1
|
Wang Z, Zhang L, Wu T, Pan X, Li L, Yang X, Zhang M, Liu Y. Mechanism of dexmedetomidine in brain injury of infant rats via the IRE1α/NF-κB/CHOP pathway. World J Biol Psychiatry 2025; 26:103-115. [PMID: 39815639 DOI: 10.1080/15622975.2024.2446817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/18/2025]
Abstract
OBJECTIVE We investigated the mechanism of Dexmedetomidine (Dex) in infant rats with brain injury. METHODS The infant rats underwent brain injury modelling. The motor function, spatial learning and memory abilities in rats, and the hippocampal CA1 region Nissl body level and apoptosis were evaluated by behavioural tests and histological stainings. Levels of the hippocampal CA1 region p-IRE1α, nuclear/cytoplasmic p65, CHOP, Bax and Bcl-2 proteins were determined by Western blot. RESULTS Propofol anaesthesia caused brain injury in infant rats. Dex increased the hippocampal CA1 region Nissl body level, abated cell apoptosis, reduced p-IRE1α, ATF6, p-PERK/PERK and CHOP levels, decreased the Bax protein level, elevated the Bcl-2 protein level, and alleviated brain injury in infant rats. After ERS induction and the NF-κB pathway inhibition, the hippocampal CA1 region nuclear/cytoplasmic p65 ratio, CHOP level, and apoptosis were reduced in infant rats with brain injury treated with Dex, while the learning and memory abilities of rats were enhanced. CONCLUSION Dex reduced the hippocampal CA1 region cell apoptosis and enhanced learning and memory abilities by inhibiting the ERS-mediated IRE1α/NF-κB/CHOP pathway, thereby alleviating brain injury in infant rats.
Collapse
Affiliation(s)
- Zhi Wang
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
| | - Lina Zhang
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
| | - Ting Wu
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
| | - Xu Pan
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
| | - Le Li
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
| | - Xin Yang
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
| | - Miao Zhang
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
| | - Ying Liu
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
| |
Collapse
|
2
|
Hou Y, Ye W, Tang Z, Li F. Anesthetics in pathological cerebrovascular conditions. J Cereb Blood Flow Metab 2025; 45:32-47. [PMID: 39450477 PMCID: PMC11563546 DOI: 10.1177/0271678x241295857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/21/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024]
Abstract
The increasing prevalence of pathological cerebrovascular conditions, including stroke, hypertensive encephalopathy, and chronic disorders, underscores the importance of anesthetic considerations for affected patients. Preserving cerebral oxygenation and blood flow during anesthesia is paramount to prevent neurological deterioration. Furthermore, protecting vulnerable neurons from damage is crucial for optimal outcomes. Recent research suggests that anesthetic agents may provide a potentially therapeutic approach for managing pathological cerebrovascular conditions. Anesthetics target neural mechanisms underlying cerebrovascular dysfunction, thereby modulating neuroinflammation, protecting neurons against ischemic injury, and improving cerebral hemodynamics. However, optimal strategies regarding mechanisms, dosage, and indications remain uncertain. This review aims to clarify the physiological effects, mechanisms of action, and reported neuroprotective benefits of anesthetics in patients with various pathological cerebrovascular conditions. Investigating anesthetic effects in cerebrovascular disease holds promise for developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Yuhui Hou
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Wei Ye
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Ziyuan Tang
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Fengxian Li
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University, Guangzhou, China
| |
Collapse
|
3
|
Zheng H, Xiao X, Han Y, Wang P, Zang L, Wang L, Zhao Y, Shi P, Yang P, Guo C, Xue J, Zhao X. Research progress of propofol in alleviating cerebral ischemia/reperfusion injury. Pharmacol Rep 2024; 76:962-980. [PMID: 38954373 DOI: 10.1007/s43440-024-00620-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/21/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024]
Abstract
Ischemic stroke is a leading cause of adult disability and death worldwide. The primary treatment for cerebral ischemia patients is to restore blood supply to the ischemic region as quickly as possible. However, in most cases, more severe tissue damage occurs, which is known as cerebral ischemia/reperfusion (I/R) injury. The pathological mechanisms of brain I/R injury include mitochondrial dysfunction, oxidative stress, excitotoxicity, calcium overload, neuroinflammation, programmed cell death and others. Propofol (2,6-diisopropylphenol), a short-acting intravenous anesthetic, possesses not only sedative and hypnotic effects but also immunomodulatory and neuroprotective effects. Numerous studies have reported the protective properties of propofol during brain I/R injury. In this review, we summarize the potential protective mechanisms of propofol to provide insights for its better clinical application in alleviating cerebral I/R injury.
Collapse
Affiliation(s)
- Haijing Zheng
- Basic Medical College, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
- Zhengzhou Central Hospital, Zhengzhou, China
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Xian Xiao
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Yiming Han
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Pengwei Wang
- Department of Pharmacy, the First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Weihui, Henan, 453100, China
| | - Lili Zang
- Department of Surgery, the First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Weihui, China
| | - Lilin Wang
- Department of Pediatric Surgery, the First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Weihui, China
| | - Yinuo Zhao
- Basic Medical College, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Peijie Shi
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Pengfei Yang
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China.
| | - Chao Guo
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China.
| | - Jintao Xue
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China.
| | - Xinghua Zhao
- Basic Medical College, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China.
| |
Collapse
|
4
|
Kulsum K, Syahrul S, Hasbalah K, Balqis U. Effect of ethanol extract of nigella sativa L seeds and propofol on BDNF protein level as neuroplasticity and neuroprotection of traumatic brain injury in rats. F1000Res 2024; 13:275. [PMID: 39810851 PMCID: PMC11729193 DOI: 10.12688/f1000research.142054.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 01/16/2025] Open
Abstract
Background Traumatic brain injury (TBI) is a change in brain function or evidence of brain pathology caused by external mechanical forces. Brain Derived Neurotrophic Factor (BDNF) is a neurotropin that functions as a neuron protective. Nigella sativa L is reported to have an antioxidant effect, administration of Nigella Sativa L to rats treated with ischemia-reperfusion brain injury. Propofol is an anesthetic agent frequently used intravenously in the management of TBI. The effect of propofol on brain tissue after TBI may be neuroprotective. We aimed to compare the potential of Nigella sativa L and propofol as neuroplasticity and neuroprotection in rats with TBI. Methods This was a laboratory experimental animal model with the post-test only control group design, namely measuring the effect of treatment by comparing the five groups of rats consisting of 30 rats. BDNF levels in rat brain tissue were collected at day 7 of treatment and measured by ELISA. Results The average BDNF protein levels per group, namely G1 (221,243 pg/mL), G2 (172,139 pg/mL), G3 (255,483 pg/mL), G4 (227,089 pg/mL), and G5 (272,603 pg/mL) respectively. Based on the ANOVA statistic, p-value = 0.032 (there was a significant difference between groups), with the Levene Test (0.077) or having variance between the same groups, sequentially the difference in average BDNF protein levels of the five groups is G5>G3>G4>G1>G2, meaning that the combination of Nigella sativa and propofol has more potential to increase BDNF protein levels than Nigella sativa, and Nigella sativa has more potential than propofol. Conclusion We concluded that both nigella sativa and propofol have the potential to increase BDNF protein levels. Nigella Sativa L had a better effect than propofol in repairing damaged neuron cells (neuroplasticity) and increasing BDNF protein levels (neuroprotection) for 7 days of administration in rat traumatic brain injury.
Collapse
Affiliation(s)
- Kulsum Kulsum
- Doctoral Program, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, 24415, Indonesia
- Anesthesiology and Intensive Therapy, Rumah Sakit Umum Daerah Dr Zainoel Abidin, Banda Aceh, Aceh, 24415, Indonesia
- Anesthesiology and Intensive Therapy, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
| | - Syahrul Syahrul
- Neurology, Rumah Sakit Umum Daerah Dr Zainoel Abidin, Banda Aceh, Aceh, 24415, Indonesia
- Neurology, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
| | - Kartini Hasbalah
- Pharmacology, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
| | - Ummu Balqis
- Pathology, Faculty of Veterinary, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
| |
Collapse
|
5
|
Ghosh A, Khanam N, Nath D. Solid lipid nanoparticle: A potent vehicle of the kaempferol for brain delivery through the blood-brain barrier in the focal cerebral ischemic rat. Chem Biol Interact 2024; 397:111084. [PMID: 38823537 DOI: 10.1016/j.cbi.2024.111084] [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: 11/29/2023] [Revised: 05/22/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
Kaempferol is major flavonoid present in Convolvulus pluricaulis. This phytochemical protects the brain against oxidative stress, neuro-inflammation, neurotoxicity, neurodegeneration and cerebral ischemia induced neuronal destruction. Kaempferol is poorly water soluble. Our study proved that solid lipid nanoparticles (SLNs) were efficient carrier of kaempferol through blood-brain barrier (BBB). Kaempferol was incorporated into SLNs prepared from stearic acid with polysorbate 80 by the process of ultrasonication. Mean particle size and zeta potential of kaempferol loaded solid lipid nanoparticles (K-SLNs) were 451.2 nm and -15.0 mV. Atomic force microscopy showed that K-SLNs were spherical in shape. Fourier transformed infrared microscopy (FTIR) showed that both stearic acid and kaempferol were present in K-SLNs. X-ray diffraction (XRD) and differential scanning calorimetry (DSC) revealed that the matrices of K-SLNs were in untidy crystalline state. Entraptment efficiency of K-SLNs was 84.92%. In-vitro drug release percentage was 93.24%. Kaempferol loaded solid lipid nanoparticles (K-SLNs) showed controlled release profile. In-vitro uptake study showed significant efficiency of K-SLNs to cross blood-brain barrier (BBB). After oral administration into the focal cerebral ischemic rat, accumulation of fluorescent labeled K-SLNs was observed in the brain cortex which confirmed its penetrability into the brain. It significantly decreased the neurological deficit, infarct volume and level of reactive oxygen species (ROS) and decreased the level of pro-inflammatory mediators like NF-κB and p-STAT3. Damaged neurons and brain texture were improved. This study indicated increased bioavailability of kaempferol into the brain tissue through SLNs formulation.
Collapse
Affiliation(s)
- Ashutosh Ghosh
- Department of Zoology, University of Kalyani, Nadia, West Bengal, 741235, India
| | - Nasima Khanam
- Department of Zoology, University of Kalyani, Nadia, West Bengal, 741235, India
| | - Debjani Nath
- Department of Zoology, University of Kalyani, Nadia, West Bengal, 741235, India.
| |
Collapse
|
6
|
Battaglini D, da Silva AL, Felix NS, Rodrigues G, Antunes MA, Rocha NN, Capelozzi VL, Morales MM, Cruz FF, Robba C, Silva PL, Pelosi P, Rocco PRM. Mild hypothermia combined with dexmedetomidine reduced brain, lung, and kidney damage in experimental acute focal ischemic stroke. Intensive Care Med Exp 2022; 10:53. [PMID: 36529842 PMCID: PMC9760586 DOI: 10.1186/s40635-022-00481-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Sedatives and mild hypothermia alone may yield neuroprotective effects in acute ischemic stroke (AIS). However, the impact of this combination is still under investigation. We compared the effects of the combination of mild hypothermia or normothermia with propofol or dexmedetomidine on brain, lung, and kidney in experimental AIS. AIS-induced Wistar rats (n = 30) were randomly assigned, after 24 h, to normothermia or mild hypothermia (32-35 °C) with propofol or dexmedetomidine. Histologic injury score and molecular biomarkers were evaluated not only in brain, but also in lung and kidney. Hemodynamics, ventilatory parameters, and carotid Doppler ultrasonography were analyzed for 60 min. RESULTS In brain: (1) hypothermia compared to normothermia, regardless of sedative, decreased tumor necrosis factor (TNF)-α expression and histologic injury score; (2) normothermia + dexmedetomidine reduced TNF-α and histologic injury score compared to normothermia + propofol; (3) hypothermia + dexmedetomidine increased zonula occludens-1 expression compared to normothermia + dexmedetomidine. In lungs: (1) hypothermia + propofol compared to normothermia + propofol reduced TNF-α and histologic injury score; (2) hypothermia + dexmedetomidine compared to normothermia + dexmedetomidine reduced histologic injury score. In kidneys: (1) hypothermia + dexmedetomidine compared to normothermia + dexmedetomidine decreased syndecan expression and histologic injury score; (2) hypothermia + dexmedetomidine compared to hypothermia + propofol decreased histologic injury score. CONCLUSIONS In experimental AIS, the combination of mild hypothermia with dexmedetomidine reduced brain, lung, and kidney damage.
