstream mammalian target of rapamycin complex 1 (mTORC1) plays an important role in autophagy following ischemic stroke. The aim of the present study was to determine whether EA exerts a neuroprotective effect through mTORC1-mediated autophagy following ischemia/reperfusion injury. Our results revealed that EA at the LI11 and ST36 acupoints attenuated motor dysfunction, improved neurological deficit outcomes and decreased the infarct volumes. The number of autophagosomes, autolysosomes and lysosomes was decreased following treatment with EA. Simultaneously, the levels of the autophagosome membrane maker, microtubule-associated protein 1 light chain 3 beta (LC3B)II/I, Unc-51-like kinase 1 (ULK1), autophagy related gene 13 Atg13) and Beclin1 (ser14) were decreased, whereas mTORC1 expression was increased in the peri-infarct cortex. These results suggest that EA protects against ischemic stroke through the inhibition of autophagosome formation and autophagy, which is mediated through the mTORC1-ULK complex-Beclin1 pathway.
Collapse
Affiliation(s)
- Weilin Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Guanhao Shang
- The Fujian Province Key Laboratory of Motor Functional Rehabilitation, Fuzhou, Fujian 350001, P.R. China
| | - Shanli Yang
- Rehabilitation Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350001, P.R. China
| | - Jia Huang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Xiehua Xue
- Rehabilitation Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350001, P.R. China
| | - Yunjiao Lin
- The Fujian Province Key Laboratory of Motor Functional Rehabilitation, Fuzhou, Fujian 350001, P.R. China
| | - Yi Zheng
- The Fujian Province Key Laboratory of Motor Functional Rehabilitation, Fuzhou, Fujian 350001, P.R. China
| | - Xian Wang
- The Fujian Province Key Laboratory of Motor Functional Rehabilitation, Fuzhou, Fujian 350001, P.R. China
| | - Lulu Wang
- The Fujian Province Key Laboratory of Motor Functional Rehabilitation, Fuzhou, Fujian 350001, P.R. China
| | - Ruhui Lin
- The Fujian Province Key Laboratory of Motor Functional Rehabilitation, Fuzhou, Fujian 350001, P.R. China
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Lidian Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| |
Collapse
|
39
|
Huang XP, Ding H, Lu JD, Tang YH, Deng BX, Deng CQ. Autophagy in cerebral ischemia and the effects of traditional Chinese medicine. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2015; 13:289-96. [PMID: 26343099 DOI: 10.1016/s2095-4964(15)60187-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Autophagy is a lysosome-mediated degradation process for non-essential or damaged cellular constituents, playing an important homeostatic role in cell survival, differentiation and development to maintain homeostasis. Autophagy is involved in tumors as well as neurodegenerative, cardiovascular and cerebrovascular diseases. Recently, active compounds from traditional Chinese medicine (TCM) have been found to modulate the levels of autophagy in tumor cells, nerve cells, myocardial cells and endothelial cells. Ischemic stroke is a major cause of neurological disability and places a heavy burden on family and society. Regaining function can significantly reduce dependence and improve the quality of life of stroke survivors. In healthy cells, autophagy plays a key role in adapting to nutritional deprivation and eliminating aggregated proteins, however inappropriate activation of autophagy may lead to cell death in cerebral ischemia. This paper reviews the process and the molecular basis of autophagy, as well as its roles in cerebral ischemia and the roles of TCM in modulating its activity.
