1
|
Yadav RK, Johnson AO, Peeples ES. The dynamic duo: Decoding the roles of hypoxia-inducible factors in neonatal hypoxic-ischemic brain injury. Exp Neurol 2025; 386:115170. [PMID: 39884332 DOI: 10.1016/j.expneurol.2025.115170] [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: 09/21/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) results in considerable mortality and neurodevelopmental disability, with a particularly high disease burden in low- and middle-income countries. Improved understanding of the pathophysiology underlying this injury could allow for improved diagnostic and therapeutic options. Specifically, hypoxia-inducible factors (HIF-1α and HIF-2α) likely play a key role, but that role is complex and remains understudied. This review analyses the recent findings seeking to uncover the impacts of HIF-1α and HIF-2α in neonatal hypoxic-ischemic brain injury (HIBI), focusing on their cell specific expression, time-dependant activities, and potential therapeutic implications. Recent findings have revealed temporal patterns of HIF-1α and HIF-2α expression following hypoxic-ischemic injury, with distinct functions for HIF-1α versus HIF-2α within the neonatal brain. Ongoing studies aimed at further revealing the relationship between HIF isoforms and developing targeted interventions offer promising avenues for therapeutic management which could improve long-term neurological outcomes in affected newborns.
Collapse
Affiliation(s)
- Rajnish Kumar Yadav
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States of America; Child Health Research Institute, Omaha, NE, United States of America
| | - Amanda O Johnson
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States of America; Child Health Research Institute, Omaha, NE, United States of America
| | - Eric S Peeples
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States of America; Child Health Research Institute, Omaha, NE, United States of America; Division of Neonatology, Children's Nebraska, Omaha, NE, United States of America.
| |
Collapse
|
2
|
Amin N, Wu F, Zhao BX, Shi Z, Abdelsadik A, Elshazly Younis A, Naz Abbasi I, Sundus J, Badry Hussein A, Geng Y, Fang M. Hif-1α ablation reduces the efficiency of NeuroD1 gene-based therapy and aggravates the brain damage following ischemic stroke. Expert Opin Drug Deliv 2025; 22:121-138. [PMID: 39632618 DOI: 10.1080/17425247.2024.2435458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 11/16/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION Hypoxia-inducible factor 1α [HIF1α] regulates gene expression, allowing the organism to respond to low oxygen levels. Meanwhile, astrocytes participate in inflammatory processes and are associated with neurotoxic chemicals that can increase stroke volume, contributing considerably to the devastating effects of a stroke. OBJECTIVE To evaluate whether Hif-1α ablation from the central nervous system is implicated in motor dysfunction and ischemic brain damage following stroke. Furthermore, to explore if Hif-1α ablation affects the therapeutic impact of NeuroD1 gene-based therapy. METHODS Endothelin-1 [ET-1] was injected to induce ischemic stroke in mice. Both wild-type and Hypoxia-inducible factor 1α conditional knockout [Hif-1α CKO] mice were used. The effect of Hif-1α ablation was assessed by the neuron numbers, astrocyte activity, vascular endothelial growth factor [VEGF] expression, and behavioral tests. Moreover, western blot, ELISA, and RNA sequencing were used. Then, we used pAAV2/9-GfaABC1D-NeuroD1-P2A-EGFP-WPRE injection to examine the impact of NeuroD1 in Hif-1α CKO mice following ischemic stroke. RESULTS We found that following stroke, motor dysfunction significantly increased in Hif-1α CKO mice. Furthermore, elevation of apoptosis and activation in both microglia and astrocytes were observed, consequently up-regulating neuroinflammation. Meanwhile, Hif-1α ablation significantly decreased the efficiency of NeuroD1 gene-based therapy. CONCLUSION Our findings demonstrate that Hif-1α ablation from the nervous system is implicated in ischemic stroke pathogenesis mainly by increasing neuron cell death and inducing astrocytes as well as decreasing the efficiency of NeuroD1. These data support the idea that manipulating HIF-1α is a viable therapeutic for ischemic stroke.
Collapse
Affiliation(s)
- Nashwa Amin
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Department of Neurology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Centre for Child Health, Hangzhou, China
- Department of Zoology, Faculty of Science, Aswan University, Aswan, Egypt
| | - Fei Wu
- Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing-Xin Zhao
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zongjie Shi
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ahmed Abdelsadik
- Department of Zoology, Faculty of Science, Aswan University, Aswan, Egypt
- Laboratory of Immunometabolism, Aswan University, Aswan, Egypt
| | | | - Irum Naz Abbasi
- Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Javaria Sundus
- Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Azhar Badry Hussein
- Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Geng
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Marong Fang
- Department of Neurology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Centre for Child Health, Hangzhou, China
- Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
3
|
Kawa H, Ahmed Z, Majid A, Chen R. Inhibition of matrix metalloproteinases to reduce blood brain barrier disruption and haemorrhagic transformation in ischaemic stroke: Go broad or go narrow? Neuropharmacology 2025; 262:110192. [PMID: 39419277 DOI: 10.1016/j.neuropharm.2024.110192] [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: 07/12/2024] [Revised: 09/19/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
Ischaemic stroke characterises impulsive cerebral-region hypoxia due to deep intracerebral arteriole blockage, often accompanied by permanent cerebral infarction and cognitive impairment. Thrombolysis with recombinant tissue plasminogen activator (rtPA) and thrombectomy remain the only guidance-approved therapies. However, emerging data draws clear links between such therapies and haemorrhage transformation, which occur when cerebral vasculature is damaged during ischaemia/reperfusion. Studies have shown that matrix metalloproteinases (MMPs) play a significant role in haemorrhage transformation, by depleting the extracellular matrix (ECM) and disrupting the blood brain barrier (BBB). Inhibitors of MMPs may be used to prevent ischaemic stroke patients from BBB disruption and haemorrhage transformation, particularly for those receiving rtPA treatment. Preclinical studies found that inhibition of MMPs with agents or in knock out mice, effectively reduced BBB disruption and infarct volume, leading to improved ischaemic stroke outcomes. At present, MMP inhibition is not an approved therapy for stroke patients. There remain concerns about timing, dosing, duration of MMP inhibition and selection of either broad spectrum or specific MMP inhibitors for stroke patients. This review aims to summarize current knowledge on MMP inhibition in ischaemic stroke and explore whether a broad spectrum or a specific MMP inhibitor should be used for ischaemic stroke patient treatment. It is crucial to inhibit MMP activities early and sufficiently to ensure BBB intact during ischaemia and reperfusion, but also to reduce side effects of MMP inhibitors to minimum. Recent advance in stroke therapy by thrombectomy could aid in such treatment with intra-arterially delivery of MMP inhibitors (and/or antioxidants).
Collapse
Affiliation(s)
- Hala Kawa
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, ST5 5BG, UK
| | - Zubair Ahmed
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Arshad Majid
- Division of Neurosciences, School of Medicine and Population Health, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Ruoli Chen
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, ST5 5BG, UK.
| |
Collapse
|
4
|
Xia Y, Zou C, Kang W, Xu T, Shao R, Zeng P, Sun B, Chen J, Qi Y, Wang Z, Lin T, Zhu H, Shen Y, Wang X, Guo S, Cui D. Invasive metastatic tumor-camouflaged ROS responsive nanosystem for targeting therapeutic brain injury after cardiac arrest. Biomaterials 2024; 311:122678. [PMID: 38917705 DOI: 10.1016/j.biomaterials.2024.122678] [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: 12/17/2023] [Revised: 05/28/2024] [Accepted: 06/22/2024] [Indexed: 06/27/2024]
Abstract
Drug transmission through the blood-brain barrier (BBB) is considered an arduous challenge for brain injury treatment following the return of spontaneous circulation after cardiac arrest (CA-ROSC). Inspired by the propensity of melanoma metastasis to the brain, B16F10 cell membranes are camouflaged on 2-methoxyestradiol (2ME2)-loaded reactive oxygen species (ROS)-triggered "Padlock" nanoparticles that are constructed by phenylboronic acid pinacol esters conjugated D-a-tocopheryl polyethylene glycol succinate (TPGS-PBAP). The biomimetic nanoparticles (BM@TP/2ME2) can be internalized, mainly mediated by the mutual recognition and interaction between CD44v6 expressed on B16F10 cell membranes and hyaluronic acid on cerebral vascular endothelial cells, and they responsively release 2ME2 by the oxidative stress microenvironment. Notably, BM@TP/2ME2 can scavenge excessive ROS to reestablish redox balance, reverse neuroinflammation, and restore autophagic flux in damaged neurons, eventually exerting a remarkable neuroprotective effect after CA-ROSC in vitro and in vivo. This biomimetic drug delivery system is a novel and promising strategy for the treatment of cerebral ischemia-reperfusion injury after CA-ROSC.
Collapse
Affiliation(s)
- Yiyang Xia
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China
| | - Chenming Zou
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Weichao Kang
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China
| | - Tianhua Xu
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China
| | - Rongjiao Shao
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, PR China
| | - Ping Zeng
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Bixi Sun
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Jie Chen
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China
| | - Yiming Qi
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Zhaozhong Wang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Tiancheng Lin
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China
| | - Haichao Zhu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Yuanyuan Shen
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Xintao Wang
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China.
| | - Shengrong Guo
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China.
| | - Derong Cui
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China.
| |
Collapse
|
5
|
Gowtham A, Chauhan C, Rahi V, Kaundal RK. An update on the role of ferroptosis in ischemic stroke: from molecular pathways to Neuroprotection. Expert Opin Ther Targets 2024; 28:1149-1175. [PMID: 39710973 DOI: 10.1080/14728222.2024.2446319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/29/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION Ischemic stroke (IS), a major cause of mortality and disability worldwide, remains a significant healthcare challenge due to limited therapeutic options. Ferroptosis, a distinct iron-dependent form of regulated cell death characterized by lipid peroxidation and oxidative stress, has emerged as a crucial mechanism in IS pathophysiology. This review explores the role of ferroptosis in IS and its potential for driving innovative therapeutic strategies. AREA COVERED This review delves into the practical implications of ferroptosis in IS, focusing on molecular mechanisms like lipid peroxidation, iron accumulation, and their interplay with inflammation, reactive oxygen species (ROS), and the Nrf2-ARE antioxidant system. It highlights ferroptotic proteins, small-molecule inhibitors, and non-coding RNA modulators as emerging therapeutic targets to mitigate neuroinflammation and neuronal cell death. Studies from PubMed (1982-2024) were identified using MeSH terms such as 'Ferroptosis' and 'Ischemic Stroke,' and only rigorously screened articles were included. EXPERT OPINION Despite preclinical evidence supporting the neuroprotective effects of ferroptosis inhibitors, clinical translation faces hurdles such as suboptimal pharmacokinetics and safety concerns. Advances in drug delivery systems, bioinformatics, and AI-driven drug discovery may optimize ferroptosis-targeting strategies, develop biomarkers, and improve therapeutic outcomes for IS patients.
Collapse
Affiliation(s)
- A Gowtham
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Lucknow, India
| | - Chandan Chauhan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Lucknow, India
| | - Vikrant Rahi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Lucknow, India
| | - Ravinder K Kaundal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Lucknow, India
| |
Collapse
|
6
|
Delgado-Martín S, Martínez-Ruiz A. The role of ferroptosis as a regulator of oxidative stress in the pathogenesis of ischemic stroke. FEBS Lett 2024; 598:2160-2173. [PMID: 38676284 DOI: 10.1002/1873-3468.14894] [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: 11/10/2023] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024]
Abstract
Ferroptosis is a unique form of cell death that was first described in 2012 and plays a significant role in various diseases, including neurodegenerative conditions. It depends on a dysregulation of cellular iron metabolism, which increases free, redox-active, iron that can trigger Fenton reactions, generating hydroxyl radicals that damage cells through oxidative stress and lipid peroxidation. Lipid peroxides, resulting mainly from unsaturated fatty acids, damage cells by disrupting membrane integrity and propagating cell death signals. Moreover, lipid peroxide degradation products can further affect cellular components such as DNA, proteins, and amines. In ischemic stroke, where blood flow to the brain is restricted, there is increased iron absorption, oxidative stress, and compromised blood-brain barrier integrity. Imbalances in iron-transport and -storage proteins increase lipid oxidation and contribute to neuronal damage, thus pointing to the possibility of brain cells, especially neurons, dying from ferroptosis. Here, we review the evidence showing a role of ferroptosis in ischemic stroke, both in recent studies directly assessing this type of cell death, as well as in previous studies showing evidence that can now be revisited with our new knowledge on ferroptosis mechanisms. We also review the efforts made to target ferroptosis in ischemic stroke as a possible treatment to mitigate cellular damage and death.