Collapse
Affiliation(s)
- Denise Battaglini
- grid.410345.70000 0004 1756 7871Anesthesiology and Critical Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, 16132 Genoa, Italy ,grid.5841.80000 0004 1937 0247Department of Medicine, University of Barcelona, 08007 Barcelona, Spain ,grid.8536.80000 0001 2294 473XLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ 21941-902 Brazil
| | - Adriana Lopes da Silva
- grid.8536.80000 0001 2294 473XLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ 21941-902 Brazil
| | - Nathane Santanna Felix
- grid.8536.80000 0001 2294 473XLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ 21941-902 Brazil
| | - Gisele Rodrigues
- grid.8536.80000 0001 2294 473XLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ 21941-902 Brazil
| | - Mariana Alves Antunes
- grid.8536.80000 0001 2294 473XLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ 21941-902 Brazil
| | - Nazareth Novaes Rocha
- grid.8536.80000 0001 2294 473XLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ 21941-902 Brazil ,grid.411173.10000 0001 2184 6919Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niterói, 24220-900 Brazil
| | - Vera Luiza Capelozzi
- grid.11899.380000 0004 1937 0722Department of Pathology, University of São Paolo, São Paolo, 05508-060 Brazil
| | - Marcelo Marcos Morales
- grid.8536.80000 0001 2294 473XLaboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901 Brazil
| | - Fernanda Ferreira Cruz
- grid.8536.80000 0001 2294 473XLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ 21941-902 Brazil
| | - Chiara Robba
- grid.410345.70000 0004 1756 7871Anesthesiology and Critical Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, 16132 Genoa, Italy ,grid.5606.50000 0001 2151 3065Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Pedro Leme Silva
- grid.8536.80000 0001 2294 473XLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ 21941-902 Brazil
| | - Paolo Pelosi
- grid.410345.70000 0004 1756 7871Anesthesiology and Critical Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, 16132 Genoa, Italy ,grid.5606.50000 0001 2151 3065Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Patricia Rieken Macedo Rocco
- grid.8536.80000 0001 2294 473XLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ 21941-902 Brazil ,grid.452991.20000 0000 8484 4876Rio de Janeiro Network On Neuroinflammation, Carlos Chagas Filho Foundation for Supporting Research in the State of Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
| |
Collapse
|
7
|
Chen K, Lu D, Yang X, Zhou R, Lan L, Wu Y, Wang C, Xu X, Jiang MH, Wei M, Feng X. Enhanced hippocampal neurogenesis mediated by PGC-1α-activated OXPHOS after neonatal low-dose Propofol exposure. Front Aging Neurosci 2022; 14:925728. [PMID: 35966788 PMCID: PMC9363786 DOI: 10.3389/fnagi.2022.925728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/28/2022] [Indexed: 11/21/2022] Open
Abstract
Background Developing brain is highly plastic and can be easily affected. Growing pediatric usage of anesthetics during painless procedures has raised concerns about the effect of low-dose anesthetics on neurodevelopment. It is urgent to ascertain the neuronal effect of low-dose Propofol, a widely used anesthetic in pediatrics, on developing brains. Methods The behavioral tests after neonatal exposure to low-dose/high-dose Propofol in mice were conducted to clarify the cognitive effect. The nascent cells undergoing proliferation and differentiation stage in the hippocampus and cultured neural stem cells (NSCs) were further identified. In addition, single-nuclei RNA sequencing (snRNA-seq), NSCs bulk RNA-seq, and metabolism trials were performed for pathway investigation. Furthermore, small interfering RNA and stereotactic adenovirus injection were, respectively, used in NSCs and hippocampal to confirm the underlying mechanism. Results Behavioral tests in mice showed enhanced spatial cognitive ability after being exposed to low-dose Propofol. Activated neurogenesis was observed both in hippocampal and cultured NSCs. Moreover, transcriptome analysis of snRNA-seq, bulk RNA-seq, and metabolism trials revealed a significantly enhanced oxidative phosphorylation (OXPHOS) level in NSCs. Furthermore, PGC-1α, a master regulator in mitochondria metabolism, was found upregulated after Propofol exposure both in vivo and in vitro. Importantly, downregulation of PGC-1α remarkably prevented the effects of low-dose Propofol in activating OXPHOS and neurogenesis. Conclusions Taken together, this study demonstrates a novel alteration of mitochondrial function in hippocampal neurogenesis after low-dose Propofol exposure, suggesting the safety, even potentially beneficial effect, of low-dose Propofol in pediatric use.
Collapse
Affiliation(s)
- Keyu Chen
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Key Laboratory for Stem Cells and Tissue Engineering, Center for Stem Cell Biology and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Dihan Lu
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Key Laboratory for Stem Cells and Tissue Engineering, Center for Stem Cell Biology and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyu Yang
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rui Zhou
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liangtian Lan
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan Wu
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chen Wang
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xuanxian Xu
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mei Hua Jiang
- Key Laboratory for Stem Cells and Tissue Engineering, Center for Stem Cell Biology and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Key Laboratory of Reproductive Medicine, Guangzhou, China
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ming Wei
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Ming Wei
| | - Xia Feng
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Xia Feng
| |
Collapse
|
8
|
Dordoe C, Wang X, Lin P, Wang Z, Hu J, Wang D, Fang Y, Liang F, Ye S, Chen J, Zhao Y, Xiong Y, Yang Y, Lin L, Li X. Non-mitogenic fibroblast growth factor 1 protects against ischemic stroke by regulating microglia/macrophage polarization through Nrf2 and NF-κB pathways. Neuropharmacology 2022; 212:109064. [PMID: 35452626 DOI: 10.1016/j.neuropharm.2022.109064] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 01/23/2023]
Abstract
Microglia are immune cells in the central nervous system (CNS) that participate in response to pathological process after ischemic injury. Non-mitogenic fibroblast growth factor 1 (nmFGF1) is an effective neuroprotective factor that is also known as a metabolic regulator. The present study aimed to investigate the effects and mechanism of the neuroprotective ability of nmFGF1 on microglia in mice after photothrombosis (PT) stroke model, to determine whether it could ameliorate ischemic injury in stroke experiment. We discovered that the intranasal administration of nmFGF1 reduced infarct size and ameliorated neurological deficits in behavioral assessment by regulating the secretion of proinflammatory and anti-inflammatory cytokines. Furthermore, in the in vitro experiments, we found that nmFGF1 regulated the expression levels of proinflammatory and anti-inflammatory cytokines in oxygen-glucose deprivation (OGD) and lipopolysaccharide (LPS) stimulation. Evidence have shown that when nuclear factor erythroid 2-related factor 2 (Nfr2) is activated, it inhibits nuclear factor-kappa B (NF-κB) activation to alleviate inflammation. Interestingly, nmFGF1 treatment in vivo remarkably inhibited NF-κB pathway activation and activated Nrf2 pathway. In addition, nmFGF1 and NF-κB inhibitor (BAY11-7082) inhibited NF-κB pathway in LPS-stimulated BV2 microglia. Moreover, in LPS-stimulated BV2 microglia, the anti-inflammatory effect produced by nmFGF1 was knocked down by Nrf2 siRNA. These results indicate that nmFGF1 promoted functional recovery in experimental stroke by modulating microglia/macrophage-mediated neuroinflammation via Nrf2 and NF-κB signaling pathways, making nmFGF1 a potential agent against ischemic stroke.
Collapse
Affiliation(s)
- Confidence Dordoe
- Pingyang Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325400, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xue Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Ping Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Zhengyi Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jian Hu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Dongxue Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China; College of Pharmacy, Chonnam National University, Gwangju, 501-190, Republic of Korea
| | - Yani Fang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Fei Liang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Shasha Ye
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jun Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yeli Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Ye Xiong
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325015, China
| | - Yunjun Yang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325015, China
| | - Li Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China; Research Units of Clinical Translation of Cell Growth Factors and Diseases Research, Chinese Academy of Medical Science, Wenzhou, Zhejiang, 325035, China.
| | - Xianfeng Li
- Pingyang Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325400, China.
| |
Collapse
|
9
|
Zhang F, Wang Z, Sun B, Huang Y, Chen C, Hu J, Li L, Xia P, Ye Z. Propofol rescued astrocytes from LPS-induced inflammatory response via blocking LncRNA-MEG3/NF-κB axis. Curr Neurovasc Res 2022; 19:5-18. [PMID: 35297349 DOI: 10.2174/1567202619666220316112509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 12/27/2021] [Accepted: 12/31/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Evidences had demonstrated that propofol attenuated neuro-inflammation following brain ischemia. Moreover, LncRNA-MEG3 was identified as an independent prognostic marker for ischemic stroke patients, and was found to be correlated with cerebral ischemia in animal models. Therefore, the current study explored the role of propofol on lipopolysaccharide (LPS)-mediated inflammation in cultured astrocytes, along with the molecular mechanism involved in LncRNA-MEG3/NF-κB axis. METHODS The primary cultured astrocytes isolated from rats were used to establish an inflammatory model, which were treated with LPS. Propofol was administrated to the primary cultured astrocytes during LPS treatment. The effect of propofol on pro-inflammatory cytokines and the LncRNA-MEG3/NF-κB pathway were detected by ELISA, qRT-PCR and Western Blot assay, respectively. Then, dual-luciferase assay, chromatin immunoprecipitation and RNA immunoprecipitation were used to determine the interaction between LncRNA-MEG3 and NF-κB. RESULTS Our study found that propofol significantly reduced LncRNA-MEG3 expression, which was elevated in LPS-stimulated astrocytes. Moreover, both propofol and LncRNA-MEG3 knockdown remarkably alleviated LPS-induced cytotoxicity by suppressing expressions and release of pro-inflammatory cytokines. Loss of LncRNA-MEG3 notably suppressed the NF-κB activity and its phosphorylated activation. Additionally, it was also observed that LncRNA-MEG3 could bind nuclear p65/p50, and promote the binding of NF-κB to IL-6 and TNF-α promoters in the nucleus, subsequently stimulating the production of inflammatory cytokines in LPS-treated astrocytes. Furthermore, a specific inhibitor of NF-κB, PDTC rescued astrocytes from LPS exposure without affecting LncRNA-MEG3 expression. CONCLUSION These findings demonstrated that LncRNA-MEG3 acted as a positive regulator of NF-κB, mediated the neuroprotection of propofol in LPS-triggered astrocytes injury.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Zhihua Wang
- Department of Anesthesiology, Hainan General Hospital, Haikou, Hainan Province, China
| | - Bei Sun
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Yan Huang
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Cheng Chen
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Jie Hu
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Longyan Li
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Pingping Xia
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Zhi Ye
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| |
Collapse
|
10
|
Schisandrin B Inhibits NLRP3 Inflammasome Pathway and Attenuates Early Brain Injury in Rats of Subarachnoid Hemorrhage. Chin J Integr Med 2022; 28:594-602. [PMID: 35015222 DOI: 10.1007/s11655-021-3348-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2021] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To determine whether Schisandrin B (Sch B) attenuates early brain injury (EBI) in rats with subarachnoid hemorrhage (SAH). METHODS Sprague-Dawley rats were divided into sham (sham operation), SAH, SAH+vehicle, and SAH+Sch B groups using a random number table. Rats underwent SAH by endovascular perforation and received Sch B (100 mg/kg) or normal saline after 2 and 12 h of SAH. SAH grading, neurological scores, brain water content, Evan's blue extravasation, and terminal transferase-mediated dUTP nick end-labeling (TUNEL) staining were carried out 24 h after SAH. Immunofluorescent staining was performed to detect the expressions of ionized calcium binding adapter molecule 1 (Iba-1) and myeloperoxidase (MPO) in the rat brain, while the expressions of B-cell lymphoma 2 (Bcl-2), Bax, Caspase-3, nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3), apoptosis-associated specklike protein containing the caspase-1 activator domain (ASC), Caspase-1, interleukin (IL)-1β, and IL-18 in the rat brains were detected by Western blot. RESULTS Compared with the SAH group, Sch B significantly improved the neurological function, reduced brain water content, Evan's blue content, and apoptotic cells number in the brain of rats (P<0.05 or P<0.01). Moreover, Sch B decreased SAH-induced expressions of Iba-1 and MPO (P<0.01). SAH caused the elevated expressions of Bax, Caspase-3, NLRP3, ASC, Caspase-1, IL-1β, and IL-18 in the rat brain (P<0.01), all of which were inhibited by Sch B (P<0.01). In addition, Sch B increased the Bcl-2 expression (P<0.01). CONCLUSION Sch B attenuated SAH-induced EBI, which might be associated with the inhibition of neuroinflammation, neuronal apoptosis, and the NLRP3 inflammatory signaling pathway.