Collapse
Affiliation(s)
- Xiao-ping Huang
- Molecular Pathology Laboratory, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Huang Ding
- Key Laboratory of Hunan Province for Prevention and Treatment of Integrated Traditional Chinese and Western Medicine on Cardio-cerebral Diseases, Changsha 410208, Hunan Province, China
| | - Jin-dong Lu
- Key Laboratory of Hunan Universities for Cell Biology and Molecular Techniques, Changsha 410208, Hunan Province, China
| | - Ying-hong Tang
- Key Laboratory of Hunan Province for Prevention and Treatment of Integrated Traditional Chinese and Western Medicine on Cardio-cerebral Diseases, Changsha 410208, Hunan Province, China
| | - Bing-xiang Deng
- Key Laboratory of Hunan Universities for Cell Biology and Molecular Techniques, Changsha 410208, Hunan Province, China
| | - Chang-qing Deng
- Molecular Pathology Laboratory, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China; E-mail:
| |
Collapse
|
40
|
Abstract
The selective degradation of damaged or excessive mitochondria by autophagy is termed mitophagy. Mitophagy is crucial for mitochondrial quality control and has been implicated in several neurodegenerative disorders as well as in ischemic brain injury. Emerging evidence suggested that the role of mitophagy in cerebral ischemia may depend on different pathological processes. In particular, a neuroprotective role of mitophagy has been proposed, and the regulation of mitophagy seems to be important in cell survival. For these reasons, extensive investigations aimed to profile the mitophagy process and its underlying molecular mechanisms have been executed in recent years. In this review, we summarize the current knowledge regarding the mitophagy process and its role in cerebral ischemia, and focus on the pathological events and molecules that regulate mitophagy in ischemic brain injury.
Collapse
Affiliation(s)
- Yang Yuan
- Department of Pharmacology, Key Laboratory of Medical Neurobiology (Ministry of Health of China), College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | | | | | | |
Collapse
|
41
|
Button RW, Luo S, Rubinsztein DC. Autophagic activity in neuronal cell death. Neurosci Bull 2015; 31:382-94. [PMID: 26077705 DOI: 10.1007/s12264-015-1528-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 05/11/2015] [Indexed: 12/19/2022] Open
Abstract
As post-mitotic cells with great energy demands, neurons depend upon the homeostatic and waste-recycling functions provided by autophagy. In addition, autophagy also promotes survival during periods of harsh stress and targets aggregate-prone proteins associated with neurodegeneration for degradation. Despite this, autophagy has also been controversially described as a mechanism of programmed cell death. Instances of autophagic cell death are typically associated with elevated numbers of cytoplasmic autophagosomes, which have been assumed to lead to excessive degradation of cellular components. Due to the high activity and reliance on autophagy in neurons, these cells may be particularly susceptible to autophagic death. In this review, we summarize and assess current evidence in support of autophagic cell death in neurons, as well as how the dysregulation of autophagy commonly seen in neurodegeneration can contribute to neuron loss. From here, we discuss potential treatment strategies relevant to such cell-death pathways.
Collapse
Affiliation(s)
- Robert W Button
- Peninsula Schools of Medicine and Dentistry, Institute of Translational and Stratified Medicine, University of Plymouth, Research Way, Plymouth, PL6 8BU, UK
| | | | | |
Collapse
|
42
|
Autophagy in spinal motor neurons of conditional ADAR2-knockout mice: An implication for a role of calcium in increased autophagy flux in ALS. Neurosci Lett 2015; 598:79-84. [DOI: 10.1016/j.neulet.2015.05.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 05/03/2015] [Accepted: 05/12/2015] [Indexed: 12/18/2022]
|
43
|
Qin H, Tan W, Zhang Z, Bao L, Shen H, Wang F, Xu F, Wang Z. 15d-prostaglandin J2 protects cortical neurons against oxygen-glucose deprivation/reoxygenation injury: involvement of inhibiting autophagy through upregulation of Bcl-2. Cell Mol Neurobiol 2015; 35:303-312. [PMID: 25349027 PMCID: PMC11486323 DOI: 10.1007/s10571-014-0125-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/17/2014] [Indexed: 01/21/2023]