Collapse
Affiliation(s)
- Susana Delgado-Martín
- Unidad de Investigación, Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Antonio Martínez-Ruiz
- Unidad de Investigación, Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Spain
| |
Collapse
|
7
|
Xie J, Zhang Z. Recent Advances and Therapeutic Implications of 2-Oxoglutarate-Dependent Dioxygenases in Ischemic Stroke. Mol Neurobiol 2024; 61:3949-3975. [PMID: 38041714 DOI: 10.1007/s12035-023-03790-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/08/2023] [Indexed: 12/03/2023]
Abstract
Ischemic stroke is a common disease with a high disability rate and mortality, which brings heavy pressure on families and medical insurance. Nowadays, the golden treatments for ischemic stroke in the acute phase mainly include endovascular therapy and intravenous thrombolysis. Some drugs are used to alleviate brain injury in patients with ischemic stroke, such as edaravone and 3-n-butylphthalide. However, no effective neuroprotective drug for ischemic stroke has been acknowledged. 2-Oxoglutarate-dependent dioxygenases (2OGDDs) are conserved and common dioxygenases whose activities depend on O2, Fe2+, and 2OG. Most 2OGDDs are expressed in the brain and are essential for the development and functions of the brain. Therefore, 2OGDDs likely play essential roles in ischemic brain injury. In this review, we briefly elucidate the functions of most 2OGDDs, particularly the effects of regulations of 2OGDDs on various cells in different phases after ischemic stroke. It would also provide promising potential therapeutic targets and directions of drug development for protecting the brain against ischemic injury and improving outcomes of ischemic stroke.
Collapse
Affiliation(s)
- Jian Xie
- Department of Neurology, Affiliated Zhongda Hospital, Research Institution of Neuropsychiatry, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Zhijun Zhang
- Department of Neurology, Affiliated Zhongda Hospital, Research Institution of Neuropsychiatry, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
8
|
Peng L, Li K, Li D, Zuo X, Zhan L, Chen M, Gong M, Sun W, Xu E. The p75 neurotrophin receptor attenuates secondary thalamic damage after cortical infarction by promoting angiogenesis. CNS Neurosci Ther 2024; 30:e14875. [PMID: 39072998 DOI: 10.1111/cns.14875] [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: 03/10/2024] [Revised: 06/23/2024] [Accepted: 07/05/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Angiogenesis is crucial in neuroprotection of secondary thalamic injury after cortical infarction. The p75 neurotrophin receptor (p75NTR) plays a key role in activating angiogenesis. However, the effects of p75NTR on angiogenesis in the thalamus after cortical infarction are largely unknown. Herein we investigate whether p75NTR facilitates angiogenesis to attenuate secondary thalamic damage via activating hypoxia-inducible factor 1α (HIF-1α)/vascular endothelial growth factor (VEGF) pathway mediated by Von Hippel-Lindau (VHL) after distal middle cerebral artery occlusion (dMCAO). METHODS The male rat model of dMCAO was established. The effects of p75NTR on the angiogenesis was evaluated using RNA-sequencing, immunohistochemistry, western blot, quantitative real-time polymerase chain reaction, magnetic resonance imaging, behavior tests, viral and pharmacological interventions. RESULTS We found that the p75NTR and vessel density were decreased in ipsilateral thalamus after dMCAO. The p75NTR-VHL interaction was reduced, which promoted the ubiquitination degradation of HIF-1α and reduced VEGF expression after dMCAO. Notably, p75NTR overexpression restrained the ubiquitination degradation of HIF-1α by inhibiting VHL-HIF-1α interaction, further promoted angiogenesis, increased cerebral blood flow of ipsilateral thalamus and improved neurological function after dMCAO. CONCLUSION For the first time, we highlighted that the enhancement of p75NTR-VHL interaction promoted angiogenesis in attenuating secondary thalamic damage after dMCAO.
Collapse
Affiliation(s)
- Linhui Peng
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Kongping Li
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Neurology, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dan Li
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xialin Zuo
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lixuan Zhan
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Meiyan Chen
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ming Gong
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Weiwen Sun
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - En Xu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Amin N, Abbasi IN, Wu F, Shi Z, Sundus J, Badry A, Yuan X, Zhao BX, Pan J, Mi XD, Luo Y, Geng Y, Fang M. The Janus face of HIF-1α in ischemic stroke and the possible associated pathways. Neurochem Int 2024; 177:105747. [PMID: 38657682 DOI: 10.1016/j.neuint.2024.105747] [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: 10/30/2023] [Revised: 03/01/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
Stroke is the most devastating disease, causing paralysis and eventually death. Many clinical and experimental trials have been done in search of a new safe and efficient medicine; nevertheless, scientists have yet to discover successful remedies that are also free of adverse effects. This is owing to the variability in intensity, localization, medication routes, and each patient's immune system reaction. HIF-1α represents the modern tool employed to treat stroke diseases due to its functions: downstream genes such as glucose metabolism, angiogenesis, erythropoiesis, and cell survival. Its role can be achieved via two downstream EPO and VEGF strongly related to apoptosis and antioxidant processes. Recently, scientists paid more attention to drugs dealing with the HIF-1 pathway. This review focuses on medicines used for ischemia treatment and their potential HIF-1α pathways. Furthermore, we discussed the interaction between HIF-1α and other biological pathways such as oxidative stress; however, a spotlight has been focused on certain potential signalling contributed to the HIF-1α pathway. HIF-1α is an essential regulator of oxygen balance within cells which affects and controls the expression of thousands of genes related to sustaining homeostasis as oxygen levels fluctuate. HIF-1α's role in ischemic stroke strongly depends on the duration and severity of brain damage after onset. HIF-1α remains difficult to investigate, particularly in ischemic stroke, due to alterations in the acute and chronic phases of the disease, as well as discrepancies between the penumbra and ischemic core. This review emphasizes these contrasts and analyzes the future of this intriguing and demanding field.
Collapse
Affiliation(s)
- Nashwa Amin
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Department of Zoology, Faculty of Science, Aswan University, Egypt; Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Irum Naz Abbasi
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Wu
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zongjie Shi
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Javaria Sundus
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Azhar Badry
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xia Yuan
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing-Xin Zhao
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jie Pan
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xiao-Dan Mi
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuhuan Luo
- Department of Pediatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Geng
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Marong Fang
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China; Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
10
|
Han Y, Ji B, Leng Y, Xie C. Inhibited hypoxia-inducible factor by intraoperative hyperglycemia increased postoperative delirium of aged patients: A review. Medicine (Baltimore) 2024; 103:e38349. [PMID: 39259057 PMCID: PMC11142828 DOI: 10.1097/md.0000000000038349] [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: 10/21/2023] [Revised: 04/24/2024] [Accepted: 05/03/2024] [Indexed: 09/12/2024] Open
Abstract
The underlying mechanism of postoperative delirium (POD) in elderly people remains unclear. Perioperative hyperglycemia (POHG) is an independent risk indicator for POD, particularly in the elderly. Under cerebral desaturation (hypoxia) during general anesthesia, hypoxia-inducible factor (HIF) is neuroprotective during cerebral hypoxia via diverse pathways, like glucose metabolism and angiogenesis. Hyperglycemia can repress HIF expression and activity. On the other hand, POHG occurred among patients undergoing surgery. For surgical stress, hypothalamic-pituitary-adrenal activation and sympathoadrenal activation may increase endogenous glucose production via gluconeogenesis and glycogenolysis. Thus, under the setting of cerebral hypoxia during general anesthesia, we speculate that POHG prevents HIF-1α levels and function in the brain of aged patients, thus exacerbating the hypoxic response of HIF-1 and potentially contributing to POD. This paper sketches the underlying mechanisms of HIF in POD in elderly patients and offers novel insights into targets for preventing or treating POD in the same way as POHG.
Collapse
Affiliation(s)
- Yutong Han
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Bing Ji
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
- Department of Pain Management, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Yulin Leng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Chunguang Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
- Traditional Chinese Medicine Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, Sichuan Province, People’s Republic of China
| |
Collapse
|
11
|
Hirayama Y, Le HPN, Hashimoto H, Ishii I, Koizumi S, Anzai N. Preconditioning-Induced Facilitation of Lactate Release from Astrocytes Is Essential for Brain Ischemic Tolerance. eNeuro 2024; 11:ENEURO.0494-23.2024. [PMID: 38604775 PMCID: PMC11064122 DOI: 10.1523/eneuro.0494-23.2024] [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: 11/26/2023] [Revised: 03/27/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024] Open
Abstract
A sublethal ischemic episode [termed preconditioning (PC)] protects neurons in the brain against a subsequent severe ischemic injury. This phenomenon is known as brain ischemic tolerance and has received much attention from researchers because of its robust neuroprotective effects. We have previously reported that PC activates astrocytes and subsequently upregulates P2X7 receptors, thereby leading to ischemic tolerance. However, the downstream signals of P2X7 receptors that are responsible for PC-induced ischemic tolerance remain unknown. Here, we show that PC-induced P2X7 receptor-mediated lactate release from astrocytes has an indispensable role in this event. Using a transient focal cerebral ischemia model caused by middle cerebral artery occlusion, extracellular lactate levels during severe ischemia were significantly increased in mice who experienced PC; this increase was dependent on P2X7 receptors. In addition, the intracerebroventricular injection of lactate protected against cerebral ischemic injury. In in vitro experiments, although stimulation of astrocytes with the P2X7 receptor agonist BzATP had no effect on the protein levels of monocarboxylate transporter (MCT) 1 and MCT4 (which are responsible for lactate release from astrocytes), BzATP induced the plasma membrane translocation of these MCTs via their chaperone CD147. Importantly, CD147 was increased in activated astrocytes after PC, and CD147-blocking antibody abolished the PC-induced facilitation of astrocytic lactate release and ischemic tolerance. Taken together, our findings suggest that astrocytes induce ischemic tolerance via P2X7 receptor-mediated lactate release.
Collapse
Affiliation(s)
- Yuri Hirayama
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Ha Pham Ngoc Le
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Hirofumi Hashimoto
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Itsuko Ishii
- Division of Pharmacy, Chiba University Hospital, Chiba 260-8677, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Naohiko Anzai
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| |
Collapse
|
12
|
Xie J, Zhang Y, Li B, Xi W, Wang Y, Li L, Liu C, Shen L, Han B, Kong Y, Yao H, Zhang Z. Inhibition of OGFOD1 by FG4592 confers neuroprotection by activating unfolded protein response and autophagy after ischemic stroke. J Transl Med 2024; 22:248. [PMID: 38454480 PMCID: PMC10921652 DOI: 10.1186/s12967-024-04993-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 02/12/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Acute ischemic stroke is a common neurological disease with a significant financial burden but lacks effective drugs. Hypoxia-inducible factor (HIF) and prolyl hydroxylases (PHDs) participate in the pathophysiological process of ischemia. However, whether FG4592, the first clinically approved PHDs inhibitor, can alleviate ischemic brain injury remains unclear. METHODS The infarct volumes and behaviour tests were first analyzed in mice after ischemic stroke with systemic administration of FG4592. The knockdown of HIF-1α and pretreatments of HIF-1/2α inhibitors were then used to verify whether the neuroprotection of FG4592 is HIF-dependent. The targets predicting and molecular docking methods were applied to find other targets of FG4592. Molecular, cell biological and gene knockdown methods were finally conducted to explore the potential neuroprotective mechanisms of FG4592. RESULTS We found that the systemic administration of FG4592 decreased infarct volume and improved neurological defects of mice after transient or permanent ischemia. Meanwhile, FG4592 also activated autophagy and inhibited apoptosis in peri-infarct tissue of mice brains. However, in vitro and in vivo results suggested that the neuroprotection of FG4592 was not classical HIF-dependent. 2-oxoglutarate and iron-dependent oxygenase domain-containing protein 1 (OGFOD1) was found to be a novel target of FG4592 and regulated the Pro-62 hydroxylation in the small ribosomal protein s23 (Rps23) with the help of target predicting and molecular docking methods. Subsequently, the knockdown of OGFOD1 protected the cell against ischemia/reperfusion injury and activated unfolded protein response (UPR) and autophagy. Moreover, FG4592 was also found to activate UPR and autophagic flux in HIF-1α independent manner. Blocking UPR attenuated the neuroprotection, pro-autophagy effect and anti-apoptosis ability of FG4592. CONCLUSION This study demonstrated that FG4592 could be a candidate drug for treating ischemic stroke. The neuroprotection of FG4592 might be mediated by inhibiting alternative target OGFOD1, which activated the UPR and autophagy and inhibited apoptosis after ischemic injury. The inhibition of OGFOD1 is a novel therapy for ischemic stroke.