Collapse
|
11
|
Huang Y, Wang X, Guan S, Lin H, Mei Z, Huang Z. Syringin protects against cerebral ischemia and reperfusion injury via suppression of inflammatory mediators and toll-like receptor/MyD88 signaling pathway in rats. Pharmacogn Mag 2022. [DOI: 10.4103/pm.pm_98_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
12
|
Xu QL, Wu J. Effects of Txk‑mediated activation of NF‑κB signaling pathway on neurological deficit and oxidative stress after ischemia‑reperfusion in rats. Mol Med Rep 2021; 24:524. [PMID: 34036382 PMCID: PMC8160475 DOI: 10.3892/mmr.2021.12163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 04/06/2021] [Indexed: 12/16/2022] Open
Abstract
Ischemic stroke is an extremely mortal cerebrovascular disease, and neuroinflammation and oxidative stress emerge as important traits of ischemic stroke. However, as an inflammation‑associated factor, Txk tyrosine kinases (Txk) has been poorly studied in neuroscience research. The aim of the present study was to investigate the role of Txk after ischemia‑reperfusion (I/R) in vivo and in vitro, observe the association between Txk knockdown and neurological deficit and oxidative stress, and to explore whether the process was mediated by the activation of nuclear factor (NF)‑κB signaling pathway. Middle cerebral artery occlusion (MCAO), oxygen and glucose deprivation/reperfusion (OGD/R) model and western blotting have been used to simulate the I/R injury to analyze the expression, and to approximate the localization of Txk, respectively. Brain infarct volume, neurological score, brain water content, apoptosis and oxidative stress assays in vivo and apoptosis, cellular viability, the LDH release and oxidative stress assays in vitro were observed using a Txk‑knockdown lentivirus. Finally, NF‑κB overexpression lentivirus was applied to discuss whether the role of Txk following I/R was regulated by the NF‑κB signaling pathway. The results show that the Txk expression peaked at 24 h after MCAO and 6 h after OGD/R, respectively. Txk molecules gradually entered the nucleus after MCAO and OGD/R. The Txk‑knockdown lentivirus resulted in decreased brain infarct volume, neurological score, brain water content, apoptosis and oxidative stress after MCAO in vivo. Besides, Txk knockdown decreased apoptosis, LDH release, oxidative stress, and increased cellular viability, after ODG in vitro. Finally, NF‑κB overexpression reversed the process of neurological deficit and oxidative stress after Txk regulation in vivo and vitro. Overall, the present study suggests that Txk potentially regulates apoptosis, neurological deficit, and oxidative stress after I/R, by entering the nucleus. NF‑κB maybe the downstream target factor of Txk.
Collapse
Affiliation(s)
- Qian-Lan Xu
- Department of Rehabilitation, Dongyang People's Hospital, Yiwu, Zhejiang 322000, P.R. China
| | - Jie Wu
- Department of Child Rehabilitation, Yiwu Maternal and Child Health Care Hospital, Yiwu, Zhejiang 322000, P.R. China
| |
Collapse
|
13
|
Yao L, Yang M, Zhang J, Wang F, Liu Q, Xie X, Liu Z, Guo Q, Su H, Zhai J, He J, Xue S, Qiu Z. Tectorigenin attenuates the OGD/R-induced HT-22 cell damage through regulation of the PI3K/AKT and the PPARγ/NF-κB pathways. Hum Exp Toxicol 2021; 40:1320-1331. [PMID: 33588632 DOI: 10.1177/0960327121993213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Tectorigenin (TEC) is an effective compound that derived from many plants, such as Iris unguicularis, Belamcanda chinensis and Pueraria thunbergiana Benth. Evidence suggested that TEC has anti-tumor, anti-oxidant activity, anti-bacterial and anti-inflammatory effects. In addition, there has some evidence indicated that TEC is a potential anti-stroke compound; however, its specific roles and associated mechanism have not yet been elucidated. In the present study, we aimed to investigate the anti-inflammatory, anti-oxidant activity and anti-apoptosis effects of TEC on oxygen-glucose deprivation/reperfusion (OGD/R)-induced HT-22 cells, and clarified the relevant mechanisms. Here, we observed that TEC significantly promoted cell survival, impeded cell apoptosis, inhibited ROS and inflammatory cytokines IL-1β, IL-6, TNF-α production in OGD/R-induced HT-22 cells. Moreover, TEC activated PI3K/AKT signal pathway, increased PPARγ expression and inhibited NF-κB pathway activation in OGD/R-induced HT-22 cells. Further studies indicated that PPARγ inhibitor GW9662 activated NF-κB pathway after TEC treatment in OGD/R-induced HT-22 cells. Also, PI3K/AKT inhibitor LY294002, PPARγ inhibitor GW9662 and NF-κB activator LPS both reversed the effects of TEC on OGD/R-induced HT-22 cell biology. Taken together, this research confirmed that TEC benefit to HT-22 cell survival and against OGD/R damage through the PI3K/AKT and PPARγ/NF-κB pathways. These results indicated that TEC might be an effective compound in the treatment for ischemic brain injury.
Collapse
Affiliation(s)
- Li Yao
- Department of Neurology, The Hospital of Xidian Group, Xi'an, China
| | - Meili Yang
- Department of Neurology, 117889The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Juanli Zhang
- Department of Neurology, The Hospital of Xidian Group, Xi'an, China
| | - Fei Wang
- Department of Neurology, The Hospital of Xidian Group, Xi'an, China
| | - Qing Liu
- Department of Neurology, The Hospital of Xidian Group, Xi'an, China
| | - Xiaojun Xie
- Department of Pathology, The Hospital of Xidian Group, Xi'an, China
| | - Zhuo Liu
- Department of Emergency, The Hospital of Xidian Group, Xi'an, China
| | - Qiang Guo
- Department of Neurology, The Hospital of Xidian Group, Xi'an, China
| | - Hang Su
- Department of Neurology, The Hospital of Xidian Group, Xi'an, China
| | - Jiemin Zhai
- Department of Neurology, The Hospital of Xidian Group, Xi'an, China
| | - Jianbo He
- Department of Neurology, The Hospital of Xidian Group, Xi'an, China
| | - Sha Xue
- Department of Anesthesiology, 117799The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Zhengguo Qiu
- Department of Anesthesiology, Affiliated Hospital of 107652Shaanxi University of Traditional Chinese Medicine, Xianyang, China
| |
Collapse
|
14
|
Bu X, Li T, Guo D, Yang C, Wang J, Wang X, Yang Z, Wang H. 1% Isoflurane and 1.2 μg/ml of Propofol: A Combination of Anesthetics That Causes the Least Damage to Hypoxic Neurons. Front Aging Neurosci 2020; 12:591938. [PMID: 33304268 PMCID: PMC7701289 DOI: 10.3389/fnagi.2020.591938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/19/2020] [Indexed: 11/22/2022] Open
Abstract
Backgrounds: Aging-related impairment of cerebral blood flow regulation leads to the disruption of neuronal micro-environmental homeostasis. Anesthetics should be carefully selected for aging patients since they have less cognition capacity. Effects and mechanisms of propofol or isoflurane have been widely investigated. However, how different combinations of propofol and isoflurane affect neurons and the mechanism still needs to be demonstrated. Methods: We cultured rat hippocampal neurons and established a hypoxic injury model to imitate the micro-environment of aging brains. Three different combinations of propofol and isoflurane were applied to find out an optimum group via Cell Counting Kit-8 (CCK8) assay, lactic acid dehydrogenase (LDH) assay, real-time qPCR, and immunofluorescence of key proteins. Then BiP was silenced by small interfering RNA (siRNA) to explore the mechanism of how isoflurane and propofol affect neurons. Endoplasmic reticulum (ER) stress was measured by Western blot and immunofluorescence. To detect GABAAR α1 subunit proteostasis and its function, real-time qPCR, immunoprecipitation, and Western blot were carried out. Results: Hypoxic neurons showed no different changes on cell viability, LDH leakage, and ER stress after treatment with 1% isoflurane and 1.2 μg/ml of propofol. Hypoxic neurons showed a sharp increase of LDH leakage and ER stress and a decrease of cell viability after treatment with 1.4% isoflurane and 0.6 μg/ml of propofol or 0.5% isoflurane and 1.8 μg/ml of propofol. After knockdown of BiP, the application of 1% isoflurane and 1.2 μg/ml of propofol led to the decrease of GABAAR α1 subunit protein content and viability of cell, as well as aggravation of ER stress. Conclusion: A combination of 1% isoflurane and 1.2 μg/ml of propofol causes the least damage than do other dosages of both two drugs, and endogenous BiP plays an important role in this process.
Collapse
Affiliation(s)
- Xinyue Bu
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Tang Li
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Di Guo
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Chenyi Yang
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China.,Department of Anesthesiology, The Third Central Hospital of Tianjin, Tianjin, China
| | - Jinxin Wang
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Xinyi Wang
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China.,Department of Anesthesiology, The Third Central Hospital of Tianjin, Tianjin, China
| | - Zhuo Yang
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin, China
| | - Haiyun Wang
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China.,Department of Anesthesiology, The Third Central Hospital of Tianjin, Tianjin, China.,Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin, China
| |
Collapse
|
15
|
Abdollahzade N, Babri S, Majidinia M. Attenuation of chronic arsenic neurotoxicity via melatonin in male offspring of maternal rats exposed to arsenic during conception: Involvement of oxidative DNA damage and inflammatory signaling cascades. Life Sci 2020; 266:118876. [PMID: 33310035 DOI: 10.1016/j.lfs.2020.118876] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/22/2020] [Accepted: 12/01/2020] [Indexed: 10/22/2022]
Abstract
Prenatal exposure to arsenic is demonstrated to elevate the risk of brain damage and neurological disorders in the fetus, mainly due to its ability for crossing through the placental barriers. Increase in oxidative stress, inflammation, and DNA damage is main mechanisms of arsenic-induced neurotoxicity. Therefore, this study aimed to evaluate the neuroprotective effects of melatonin, as a potent anti-oxidant and anti-inflammatory agent against arsenic toxicity in the brains of male offspring rats. Pregnant mother rats were randomly assigned into four groups including group I, as control, group II received 10 mg/kg melatonin, group III received arsenic at 50 mg/kg, and group IV received melatonin and arsenic. After a two-month period, oxidative stress, DNA damage, inflammation and apoptosis were assessed in the male offspring rats. Exposure to arsenic significantly increased the pro-inflammatory and oxidative factors resulting in DNA damage and apoptosis in the brain tissues of offspring rats in comparison to controls (p < 0.05). Exogenous administration of melatonin showed a significant increase in the tissue levels of acetylcholine esterase, decrease in the lactate dehydrogenase and myeloperoxidase, when compared to arsenic group (p < 0.05). Melatonin also overcame the arsenic-induced oxidative stress and suppressed inflammation, DNA damage and apoptosis. Our results suggested that melatonin may be a promising neuro-protective agent and momentous therapy for the treatment of arsenic-toxicity in clinical conditions.