Abstract
We have previously shown that PPAR-γ agonist 15d-PGJ2 inhibited neuronal autophagy after cerebral ischemia/reperfusion injury. However, the underlying mechanism of its regulatory role in neuronal autophagy remains unclear. This study was designed to test the hypothesis that 15d-PGJ2 upregulated Bcl-2 which binds to Beclin 1, and thereby inhibits autophagy. We performed cell viability assay, cytotoxicity assay, western blot, and co-immunoprecipitation to analyze autophagy activities in vitro model of oxygen-glucose deprivation/reoxygenation (OGD/R). OGD/R induced autophagy in cultured cortical neurons. 15d-PGJ2 treatment significantly decreased LC3-II/LC3-I ratio and Beclin 1 expression, but increased p62 expression. Autophagic inhibitor 3-methyladenine decreased LC3-II levels, increased neuronal cell viability, and mimicked some protective effect of 15d-PGJ2 against OGD/R injury. OGD/R-induced autophagy coincided with decreases in Bcl-2 expression and increases in Beclin 1 expression. 15d-PGJ2 treatment upregulated Bcl-2 expression and decreased Beclin 1 expression, and inhibit the dissociation of Beclin1 from Bcl-2 significantly. Bcl-2 siRNA abrogated the effect of 15d-PGJ2 on Beclin 1, LC3-II and p62, and influence cell viability and LDH level, while scRNA did not. PPAR-γ agonist 15d-PGJ2 exerts neuroprotection partially via inhibiting neuronal autophagy after OGD/R injury. The inhibition of autophagy by 15d-PGJ2 is mediated through upregulation of Bcl-2.
Collapse
Affiliation(s)
- Haidong Qin
- Department of Emergency Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Weiguo Tan
- Department of Neurosurgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, 215300, Jiangsu, China
| | - Zheng Zhang
- Department of Emergency Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Lei Bao
- Department of Emergency Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Hua Shen
- Department of Emergency Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Feng Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, 68 of Changle Road, Nanjing, 210006, Jiangsu, China
| | - Feng Xu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Zizheng Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, 68 of Changle Road, Nanjing, 210006, Jiangsu, China.
| |
Collapse
|
44
|
Wang W, Guo Z, Xu Z, Meng Q, Chen C, Zhang Y, Cao X. Effect of pollen typhae on inhibiting autophagy in spinal cord injury of rats and its mechanisms. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:2375-2383. [PMID: 26045744 PMCID: PMC4440053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/22/2015] [Indexed: 06/04/2023]
Abstract
PURPOSE To investigate the effects and mechanism of pollen typhae on spinal cord injury (SCI) in rats. METHODS The SCI model was built and animals were randomly divided into three groups according to different concentrations of pollen typhae. Protein, mRNA, and fluorescence expression levels of light-chain-3 (LC-3) and Beclin-1 were determined by western blotting (WB), real-time PCR and immunofluorescence, along as Akt and mammalian target of rapamycin (mROT) by WB. The demyelination area and integrated optical density (IOD) were analyzed by luxol fast blue (LFB) and Nissl staining, respectively; Behavioral assessments were assessed by Basso Beattie Bresnahan (BBB) scale. RESULTS Protein, mRNA, and fluorescence expression levels of LC-3 and Beclin-1 were significantly increased after SCI, while were obviously decreased by administration of pollen typhae, along with protein level of Akt and mROT. The demyelination area was significantly reduced, while IOD and BBB were significantly increased compared with the model group. CONCLUSION Autophagic activity increased in damaged neural tissue after SCI, and pollen typhae have certain therapeutic effect on SCI, the higher concentration of pollen typhae, the more effective. Besides, pollen typhae also provided neuroprotective effect and improved locomotor function. The effects may be produced by blockade of Akt/mTOR pathway.