Collapse
Affiliation(s)
- Jian Xie
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Institution of Neuropsychiatry, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Yuan Zhang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Bin Li
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Wen Xi
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Yu Wang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Lu Li
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Chenchen Liu
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Ling Shen
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Bing Han
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Yan Kong
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, No. 87 Dingjiaqiao Road, Nanjing, 210009, Jiangsu, China
| | - HongHong Yao
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.
| | - Zhijun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Institution of Neuropsychiatry, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, 210009, Jiangsu, China.
- The Brain Cognition and Brain Disease Institute of Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
13
|
Koukalova L, Chmelova M, Amlerova Z, Vargova L. Out of the core: the impact of focal ischemia in regions beyond the penumbra. Front Cell Neurosci 2024; 18:1336886. [PMID: 38504666 PMCID: PMC10948541 DOI: 10.3389/fncel.2024.1336886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/08/2024] [Indexed: 03/21/2024] Open
Abstract
The changes in the necrotic core and the penumbra following induction of focal ischemia have been the focus of attention for some time. However, evidence shows, that ischemic injury is not confined to the primarily affected structures and may influence the remote areas as well. Yet many studies fail to probe into the structures beyond the penumbra, and possibly do not even find any significant results due to their short-term design, as secondary damage occurs later. This slower reaction can be perceived as a therapeutic opportunity, in contrast to the ischemic core defined as irreversibly damaged tissue, where the window for salvation is comparatively short. The pathologies in remote structures occur relatively frequently and are clearly linked to the post-stroke neurological outcome. In order to develop efficient therapies, a deeper understanding of what exactly happens in the exo-focal regions is necessary. The mechanisms of glia contribution to the ischemic damage in core/penumbra are relatively well described and include impaired ion homeostasis, excessive cell swelling, glutamate excitotoxic mechanism, release of pro-inflammatory cytokines and phagocytosis or damage propagation via astrocytic syncytia. However, little is known about glia involvement in post-ischemic processes in remote areas. In this literature review, we discuss the definitions of the terms "ischemic core", "penumbra" and "remote areas." Furthermore, we present evidence showing the array of structural and functional changes in the more remote regions from the primary site of focal ischemia, with a special focus on glia and the extracellular matrix. The collected information is compared with the processes commonly occurring in the ischemic core or in the penumbra. Moreover, the possible causes of this phenomenon and the approaches for investigation are described, and finally, we evaluate the efficacy of therapies, which have been studied for their anti-ischemic effect in remote areas in recent years.
Collapse
Affiliation(s)
- Ludmila Koukalova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Martina Chmelova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Zuzana Amlerova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Lydia Vargova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
14
|
Ferenczi S, Mogor F, Takacs P, Kovacs T, Toth VE, Varga ZV, Kovács K, Lohinai Z, Vass KC, Nagy N, Dora D. Depletion of muscularis macrophages ameliorates inflammation-driven dysmotility in murine colitis model. Sci Rep 2023; 13:22451. [PMID: 38105266 PMCID: PMC10725888 DOI: 10.1038/s41598-023-50059-7] [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: 06/27/2023] [Accepted: 12/14/2023] [Indexed: 12/19/2023] Open
Abstract
Previously, the presence of a blood-myenteric plexus barrier and its disruption was reported in experimentally induced colitis via a macrophage-dependent process. The aim of this study is to reveal how myenteric barrier disruption and subsequent neuronal injury affects gut motility in vivo in a murine colitis model. We induced colitis with dextran sulfate sodium (DSS), with the co-administration of liposome-encapsulated clodronate (L-clodronate) to simultaneously deplete blood monocytes contributing to macrophage infiltration in the inflamed muscularis of experimental mice. DSS-treated animals receiving concurrent L-clodronate injection showed significantly decreased blood monocyte numbers and colon muscularis macrophage (MM) density compared to DSS-treated control (DSS-vehicle). DSS-clodronate-treated mice exhibited significantly slower whole gut transit time than DSS-vehicle-treated animals and comparable to that of controls. Experiments with oral gavage-fed Evans-blue dye showed similar whole gut transit times in DSS-clodronate-treated mice as in control animals. Furthermore, qPCR-analysis and immunofluorescence on colon muscularis samples revealed that factors associated with neuroinflammation and neurodegeneration, including Bax1, Hdac4, IL-18, Casp8 and Hif1a are overexpressed after DSS-treatment, but not in the case of concurrent L-clodronate administration. Our findings highlight that MM-infiltration in the muscularis layer is responsible for colitis-associated dysmotility and enteric neuronal dysfunction along with the release of mediators associated with neurodegeneration in a murine experimental model.
Collapse
Affiliation(s)
- Szilamér Ferenczi
- Institute of Experimental Medicine, Laboratory of Molecular Neuroendocrinology, Budapest, Hungary
- Institute of Genetics and Biotechnology, Department of Microbiology and Applied Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, Hungary
| | - Fruzsina Mogor
- Department of Anatomy, Histology and Embryology, Semmelweis University, Tuzolto St. 58, Budapest, 1094, Hungary
| | - Peter Takacs
- Department of Anatomy, Histology and Embryology, Semmelweis University, Tuzolto St. 58, Budapest, 1094, Hungary
| | - Tamas Kovacs
- Department of Anatomy, Histology and Embryology, Semmelweis University, Tuzolto St. 58, Budapest, 1094, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Viktoria E Toth
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Krisztina Kovács
- Institute of Experimental Medicine, Laboratory of Molecular Neuroendocrinology, Budapest, Hungary
| | - Zoltan Lohinai
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| | - Koppány Csaba Vass
- Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary
| | - Nandor Nagy
- Department of Anatomy, Histology and Embryology, Semmelweis University, Tuzolto St. 58, Budapest, 1094, Hungary
| | - David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, Tuzolto St. 58, Budapest, 1094, Hungary.
| |
Collapse
|
15
|
Liao W, Wang M, Wu Y, Du J, Li Y, Su A, Zhong L, Xie Z, Gong M, Liang J, Wang P, Liu Z, Wang L. The mechanisms of Huangqi Guizhi Wuwu decoction in treating ischaemic stroke based on network pharmacology and experiment verification. PHARMACEUTICAL BIOLOGY 2023; 61:1014-1029. [PMID: 37410583 DOI: 10.1080/13880209.2023.2230477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 05/31/2023] [Accepted: 06/22/2023] [Indexed: 07/08/2023]
Abstract
CONTEXT Huangqi Guizhi Wuwu Decoction (HGWD) is effective in treating ischaemic stroke (IS). However, its mechanism of action is still unclear. OBJECTIVE Network pharmacology integrated with in vivo experiments were used to clarify the underlying mechanisms of HGWD for treating IS. MATERIALS AND METHODS TCMSP, GeneCards, OMIM and STRING were used to retrieve and construct visual protein interaction networks for the key targets. The AutoDock tool was used for molecular docking between key targets and active compounds. The neuroprotective effect of HGWD were verified in a middle cerebral artery occlusion (MCAO) model rat. The Sprague-Dawley (SD) rats were divided into sham, model, low-dose (5 g/kg, i.g.), high-dose (20 g/kg, i.g.), and nimodipine (20 mg/kg, i.g.) groups once daily for 7 days. The neurological scores, brain infarct volumes, lipid peroxidation, inflammatory cytokines, Nissl bodies, apoptotic neurons, and signalling pathways were all investigated and evaluated in vivo. RESULTS Network pharmacology identified 117 HGWD targets related to IS and 36 candidate compounds. GO and KEGG analyses showed that HGWD anti-IS effects were mainly associated with PI3K-Akt and HIF-1 signalling pathways. HGWD effectively reduced the cerebral infarct volumes (19.19%), the number of apoptotic neurons (16.78%), and the release of inflammatory cytokines, etc. in MCAO rats. Furthermore, HGWD decreased the levels of HIF-1A, VEGFA, Bax, cleaved caspase-3, p-MAPK1, and p-c-Jun while increasing the expression of p-PI3K, p-AKT1, and Bcl-2. DISCUSSION AND CONCLUSION This study initially elucidated the mechanism of HGWD anti-IS, which contributed to the further promotion and secondary development of HGWD in clinical practice.
Collapse
Affiliation(s)
- Weiguo Liao
- College of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Minchun Wang
- College of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ying Wu
- College of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jinyan Du
- College of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yaxin Li
- College of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Anyu Su
- College of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lanying Zhong
- College of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zi Xie
- College of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Mingyu Gong
- College of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Junhui Liang
- College of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Pengcheng Wang
- College of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zai Liu
- Pharmacy Department, Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong, China
| | - Lisheng Wang
- College of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
16
|
Hao H, Hou Y, Li A, Niu L, Li S, He B, Zhang X, Song H, Cai R, Zhou Y, Yao C, Wang Y, Wang Y. HIF-1α promotes astrocytic production of macrophage migration inhibitory factor following spinal cord injury. CNS Neurosci Ther 2023; 29:3802-3814. [PMID: 37334735 PMCID: PMC10651974 DOI: 10.1111/cns.14300] [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: 03/15/2023] [Revised: 05/16/2023] [Accepted: 05/28/2023] [Indexed: 06/20/2023] Open
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF) is an important mediator of neuropathology in various central nervous system (CNS) diseases. However, little is known about its inducers for production from the nerve cells, as well as the underlying regulatory mechanism. Injury-induced HIF-1α has been shown to exacerbate neuroinflammation by activating multiple downstream target molecules. It is postulated that HIF-1α is involved in the regulation of MIF following spinal cord injury (SCI). METHODS SCI model of Sprague-Dawley rats was established by cord contusion at T8-T10. The dynamic changes of HIF-1α and MIF protein levels at lesion site of rat spinal cord were determined by Western blot. The specific cell types of HIF-1α and MIF expression were examined by immunostaining. Primary astrocytes were isolated from the spinal cord, cultured and stimulated with various agonist or inhibitor of HIF-1α for analysis of HIF-1α-mediated expression of MIF. Luciferase report assay was used to determine the relationship between HIF-1α and MIF. The Basso, Beattie, and Bresnahan (BBB) locomotor scale was used to assess the locomotor function following SCI. RESULTS The protein levels of HIF-1α and MIF at lesion site were significantly elevated by SCI. Immunofluorescence demonstrated that both HIF-1α and MIF were abundantly expressed in the astrocytes of the spinal cord. By using various agonists or inhibitors of HIF-1α, it was shown that HIF-1α sufficiently induced astrocytic production of MIF. Mechanistically, HIF-1α promoted MIF expression through interaction with MIF promoter. Inhibition of HIF-1α activity using specific inhibitor markedly reduced the protein levels of MIF at lesion site following SCI, which in turn favored for the functional recovery. CONCLUSION SCI-induced activation of HIF-1α is able to promote MIF production from astrocytes. Our results have provided new clues for SCI-induced production of DAMPs, which may be helpful for clinical treatment of neuroinflammation.
Collapse
Affiliation(s)
- Huifei Hao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Yuxuan Hou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Aicheng Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Li Niu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Shaolan Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Bingqiang He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Xingyuan Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Honghua Song
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Rixin Cai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Yue Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Chun Yao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Yongjun Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Yingjie Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| |
Collapse
|
17
|
Zhang Y, Yu L, Zhou S, He Y, Jin W, Wan H, Yang J. A comparative study of the protective effects of Guhong injection and its component on cerebral ischemia-reperfusion injury based on the oxidation index. Brain Res 2023; 1819:148532. [PMID: 37586676 DOI: 10.1016/j.brainres.2023.148532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/18/2023]
Abstract
Guhong injection (GHI), a compound preparation of Chinese and Western medicine, is composed of safflower water extract and aceglutamide, and has a certain therapeutic effect on cerebral ischemia diseases. In this study, we investigated and compared the protective effects of GHI, Honghua injection (HHI), and aceglutamide (ACG) on cerebral ischemia-reperfusion injury in Sprague-Dawley (SD) rats randomly assigned to the following 5 groups: Sham, MCAO, MCAO + GHI, MCAO + HHI, and MCAO + ACG. The results revealed that GHI, HHI, and ACG improved neurological functions and reduced the infarct volume, the contents of HIF-1α, PKC, and EPO, and the expression of NOX-4 and HIF-1α mRNA. The protein expression of HIF-1α and iNOS treated with GHI, HHI, and ACG was decreased, while that of PHD2 was increased. Meanwhile, the BrdU+/NeuN+ cell counts of SGZ and SVZ areas in the brain tissues of the GHI, HHI, and ACG groups were greater than those of the MCAO rats. Thus, GHI, HHI, and ACG can confer protection against cerebral ischemia-reperfusion injury, possibly through antioxidation. Our research findings may provide evidence for the effectiveness of the combination of traditional Chinese and Western medicine.