Collapse
Affiliation(s)
- Naseh Abdollahzade
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Physiology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shirin Babri
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Physiology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
16
|
Yu S, Xin W, Jiang Q, Li A. Propofol exerts neuroprotective functions by down-regulating microRNA-19a in glutamic acid-induced PC12 cells. Biofactors 2020; 46:934-942. [PMID: 31913544 DOI: 10.1002/biof.1607] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 12/17/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Propofol, a kind of intravenous sedative drug, is certified that exerts anti-inflammation and antitumor functions. However, the influence of propofol in cerebral injury and the corresponding mechanism remains unexplained, that our article focuses on. METHODS PC12 cells were treated with propofol and exposed in glutamic acid (Glu) solutions. Cell viability, apoptotic potential, apoptosis-related and autophagy-linked proteins were tested via CCK-8, flow cytometry, and western blot assays. Reverse transcription-quantitative real-time PCR was utilized to test miR-19a expression in Glu-stimulated cells. Next, miR-19a mimic transfection was used to assess the effects of miR-19a on cell apoptosis and autophagy in Glu or propofol treated cells. Finally, western blot was performed to test AMPK and mTOR pathways. RESULTS Glu exposure promoted cell apoptosis and autophagy of PC12 cells, while propofol attenuated cell apoptosis and autophagy triggered by Glu. Additionally, propofol decreased the miR-19a expression in Glu-stimulated PC12 cells. Meanwhile, over-expression of miR-19a reversed the effects of propofol on Glu-induced cell apoptosis and autophagy. Moreover, propofol potentiated AMPK and mTOR pathways in Glu-stimulated PC12 cells via impeding miR-19a expression. CONCLUSIONS These finding revealed that propofol relieved Glu-triggered apoptosis and autophagy of PC12, and activated AMPK and mTOR pathways by suppressing miR-19a expression.
Collapse
Affiliation(s)
- Shashuang Yu
- Department of Anesthesiology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Wenqi Xin
- Department of Anesthesiology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Qiliang Jiang
- Department of Anesthesiology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Aixiang Li
- Department of Anesthesiology, Huaihe Hospital of Henan University, Kaifeng, China
| |
Collapse
|
17
|
Wang J, Mao J, Wang R, Li S, Wu B, Yuan Y. Kaempferol Protects Against Cerebral Ischemia Reperfusion Injury Through Intervening Oxidative and Inflammatory Stress Induced Apoptosis. Front Pharmacol 2020; 11:424. [PMID: 32351385 PMCID: PMC7174640 DOI: 10.3389/fphar.2020.00424] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 03/19/2020] [Indexed: 01/04/2023] Open
Abstract
The aim of this research is to investigate the potential neuro-protective effect of kaempferol which with anti-oxidant, anti-inflammatory, and immune modulatory properties, and understand the effect of kaempferol on reducing cerebral ischemia reperfusion (I/R) injury in vivo. Male adult Sprague Dawley (SD) rats were pretreated with kaempferol for one week via gavage before cerebral I/R injury operation. We found that kaempferol treatment can reduce the cerebral infarct volume and neurological score after cerebral I/R. Rats were sacrificed after 24 h reperfusion. We observed that kaempferol improved the arrangement, distribution, and morphological structure of neurons, as well as attenuated cell apoptosis in brain tissue via hematoxylin and eosin (H&E) staining, Nissl staining and TUNEL staining. Superoxide dismutase (SOD), malondialdehyde (MDA), and glutathione peroxidase (GSH) kit analysis, enzyme-linked immunosorbent (ELISA) assay, real-time PCR, Western blot, and immunohistochemical examination indicated that kaempferol mitigated oxidative and inflammatory stress via regulating the expression of proteins, p-Akt, p-GSK-3β, nuclear factor erythroid2-related factor 2 (Nrf-2), and p-NF-κB during cerebral I/R, thus increasing the activity of SOD and GSH, meanwhile decreasing the content of MDA in serum and brain tissue, as well as restoring the expression levels of tumor necrosis factor alpha (TNF-α), interleukin-1β (IL-1β), and IL-6 in vivo. Taken together, this study suggested that kaempferol protects against cerebral I/R induced brain damage. The possible mechanism is related with inhibiting oxidative and inflammatory stress induced apoptosis.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junqin Mao
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Rong Wang
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengnan Li
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Wu
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongfang Yuan
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Ruan H, Li W, Wang J, Chen G, Xia B, Wang Z, Zhang M. Propofol alleviates ventilator-induced lung injury through regulating the Nrf2/NLRP3 signaling pathway. Exp Mol Pathol 2020; 114:104427. [PMID: 32199914 DOI: 10.1016/j.yexmp.2020.104427] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 02/17/2020] [Accepted: 03/17/2020] [Indexed: 12/17/2022]
Abstract
Ventilator-induced lung injury (VILI) causes problems during acute lung injury treatment, and propofol is a well-known drug to prevent VILI. Herein, we discussed how propofol protects against VILI-induced inflammation with the interaction of nuclear factor E2-related factor 2 (Nrf2)/NOD-like receptor protein 3 (NLRP3). We established VILI mouse models for collecting lung tissues, and these mice were later treated with propofol and Nrf2/NLRP3 activator or inhibitor to observe their effects on VILI with inflammatory factors, 8-hydroxy-2 deoxyguanosine, malondialchehyche level, mitochondrial reactive oxygen species production rate, lung wet/dry weight ratio, lung permeability index measured. Propofol treatment improved VILI, alleviated pulmonary inflammation induced by mechanical ventilation. Propofol up-regulated Nrf2 and down-regulated NLRP3 in VILI model. Activating Nrf2 or inhibiting NLRP3 downregulated pro-inflammatory factors in lung tissues in VILI mice. Above all, we can conclude that propofol exerts it protective function against VILI and the subsequent inflammatory responses through activating Nrf2 and inhibiting NLRP3 expression. Therefore, Nrf2 activator and NLRP3 inhibitor might be latent targets in the VILI prevention.
Collapse
Affiliation(s)
- Hongyan Ruan
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong, PR China
| | - Wei Li
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong, PR China
| | - Jilan Wang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong, PR China
| | - Gang Chen
- Department of thoracic surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong, PR China
| | - Bin Xia
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong, PR China
| | - Zhou Wang
- Department of thoracic surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong, PR China.
| | - Mengyuan Zhang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong, PR China.
| |
Collapse
|
19
|
Zhang HB, Tu XK, Song SW, Liang RS, Shi SS. Baicalin Reduces Early Brain Injury after Subarachnoid Hemorrhage in Rats. Chin J Integr Med 2020; 26:510-518. [DOI: 10.1007/s11655-020-3183-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2018] [Indexed: 10/25/2022]
|
20
|
Hua S, Liu J, Zhang Y, Li J, Zhang X, Dong L, Zhao Y, Fu X. Piperine as a neuroprotective functional component in rats with cerebral ischemic injury. Food Sci Nutr 2019; 7:3443-3451. [PMID: 31762997 PMCID: PMC6848843 DOI: 10.1002/fsn3.1185] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 08/01/2019] [Accepted: 08/12/2019] [Indexed: 01/27/2023] Open
Abstract
Long pepper (Piper longum L.) and black pepper (Piper nigrum L.) plants are commonly used as spices around the world and have also been postulated to have medicinal effects. Piperine, as the major alkaloid of P. nigrum and P. longum, has gained wide attention of the medical community and culinary enthusiasts. This study seeks to determine the effects of piperine on neuronal apoptosis in peri-infarcted cerebral cortices of rats with permanent middle cerebral artery occlusion (pMCAO) injury. Evaluation of the different behavioral components was conducted after pMCAO. 2, 3, 5-Triphenyltetrazolium chloride (TTC) was used to evaluate the area of cortical ischemia. Gross histopathological changes, as well as microscopic neuronal changes, were observed in brain tissue samples. The protein expression of Caspase-3, Caspase-9, Bax, Bcl-2, and Cytochrome C (Cyt-c) was analyzed using western blotting. The findings reveal that rats that received piperine treatment show markedly decreased neurological deficit, less ischemia-induced cellular damage, as well as smaller areas of cerebral infarction, with less severe macro and microcellular cerebral structural changes. Western blotting analysis reveals that piperine administration inhibits Bax, while enhancing Bcl-2 expression. The protein expression of Caspase-3, Caspase-9, and Cyt-c was also found to be significantly inhibited. We conclude that piperine may provide several beneficial neuroprotective effects that warrant further investigation.
Collapse
Affiliation(s)
- Shiyao Hua
- School of PharmacyNingxia Medical UniversityYinchuanChina
| | - Jiayue Liu
- School of PharmacyNingxia Medical UniversityYinchuanChina
| | - Yiwei Zhang
- School of Basic Medical SciencesNingxia Medical UniversityYinchuanChina
| | - Juan Li
- School of PharmacyNingxia Medical UniversityYinchuanChina
- Ningxia Engineering and Technology Research Center for Modernization of Hui MedicineYinchuanChina
- Key Laboratory of Hui Ethnic Medicine ModernizationMinistry of Education (Ningxia Medical University)YinchuanChina
| | - Xinhui Zhang
- School of PharmacyNingxia Medical UniversityYinchuanChina
- Ningxia Engineering and Technology Research Center for Modernization of Hui MedicineYinchuanChina
- Key Laboratory of Hui Ethnic Medicine ModernizationMinistry of Education (Ningxia Medical University)YinchuanChina
| | - Lin Dong
- School of PharmacyNingxia Medical UniversityYinchuanChina
- Ningxia Engineering and Technology Research Center for Modernization of Hui MedicineYinchuanChina
- Key Laboratory of Hui Ethnic Medicine ModernizationMinistry of Education (Ningxia Medical University)YinchuanChina
| | - Yunsheng Zhao
- School of PharmacyNingxia Medical UniversityYinchuanChina
- Ningxia Engineering and Technology Research Center for Modernization of Hui MedicineYinchuanChina
- Key Laboratory of Hui Ethnic Medicine ModernizationMinistry of Education (Ningxia Medical University)YinchuanChina
| | - Xueyan Fu
- School of PharmacyNingxia Medical UniversityYinchuanChina
- Ningxia Engineering and Technology Research Center for Modernization of Hui MedicineYinchuanChina
- Key Laboratory of Hui Ethnic Medicine ModernizationMinistry of Education (Ningxia Medical University)YinchuanChina
| |
Collapse
|
21
|
Propofol Reduces Inflammatory Brain Injury after Subarachnoid Hemorrhage: Involvement of PI3K/Akt Pathway. J Stroke Cerebrovasc Dis 2019; 28:104375. [PMID: 31590996 DOI: 10.1016/j.jstrokecerebrovasdis.2019.104375] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/04/2019] [Accepted: 08/26/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Our previous study showed that propofol, one of the widely used anesthetic agents, can attenuate subarachnoid hemorrhage (SAH)-induced early brain injury (EBI) via inhibiting inflammatory and oxidative reaction. However, it is perplexing whether propofol attenuates inflammatory and oxidative reaction through modulating PI3K/Akt pathway. The present study investigated whether PI3K/Akt pathway is involved in propofol's anti-inflammation, antioxidation, and neuroprotection against SAH-induced EBI. MATERIALS AND METHODS Adult Sprague-Dawley rats underwent SAH and received treatment with propofol or vehicle after 2 and 12 hours of SAH. LY294002 was injected intracerebroventricularly to selectively inhibit PI3K/Akt signaling. Mortality, SAH grading, neurological scores, brain water content, evans blue extravasation, myeloperoxidase, malondialdehyde, superoxide dismutase, and glutathione peroxidase were measured 24 hours after SAH. Immunoreactivity of p-Akt, t-Akt, nuclear factor- kappa B (NF-κB) p65, nuclear factor erythroid-related factor 2 (Nrf2), NAD(P)H:quinone oxidoreductase (NQO1), and cyclooxygenase-2 (COX-2) in rat brain was determined by western blot. Tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in rat brain were examined by ELISA. RESULTS Propofol significantly reduces neurological dysfunction, BBB permeability, brain edema, inflammation, and oxidative stress, all of which were reversed by LY294002. Propofol significantly upregulates the immunoreactivity of p-Akt, Nrf2, and NQO1, all of which were abolished by LY294002. Propofol significantly downregulates the overexpression of NF-κB p65, COX-2, TNF-α, and IL-1β, all of which were inhibited by LY294002. CONCLUSION These results suggest that propofol attenuates SAH-induced EBI by inhibiting inflammatory reaction and oxidative stress, which might be associated with the activation of PI3K/Akt signaling pathway.