Collapse
Affiliation(s)
- Weiguo Wang
- Department of Orthopaedic Surgery, General Hospital of Jinan Military CommandJinan 250031, China
| | - Zhihong Guo
- Department of Anesthesiology, General Hospital of Jinan Military CommandJinan 250031, China
| | - Zhanwang Xu
- Department of Orthopaedics, The First Affiliated Hospital of Shandong University of Traditional Chinese MedicineJinan 250014, China
| | - Qingxi Meng
- Department of Spinal Cord Repair, General Hospital of Jinan Military CommandJinan 250031, China
| | - Chen Chen
- Department of Orthopaedic Surgery, General Hospital of Jinan Military CommandJinan 250031, China
| | - Yaoguang Zhang
- Department of Emergency Surgery, General Hospital of Jinan Military CommandJinan 250031, China
| | - Xuecheng Cao
- Department of Orthopaedic Surgery, General Hospital of Jinan Military CommandJinan 250031, China
| |
Collapse
|
45
|
Autophagy in neuronal cells: general principles and physiological and pathological functions. Acta Neuropathol 2015; 129:337-62. [PMID: 25367385 DOI: 10.1007/s00401-014-1361-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 10/21/2014] [Accepted: 10/25/2014] [Indexed: 12/12/2022]
Abstract
Autophagy delivers cytoplasmic components and organelles to lysosomes for degradation. This pathway serves to degrade nonfunctional or unnecessary organelles and aggregate-prone and oxidized proteins to produce substrates for energy production and biosynthesis. Macroautophagy delivers large aggregates and whole organelles to lysosomes by first enveloping them into autophagosomes that then fuse with lysosomes. Chaperone-mediated autophagy (CMA) degrades proteins containing the KFERQ-like motif in their amino acid sequence, by transporting them from the cytosol across the lysosomal membrane into the lysosomal lumen. Autophagy is especially important for the survival and homeostasis of postmitotic cells like neurons, because these cells are not able to dilute accumulating detrimental substances and damaged organelles by cell division. Our current knowledge on the autophagic pathways and molecular mechanisms and regulation of autophagy will be summarized in this review. We will describe the physiological functions of macroautophagy and CMA in neuronal cells. Finally, we will summarize the current evidence showing that dysfunction of macroautophagy and/or CMA contributes to neuronal diseases. We will give an overview of our current knowledge on the role of autophagy in aging neurons, and focus on the role of autophagy in four types of neurodegenerative diseases, i.e., amyotrophic lateral sclerosis and frontotemporal dementia, prion diseases, lysosomal storage diseases, and Parkinson's disease.
Collapse
|
46
|
Liao J, Xia X, Wang GZ, Shi YM, Ge JW. Naotaifang extract treatment results in increased ferroportin expression in the hippocampus of rats subjected to cerebral ischemia. Mol Med Rep 2015; 11:4047-52. [PMID: 25672910 PMCID: PMC4394947 DOI: 10.3892/mmr.2015.3309] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 09/09/2014] [Indexed: 01/06/2023] Open
Abstract
The expression of Ferroportin (Fpn) was examined at different time points in rats following focal cerebral ischemia treated with or without the traditional Chinese medicine Naotaifang. Initially, rats were randomly divided into 2, 6, 12, 24 and 72 h groups following middle cerebral artery occlusion (MCAO) and the mRNA and protein level of Fpn was detected by immunohistochemistry and reverse transcription polymerase chain reaction (RT‑PCR) at the above time points. Secondly, the rats were randomly divided into five groups as follows: Sham surgery group, model group, low‑dose group (3 g/kg NTE), medium dose group (9 g/kg NTE) and the high‑dose group (27 g/kg NTE). After 3 days of corresponding therapy by intragastric administration once a day, the regional cerebral ischemia model was reproduced by the MCAO suture method. On the third day, the neurological behavior of the rats was analyzed by neurobehavioral assessment. Fpn in the hippocampal CA2 region was measured by immunohistochemistry and the mRNA level of Fpn was detected by RT‑PCR. Expression of Fpn in the hippocampal CA2 region reached a peak 12 h after surgery (P<0.05, compared with the model group). The high‑dose group (27 g/kg NTE) exhibited a lower neurological behavior score (P<0.05) and a higher level of expression of Fpn at the mRNA and protein level compared with the sham surgery group and model group (P<0.05). Dysregulation of intracellular iron balance is possibly a new mechanism underlying cerebral ischemia. NTE can protect the neuronal population in the hippocampal CA2 region by adjusting the expression of Fpn to balance iron levels following cerebral ischemia.