Collapse
Affiliation(s)
- Yangyang Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| | - Li Yu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| | - Saiya Zhou
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| | - Weifeng Jin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| | - Haitong Wan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| | - Jiehong Yang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| |
Collapse
|
18
|
Jiang G, Ayaki T, Maki T, Yasuda K, Yoshii D, Kaji S, Takahashi R. Evaluation of BCAS1-positive immature oligodendrocytes after cerebral ischemic stroke and SVD. Neurosci Lett 2023; 812:137405. [PMID: 37479175 DOI: 10.1016/j.neulet.2023.137405] [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: 05/02/2023] [Revised: 07/02/2023] [Accepted: 07/18/2023] [Indexed: 07/23/2023]
Abstract
Ischemic cerebrovascular disease is an important cause of physical disability and dementia. Oligodendrocytes (OLGs), which differentiate from oligodendrocyte precursor cells (OPCs), are crucial for remyelination of the damaged brain and functional recovery. Breast carcinoma amplified sequence 1 (BCAS1) has recently been shown to be highly expressed in newly formed pre-myelinating oligodendrocytes (pre-mOLGs), while its expression level is reduced in mature OLGs. In this study, we analyzed BCAS1 expression by immunohistochemical analysis of human post-mortem brain tissue from six stroke patients (death within 2 months after stroke onset) and eight small vessel disease (SVD) patients. Control post-mortem brain tissue was from eight age-matched patients without any obvious central nervous system (CNS) pathology. The Olig2 expression in the area corresponding to the same section of the BCAS1-stained slice was analyzed to determine the total oligodendrocyte lineage. The percentage of differentiating OPCs in the oligodendrocyte lineage was calculated as the ratio of BCAS1+ to Olig2+ cells (BCAS1+/Olig2+). The stroke and SVD cases showed demyelination with decreased expression of myelin basic protein (MBP, a mature OLG marker). The stroke cases showed significantly increased numbers of early-stage BCAS1+ cells with an immature morphology and Olig2+ cells (pan-oligodendrocyte lineages) in the peri-infarct areas in both the cortex and white matter, but showed no increase in the number of late-stage BCAS1+ cells with a mature morphology. In contrast, the SVD cases showed no significant increase in Olig2+ and BCAS1+ cells. These results indicated that remyelination dysfunction could be attributed to insufficient maturation of OPCs in stroke and impaired recruitment of OPCs in SVD.
Collapse
Affiliation(s)
- Guanhua Jiang
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takashi Ayaki
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Takakuni Maki
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ken Yasuda
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Daisuke Yoshii
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Seiji Kaji
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
19
|
Singh M, Zhao Y, Gastaldi VD, Wojcik SM, Curto Y, Kawaguchi R, Merino RM, Garcia-Agudo LF, Taschenberger H, Brose N, Geschwind D, Nave KA, Ehrenreich H. Erythropoietin re-wires cognition-associated transcriptional networks. Nat Commun 2023; 14:4777. [PMID: 37604818 PMCID: PMC10442354 DOI: 10.1038/s41467-023-40332-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/18/2023] [Indexed: 08/23/2023] Open
Abstract
Recombinant human erythropoietin (rhEPO) has potent procognitive effects, likely hematopoiesis-independent, but underlying mechanisms and physiological role of brain-expressed EPO remained obscure. Here, we provide transcriptional hippocampal profiling of male mice treated with rhEPO. Based on ~108,000 single nuclei, we unmask multiple pyramidal lineages with their comprehensive molecular signatures. By temporal profiling and gene regulatory analysis, we build developmental trajectory of CA1 pyramidal neurons derived from multiple predecessor lineages and elucidate gene regulatory networks underlying their fate determination. With EPO as 'tool', we discover populations of newly differentiating pyramidal neurons, overpopulating to ~200% upon rhEPO with upregulation of genes crucial for neurodifferentiation, dendrite growth, synaptogenesis, memory formation, and cognition. Using a Cre-based approach to visually distinguish pre-existing from newly formed pyramidal neurons for patch-clamp recordings, we learn that rhEPO treatment differentially affects excitatory and inhibitory inputs. Our findings provide mechanistic insight into how EPO modulates neuronal functions and networks.
Collapse
Affiliation(s)
- Manvendra Singh
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany.
| | - Ying Zhao
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Vinicius Daguano Gastaldi
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Sonja M Wojcik
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Yasmina Curto
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Riki Kawaguchi
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ricardo M Merino
- Max Planck Institute for Dynamics and Self-Organization and Campus Institute for Dynamics of Biological Networks, Georg-August-University, Göttingen, Germany
| | | | - Holger Taschenberger
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Daniel Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Hannelore Ehrenreich
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany.
| |
Collapse
|
20
|
Aboouf MA, Thiersch M, Soliz J, Gassmann M, Schneider Gasser EM. The Brain at High Altitude: From Molecular Signaling to Cognitive Performance. Int J Mol Sci 2023; 24:10179. [PMID: 37373327 DOI: 10.3390/ijms241210179] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/13/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
The brain requires over one-fifth of the total body oxygen demand for normal functioning. At high altitude (HA), the lower atmospheric oxygen pressure inevitably challenges the brain, affecting voluntary spatial attention, cognitive processing, and attention speed after short-term, long-term, or lifespan exposure. Molecular responses to HA are controlled mainly by hypoxia-inducible factors. This review aims to summarize the cellular, metabolic, and functional alterations in the brain at HA with a focus on the role of hypoxia-inducible factors in controlling the hypoxic ventilatory response, neuronal survival, metabolism, neurogenesis, synaptogenesis, and plasticity.
Collapse
Affiliation(s)
- Mostafa A Aboouf
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, 8057 Zurich, Switzerland
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| | - Markus Thiersch
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, 8057 Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| | - Jorge Soliz
- Institute Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Faculty of Medicine, Université Laval, Québec, QC G1V 4G5, Canada
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, 8057 Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| | - Edith M Schneider Gasser
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, 8057 Zurich, Switzerland
- Institute Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Faculty of Medicine, Université Laval, Québec, QC G1V 4G5, Canada
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
21
|
Huang T, Fakurazi S, Cheah PS, Ling KH. REST Targets JAK-STAT and HIF-1 Signaling Pathways in Human Down Syndrome Brain and Neural Cells. Int J Mol Sci 2023; 24:9980. [PMID: 37373133 DOI: 10.3390/ijms24129980] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Down syndrome (DS) is the most frequently diagnosed chromosomal disorder of chromosome 21 (HSA21) aneuploidy, characterized by intellectual disability and reduced lifespan. The transcription repressor, Repressor Element-1 Silencing Transcription factor (REST), which acts as an epigenetic regulator, is a crucial regulator of neuronal and glial gene expression. In this study, we identified and investigated the role of REST-target genes in human brain tissues, cerebral organoids, and neural cells in Down syndrome. Gene expression datasets generated from healthy controls and DS samples of human brain tissues, cerebral organoids, NPC, neurons, and astrocytes were retrieved from the Gene Ontology (GEO) and Sequence Read Archive (SRA) databases. Differential expression analysis was performed on all datasets to produce differential expression genes (DEGs) between DS and control groups. REST-targeted DEGs were subjected to functional ontologies, pathways, and network analyses. We found that REST-targeted DEGs in DS were enriched for the JAK-STAT and HIF-1 signaling pathways across multiple distinct brain regions, ages, and neural cell types. We also identified REST-targeted DEGs involved in nervous system development, cell differentiation, fatty acid metabolism and inflammation in the DS brain. Based on the findings, we propose REST as the critical regulator and a promising therapeutic target to modulate homeostatic gene expression in the DS brain.
Collapse
Affiliation(s)
- Tan Huang
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Sharida Fakurazi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Pike-See Cheah
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - King-Hwa Ling
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
- Malaysian Research Institute on Ageing (MyAgeingTM), Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| |
Collapse
|
22
|
Kim H, Moon S, Lee D, Park J, Kim CH, Kim YM, Choi YK. Korean Red Ginseng-Induced SIRT3 Promotes the Tom22-HIF-1α Circuit in Normoxic Astrocytes. Cells 2023; 12:1512. [PMID: 37296633 PMCID: PMC10252242 DOI: 10.3390/cells12111512] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/25/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Astrocytes play a key role in brain functioning by providing energy to neurons. Increased astrocytic mitochondrial functions by Korean red ginseng extract (KRGE) have been investigated in previous studies. KRGE administration induces hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF) in astrocytes in the adult mouse brain cortex. VEGF expression can be controlled by transcription factors, such as the HIF-1α and estrogen-related receptor α (ERRα). However, the expression of ERRα is unchanged by KRGE in astrocytes of the mouse brain cortex. Instead, sirtuin 3 (SIRT3) expression is induced by KRGE in astrocytes. SIRT3 is a nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase that resides in the mitochondria and maintains mitochondrial homeostasis. Mitochondrial maintenance requires oxygen, and active mitochondria enhance oxygen consumption, resulting in hypoxia. The effects of SIRT3 on HIF-1α-mediated mitochondria functions induced by KRGE are not well established. We aimed to investigate the relationship between SIRT3 and HIF-1α in KRGE-treated normoxic astrocyte cells. Without changing the expression of the ERRα, small interfering ribonucleic acid targeted for SIRT3 in astrocytes substantially lowers the amount of KRGE-induced HIF-1α proteins. Reduced proline hydroxylase 2 (PHD2) expression restores HIF-1α protein levels in SIRT3-depleted astrocytes in normoxic cells treated with KRGE. The translocation of outer mitochondrial membranes 22 (Tom22) and Tom20 is controlled by the SIRT3-HIF-1α axis, which is activated by KRGE. KRGE-induced Tom22 increased oxygen consumption and mitochondrial membrane potential, as well as HIF-1α stability through PHD2. Taken together, in normoxic astrocytes, KRGE-induced SIRT3 activated the Tom22-HIF-1α circuit by increasing oxygen consumption in an ERRα-independent manner.
Collapse
Affiliation(s)
- Hyungsu Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (H.K.); (S.M.); (D.L.); (J.P.)
| | - Sunhong Moon
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (H.K.); (S.M.); (D.L.); (J.P.)
| | - Dohyung Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (H.K.); (S.M.); (D.L.); (J.P.)
| | - Jinhong Park
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (H.K.); (S.M.); (D.L.); (J.P.)
- Department of Otorhinolaryngology-Head and Neck Surgery, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05030, Republic of Korea;
| | - Chang-Hee Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05030, Republic of Korea;
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea;
| | - Yoon Kyung Choi
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (H.K.); (S.M.); (D.L.); (J.P.)
| |
Collapse
|
23
|
Waseem A, Rashid S, Rashid K, Khan MA, Khan R, Haque R, Seth P, Raza SS. Insight into the transcription factors regulating Ischemic Stroke and Glioma in Response to Shared Stimuli. Semin Cancer Biol 2023; 92:102-127. [PMID: 37054904 DOI: 10.1016/j.semcancer.2023.04.006] [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: 11/23/2022] [Revised: 03/28/2023] [Accepted: 04/09/2023] [Indexed: 04/15/2023]
Abstract
Cerebral ischemic stroke and glioma are the two leading causes of patient mortality globally. Despite physiological variations, 1 in 10 people who have an ischemic stroke go on to develop brain cancer, most notably gliomas. In addition, glioma treatments have also been shown to increase the risk of ischemic strokes. Stroke occurs more frequently in cancer patients than in the general population, according to traditional literature. Unbelievably, these events share multiple pathways, but the precise mechanism underlying their co-occurrence remains unknown. Transcription factors (TFs), the main components of gene expression programmes, finally determine the fate of cells and homeostasis. Both ischemic stroke and glioma exhibit aberrant expression of a large number of TFs, which are strongly linked to the pathophysiology and progression of both diseases. The precise genomic binding locations of TFs and how TF binding ultimately relates to transcriptional regulation remain elusive despite a strong interest in understanding how TFs regulate gene expression in both stroke and glioma. As a result, the importance of continuing efforts to understand TF-mediated gene regulation is highlighted in this review, along with some of the primary shared events in stroke and glioma.