Collapse
|
22
|
N-Palmitoylethanolamide-Oxazoline Protects against Middle Cerebral Artery Occlusion Injury in Diabetic Rats by Regulating the SIRT1 Pathway. Int J Mol Sci 2019; 20:ijms20194845. [PMID: 31569558 PMCID: PMC6801841 DOI: 10.3390/ijms20194845] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 12/19/2022] Open
Abstract
Diabetes causes various macrovascular and microvascular alterations, often culminating in major clinical complications (first of all, stroke) that lack an effective therapeutic intervention. N-palmitoylethanolamide-oxazoline (PEA-OXA) possesses anti-inflammatory and potent neuroprotective effects. Although recent studies have explained the neuroprotective properties of PEA-OXA, nothing is known about its effects in treating cerebral ischemia. Methods: Focal cerebral ischemia was induced by transient middle cerebral artery occlusion (MCAo) in the right hemisphere. Middle cerebral artery (MCA) occlusion was provided by introducing a 4–0 nylon monofilament (Ethilon; Johnson & Johnson, Somerville, NJ, USA) precoated with silicone via the external carotid artery into the internal carotid artery to occlude the MCA. Results: A neurological severity score and infarct volumes were carried out to assess the neuroprotective effects of PEA-OXA. Moreover, we observed PEA-OXA-mediated improvements in tissue histology shown by a reduction in lesion size and an improvement in apoptosis level (assessed by caspases, Bax, and Bcl-2 modulation and a TUNEL assay), which further supported the efficacy of PEA-OXA therapy. We also found that PEA-OXA treatment was able to reduce mast cell degranulation and reduce the MCAo-induced expression of NF-κB pathways, cytokines, and neurotrophic factors. Conclusions: based on these findings, we propose that PEA-OXA could be useful in decreasing the risk of impairment or improving function in ischemia/reperfusion brain injury-related disorders.
Collapse
|
23
|
Propofol can suppress renal ischemia-reperfusion injury through the activation of PI3K/AKT/mTOR signal pathway. Gene 2019; 708:14-20. [DOI: 10.1016/j.gene.2019.05.023] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 05/04/2019] [Accepted: 05/09/2019] [Indexed: 12/14/2022]
|
24
|
Mohamed SK, Ahmed AAE, Elmorsy EM, Nofal S. ERK activation by zeranol has neuroprotective effect in cerebral ischemia reperfusion. Life Sci 2019; 227:137-144. [PMID: 31005550 DOI: 10.1016/j.lfs.2019.04.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/02/2019] [Accepted: 04/15/2019] [Indexed: 11/29/2022]
Abstract
AIMS Incidence of stroke increases in postmenopausal women with dangerous consequences. In this study we used zeranol to protect ovariectomized (OVX) rats against cerebral I/R damage and our target is to identify the mechanism of its protection, in addition to investigating whether this mechanism inhibits inflammation (by preventing glial cell activation) and apoptosis. MAIN METHODS First 18 ovariectomized rats were allocated into 3 groups: I/R group, zeranol+ I/R group and U0126, MEK1/2 inhibitor + zeranol+ I/R group. After 24 h reperfusion, protein expression of total extracellular signal-regulated protein kinase (t-ERK1/2), phosphorylated extracellular signal-regulated protein kinase (p-ERK1/2), Bcl-2, and Bax were quantified. Second 36 female rats were allocated into 3 groups: sham group, I/R group (after ovariectomy by 7 weeks, rats exposed to cerebral I/R) and zeranol group (after ovariectomy by 2 weeks, rats received zeranol for 5 weeks). After 24 h of reperfusion, the following parameters were measured; total nitrate/nitrite, interleukin-10, myeloperoxidase, caspase-3, and finally immunohistochemistry analysis of glial fibrillary acidic protein, cyclooxygenase-2 in cortex and hippocampus (CA1) regions were performed. KEY FINDINGS U-0126 administration reversed the neuroprotective effect induced by zeranol through decreasing ratio of p-ERK1/2:ERK1/2 and Bcl-2/Bax in brain tissue. Activation of ERK signaling pathway by zeranol caused reduction in brain apoptosis and inflammation. SIGNIFICANCE Zeranol showed protective effect in OVX rats that were exposed to cerebral I/R by activation of ERK signaling pathway which was blocked by U0126. This protective effect in turns led to decrease inflammation and apoptosis.
Collapse
Affiliation(s)
- Shimaa K Mohamed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt.
| | - Amany A E Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Engy M Elmorsy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Shahira Nofal
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| |
Collapse
|
25
|
Li Q, Cui S, Jing G, Ding H, Xia Z, He X. The role of PI3K/Akt signal pathway in the protective effects of propofol on intestinal and lung injury induced by intestinal ischemia/reperfusion1. Acta Cir Bras 2019; 34:e20190010000005. [PMID: 30785506 PMCID: PMC6585923 DOI: 10.1590/s0102-865020190010000005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/15/2019] [Indexed: 12/15/2022] Open
Abstract
Purpose To investigate the role of PI3k/Akt signal pathway in the protective effects
of propofol on intestinal and lung injury induced by intestinal
ischemia/reperfusion(I/R). Methods Male Sprague-Dawley rats were subjected to 45 min of ischemia by occluding
the superior mesenteric artery and to 2h of reperfusion to establish the
model of I/R. Twenty four rats were randomly divided into four groups: Sham,
intestinal I/R (II/R), propofol (P), wortmannin (W). In groups P, W,
propofol was injected intravenously and continuously at the onset of
reperfusion via infusion pump. PI3K inhibitor (wortmannin) was administered
intravenously in group W 25 min before ischemia. Intestinal tissues and lung
tissues were obtained for determination of histologic injury, wet/dry weight
ratio, malondialdehyde (MDA) levels, superoxide dismutase (SOD) and
myeloperoxidase (MPO) activities. Meanwhile, the expressions of caspase-3
and phosphorylated Akt (p-Akt) in intestines and lungs were detected by
western blot. Results Propofol treatment alleviated intestinal and lung morphological changes which
were observed in II/R group , Moreover, wet/dry weight ratio, the MDA level,
MPO activity and expression of caspase-3 were significantly decreased
whereas the SOD activity and p-Akt expression were significantly increased.
Notably, the protections were significantly reversed by pretreatment of
wortmannin. Conclusion: PI3K/Akt pathway activation play a critical role in the protective effects of
propofol on intestinal and lung injury induced by ischemia/reperfusion.
Collapse
Affiliation(s)
- Qingwen Li
- MD, Department of Anesthesiology, Renmin Hospital of Wuhan University, Hubei, China. Manuscript preparation and writng
| | - Shanshan Cui
- MD, Department of Anesthesiology, Renmin Hospital of Wuhan University, Hubei, China. Conception and design of the study, acquisiton of data
| | - Guoqing Jing
- MD, Department of Anesthesiology, Renmin Hospital of Wuhan University, Hubei, China. Analysis and interpretation of data, technical procedures
| | - Huang Ding
- MD, Department of Anesthesiology, Renmin Hospital of Wuhan University, Hubei, China. Histopathological examinations, statistics analysis
| | - Zhongyuan Xia
- MD, Department of Anesthesiology, Renmin Hospital of Wuhan University, Hubei, China. Final approval
| | - Xianghu He
- MD, Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Hubei, China. Critical revision
| |
Collapse
|
26
|
Naomaitai Ameliorated Brain Damage in Rats with Vascular Dementia by PI3K/PDK1/AKT Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:2702068. [PMID: 30867669 PMCID: PMC6379870 DOI: 10.1155/2019/2702068] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/21/2018] [Accepted: 12/13/2018] [Indexed: 12/30/2022]
Abstract
Background/Aims Naomaitai can improve blood perfusion and ameliorate the damage in the paraventricular white matter. This study was focused on observing the neuroprotective effect of Naomaitai on the vascular dementia of rat and exploring the action mechanism of PI3K/PDK1/AKT signaling pathway. Methods A vascular dementia model of rats was established by permanent, bilateral common carotid artery occlusion. Rats' behavior was tested by Neurological deficit score and the Morris water maze. The pathology and apoptosis were detected through HE staining and TUNEL assay. Myelin sheath loss and nerve fiber damage were detected by LFB staining. Inflammatory factors, oxidative stress, and brain damage markers were detected through ELISA. The expression of apoptosis-related proteins and PI3K/PDK1/AKT signaling pathway related proteins were measured by western blot. The expressions of PI3K, PDK1, AKT, and MBP in paraventricular white matter cells were detected by immunofluorescence. Results Naomaitai treatment decreased neurological function score in rats with vascular dementia, ameliorated paraventricular white matter damage caused by long-term hypoxia, and hypoperfusion reduced the brain injury markers S-100β and NSE contents, suppressed inflammatory reaction and oxidative stress, reduced IL-1β, IL-6, TNF-α, and MDA contents, and remarkably increased IL-10 and SOD contents. TUNEL and western blot assay showed that Naomaitai treatment decreased neuronal cell apoptosis, increased Bcl-2 expression, and reduced caspase-3 and Bax expression. Furthermore, we found Naomaitai inhibited PI3K and PDK1 expression and activated phosphorylated AKT protein in rats with vascular dementia. However, the protective effect of Naomatai in rats with vascular dementia was inhibited, and expression of PI3K signaling pathway-related proteins was blocked after administration of PI3K inhibitor. Conclusion Naomaitai can ameliorate brain damage in rats with vascular dementia, inhibit neuronal apoptosis, and have anti-inflammatory and antioxidative stress effects, which may be regulated by the PI3K/PDK1/AKT signaling pathway.
Collapse
|
27
|
Luteoloside attenuates neuroinflammation in focal cerebral ischemia in rats via regulation of the PPARγ/Nrf2/NF-κB signaling pathway. Int Immunopharmacol 2018; 66:309-316. [PMID: 30502652 DOI: 10.1016/j.intimp.2018.11.044] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 11/20/2018] [Accepted: 11/26/2018] [Indexed: 11/24/2022]
Abstract
Luteoloside, a flavonoid compound, has been reported to have anti-inflammatory, anti-oxidative, antibacterial, antiviral, anticancer, and cardioprotective effects, among others, but its neuroprotective effects have rarely been studied. The purpose of this study was to investigate the protective effect of luteoloside on cerebral ischemia and explore its potential mechanism. Middle cerebral artery occlusion (MCAO) was performed to investigate the effects of luteoloside on cerebral ischemia-reperfusion (I/R). Male Sprague-Dawley rats were randomly divided into six groups: sham, MCAO, luteoloside (20 mg/kg, 40 mg/kg, 80 mg/kg) and nimodipine (4 mg/kg). The results showed that luteoloside alleviated neurologic deficits and cerebral edema as well as improved cerebral infarction and histopathological changes in MCAO rats. Luteoloside significantly inhibited I/R-induced neuroinflammation, as demonstrated by reduced levels of interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in the brain tissues of MCAO rats. Furthermore, our results demonstrated that luteoloside significantly suppressed the activation of nuclear factor-kappa B (NF-κB) signaling, upregulated the protein expression of peroxisome proliferator activated receptor gamma (PPARγ) and increased NF-E2-related factor (Nrf2) nuclear accumulation in MCAO rats. Collectively, our findings suggested that luteoloside played a crucial neuroprotective role by inhibiting NF-κB signaling in focal cerebral ischemia in rats. Furthermore, PPARγ and Nrf2 were also important for the anti-inflammatory effect of luteoloside. In addition, our data suggested that luteoloside might be an effective treatment for cerebral ischemia and other neurological disorders.