Collapse
Affiliation(s)
- Jun Liao
- Department of Anatomy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Xing Xia
- Campus Network Center, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Guo-Zuo Wang
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Yong-Mei Shi
- Department of Anatomy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Jin-Wen Ge
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| |
Collapse
|
47
|
Liang HJ, Chai RC, Li X, Kong JG, Jiang JH, Ma J, Vatcher G, Yu ACH. Astrocytic exportin-7 responds to ischemia through mediating LKB1 translocation from the nucleus to the cytoplasm. J Neurosci Res 2014; 93:253-67. [PMID: 25250856 DOI: 10.1002/jnr.23486] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 07/16/2014] [Accepted: 08/18/2014] [Indexed: 11/06/2022]
Abstract
The superfamily of importin-β-related proteins is the largest class of nuclear transport receptors and can be generally divided into importins and exportins according to their transport directions. Eleven importins and seven exportins have been identified, and the expression patterns of both classes are important for their functions in nucleocytoplasmic transport activities. This study demonstrates that all of the importins (importin-β; transportin-1, -2, and -3; and importin-4, -5, -7, -8, -9, -11, and -13) and all the exportins (exportin-1, -2, -4, -5, -6, -7, and -t) are differentially expressed in the cerebral cortex, cerebellum, hippocampus, and brainstem and in primary cultures of cerebral cortical astrocytes and neurons. For astrocytes, we observed that different importins and exportins displayed different expression changes during 0-6 hr of ischemia treatment, especially an increase of both the mRNA and the protein of exportin-7. Immunostaining showed that exportin-7 accumulated inside the nucleus and around the nuclear envelope. In addition, we noticed an increased cytoplasmic distribution of one of the cargo proteins of exportin-7, LKB1, an important element in maintaining energy homeostasis. This increased cytoplasmic distribution was accompanied by an increased expression of exportin-7 under ischemia in astrocytes. We demonstrate that exportin-7 responds to ischemia in astrocytes and that this response involves translocation of LKB1, a protein that plays important roles during metabolic stress, from the nucleus to the cytoplasm.
Collapse
Affiliation(s)
- Hai Jie Liang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China; Key Laboratory for Neuroscience of the Ministry of Education and Key Laboratory for Neuroscience of the National Health and Family Planning Commission, Peking University, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Chen W, Sun Y, Liu K, Sun X. Autophagy: a double-edged sword for neuronal survival after cerebral ischemia. Neural Regen Res 2014; 9:1210-6. [PMID: 25206784 PMCID: PMC4146291 DOI: 10.4103/1673-5374.135329] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2014] [Indexed: 12/19/2022] Open
Abstract
Evidence suggests that autophagy may be a new therapeutic target for stroke, but whether activation of autophagy increases or decreases the rate of neuronal death is still under debate. This review summarizes the potential role and possible signaling pathway of autophagy in neuronal survival after cerebral ischemia and proposes that autophagy has dual effects.