Collapse
Affiliation(s)
- Arshi Waseem
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Lucknow-226003, India
| | - Sumaiya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Khalid Rashid
- Department of Cancer Biology, Vontz Center for Molecular Studies, Cincinnati, OH 45267-0521
| | | | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City,Mohali, Punjab 140306, India
| | - Rizwanul Haque
- Department of Biotechnology, Central University of South Bihar, Gaya -824236, India
| | - Pankaj Seth
- Molecular and Cellular Neuroscience, Neurovirology Section, National Brain Research Centre, Manesar, Haryana-122052, India
| | - Syed Shadab Raza
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Lucknow-226003, India; Department of Stem Cell Biology and Regenerative Medicine, Era's Lucknow Medical College Hospital, Era University, Sarfarazganj, Lucknow-226003, India
| |
Collapse
|
24
|
Wang M, Yu X, Li B, Gao C, Chen Y, Zhang X, Li W, Yang L, Fan Z. miR-211-5p targeting MMP9 regulates the expressions of AQP4 in traumatic brain injury. Acta Neurol Belg 2023:10.1007/s13760-023-02205-1. [PMID: 37020131 DOI: 10.1007/s13760-023-02205-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 01/30/2023] [Indexed: 04/07/2023]
Abstract
OBJECTIVE The abnormal expression of matrix metalloproteinase 9 (MMP9) and Aquaporin 4 (AQP4) closely associates with the traumatic brain injury (TBI) development. METHODS Here, we investigated the relationship between miR-211-5p and MMP9/AQP4 axis in TBI patients and astrocyte cells. Demographics, clinical features, and cerebrospinal fluid (CSF) samples were collected from traumatic brain injury (TBI) patients (n = 96) and controls (n = 30) for pathological and gene expression analyses. Luciferase activity assay and gene expression analyses were performed to dissect the regulatory mechanism of miR-211-5p on MMP9/AQP4 in human astrocyte cells. RESULTS miR-211-5p mRNA was significantly decreased in the CSF of TBI patients, which positively correlated with the expression of both MMP9 and AQP4. miR-211-5p could target MMP9 directly in SVG P12 cells. Overexpression of miR-211-5p decreased the expression of MMP9, on the contrary, knockdown miR-211-5p through inhibitors increased the expression of both MMP9 and AQP4. CONCLUSION miR-211-5p inhibits the MMP9/AQP4 axis in human astrocyte cells, which represents a promising approach for the TBI treatment.
Collapse
Affiliation(s)
- Meng Wang
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Xin Yu
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Bin Li
- Department of Neurosurgery, North China Oilfield General Hospital, Renqiu, 062552, Hebei, China
| | - Chensong Gao
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Yan Chen
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Xiaoyang Zhang
- Department of Neurosurgery, Hebei General Hospital, Shijiazhuang, 050000, Hebei, China
| | - Wenling Li
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Lijun Yang
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China.
| | - Zhenzeng Fan
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
25
|
Xu S, Zhang N, Cao L, Liu L, Deng H, Hua S, Zhang Y. Tetramethylpyrazine Attenuates Oxygen-glucose Deprivation-induced Neuronal Damage through Inhibition of the HIF-1α/BNIP3 Pathway: from Network Pharmacological Finding to Experimental Validation. Curr Pharm Des 2023; 29:543-554. [PMID: 36790003 DOI: 10.2174/1381612829666230215100507] [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: 08/26/2022] [Revised: 12/23/2022] [Accepted: 01/02/2023] [Indexed: 02/16/2023]
Abstract
AIMS A network pharmacological analysis combined with experimental validation was used to investigate the neuroprotective mechanism of the natural product Tetramethylpyrazine(TMP). BACKGROUND Protecting neurons is critical for acute ischemic stroke treatment. Tetramethylpyrazine is a bioactive component extracted from Chuanxiong. The neuroprotective potential of TMP has been reported, but a systematic analysis of its mechanism has not been performed. OBJECTIVE Based on the hints of network pharmacology and bioinformatics analysis, the mechanism by which TMP alleviates oxygen-glucose deprivation-induced neuronal damage through inhibition of the HIF-1α/BNIP3 pathway was verified. METHOD In this study, we initially used network pharmacology and bioinformatics analyses to elucidate the mechanisms involved in TMP's predictive targets on a system level. The HIF-1α/BNIP3 pathway mediating the cellular response to hypoxia and apoptosis was considered worthy of focus in the bioinformatic analysis. An oxygen-glucose deprivation (OGD)-induced PC12 cell injury model was established for functional and mechanical validation. Cell viability, lactate dehydrogenase leakage, intracellular reactive oxygen species, percentage of apoptotic cells, and Caspase-3 activity were determined to assess the TMP's protective effects. Transfection with siRNA/HIF-1α or pcDNA/HIF-1α plasmids to silence or overexpress hypoxia-inducible factor 1α(HIF-1α). The role of HIF-1α in OGD-injured cells was observed first. After that, TMP's regulation of the HIF-1α/BNIP3 pathway was investigated. The pcDNA3.1/HIF-1α-positive plasmids were applied in rescue experiments. RESULT The results showed that TMP dose-dependently attenuated OGD-induced cell injury. The expression levels of HIF-1α, BNIP3, and the Bax/Bcl-2 increased significantly with increasing OGD duration. Overexpression of HIF-1α decreased cell viability, increased BNIP3 expression, and Bax/Bcl-2 ratio; siRNA-HIF-1α showed the opposite effect. TMP treatment suppressed HIF-1α, BNIP3 expression, and the Bax/Bcl-2 ratio and was reversed by HIF-1α overexpression. CONCLUSION Our study shows that TMP protects OGD-damaged PC12 cells by inhibiting the HIF-1α/BNIP3 pathway, which provides new insights into the mechanism of TMP and its neuroprotective potential.
Collapse
Affiliation(s)
- Shixin Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Nannan Zhang
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Dongcheng District Community Health Service Management Center, Beijing, China
| | - Lanlan Cao
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Lu Liu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Binhai New Area Hospital of TCM. Tian Jin, Fourth Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hao Deng
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Shengyu Hua
- Institute of traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunsha Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
26
|
Fan G, Liu M, Liu J, Huang Y. The initiator of neuroexcitotoxicity and ferroptosis in ischemic stroke: Glutamate accumulation. Front Mol Neurosci 2023; 16:1113081. [PMID: 37033381 PMCID: PMC10076579 DOI: 10.3389/fnmol.2023.1113081] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
Glutamate plays an important role in excitotoxicity and ferroptosis. Excitotoxicity occurs through over-stimulation of glutamate receptors, specifically NMDAR, while in the non-receptor-mediated pathway, high glutamate concentrations reduce cystine uptake by inhibiting the System Xc-, leading to intracellular glutathione depletion and resulting in ROS accumulation, which contributes to increased lipid peroxidation, mitochondrial damage, and ultimately ferroptosis. Oxidative stress appears to crosstalk between excitotoxicity and ferroptosis, and it is essential to maintain glutamate homeostasis and inhibit oxidative stress responses in vivo. As researchers work to develop natural compounds to further investigate the complex mechanisms and regulatory functions of ferroptosis and excitotoxicity, new avenues will be available for the effective treatment of ischaemic stroke. Therefore, this paper provides a review of the molecular mechanisms and treatment of glutamate-mediated excitotoxicity and ferroptosis.
Collapse
Affiliation(s)
- Genhao Fan
- Graduate School, Tianjin University of Chinese Medicine, Tianjin, China
| | - Menglin Liu
- Graduate School, Tianjin University of Chinese Medicine, Tianjin, China
| | - Jia Liu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Tianjin University of Chinese Medicine, Tianjin, China
| | - Yuhong Huang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Tianjin University of Chinese Medicine, Tianjin, China
- *Correspondence: Yuhong Huang,
| |
Collapse
|
27
|
Jin J, Duan J, Du L, Xing W, Peng X, Zhao Q. Inflammation and immune cell abnormalities in intracranial aneurysm subarachnoid hemorrhage (SAH): Relevant signaling pathways and therapeutic strategies. Front Immunol 2022; 13:1027756. [PMID: 36505409 PMCID: PMC9727248 DOI: 10.3389/fimmu.2022.1027756] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/31/2022] [Indexed: 11/25/2022] Open
Abstract
Intracranial aneurysm subarachnoid hemorrhage (SAH) is a cerebrovascular disorder associated with high overall mortality. Currently, the underlying mechanisms of pathological reaction after aneurysm rupture are still unclear, especially in the immune microenvironment, inflammation, and relevant signaling pathways. SAH-induced immune cell population alteration, immune inflammatory signaling pathway activation, and active substance generation are associated with pro-inflammatory cytokines, immunosuppression, and brain injury. Crosstalk between immune disorders and hyperactivation of inflammatory signals aggravated the devastating consequences of brain injury and cerebral vasospasm and increased the risk of infection. In this review, we discussed the role of inflammation and immune cell responses in the occurrence and development of aneurysm SAH, as well as the most relevant immune inflammatory signaling pathways [PI3K/Akt, extracellular signal-regulated kinase (ERK), hypoxia-inducible factor-1α (HIF-1α), STAT, SIRT, mammalian target of rapamycin (mTOR), NLRP3, TLR4/nuclear factor-κB (NF-κB), and Keap1/nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/ARE cascades] and biomarkers in aneurysm SAH. In addition, we also summarized potential therapeutic drugs targeting the aneurysm SAH immune inflammatory responses, such as nimodipine, dexmedetomidine (DEX), fingolimod, and genomic variation-related aneurysm prophylactic agent sunitinib. The intervention of immune inflammatory responses and immune microenvironment significantly reduces the secondary brain injury, thereby improving the prognosis of patients admitted to SAH. Future studies should focus on exploring potential immune inflammatory mechanisms and developing additional therapeutic strategies for precise aneurysm SAH immune inflammatory regulation and genomic variants associated with aneurysm formation.
Collapse
Affiliation(s)
- Jing Jin
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China,Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jian Duan
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Leiya Du
- 4Department of Oncology, The Second People Hospital of Yibin, Yibin, Sichuan, China
| | - Wenli Xing
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China,*Correspondence: Qijie Zhao, ; Xingchen Peng,
| | - Qijie Zhao
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China,*Correspondence: Qijie Zhao, ; Xingchen Peng,
| |
Collapse
|
28
|
Asgari R, Yarani R, Mohammadi P, Emami Aleagha MS. HIF-1α in the Crosstalk Between Reactive Oxygen Species and Autophagy Process: A Review in Multiple Sclerosis. Cell Mol Neurobiol 2022; 42:2121-2129. [PMID: 34089426 PMCID: PMC11421632 DOI: 10.1007/s10571-021-01111-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/27/2021] [Indexed: 12/16/2022]
Abstract
Cellular stress can lead to the production of reactive oxygen species (ROS) while autophagy, as a catabolic pathway, protects the cells against stress. Autophagy in its turn plays a pivotal role in the pathophysiology of multiple sclerosis (MS). In the current review, we first summarized the contribution of ROS and autophagy to MS pathogenesis. Then probable crosstalk between these two pathways through HIF-1α for the first time has been proposed with the hope of employing a better understanding of MS pathophysiology and probable therapeutic approaches.
Collapse
Affiliation(s)
- Rezvan Asgari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Yarani
- Translational Type 1 Diabetes Biology, Department of Clinical Research, Steno Diabetes Center Copenhagen, Copenhagen, Denmark
| | - Pantea Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | |
Collapse
|
29
|
Chen H, Ma D, Yue F, Qi Y, Dou M, Cui L, Xing Y. The Potential Role of Hypoxia-Inducible Factor-1 in the Progression and Therapy of Central Nervous System Diseases. Curr Neuropharmacol 2022; 20:1651-1666. [PMID: 34325641 PMCID: PMC9881070 DOI: 10.2174/1570159x19666210729123137] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/19/2021] [Accepted: 07/16/2021] [Indexed: 11/22/2022] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1) is a heterodimer protein composed of an oxygenregulated functional subunit, HIF-1α, and a structural subunit, HIF-1β, belonging to the basic helixloop- helix family. Strict regulation of HIF-1 protein stability and subsequent transcriptional activity involves various molecular interactions and is primarily controlled by post-transcriptional modifications. Hypoxia, owing to impaired cerebral blood flow, has been implicated in a range of central nervous system (CNS) diseases by exerting a deleterious effect on brain function. As a master oxygen- sensitive transcription regulator, HIF-1 is responsible for upregulating a wide spectrum of target genes involved in glucose metabolism, angiogenesis, and erythropoiesis to generate the adaptive response to avoid, or at least minimize, hypoxic brain injury. However, prolonged, severe oxygen deprivation may directly contribute to the role-conversion of HIF-1, namely, from neuroprotection to the promotion of cell death. Currently, an increasing number of studies support the fact HIF-1 is involved in a variety of CNS-related diseases, such as intracranial atherosclerosis, stroke, and neurodegenerative diseases. This review article chiefly focuses on the effect of HIF-1 on the pathogenesis and mechanism of progression of numerous CNS-related disorders by mediating the expression of various downstream genes and extensive biological functional events and presents robust evidence that HIF-1 may represent a potential therapeutic target for CNS-related diseases.