Collapse
|
28
|
Yu H, Wang X, Kang F, Chen Z, Meng Y, Dai M. Propofol attenuates inflammatory damage on neurons following cerebral infarction by inhibiting excessive activation of microglia. Int J Mol Med 2018; 43:452-460. [PMID: 30431058 DOI: 10.3892/ijmm.2018.3974] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 02/02/2018] [Indexed: 11/06/2022] Open
Affiliation(s)
- Hang Yu
- Intensive Care Unit, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Xiaozhi Wang
- Intensive Care Unit, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Fuxin Kang
- Intensive Care Unit, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Zhile Chen
- Intensive Care Unit, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Yunxia Meng
- Intensive Care Unit, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Mingming Dai
- Department of Internal Neurology, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570311, P.R. China
| |
Collapse
|
29
|
Angiotensin II-mediated suppression of synaptic proteins in mouse hippocampal neuronal HT22 cell was inhibited by propofol: role of calcium signaling pathway. J Anesth 2018; 32:856-865. [DOI: 10.1007/s00540-018-2565-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 10/03/2018] [Indexed: 12/11/2022]
|
30
|
Propofol inhibits parthanatos via ROS-ER-calcium-mitochondria signal pathway in vivo and vitro. Cell Death Dis 2018; 9:932. [PMID: 30224699 PMCID: PMC6141459 DOI: 10.1038/s41419-018-0996-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 08/20/2018] [Accepted: 08/27/2018] [Indexed: 12/14/2022]
Abstract
Parthanatos is a new form of programmed cell death. It has been recognized to be critical in cerebral ischemia–reperfusion injury, and reactive oxygen species (ROS) can induce parthanatos. Recent studies found that propofol, a widely used intravenous anesthetic agent, has an inhibitory effect on ROS and has neuroprotective in many neurological diseases. However, the functional roles and mechanisms of propofol in parthanatos remain unclear. Here, we discovered that the ROS–ER–calcium–mitochondria signal pathway mediated parthanatos and the significance of propofol in parthanatos. Next, we found that ROS overproduction would cause endoplasmic reticulum (ER) calcium release, leading to mitochondria depolarization with the loss of mitochondrial membrane potential. Mitochondria depolarization caused mitochondria to release more ROS, which, in turn, contributed to parthanatos. Also, we found that propofol inhibited parthanatos through impeding ROS overproduction, calcium release from ER, and mitochondrial depolarization in parthanatos. Importantly, our results indicated that propofol protected cerebral ischemia–reperfusion via parthanatos suppression, amelioration of mitochondria, and ER swelling. Our findings provide new insights into the mechanisms of how ER and mitochondria contribute to parthanatos. Furthermore, our studies elucidated that propofol has a vital role in parthanatos prevention in vivo and in vitro, and propofol can be a promising therapeutic approach for nerve injury patients.
Collapse
|
31
|
Li Y, Liu Y, Fan J, Zhou Q, Song X, Peng Z, Qin Z, Tao T. Validation and bioinformatic analysis of propofol-induced differentially expressed microRNAs in primary cultured neural stem cells. Gene 2018; 664:90-100. [PMID: 29679758 DOI: 10.1016/j.gene.2018.04.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/26/2018] [Accepted: 04/16/2018] [Indexed: 11/27/2022]
Abstract
Propofol, a widely used intravenous anesthetic, was previously considered as a neuroprotective agent. Recently, however, accumulating evidence suggests that it may cause neurotoxicity, especially in the development of neural stem cells (NSCs). The potential mechanisms contributing to propofol-induced neurotoxicity during neurogenesis, such as those involving microRNAs (miRNAs), are still unknown. In this study, a total of 27 differentially expressed miRNAs were identified in our initial screen and 6 miRNAs were validated by qRT-PCR. Three miRNAs were up-regulated (miR-377-5p, miR-194-3p and miR-143-5p), and three were down-regulated (miR-3583-3p, miR-466b-5p and miR-410-5p). Following gene ontology and KEGG pathway enrichment analysis, Gabbr1, Canca1b and Gabbr2, which are enriched in the GABAergic synapse pathway, were selected as genes potentially playing a role in propofol-induced neurotoxicity. Gabbr1 and Cacna1b, which are targeted by miRNAs that are up-regulated following propofol exposure, showed decreased expression at the mRNA and protein levels. Gabbr2, targeted by miRNAs that were down-regulated following treatment with propofol, was up-regulated at both the levels of mRNA and protein expression. The two clusters of miRNAs that show differential expression following propofol exposure may act in a synergistic manner to regulate several genes simultaneously during the development of NSCs. Our results may contribute to clarify the molecular mechanism and provide potential therapeutic targets for propofol induced neurotoxicity.
Collapse
Affiliation(s)
- Yan Li
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youtan Liu
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Jun Fan
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Key Laboratory of Psychiatric Disorders of Guangdong Province, Guangzhou, China
| | - Quan Zhou
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiuling Song
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiyong Peng
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zaisheng Qin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Tao Tao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
32
|
Saha S, Sadhukhan P, Mahalanobish S, Dutta S, Sil PC. Ameliorative role of genistein against age-dependent chronic arsenic toxicity in murine brains via the regulation of oxidative stress and inflammatory signaling cascades. J Nutr Biochem 2018; 55:26-40. [PMID: 29331881 DOI: 10.1016/j.jnutbio.2017.11.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 10/11/2017] [Accepted: 11/14/2017] [Indexed: 01/22/2023]
Abstract
Brain is highly prone to oxidative damage due to its huge lipid content and extensive energy requirements. Exogenous insult in brain via oxidative injury can lead to severe pathophysiological conditions. Age-dependent deterioration of normal brain functions is also noteworthy. Genistein, a polyphenolic isoflavonoid, obtained from the soy plant, is well known to protect against several diseased conditions. Here, in this study chronic brain toxicity model was developed using oral administration of arsenic for 90 days in adult and aged murines. We observed that intraperitoneal administration of genistein improved the arsenic induced behavioral abnormalities in the rats. It was also evident from the histopathological studies that the extent of tissue damage due to arsenic exposure was more in aged rats compared to the adults. Evaluation of different stress markers, intracellular ROS level and mitochondrial membrane potential revealed the involvement of oxidative stress and mitochondrial dysfunction in inducing brain damage in arsenic exposed murines. It was observed that genistein can significantly ameliorate the stressed condition in both the animal groups but the protective effect of genistein was more significant in the adult animals. The underlying signalling mechanism behind the cytotoxicity of arsenic was investigated and revealed that genistein exhibited neuroprotection significantly by modulating the JNK3 mediated apoptosis, ERK1/2 mediated autophagy and TNFα associated inflammatory pathways. Overall study infers that genistein has significant ameliorative effect of against age-dependent cytotoxicity of arsenic in murine brains.
Collapse
Affiliation(s)
- Sukanya Saha
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Pritam Sadhukhan
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Sushweta Mahalanobish
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Sayanta Dutta
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, 700054, India.
| |
Collapse
|
33
|
Liraglutide Activates the Nrf2/HO-1 Antioxidant Pathway and Protects Brain Nerve Cells against Cerebral Ischemia in Diabetic Rats. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2018; 2018:3094504. [PMID: 29623090 PMCID: PMC5829331 DOI: 10.1155/2018/3094504] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 12/15/2017] [Accepted: 12/27/2017] [Indexed: 01/10/2023]
Abstract
This study aimed to determine the effect of liraglutide pretreatment and to elucidate the mechanism of nuclear factor erythroid 2-related factor (Nrf2)/heme oxygenase-1 (HO-1) signaling after focal cerebral ischemia injury in diabetic rats model. Adult male Sprague-Dawley rats were randomly divided into the sham-operated (S) group, diabetes mellitus ischemia (DM + MCAO) group, liraglutide pretreatment normal blood glucose ischemia (NDM+MCAO+L) group, and liraglutide pretreatment diabetes ischemia (DM + MCAO + L) group. At 48 h after middle cerebral artery occlusion (MCAO), neurological deficits and infarct volume of brain were measured. Oxidative stress brain tissue was determined by superoxide dismutase (SOD) and myeloperoxidase (MPO) activities. The expression levels of Nrf2 and HO-1 of brain tissue were analyzed by western blotting. In the DM + MCAO + L group, neurological deficits scores and cerebral infarct volume seemed to decrease at 48 h after MCAO cerebral ischemia compared with those in DM + MCAO group (P < 0.05). In addition, the expression of Nrf2 and HO-1 increased in 48 h at liraglutide pretreatment groups after MCAO cerebral ischemia if compared with those in the DM + MCAO group (P < 0.05). Furthermore, the DM + MCAO + L group has no significant difference compared with the NDM + MCAO + L group (P > 0.05). To sum up, alleviating effects of liraglutide on diabetes complicated with cerebral ischemia injury rats would be related to Nrf2/HO-1 signaling pathway.
Collapse
|
34
|
Propofol, but not ketamine or midazolam, exerts neuroprotection after ischaemic injury by inhibition of Toll-like receptor 4 and nuclear factor kappa-light-chain-enhancer of activated B-cell signalling: A combined in vitro and animal study. Eur J Anaesthesiol 2018; 33:670-80. [PMID: 26981881 DOI: 10.1097/eja.0000000000000449] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Propofol, midazolam and ketamine are widely used in today's anaesthesia practice. Both neuroprotective and neurotoxic effects have been attributed to all three agents. OBJECTIVE To establish whether propofol, midazolam and ketamine in the same neuronal injury model exert neuroprotective effects on injured neurones in vitro and in vivo by modulation of the Toll-like receptor 4-nuclear factor kappa-light-chain-enhancer of activated B cells (TLR-4-NF-κB) pathway. DESIGN AND SETTING Cell-based laboratory (n = 6 repetitions per experiment) and animal (n = 6 per group) studies using a neuronal cell line (SH-SY5Y cells) and adult Sprague-Dawley rats. INTERVENTIONS Cells were exposed to oxygen-glucose deprivation before or after treatment using escalating, clinically relevant doses of propofol, midazolam and ketamine. In animals, retinal ischaemia (60 min) was induced followed by reperfusion and randomised treatment with saline or propofol. MAIN OUTCOME MEASURES Neuronal cell death was determined using flow-cytometry (mitochondrial membrane potential) and lactate dehydrogenase (LDH) release. Nuclear factor NF-κB and hypoxia-inducible factor 1 α-activity were analysed by DNA-binding ELISA, expression of NF-κB-dependent genes and TLR-4 by luciferase-assay and flow-cytometry, respectively. In animals, retinal ganglion cell density, caspase-3 activation and gene expression (TLR-4, NF-κB) were used to determine in vivo effects of propofol. Results were compared using ANOVA (Analysis of Variance) and t test. A P value less than 0.05 was considered statistically significant. RESULTS Post-treatment with clinically relevant concentrations of propofol (1 to 10 μg ml) preserved the mitochondrial membrane potential in oxygen-glucose deprivation-injured cells by 54% and reduced LDH release by 21%. Propofol diminished TLR-4 surface expression and preserved the DNA-binding activity of the protective hypoxia-inducible factor 1 α transcription factor. DNA-binding and transcriptional NF-κB-activity were inhibited by propofol. Neuronal protection and inhibition of TLR-4-NF-κB signalling were not consistently seen with midazolam or ketamine. In vivo, propofol treatment preserved rat retinal ganglion cell densities (cells mm, saline 1504 ± 251 vs propofol 2088 ± 144, P = 0.0001), which was accompanied by reduced neuronal caspase-3, TLR-4 and NF-κB expression. CONCLUSION Propofol, but neither midazolam nor ketamine, provides neuroprotection to injured neuronal cells via inhibition of TLR-4-NF-κB-dependent signalling.