Collapse
Affiliation(s)
- Wenqi Chen
- Department of Neurology, the Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Yinyi Sun
- Department of Neurology, the Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Kangyong Liu
- Zhoupu Hospital, Pudong New District, Shanghai, China
| | - Xiaojiang Sun
- Department of Neurology, the Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
49
|
Sheldon RA, Lee CL, Jiang X, Knox RN, Ferriero DM. Hypoxic preconditioning protection is eliminated in HIF-1α knockout mice subjected to neonatal hypoxia-ischemia. Pediatr Res 2014; 76:46-53. [PMID: 24713818 PMCID: PMC4167022 DOI: 10.1038/pr.2014.53] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 01/16/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND Hypoxic preconditioning (HPc) protects the neonatal brain in the setting of hypoxia-ischemia (HI). The mechanisms of protection may depend on activation of hypoxia-inducible factor (HIF-1α). This study sought to clarify the role of HIF-1α after HPc and HI. METHODS To induce HPc, HIF-1α knockout and wild-type (WT) mice were exposed to hypoxia at postnatal day 6. At day 7, the mice underwent HI. Brain injury was determined by histology. HIF-1α, downstream targets, and markers of cell death were measured by western blot. RESULTS HPc protected the WT brain compared with WT without HPc, but did not protect the HIF-1α knockout brain. In WT, HIF-1α increased after hypoxia and after HI, but not with HPc. The HIF-1α knockout showed no change in HIF-1α after hypoxia, HI, or HPc/HI. After HI, spectrin 145/150 was higher in HIF-1α knockout, but after HPc/HI, it was higher in WT. Lysosome-associated membrane protein was higher in WT early after HI, but not later. After HPc/HI, lysosome-associated membrane protein was higher in HIF-1α knockout. CONCLUSION These results indicate that HIF-1α is necessary for HPc protection in the neonatal brain and may affect cell death after HI. Different death and repair mechanisms depend on the timing of HPc.
Collapse
Affiliation(s)
- R. Ann Sheldon
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, US
- Newborn Brain Research Institute, University of California San Francisco, San Francisco, CA, US
| | - Christina L. Lee
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, US
| | - Xiangning Jiang
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, US
- Newborn Brain Research Institute, University of California San Francisco, San Francisco, CA, US
| | - Renatta N. Knox
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, US
| | - Donna M. Ferriero
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, US
- Newborn Brain Research Institute, University of California San Francisco, San Francisco, CA, US
- Department of Neurology, University of California San Francisco, San Francisco, CA, US
| |
Collapse
|
50
|
Guo Z, Cao G, Yang H, Zhou H, Li L, Cao Z, Yu B, Kou J. A combination of four active compounds alleviates cerebral ischemia-reperfusion injury in correlation with inhibition of autophagy and modulation of AMPK/mTOR and JNK pathways. J Neurosci Res 2014; 92:1295-306. [PMID: 24801159 DOI: 10.1002/jnr.23400] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 03/21/2014] [Accepted: 03/30/2014] [Indexed: 01/10/2023]
Abstract
SMXZF is a combination of Rb1, Rg1, schizandrin, and DT-13 (6:9:5:4) derived from Sheng-mai San, a widely used Chinese traditional medicine for the treatment of cardiovascular and cerebral diseases. The present study explores the inhibitory effects and signaling pathways of SMXZF on autophagy induced by cerebral ischemia-reperfusion injury. Male C57BL/6 mice were subjected to ischemia-reperfusion insult by right middle cerebral artery occlusion (MCAO) for 1 hr with subsequent 24 hr reperfusion. Three doses of SMXZF (4.5, 9, and 18 mg/kg) were administered intraperitoneally (i.p.) after ischemia for 1 hr. An autophagic inhibitor, 3-methyladenine (3-MA; 300 μg/kg), was administered i.p. 20 min before ischemia as a positive drug. We found that SMXZF significantly increased cerebral blood flow and reduced the infarct volume, brain water content, and the neurological deficits in a dose-dependent manner. Similar to the positive control, SMXZF at 18 mg/kg also significantly inhibited autophagosome formation. Immunofluorescence staining and Western blotting demonstrated that SMXZF could significantly decrease the expression levels of beclin1 and microtubule-associated protein 1 light chain 3. SMXZF also remarkably inhibited the phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) as well as the expression of c-Jun N-terminal kinase (JNK) and its phosphorylation induced by 24 hr reperfusion. Finally, we demonstrated that the optimal administration time of SMXZF was at the early period of reperfusion. This study reveals that SMXZF displays neuroprotective effect against focal ischemia-reperfusion injury, possibly associated with autophagy inactivation through AMPK/mTOR and JNK pathways.
Collapse
Affiliation(s)
- Zhongshun Guo
- State Key Laboratory of Natural Medicines, Department of Complex Prescription of TCM, China Pharmaceutical University, Nanjing, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|