Collapse
Affiliation(s)
- Hongxiu Chen
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China; ,Beijing Diagnostic Center of Vascular Ultrasound, Beijing, China; ,Center of Vascular Ultrasonography, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, 45 Changchun Road, Xicheng District, Beijing, 100053, China; ,Hongxiu Chen and Di Ma contributed equally to this work.
| | - Di Ma
- Department of Neurology, The First Hospital of Jilin University, Changchun, China,Hongxiu Chen and Di Ma contributed equally to this work.
| | - Feixue Yue
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yajie Qi
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Manman Dou
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Liuping Cui
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yingqi Xing
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China; ,Beijing Diagnostic Center of Vascular Ultrasound, Beijing, China; ,Center of Vascular Ultrasonography, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, 45 Changchun Road, Xicheng District, Beijing, 100053, China; ,Address correspondence to this author at the Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing Diagnostic Center of Vascular Ultrasound, Center of Vascular Ultrasonography, Beijing Institute of Brain Disorders, 45 Changchun Road, Xicheng District, Beijing, 100053, China; E-mail: This work is recommended by Pro Jiachun Feng, The First Hospital of Jilin University.
| |
Collapse
|
30
|
Neag MA, Mitre AO, Burlacu CC, Inceu AI, Mihu C, Melincovici CS, Bichescu M, Buzoianu AD. miRNA Involvement in Cerebral Ischemia-Reperfusion Injury. Front Neurosci 2022; 16:901360. [PMID: 35757539 PMCID: PMC9226476 DOI: 10.3389/fnins.2022.901360] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Cerebral ischemia reperfusion injury is a debilitating medical condition, currently with only a limited amount of therapies aimed at protecting the cerebral parenchyma. Micro RNAs (miRNAs) are small, non-coding RNA molecules that via the RNA-induced silencing complex either degrade or prevent target messenger RNAs from being translated and thus, can modulate the synthesis of target proteins. In the neurological field, miRNAs have been evaluated as potential regulators in brain development processes and pathological events. Following ischemic hypoxic stress, the cellular and molecular events initiated dysregulate different miRNAs, responsible for long-terming progression and extension of neuronal damage. Because of their ability to regulate the synthesis of target proteins, miRNAs emerge as a possible therapeutic strategy in limiting the neuronal damage following a cerebral ischemic event. This review aims to summarize the recent literature evidence of the miRNAs involved in signaling and modulating cerebral ischemia-reperfusion injuries, thus pointing their potential in limiting neuronal damage and repair mechanisms. An in-depth overview of the molecular pathways involved in ischemia reperfusion injury and the involvement of specific miRNAs, could provide future perspectives in the development of neuroprotective agents targeting these specific miRNAs.
Collapse
Affiliation(s)
- Maria-Adriana Neag
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Andrei-Otto Mitre
- Faculty of Medicine, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | | | - Andreea-Ioana Inceu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Carina Mihu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Carmen-Stanca Melincovici
- Department of Morphological Sciences, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Marius Bichescu
- Faculty of Medicine, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Anca-Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
31
|
Group II Metabotropic Glutamate Receptors Reduce Apoptosis and Regulate BDNF and GDNF Levels in Hypoxic-Ischemic Injury in Neonatal Rats. Int J Mol Sci 2022; 23:ijms23137000. [PMID: 35806000 PMCID: PMC9266366 DOI: 10.3390/ijms23137000] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 12/13/2022] Open
Abstract
Birth asphyxia causes brain injury in neonates, but a fully successful treatment has yet to be developed. This study aimed to investigate the effect of group II mGlu receptors activation after experimental birth asphyxia (hypoxia-ischemia) on the expression of factors involved in apoptosis and neuroprotective neurotrophins. Hypoxia-ischemia (HI) on 7-day-old rats was used as an experimental model. The effects of intraperitoneal application of mGluR2 agonist LY379268 (5 mg/kg) and the specific mGluR3 agonist NAAG (5 mg/kg) (1 h or 6 h after HI) on apoptotic processes and initiation of the neuroprotective mechanism were investigated. LY379268 and NAAG applied shortly after HI prevented brain damage and significantly decreased pro-apoptotic Bax and HtrA2/Omi expression, increasing expression of anti-apoptotic Bcl-2. NAAG or LY379268 applied at both times also decreased HIF-1α formation. HI caused a significant decrease in BDNF concentration, which was restored after LY379268 or NAAG administration. HI-induced increase in GDNF concentration was decreased after administration of LY379268 or NAAG. Our results show that activation of mGluR2/3 receptors shortly after HI prevents brain damage by the inhibition of excessive glutamate release and apoptotic damage decrease. mGluR2 and mGluR3 agonists produced comparable results, indicating that both receptors may be a potential target for early treatment in neonatal HI.
Collapse
|
32
|
Liu J, Guo X, Yang L, Tao T, Cao J, Hong Z, Zeng F, Lu Y, Lin C, Qin Z. Effect of Celastrol on LncRNAs and mRNAs Profiles of Cerebral Ischemia-Reperfusion Injury in Transient Middle Cerebral Artery Occlusion Mice Model. Front Neurosci 2022; 16:889292. [PMID: 35677353 PMCID: PMC9169531 DOI: 10.3389/fnins.2022.889292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/28/2022] [Indexed: 12/15/2022] Open
Abstract
Celastrol plays a significant role in cerebral ischemia-reperfusion injury. Although previous studies have confirmed that celastrol post-treatment has a protective effect on ischemic stroke, the therapeutic effect of celastrol on ischemic stroke and the underlying molecular mechanism remain unclear. In the present study, focal transient cerebral ischemia was induced by transient middle cerebral artery occlusion (tMCAO) in mice and celastrol was administered immediately after reperfusion. We performed lncRNA and mRNA analysis in the ischemic hemisphere of adult mice with celastrol post-treatment through RNA-Sequencing (RNA-Seq). A total of 50 differentially expressed lncRNAs (DE lncRNAs) and 696 differentially expressed mRNAs (DE mRNAs) were identified between the sham and tMCAO group, and a total of 544 DE lncRNAs and 324 DE mRNAs were identified between the tMCAO and tMCAO + celastrol group. Bioinformatic analysis was done on the identified deregulated genes through gene ontology (GO) analysis, KEGG pathway analysis and network analysis. Pathway analysis indicated that inflammation-related signaling pathways played vital roles in the treatment of ischemic stroke by celastrol. Four DE lncRNAs and 5 DE mRNAs were selected for further validation by qRT-PCR in brain tissue, primary neurons, primary astrocytes, and BV2 cells. The results of qRT-PCR suggested that most of selected differentially expressed genes showed the same fold change patterns as those in RNA-Seq results. Our study suggests celastrol treatment can effectively reduce cerebral ischemia-reperfusion injury. The bioinformatics analysis of lnRNAs and mRNAs profiles in the ischemic hemisphere of adult mice provides a new perspective in the neuroprotective effects of celastrol, particularly with regards to ischemic stroke.
Collapse
Affiliation(s)
- Jiandong Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Anesthesiology, The Affiliated Dongnan Hospital of Xiamen University, School of Medicine, Xiamen University, Zhangzhou, China
| | - Xiangna Guo
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lu Yang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tao Tao
- Department of Anesthesiology, The Central People’s Hospital of Zhanjiang, Zhanjiang, China
| | - Jun Cao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Anesthesiology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Zexuan Hong
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fanning Zeng
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yitian Lu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chunshui Lin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Chunshui Lin,
| | - Zaisheng Qin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Zaisheng Qin,
| |
Collapse
|
33
|
Naoluo Xintong Decoction Ameliorates Cerebral Ischemia-Reperfusion Injury by Promoting Angiogenesis through Activating the HIF-1α/VEGF Signaling Pathway in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:9341466. [PMID: 35449809 PMCID: PMC9017488 DOI: 10.1155/2022/9341466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 02/12/2022] [Accepted: 02/22/2022] [Indexed: 11/17/2022]
Abstract
Background Naoluo Xintong decoction (NLXTD) is a traditional Chinese medicine (TCM) formula which has been used to improve neuronal functional recovery after cerebral ischemic stroke. However, the molecular mechanism underlying NLXTD's amelioration of ischemic stroke remains unclear. The present study was designed to explore the effect and mechanism of NLXTD on brain angiogenesis in a rat model with cerebral ischemia-reperfusion (I/R) injury targeting the hypoxia-inducible factor-1α (HIF-1α)/vascular endothelial growth factor (VEGF) pathway. Materials and Methods Cerebral I/R model was established by the classical middle cerebral artery occlusion (MCAO) method. Sprague-Dawley (SD) male rats (n = 80) were randomly divided into the sham-operation group, the model group, the HIF-1α inhibitor 2-methoxyestradiol (2ME2) group, the 2ME2 with NLXTD group, and the NLXTD group. Neurological deficit test, TTC staining, H&E staining, TUNEL staining, immunohistochemistry (IH), immunofluorescence (IF), western blot, and quantitative RT-PCR were performed to evaluate the effect of NLXTD after MCAO. Results Administration of NLXTD significantly decreased neuron deficiency scores, reduced brain infarct volume, and lowered damaged and apoptotic cells after brain I/R injury in rats. Meanwhile, NLXTD had a protective effect on angiogenesis by increasing the MVD and the expressions of BrdU and CD34, which enhanced the number of endothelial cells in the ischemic penumbra brain. NLXTD treatment significantly raised the protein and mRNA levels of HIF-1α, VEGF, VEGFR2, and Notch1 compared with the model treatment. In contrast, a specific HIF-1α inhibitor, 2ME2, inhibited the improvement of neurological function and angiogenesis in NLXTD-induced rats with cerebral I/R injury, suggesting that NLXTD played a positive role in ischemic brain injury by activating the HIF-1α/VEGF signaling pathway. Conclusions NLXTD exerts neuroprotection targeting angiogenesis by upregulating the HIF-1α/VEGF signaling pathway on cerebral I/R injury rats.
Collapse
|
34
|
Li YC, Wang Y, Zou W. Exploration on the Mechanism of Ubiquitin Proteasome System in Cerebral Stroke. Front Aging Neurosci 2022; 14:814463. [PMID: 35462700 PMCID: PMC9022456 DOI: 10.3389/fnagi.2022.814463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 03/14/2022] [Indexed: 12/23/2022] Open
Abstract
Stroke’s secondary damage, such as inflammation, oxidative stress, and mitochondrial dysfunction, are thought to be crucial factors in the disease’s progression. Despite the fact that there are numerous treatments for secondary damage following stroke, such as antiplatelet therapy, anticoagulant therapy, surgery, and so on, the results are disappointing and the side effects are numerous. It is critical to develop novel and effective strategies for improving patient prognosis. The ubiquitin proteasome system (UPS) is the hub for the processing and metabolism of a wide range of functional regulatory proteins in cells. It is critical for the maintenance of cell homeostasis. With the advancement of UPS research in recent years, it has been discovered that UPS is engaged in a variety of physiological and pathological processes in the human body. UPS is expected to play a role in the onset and progression of stroke via multiple targets and pathways. This paper explores the method by which UPS participates in the linked pathogenic process following stroke, in order to give a theoretical foundation for further research into UPS and stroke treatment.