Collapse
|
35
|
Berberine Ameliorates MCAO Induced Cerebral Ischemia/Reperfusion Injury via Activation of the BDNF–TrkB–PI3K/Akt Signaling Pathway. Neurochem Res 2018; 43:702-710. [DOI: 10.1007/s11064-018-2472-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 12/25/2017] [Accepted: 01/09/2018] [Indexed: 11/25/2022]
|
36
|
Wang M, Meng F, Song Q, Zhang J, Dai C, Zhao Q. Changes in transcranial electrical motor-evoked potentials during the early and reversible stage of permanent spinal cord ischemia predict spinal cord injury in a rabbit animal model. Exp Ther Med 2017; 14:5429-5437. [PMID: 29285072 PMCID: PMC5740705 DOI: 10.3892/etm.2017.5215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 04/07/2017] [Indexed: 12/30/2022] Open
Abstract
The present study examined changes in the transcranial electrical motor-evoked potentials (TceMEP) waveform to predict neurological deficits and histopathological changes during the early and reversible stage of different levels of permanent spinal cord ischemic injury in a rabbit animal model. A total of 24 New Zealand rabbits were randomly divided into four groups of 6 rabbits each. Group 1 underwent a ligation of the lumbar artery at three levels (L1-L3), group 2 underwent a ligation of the lumbar artery at four levels (L1-L4) and group 3 underwent a ligation of the lumbar artery at five levels (L1-L5). The sham group contained 6 rabbits and did not receive ligation. TceMEP was recorded within 5 min of ligation and, 2 days later, motor function was assessed and the spinal cords were removed for histological examination. Following spinal cord injury, the relationship between variations in the TceMEP waveform and motor function and pathological damage was analyzed. It was observed that the amplitude of TceMEP began to decrease within 1 min of lumbar artery ligation and that the amplitude stabilized within 5 min. These amplitude changes that occurred within 5 min of different levels of permanent spinal cord ischemic injury were positively related to changes in motor function following recovery from anesthesia and 2 days after ligation. The Pearson correlation coefficient was 0.980 and 0.923 for these two time points, respectively (P<0.001). In addition, the amplitude changes were positively related to pathological damage, with a Pearson correlation coefficient of 0.945 (P<0.001). The results of the present study suggested that amplitude changes in TceMEP are particularly sensitive to ischemia. Ischemia may be detected within 1 min and the amplitude changes begin to stabilize within 5 min following ligation of the lumbar artery. The use of intraoperative monitoring of TceMEP allows for the detection of spinal cord ischemic injury with no time delay, which may allow for protective measures to be taken to prevent the occurrence of irreversible spinal cord injury.
Collapse
Affiliation(s)
- Mingguang Wang
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Fanguo Meng
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Qimin Song
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Jian Zhang
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Chao Dai
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Qingyan Zhao
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| |
Collapse
|
37
|
Su M, Ren S, Zhong W, Han X. Impact of propofol on renal ischemia/reperfusion endoplasmic reticulum stress. Acta Cir Bras 2017; 32:533-539. [PMID: 28793037 DOI: 10.1590/s0102-865020170070000004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 06/26/2017] [Indexed: 01/22/2023] Open
Abstract
Purpose: To investigate the protective mechanisms of propofol (Pro) on renal ischemia/reperfusion (I/R) injury by studying its impact on renal I/R endoplasmic reticulum stress. Methods: Eighteen male Sprague-Dawley rats (SD rats) were randomly divided into three groups: the I/R group, the Pro pretreatment group, and the control group, and corresponding treatments were performed. The levels of serum creatinine (Cr) and blood urea nitrogen (BUN) of each group were detected. The expression levels of CCAAT-enhancer-binding protein (C/EBP) homology protein (CHOP) and caspase-12 protein within renal tissue samples were detected by western blot. Results: The periodic acid-Schiff (PAS) staining was performed to observe the morphological changes within the renal tissues, and the terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) assay was performed to detect the presence of renal apoptosis. The Pro pretreatment significantly reduced the serum Cr and BUN levels, as well as the expressions levels of CHOP and caspase-12 protein inside the kidney of I/R rats, improving renal pathological injury and reducing the I/R-induced renal apoptosis. Conclusion: Propofol could downregulate the expression of stress-apoptotic proteins CHOP and caspase-12 in the endoplasmic reticulum, thus reducing renal I/R injury.
Collapse
Affiliation(s)
- Mengqin Su
- PhD, Department of Anesthesiology, Henan Provincial Chest Hospital, Zhengzhou, China. Conception and design of the study, analysis and interpretation of data, manuscript writing, critical revision
| | - Sueng Ren
- PhD, Department of Anesthesiology, Henan Provincial Chest Hospital, Zhengzhou, China. Conception and design of the study, analysis and interpretation of data, manuscript writing, critical revision
| | - Wei Zhong
- PhD, Department of Anesthesiology, Henan Provincial Chest Hospital, Zhengzhou, China. Conception and design of the study, analysis and interpretation of data, manuscript writing, critical revision
| | - Xueping Han
- Professor, Department of Anesthesiology, First Affiliated Hospital, Zhengzhou University; Institute of Clinical Medical Research, Henan Universities, Zhengzhou, China. Conception, design and intellectual content of the study, supervised all phases of the study
| |
Collapse
|
38
|
Liu F, Chen MR, Liu J, Zou Y, Wang TY, Zuo YX, Wang TH. Propofol administration improves neurological function associated with inhibition of pro-inflammatory cytokines in adult rats after traumatic brain injury. Neuropeptides 2016; 58:1-6. [PMID: 27045803 DOI: 10.1016/j.npep.2016.03.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 12/22/2015] [Accepted: 03/21/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND Neurological deficits following traumatic brain injury (TBI) result in dramatic impacts on the survivors, but the effect of propofol and associated mechanism are waiting to be determined. METHODS Adult male Sprague-Dawley rats were randomly assigned into Sham, TBI, TBI+Intralipid and TBI+Propofol group. Modified Feeney method was adopted to generate TBI model from free hammer fall injury, and animals in TBI+Propofol group were immediately treated with propofol administration for 2hours after TBI, rats after TBI without propofol treatment was used as injury control, intralipid as vehicle in propofol was injected in TBI+intralipid group. Then, neurological severity scores (NSS) were evaluated at 1, 3, 7 and 14days. Moreover, the expressions of IL-1β, IL-6 and TNF-α mRNA and protein were examined using quantitative real time-polymerase chain reaction and Western blot, immunohistochemical staining was used to localize cytokines. RESULTS The NSS increased greatly in the rats induced by TBI, while propofol could effectively decreased NSS, confirming the neuroprotective effect of propofol. Moreover, the mRNA expressions of IL-1β, IL-6 and TNF-α, at 1, 3, 7days after operation (dpo), were significantly augmented in the injured cortex, compared with sham one. But there was no difference between TBI and TBI+Intralipid group, but markedly decreased after propofol treatment. Additionally, the protein level of IL-1β, IL-6 and TNF-α in four groups determined by Western blot and immunohistochemistry showed the similar change with mRNA expression. CONCLUSION Propofol treatment could elicit a robust neuroprotective response, resulting in significant neurological function improvement for TBI rats, which was independent with intralipid. The underlying molecular mechanism may be partially associated with an inhibition of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Fei Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Mei-Rong Chen
- Institute of Neuroscience, Kunming Medical University, Kunming 650031, China
| | - Jia Liu
- Institute of Neuroscience, Kunming Medical University, Kunming 650031, China
| | - Yu Zou
- Institute of Neuroscience, Kunming Medical University, Kunming 650031, China
| | - Ting-Yong Wang
- Institute of Neuroscience, Kunming Medical University, Kunming 650031, China
| | - Yun-Xia Zuo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Ting-Hua Wang
- Institute of Neuroscience, Kunming Medical University, Kunming 650031, China; Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
39
|
Yuan J, Cui G, Li W, Zhang X, Wang X, Zheng H, Zhang J, Xiang S, Xie Z. Propofol Enhances Hemoglobin-Induced Cytotoxicity in Neurons. Anesth Analg 2016; 122:1024-30. [PMID: 26771264 DOI: 10.1213/ane.0000000000001123] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND It has been increasingly suggested that propofol protects against hypoxic-/ischemic-induced neuronal injury. As evidenced by hemorrhage-induced stroke, hemorrhage into the brain may also cause brain damage. Whether propofol protects against hemorrhage-induced brain damage remains unknown. Therefore, in this study, we investigated the effects of propofol on hemoglobin-induced cytotoxicity in cultured mouse cortical neurons. METHODS Neurons were prepared from the cortex of embryonic 15-day-old mice. Hemoglobin was used to induce cytotoxicity in the neurons. The neurons were then treated with propofol for 4 hours. Cytotoxicity was determined by lactate dehydrogenase release assay. Caspase-3 activation was examined by Western blot analysis. Finally, the free radical scavenger U83836E was used to examine the potential involvement of oxidative stress in propofol's effects on hemoglobin-induced cytotoxicity. RESULTS We found that treatment with hemoglobin induced cytotoxicity in the neurons. Propofol enhanced hemoglobin-induced cytotoxicity. Specifically, there was a significant difference in the amount of lactate dehydrogenase release between hemoglobin plus saline (19.84% ± 5.38%) and hemoglobin plus propofol (35.79% ± 4.41%) in mouse cortical neurons (P = 0.00058, Wilcoxon Mann-Whitney U test, n = 8 in the control group or the treatment group). U83836E did not attenuate the enhancing effects of propofol on hemoglobin-induced cytotoxicity in the neurons, and propofol did not significantly affect caspase-3 activation induced by hemoglobin. These data suggested that caspase-3 activation and oxidative stress might not be the underlying mechanisms by which propofol enhanced hemoglobin-induced cytotoxicity. Moreover, these data suggested that the neuroprotective effects of propofol would be dependent on the condition of the brain injury, which will need to be confirmed in future studies. CONCLUSIONS These results from our current proof-of-concept study should promote more research in vitro and in vivo to develop better anesthesia care for patients with hemorrhagic stroke.
Collapse
Affiliation(s)
- Jing Yuan
- From the *Key Laboratory of Protein Biochemistry and Developmental Biology of State Education Ministry, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China; †Neuroprotection Research Laboratory, Massachusetts General Hospital, Charlestown, Massachusetts; Departments of ‡Radiology and §Neurology, Massachusetts General Hospital, Charlestown, Massachusetts; ‖Program in Neuroscience, Harvard Medical School, Boston, Massachusetts; ¶Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China; #Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and **Massachusetts General Hospital Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ferreira EDO, Fernandes MYSD, Lima NMRD, Neves KRT, Carmo MRSD, Lima FAV, Fonteles AA, Menezes APF, Andrade GMD. Neuroinflammatory response to experimental stroke is inhibited by eriodictyol. Behav Brain Res 2016; 312:321-32. [PMID: 27353856 DOI: 10.1016/j.bbr.2016.06.046] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 06/21/2016] [Accepted: 06/24/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND Cerebral ischemia is a common disease and one of the most common causes of death and disability worldwide. The lack of glucose and oxygen in neuronal tissue leads to a series of inflammatory events, culminating in neuronal death. Eriodictyol is a flavonoid isolated from the Chinese herb Dracocephalum rupestre that has been proven to have anti-inflammatory properties. HYPOTHESIS/PURPOSE Thus, the present study was designed to explore whether eriodictyol has neuroprotective effects against the neuronal damage, motor and memory deficits induced by permanent middle cerebral artery occlusion (pMCAO) in mice. STUDY DESIGN Animals were orally treated with eriodictyol (1, 2 and 4mg/kg) or vehicle (saline) 30min before pMCAO, 2h after, and then once daily for the following five days. METHODS The parameters studied were neuronal viability, brain infarcted area; sensorimotor deficits; exploratory activity; working and aversive memory; myeloperoxidase (MPO) activity; TNFα, iNOS and GFAP immunoreactivity. RESULTS The treatment with eriodictyol prevented neuronal death, reduced infarct area and improved neurological and memory deficits induced by brain ischemia. The increase of MPO activity and TNF-α, iNOS and GFAP expression were also reduced by eriodictyol treatment. CONCLUSION These findings demonstrate that eriodictyol exhibit promising neuroprotection effects against the permanent focal ischemia cerebral injury in the mice experimental model and the underlying mechanisms might be mediated through inhibition of neuroinflammation.