Collapse
Affiliation(s)
- Yu-Chao Li
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yan Wang
- School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, China
| | - Wei Zou
- Heilongjiang University of Chinese Medicine, Harbin, China
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
- *Correspondence: Wei Zou,
| |
Collapse
|
35
|
Ni H, Li J, Zheng J, Zhou B. Cardamonin attenuates cerebral ischemia/reperfusion injury by activating the HIF-1α/VEGFA pathway. Phytother Res 2022; 36:1736-1747. [PMID: 35142404 DOI: 10.1002/ptr.7409] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 01/17/2022] [Accepted: 01/27/2022] [Indexed: 12/13/2022]
Abstract
Cardamonin is a chalcone with neuroprotective activity. The aim of our study was to explore the functions and mechanism of action of cardamonin in ischemic stroke. Oxygen-glucose deprivation and reperfusion (OGD/R)-induced human brain microvascular endothelial cells (HBMECs) and middle cerebral artery occlusion (MCAO) mouse model were utilized to mimic ischemic stroke. Cell viability was analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide. Permeability was investigated via fluorescein isothiocyanate-dextran assay. Apoptosis was detected by TdT-Mediated dUTP Nick End Labeling staining. Hypoxia-inducible factor (HIF)-1α and vascular endothelial growth factor A (VEGFA) protein levels were measured using Western blotting. Brain injury was evaluated by 2,3,5-triphenyltetrazolium chloride staining, neurological score and brain water content. The 37 overlapping targets of ischemic stroke and cardamonin were predicted to be associated with the HIF-1/VEGFA signaling. Cardamonin alleviated OGD/R-induced viability reduction and increase of permeability and apoptosis in HBMECs. Cardamonin increased OGD/R-induced activation of the HIF-1α/VEGFA pathway. Inhibition of the HIF-1α/VEGFA signaling using inhibitor relieved the effect of cardamonin on cell viability, permeability and apoptosis in HBMECs under OGD/R. Cardamonin mitigated brain injury and promoted activation of the HIF-1α/VEGFA signaling in MCAO-treated mice. Overall, cardamonin protected against OGD/R-induced HBMEC damage and MACO-induced brain injury through activating the HIF-1α/VEGFA pathway.
Collapse
Affiliation(s)
- Hongzao Ni
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an, China
| | - Jinxiao Li
- Department of Neurosurgery, Xinyi People's Hospital, Xuzhou, China
| | - Jinyu Zheng
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an, China
| | - Botao Zhou
- Department of Neurosurgery, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
36
|
Intracellular Signaling. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00006-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
37
|
Abstract
Patients with type 2 diabetes mellitus (T2D) are at increased risk of cardiovascular (CV) disease. Sodium glucose cotransporter 2 (SGLT2) inhibitors, also known as gliflozins, are a class of medications used to treat T2D by preventing the reabsorption of glucose filtered through the kidney and thereby facilitating glucose excretion in the urine. Over the past 5 years, many cardiovascular outcome trials (CVOTs) have evaluated the safety and efficacy of SGLT2 inhibitors in preventing CV events. The results of 7 CVOTs have provided solid evidence that the use of SGLT2 in patients with T2D and at high CV risk significantly reduced the risk of death from CV causes. Moreover, in patient with heart failure with reduced ejection fraction, regardless of the presence or absence of T2D, SGLT2 inhibitors use significantly reduced the risk of worsening heart failure and death from CV causes. Although the exact mechanism of the cardiorenal benefit of SGLT2 inhibitors is still unknown, studies have shown that the beneficial effect of these drugs cannot be exclusively explained by their glucose lowering effect, and several possible mechanisms have been proposed. This review will explore the changing role of SGLT2 inhibitors from a diabetes drug to clinical practice guideline-supported therapy for the prevention and treatment of CV diseases, including heart failure.
Collapse
Affiliation(s)
- Reza Mohebi
- Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - James L Januzzi
- Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Baim Institute for Clinical Research, Boston, MA, USA
| |
Collapse
|
38
|
Beker MC, Caglayan AB, Altunay S, Ozbay E, Ates N, Kelestemur T, Caglayan B, Kilic U, Doeppner TR, Hermann DM, Kilic E. Phosphodiesterase 10A Is a Critical Target for Neuroprotection in a Mouse Model of Ischemic Stroke. Mol Neurobiol 2021; 59:574-589. [PMID: 34735672 DOI: 10.1007/s12035-021-02621-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 10/26/2021] [Indexed: 12/14/2022]
Abstract
Phosphodiesterase 10A (PDE10A) hydrolyzes adenosine 3',5'-cyclic monophosphate (cAMP) and guanosine 3',5'-cyclic monophosphate (cGMP). It is highly expressed in the striatum. Recent evidence implied that PDE10A may be involved in the inflammatory processes following injury, such as ischemic stroke. Its role in ischemic injury was unknown. Herein, we exposed mice to 90 or 30-min middle cerebral artery occlusion, followed by the delivery of the highly selective PDE10A inhibitor TAK-063 (0.3 mg/kg or 3 mg/kg) immediately after reperfusion. Animals were sacrificed after 24 or 72 h, respectively. Both TAK-063 doses enhanced neurological function, reduced infarct volume, increased neuronal survival, reduced brain edema, and increased blood-brain barrier integrity, alongside cerebral microcirculation improvements. Post-ischemic neuroprotection was associated with increased phosphorylation (i.e., activation) of pro-survival Akt, Erk-1/2, GSK-3α/β and anti-apoptotic Bcl-xL abundance, decreased phosphorylation of pro-survival mTOR, and HIF-1α, MMP-9 and pro-apoptotic Bax abundance. Interestingly, PDE10A inhibition reduced inflammatory cytokines/chemokines, including IFN-γ and TNF-α, analyzed by planar surface immunoassay. In addition, liquid chromatography-tandem mass spectrometry revealed 40 proteins were significantly altered by TAK-063. Our study established PDE10A as a target for ischemic stroke therapy.
Collapse
Affiliation(s)
- Mustafa C Beker
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey. .,Regenerative and Restorative Medical Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey.
| | - Ahmet B Caglayan
- Regenerative and Restorative Medical Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey.,Department of Physiology, International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Serdar Altunay
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey.,Regenerative and Restorative Medical Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Elif Ozbay
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey.,Regenerative and Restorative Medical Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Nilay Ates
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey.,Regenerative and Restorative Medical Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Taha Kelestemur
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey.,Regenerative and Restorative Medical Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Berrak Caglayan
- Regenerative and Restorative Medical Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey.,Department of Medical Genetics, International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ulkan Kilic
- Department of Medical Biology, International School of Medicine, University of Health Sciences Turkey, Istanbul, Turkey
| | - Thorsten R Doeppner
- Department of Neurology, University Medicine Göttingen, University of Göttingen, Göttingen, Germany
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ertugrul Kilic
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey.,Regenerative and Restorative Medical Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
39
|
Li K, Zheng Y, Wang X. The Potential Relationship Between HIF-1α and Amino Acid Metabolism After Hypoxic Ischemia and Dual Effects on Neurons. Front Neurosci 2021; 15:676553. [PMID: 34483819 PMCID: PMC8416424 DOI: 10.3389/fnins.2021.676553] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 07/30/2021] [Indexed: 12/11/2022] Open
Abstract
Hypoxia inducible factor (HIF) is one of the major transcription factors through which cells and tissues adapt to hypoxic-ischemic injury. However, the specific mechanism by which HIF regulates amino acid metabolism and its effect on neurons during hypoxic ischemia (HI) have remained unclear. This study analyzed the changes in cerebral metabolism of amino acids after HI by using 1H-MRS and investigated the relationship between the changes in cerebral metabolism of amino acids and HIF-1α as well as the potential effects on neurons. Newborn pigs were used as an HI model in this study. Twenty-eight newborn Yorkshire pigs (male, 1.0-1.5 kg) aged 3-5 days were selected and randomly divided into experimental groups tested at 0-2 h (n = 4), 2-6 h (n = 4), 6-12 h (n = 4), 12-24 h (n = 4), 24-48 h (n = 4), and 48-72 h (n = 4) after HI, and a control group (n = 4). After the modeling was completed, 1H-MRS imaging was conducted, followed by immunohistochemical staining of HIF-1α, NeuN, and doublecortin (DCX), and immunofluorescence of glutamic oxaloacetic transaminase (GOT)-1, GOT2, glutathione synthase (GS), glutamate-cysteine ligase catalytic subunit (GCLC), and glutamate-cysteine ligase modifier subunit (GCLM) in brain tissues. The expression of HIF-1α exhibited two increases after HI injury. The first time was opposite to the trends of change of GOT2, aspartic acid, and the number of neurons, while the second was consistent with these trends, suggesting that HIF-1α may have a two-way induction effect on neurons by regulating GOT2 after HI. HIF-1α was closely related to GCLM expression, and GSH level was correlated with the number of hippocampal neurons, indicating that HIF-1α may regulate GCLM to promote GSH synthesis and additionally play a neuroprotective role.
Collapse
Affiliation(s)
- Kexin Li
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yang Zheng
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaoming Wang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
40
|
Lv QW, Zheng ZQ, Zhang H, Guo M, Shen LJ. Serum hypoxia-inducible factor 1alpha emerges as a prognostic factor for severe traumatic brain injury. Clin Chim Acta 2021; 522:77-82. [PMID: 34411556 DOI: 10.1016/j.cca.2021.08.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/06/2021] [Accepted: 08/13/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Hypoxia-inducible factor 1alpha (HIF-1α) is implicated in the cell's response to hypoxia. We investigated whether serum HIF-1α concentrations are correlated with the severity and clinical outcome of severe traumatic brain injury (sTBI). METHODS Serum HIF-1α concentrations were quantified in 104 sTBI patients and 80 healthy controls. Trauma severity was assessed using Glasgow coma scale (GCS). Glasgow outcome scale (GOS) score of 1-3 at post-trauma 90 days was defined as a poor outcome. Multivariate analyses were performed to discern the relationship between serum HIF-1α concentrations and outcome. RESULTS Patients displayed significantly higher serum HIF-1α concentrations than controls (median, 294.9 versus 102.7 pg/ml). HIF-1α concentrations were intimately related to GCS scores (r = -0.62) and GOS scores (r = -0.64). 48 patients (46.2%) experienced a poor outcome. Serum HIF-1α concentrations > 280.2 pg/ml significantly distinguished patients with the development of poor outcome with 77.1% sensitivity and 69.6% specificity (AUC, 0.750; 95% CI: 0.655-0.829). Serum HIF-1α concentrations > 280.2 pg/ml emerged as an independent predictor for poor outcome (OR: 4.179; 95% CI: 1.024-17.052). CONCLUSIONS Serum HIF-1α concentrations are tightly associated with trauma severity and poor 90-day outcome, substantializing serum HIF-1α as a promising prognostic biomarker for sTBI.
Collapse
Affiliation(s)
- Qing-Wei Lv
- Department of Neurosurgery, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou branch), No. 666 Dangui Road, Shengzhou 312400, Zhejiang, China.
| | - Zi-Qiang Zheng
- Department of Neurosurgery, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou branch), No. 666 Dangui Road, Shengzhou 312400, Zhejiang, China
| | - Han Zhang
- Department of Neurosurgery, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou branch), No. 666 Dangui Road, Shengzhou 312400, Zhejiang, China
| | - Mi Guo
- Department of Neurosurgery, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou branch), No. 666 Dangui Road, Shengzhou 312400, Zhejiang, China
| | - Liang-Jun Shen
- Department of Neurosurgery, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou branch), No. 666 Dangui Road, Shengzhou 312400, Zhejiang, China
| |
Collapse
|
41
|
Acteoside isolated from Colebrookea oppositifolia attenuates I/R brain injury in Wistar rats via modulation of HIF-1α, NF-κB, and VEGF pathways. Inflammopharmacology 2021; 29:1565-1577. [PMID: 34365555 DOI: 10.1007/s10787-021-00851-6] [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: 07/06/2021] [Accepted: 07/18/2021] [Indexed: 10/20/2022]
Abstract
AIMS The objective of this study was to assess the anti-stroke activity of acteoside isolated from methanolic root extract of C. oppositifolia METHODS: Ischemia-reperfusion(I/R) brain injury was induced in Wistar rats to assess the anti-stroke activity of acteoside. Rats were pretreated with acteoside (10, 25 & 50 mg/kg, p.o.) before the induction of I/R injury. Parameters such as neurological, motor-cognitive functions were evaluated along with morphological (brain volume, infarct size), biochemical (SOD, Catalase, GSH, lipid peroxidation, TNF-α, IL-6, IL-10, ICAM-1, HIF-1α, VEGF, and NF-κB), histopathological, and gene expression studies (HIF-1α, VEGF) were performed to study the protective effect of acteoside against I/R induced brain injury. RESULTS I/R injury caused significant deterioration of neurological (p < 0.01), motor (p < 0.01) and cognitive (p < 0.01) functions, associated with increase in the brain volume (p < 0.01), and infarct size (p < 0.01); increase in the levels of MDA, TNF-α, IL-6, ICAM-1, HIF-1α, VEGF, and NF-κB along with significant decrease in SOD, catalase, GSH, and IL-10 (p < 0.01 for all parameters) compared to Sham control group. Histology of brain tissue of disease control group exhibited significant vascular changes, neutrophil infiltration, cerebral oedema, and necrosis of the neuronal cells. Further, the gene-expression studies showed significant increase in the HIF-1α (p < 0.01) and VEGF (p < 0.01) mRNA levels in the I/R control compared to Sham control. Interestingly, the acteoside (10, 25 & 50 mg/kg) has prevented the neurological, motor and cognitive dysfunctions, along with inhibiting the morphological, biochemical, histological and gene expression changes induced by I/R-injury (p < 0.05 for 10 mg; p < 0.01 for 25 & 50 mg/kg of acteoside for all the parameters). CONCLUSION These findings suggest that acteoside possess potent anti-stroke activity through modulation of HIF-1α, NF-κB, and VEGF pathway along with its potent antioxidant activity.