Collapse
Affiliation(s)
- Emerson de Oliveira Ferreira
- Post-Graduate Programme in Medical Sciences, Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Brazil.
| | - Mara Yone Soares Dias Fernandes
- Post-Graduate Programme in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Brazil.
| | - Neila Maria Rocha de Lima
- Post-Graduate Programme in Medical Sciences, Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Brazil.
| | - Kelly Rose Tavares Neves
- Post-Graduate Programme in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Brazil.
| | - Marta Regina Santos do Carmo
- Post-Graduate Programme in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Brazil.
| | - Francisco Arnaldo Viana Lima
- Post-Graduate Programme in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Brazil.
| | - Analu Aragão Fonteles
- Post-Graduate Programme in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Brazil.
| | - Ana Paula Fontenele Menezes
- Post-Graduate Programme in Medical Sciences, Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Brazil.
| | - Geanne Matos de Andrade
- Post-Graduate Programme in Medical Sciences, Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Brazil; Post-Graduate Programme in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Brazil; Institute of Biomedicine of Brazilian Semi-arid, Brazil.
| |
Collapse
|
41
|
Wong JK, Nikravan S, Maxwell BG, Marques MA, Pearl RG. Nocturnal Low-Dose Propofol Infusion for the Management of ICU Delirium: A Case Series in Nonintubated Cardiac Surgery Patients. J Cardiothorac Vasc Anesth 2016; 30:1340-3. [PMID: 27423473 DOI: 10.1053/j.jvca.2016.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Jim K Wong
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA.
| | - Sara Nikravan
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA
| | - Bryan G Maxwell
- Department of Anesthesiology and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Michael A Marques
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA
| | - Ronald G Pearl
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
42
|
Angiotensin II-induced mouse hippocampal neuronal HT22 cell apoptosis was inhibited by propofol: Role of neuronal nitric oxide synthase and metallothinonein-3. Neuroscience 2015; 305:117-27. [DOI: 10.1016/j.neuroscience.2015.07.076] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 07/19/2015] [Accepted: 07/28/2015] [Indexed: 01/26/2023]
|
43
|
Shi SS, Zhang HB, Wang CH, Yang WZ, Liang RS, Chen Y, Tu XK. Propofol Attenuates Early Brain Injury After Subarachnoid Hemorrhage in Rats. J Mol Neurosci 2015; 57:538-45. [PMID: 26342279 DOI: 10.1007/s12031-015-0634-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 07/28/2015] [Indexed: 02/01/2023]
Abstract
Our previous studies demonstrated that propofol protects rat brain against focal cerebral ischemia. However, whether propofol attenuates early brain injury after subarachnoid hemorrhage in rats remains unknown until now. The present study was performed to evaluate the effect of propofol on early brain injury after subarachnoid hemorrhage in rats and further explore the potential mechanisms. Sprague-Dawley rats underwent subarachnoid hemorrhage (SAH) by endovascular perforation then received treatment with propofol (10 or 50 mg/kg) or vehicle after 2 and 12 h of SAH. SAH grading, neurological scores, brain water content, Evans blue extravasation, the myeloperoxidase activity, and malondialdehyde (MDA) content were measured 24 h after SAH. Expression of nuclear factor erythroid-related factor 2 (Nrf2), nuclear factor-kappa B (NF-κB) p65, and aquaporin 4 (AQP4) expression in rat brain were detected by Western blot. Expression of cyclooxygenase-2 (COX-2) and matrix metalloproteinase-9 (MMP-9) were determined by reverse transcription-polymerase chain reaction (RT-PCR). Expressions of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) were assessed by ELISA. Neurological scores, brain water content, Evans blue extravasation, the myeloperoxidase activity, and MDA content were significantly reduced by propofol. Furthermore, expression of Nrf2 in rat brain was upregulated by propofol, and expression of NF-κB p65, AQP4, COX-2, MMP-9, TNF-α, and IL-1β in rat brain were attenuated by propofol. Our results demonstrated that propofol improves neurological scores, reduces brain edema, blood-brain barrier (BBB) permeability, inflammatory reaction, and lipid peroxidation in rats of SAH. Propofol exerts neuroprotection against SAH-induced early brain injury, which might be associated with the inhibition of inflammation and lipid peroxidation.
Collapse
Affiliation(s)
- Song-sheng Shi
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Hua-bin Zhang
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Chun-hua Wang
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Wei-zhong Yang
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Ri-sheng Liang
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Ye Chen
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Xian-kun Tu
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China.
| |
Collapse
|
44
|
Tian Y, Guo S, Guo Y, Jian L. Anesthetic Propofol Attenuates Apoptosis, Aβ Accumulation, and Inflammation Induced by Sevoflurane Through NF-κB Pathway in Human Neuroglioma Cells. Cell Mol Neurobiol 2015; 35:891-8. [PMID: 25809614 PMCID: PMC11486239 DOI: 10.1007/s10571-015-0184-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 03/17/2015] [Indexed: 01/22/2023]
Abstract
Anesthetics have been reported to promote Alzheimer's disease neuropathogenesis by inducing amyloid beta (Aβ) protein accumulation and apoptosis. The aim of this study was to evaluate the effect of propofol on the apoptosis, Aβ accumulation, and inflammation induced by sevoflurane in human neuroglioma cells. Human neuroglioma cells were treated with or without sevoflurane and then co-incubated with or without propofol. Cell apoptosis was evaluated by fluorescence-activated cell sorting analysis (FACS) using AV-PI kits, and data showed that apoptosis induced by sevoflurane was significantly attenuated by propofol treatment. In addition, with the reactive oxygen species (ROS) production measured by FACS after staining with dichloro-dihydrofluorescein diacetate, propofol could significantly reduce the production of ROS as well as the accumulation of Aβ induced by sevoflurane assessed by enzyme-linked immuno sorbent assay (ELISA) analysis. On the other hand, the same treatment decreased the inflammation factor production of interleukin-6. Moreover, the level of nuclear factor-kappa B (NF-κB) was tested by Western blot and immunofluorescence assay. We found that the activation of NF-κB pathway was suppressed by propofol. The results suggest that propofol can effectively attenuate the apoptosis, Aβ accumulation, and inflammation induced by sevoflurane in human neuroglioma cells through NF-κB signal pathway.
Collapse
Affiliation(s)
- Yue Tian
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004 People’s Republic of China
| | - Shanbin Guo
- Department of Pharmacy, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004 People’s Republic of China
| | - Yao Guo
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004 People’s Republic of China
| | - Lingyan Jian
- Department of Pharmacy, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004 People’s Republic of China
| |
Collapse
|
45
|
|
46
|
Tian KY, Liu XJ, Xu JD, Deng LJ, Wang G. Propofol inhibits burn injury-induced hyperpermeability through an apoptotic signal pathway in microvascular endothelial cells. ACTA ACUST UNITED AC 2015; 48:401-7. [PMID: 25760023 PMCID: PMC4445662 DOI: 10.1590/1414-431x20144107] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 11/11/2014] [Indexed: 01/10/2023]
Abstract
Recent studies have revealed that an intrinsic apoptotic signaling cascade is involved in vascular hyperpermeability and endothelial barrier dysfunction. Propofol (2,6-diisopropylphenol) has also been reported to inhibit apoptotic signaling by regulating mitochondrial permeability transition pore (mPTP) opening and caspase-3 activation. Here, we investigated whether propofol could alleviate burn serum-induced endothelial hyperpermeability through the inhibition of the intrinsic apoptotic signaling cascade. Rat lung microvascular endothelial cells (RLMVECs) were pretreated with propofol at various concentrations, followed by stimulation with burn serum, obtained from burn-injury rats. Monolayer permeability was determined by transendothelial electrical resistance. Mitochondrial release of cytochrome C was measured by ELISA. Bax and Bcl-2 expression and mitochondrial release of second mitochondrial-derived activator of caspases (smac) were detected by Western blotting. Caspase-3 activity was assessed by fluorometric assay; mitochondrial membrane potential (Δψm) was determined with JC-1 (a potential-sensitive fluorescent dye). Intracellular ATP content was assayed using a commercial kit, and reactive oxygen species (ROS) were measured by dichlorodihydrofluorescein diacetate (DCFH-DA). Burn serum significantly increased monolayer permeability (P<0.05), and this effect could be inhibited by propofol (P<0.05). Compared with a sham treatment group, intrinsic apoptotic signaling activation - indicated by Bax overexpression, Bcl-2 downregulation, Δψm reduction, decreased intracellular ATP level, increased cytosolic cytochrome C and smac, and caspase-3 activation - was observed in the vehicle group. Propofol not only attenuated these alterations (P<0.05 for all), but also significantly decreased burn-induced ROS production (P<0.05). Propofol attenuated burn-induced RLMVEC monolayer hyperpermeability by regulating the intrinsic apoptotic signaling pathway.
Collapse
Affiliation(s)
- K Y Tian
- Department of Anesthesiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - X J Liu
- Department of Anesthesia, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - J D Xu
- Department of Anesthesiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - L J Deng
- Department of Anesthesiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - G Wang
- Department of Anesthesiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
47
|
Abstract
It has been determined that there is extensive communication between the immune system and the central nervous system (CNS). Proinflammatory cytokines play a key role in this communication. There is an emerging realization that glia and microglia, in particular, (which are the brain’s resident macrophages), are an important source of inflammatory mediators and may have fundamental roles in CNS disorders. Microglia respond also to proinflammatory signals released from other non-neuronal cells, principally those of immune origin, such as mast cells. Mast cells reside in the CNS and are capable of migrating across the blood-brain barrier (BBB) in situations where the barrier is compromised as a result of CNS pathology. Mast cells are both sensors and effectors in communication among nervous, vascular, and immune systems. In the brain, they reside on the brain side of the BBB, and interact with astrocytes, microglia, and blood vessels via their neuroactive stored and newly synthesized chemicals. They are first responders, acting as catalysts and recruiters to initiate, amplify, and prolong other immune and nervous responses upon activation. Mast cells both promote deleterious outcomes in brain function and contribute to normative behavioral functioning, particularly cognition and emotion. Mast cells may play a key role in treating systemic inflammation or blockade of signaling pathways from the periphery to the brain.
Collapse
Affiliation(s)
- Hongquan Dong
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Xiang Zhang
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Yanning Qian
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| |
Collapse
|
48
|
Saad MA, Abdelsalam RM, Kenawy SA, Attia AS. Montelukast, a cysteinyl leukotriene receptor-1 antagonist protects against hippocampal injury induced by transient global cerebral ischemia and reperfusion in rats. Neurochem Res 2014; 40:139-50. [PMID: 25403620 DOI: 10.1007/s11064-014-1478-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Revised: 10/11/2014] [Accepted: 11/11/2014] [Indexed: 12/12/2022]
Abstract
Cysteinyl leukotrienes (CysLTs) are potent pro-inflammatory and immune modulating lipid mediators involved in inflammatory diseases and were boosted in human brain after acute phase of cerebral ischemia. The antagonism of CysLTs receptors may offer protection against ischemic damage. Therefore it seemed interesting to study the possible neuroprotective effect of Montelukast, a CysLTR1 antagonist in global cerebral ischemia/reperfusion (IR) injury in rats. Global cerebral ischemia-reperfusion was induced by bilateral carotid artery occlusion for 15 min followed by 60 min reperfusion period. Animals were randomly allocated into three groups (n = 30 per group): Sham operated, I/R control and rats treated with montelukast (0.5 mg/kg, po) daily for 7 days then I/R was induced 1 h after the last dose of montelukast. After reperfusion rats were killed by decapitation, brains were removed and both hippocampi separated and the following biochemical parameters were estimated; lactate dehydrogenase activity, oxidative stress markers (lipid peroxides, nitric oxide and reduced glutathione), inflammatory markers (myeloperoxidase, tumor necrosis factor-alpha, nuclear factor kappa-B, interleukin-6 and interleukin-10), apoptotic biomarkers (caspase 3 and cytochrome C), neurotransmitters (glutamate, gamma aminobutyric acid), Cys-LTs contents and CysLT1 receptor expression; as well as total brain infarct size and histopathological examination of the hippocampus were assessed. Montelukast protected hippocampal tissue by reducing oxidative stress, inflammatory and apoptotic markers. Furthermore, it reduced glutamate and lactate dehydrogenase activity as well as infarct size elevated by I/R. These results were consistent with the histopathological findings. Montelukast showed a neuroprotective effects through antioxidant, anti-inflammatory and antiapoptotic mechanisms.
Collapse
Affiliation(s)
- M A Saad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt,
| | | | | | | |
Collapse
|