Collapse
|
42
|
Mitroshina EV, Savyuk MO, Ponimaskin E, Vedunova MV. Hypoxia-Inducible Factor (HIF) in Ischemic Stroke and Neurodegenerative Disease. Front Cell Dev Biol 2021; 9:703084. [PMID: 34395432 PMCID: PMC8355741 DOI: 10.3389/fcell.2021.703084] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/05/2021] [Indexed: 01/09/2023] Open
Abstract
Hypoxia is one of the most common pathological conditions, which can be induced by multiple events, including ischemic injury, trauma, inflammation, tumors, etc. The body's adaptation to hypoxia is a highly important phenomenon in both health and disease. Most cellular responses to hypoxia are associated with a family of transcription factors called hypoxia-inducible factors (HIFs), which induce the expression of a wide range of genes that help cells adapt to a hypoxic environment. Basic mechanisms of adaptation to hypoxia, and particularly HIF functions, have being extensively studied over recent decades, leading to the 2019 Nobel Prize in Physiology or Medicine. Based on their pivotal physiological importance, HIFs are attracting increasing attention as a new potential target for treating a large number of hypoxia-associated diseases. Most of the experimental work related to HIFs has focused on roles in the liver and kidney. However, increasing evidence clearly demonstrates that HIF-based responses represent an universal adaptation mechanism in all tissue types, including the central nervous system (CNS). In the CNS, HIFs are critically involved in the regulation of neurogenesis, nerve cell differentiation, and neuronal apoptosis. In this mini-review, we provide an overview of the complex role of HIF-1 in the adaptation of neurons and glia cells to hypoxia, with a focus on its potential involvement into various neuronal pathologies and on its possible role as a novel therapeutic target.
Collapse
Affiliation(s)
- Elena V. Mitroshina
- Department of Neurotechnologe, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, Russia
| | - Maria O. Savyuk
- Department of Neurotechnologe, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, Russia
| | - Evgeni Ponimaskin
- Department of Neurotechnologe, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, Russia
- Department of Cellular Neurophysiology, Hannover Medical School, Hanover, Germany
| | - Maria V. Vedunova
- Department of Neurotechnologe, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, Russia
| |
Collapse
|
43
|
Wierońska JM, Cieślik P, Kalinowski L. Nitric Oxide-Dependent Pathways as Critical Factors in the Consequences and Recovery after Brain Ischemic Hypoxia. Biomolecules 2021; 11:biom11081097. [PMID: 34439764 PMCID: PMC8392725 DOI: 10.3390/biom11081097] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 12/14/2022] Open
Abstract
Brain ischemia is one of the leading causes of disability and mortality worldwide. Nitric oxide (NO•), a molecule that is involved in the regulation of proper blood flow, vasodilation, neuronal and glial activity constitutes the crucial factor that contributes to the development of pathological changes after stroke. One of the early consequences of a sudden interruption in the cerebral blood flow is the massive production of reactive oxygen and nitrogen species (ROS/RNS) in neurons due to NO• synthase uncoupling, which leads to neurotoxicity. Progression of apoptotic or necrotic neuronal damage activates reactive astrocytes and attracts microglia or lymphocytes to migrate to place of inflammation. Those inflammatory cells start to produce large amounts of inflammatory proteins, including pathological, inducible form of NOS (iNOS), which generates nitrosative stress that further contributes to brain tissue damage, forming vicious circle of detrimental processes in the late stage of ischemia. S-nitrosylation, hypoxia-inducible factor 1α (HIF-1α) and HIF-1α-dependent genes activated in reactive astrocytes play essential roles in this process. The review summarizes the roles of NO•-dependent pathways in the early and late aftermath of stroke and treatments based on the stimulation or inhibition of particular NO• synthases and the stabilization of HIF-1α activity.
Collapse
Affiliation(s)
- Joanna M Wierońska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna Street 12, 31-343 Kraków, Poland; (J.M.W.); (P.C.)
| | - Paulina Cieślik
- Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna Street 12, 31-343 Kraków, Poland; (J.M.W.); (P.C.)
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics—Biobank Fahrenheit BBMRI.pl, Medical University of Gdansk, Debinki Street 7, 80-211 Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.PL), Debinki Street 7, 80-211 Gdansk, Poland
- BioTechMed Center/Department of Mechanics of Materials and Structures, Gdansk University of Technology, Narutowicza 11/12, 80-223 Gdansk, Poland
- Correspondence: ; Tel.: +48-58-349-1182
| |
Collapse
|
44
|
Adiponectin Treatment Attenuates Cerebral Ischemia-Reperfusion Injury through HIF-1 α-Mediated Antioxidation in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5531048. [PMID: 34336097 PMCID: PMC8298180 DOI: 10.1155/2021/5531048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/01/2021] [Accepted: 05/10/2021] [Indexed: 01/01/2023]
Abstract
Adiponectin (ADPN) plays an important role in cerebral ischemia-reperfusion injury. Although previous studies have confirmed that ADPN pretreatment has a protective effect on ischemic stroke, the therapeutic effect of ADPN on ischemic stroke and the underlying mechanism are still unclear. In order to clarify these questions, focal transient cerebral ischemia was induced by middle cerebral artery occlusion (MCAO) in mice and ADPN was administered for three times at 6 h, 24 h, and 48 h after reperfusion. Meanwhile, a virus-delivered HIF-1α siRNA was used before ADPN administration. The infarct volume, neurological score, cellular apoptosis, and oxidative stress were assessed at 72 h after reperfusion. The long-term outcome of mice after stroke was recorded as well. The results indicated that ADPN treatment reduced the infarct volume (P = 0.032), neurological deficits (P = 0.047), cellular apoptosis (P = 0.041), and oxidative responses (P = 0.031) at 72 h after MCAO. Moreover, ADPN increased both the protein level and transcriptional activity of HIF-1α as evidenced by the transcription levels of VEGF (P = 0.046) and EPO (P = 0.043) at 72 h after MCAO. However, knockdown of HIF-1α partially reversed the antioxidant and treatment effect of ADPN after cerebral ischemia. In the observation of long-term outcome after ADPN treatment, it demonstrated that ADPN not only prevented the cerebral atrophy (P = 0.031) and the neurological function decline (P = 0.048), but also promoted angiogenesis (P = 0.028) after stroke. In conclusion, our findings suggest that ADPN is effective in treatment of ischemic stroke which could be attributed to the increased antioxidant capacity regulated by HIF-1α.
Collapse
|
45
|
Cai Y, Zhuang YK, Wu XY, Dong XQ, Du Q, Yu WH, Wang KY, Hu W, Zheng YK. Serum Hypoxia-Inducible Factor 1alpha Levels Correlate with Outcomes After Intracerebral Hemorrhage. Ther Clin Risk Manag 2021; 17:717-726. [PMID: 34285494 PMCID: PMC8286156 DOI: 10.2147/tcrm.s313433] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/25/2021] [Indexed: 12/02/2022] Open
Abstract
Background Serum hypoxia-inducible factor 1alpha (HIF-1α) is a key regulator in hypoxic and ischemic brain injury. We determined the relationship between serum HIF-1α levels and long-term prognosis plus severity of intracerebral hemorrhage (ICH). Methods A total of 97 ICH cases and 97 healthy controls were enrolled. Glasgow Coma Scale (GCS) score and hematoma volume were used to assess hemorrhagic severity. Glasgow Outcome Scale (GOS) score of 1–3 at post-stroke 90 days was defined as a poor outcome. Results Serum HIF-1α levels of ICH patients were significantly higher than those of healthy controls (median, 218.8 vs 105.4 pg/mL; P<0.001) and were substantially correlated with GCS score (r=−0.485, P<0.001), hematoma volume (r=0.357, P<0.001) and GOS score (r=−0.436, P<0.001). Serum HIF-1α levels >239.4 pg/mL discriminated patients at risk of 90-day poor outcome with sensitivity of 65.9% and specificity of 79.3% (area under the receiver operating characteristic curve, 0.725; 95% confidence interval, 0.625–0.811; P<0.001). Moreover, serum HIF-1α levels >239.4 pg/mL were independently associated with a poor 90-day outcome (odds ratio, 5.133; 95% confidence interval, 1.117–23.593; P=0.036). Conclusion Serum HIF-1α, in close correlation with hemorrhagic severity and poor 90-day outcome, may serve as a potential prognostic biomarker for ICH.
Collapse
Affiliation(s)
- Yong Cai
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang Province, People's Republic of China
| | - Yao-Kun Zhuang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang Province, People's Republic of China
| | - Xiao-Yu Wu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang Province, People's Republic of China
| | - Xiao-Qiao Dong
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang Province, People's Republic of China
| | - Quan Du
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang Province, People's Republic of China
| | - Wen-Hua Yu
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang Province, People's Republic of China
| | - Ke-Yi Wang
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang Province, People's Republic of China
| | - Wei Hu
- Department of Intensive Care Unit, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang Province, People's Republic of China
| | - Yong-Ke Zheng
- Department of Intensive Care Unit, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang Province, People's Republic of China
| |
Collapse
|
46
|
Schneider Gasser EM, Gassmann M, Thiersch M. HIF-2: an important player in neuronal response to ischemia. Pflugers Arch 2021; 473:1175-1176. [PMID: 34245376 DOI: 10.1007/s00424-021-02601-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 10/20/2022]
Affiliation(s)
- Edith M Schneider Gasser
- Institute of Veterinary Physiology, Vetsuisse-Faculty, University of Zurich, Zurich, Switzerland. .,Center for Neuroscience Zurich (ZNZ), Zurich, Switzerland.
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse-Faculty, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIPH), University of Zurich, Zurich, Switzerland
| | - Markus Thiersch
- Institute of Veterinary Physiology, Vetsuisse-Faculty, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIPH), University of Zurich, Zurich, Switzerland
| |
Collapse
|
47
|
Xu C, Dai Y, Bai J, Ren B, Xu J, Gao F, Wang L, Zhang W, Wang R. 17β-oestradiol alleviates endoplasmic reticulum stress injury induced by chronic cerebral hypoperfusion through the Haemoglobin/HIF 1α signalling pathway in ovariectomized rats. Neurochem Int 2021; 148:105119. [PMID: 34224805 DOI: 10.1016/j.neuint.2021.105119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/04/2021] [Accepted: 06/27/2021] [Indexed: 11/28/2022]
Abstract
Endoplasmic reticulum stress (ERS) is known to be an essential target in protecting against ischaemic brain injury. In this study, using a vascular dementia (VaD) animal model induced by bilateral common carotid artery occlusion (BCCAO), we evaluated the effect and mechanism of 17β-oestradiol (E2) against VaD by inhibiting ERS at the early stage (14 d, 21 d, 28 d) and late stage (3 m) after BCCAO in the hippocampal CA1 region of ovariectomized rats. The results showed that the activation of the PERK-eIF2α-ATF4-CHOP axis, a typical ERS pathway, was significantly increased at the early and late stages after BCCAO. JNK (c-Jun N-terminal kinase)-cJun, a pro-death pathway, also displayed the same pattern as the ERS axis. E2 treatment profoundly suppressed the impairments caused by BCCAO. Further mechanistic studies revealed that cerebral blood flow (CBF) was sharply decreased at 14 d and returned to the normal level at 21 d after BCCAO. E2 could not change CBF, while it unexpectedly enhanced the ability to carry oxygen. This is evidenced by the fact that the protein expression of haemoglobin α/β (Hα/β), an oxygen carrier, robustly increased at BCCAO 21 d and 3 m after E2 treatment. The oxygen carrier increased strongly after 21 d and 3 m of BCCAO treated with E2. Moreover, E2 correspondingly enhanced the protein expression of hypoxia-inducible factor 1α (HIF 1α) in both the early and late stage after BCCAO in the hippocampal CA1 region. Finally, E2 administration markedly decreased the activities of caspase-8, caspase-3, and caspase-12 and increased the number of NeuN-positive cells. These findings suggest that E2 serves as a neuroprotectant to alleviate VaD by suppressing ERS injury involving the haemoglobin/HIF 1α signalling pathway.
Collapse
Affiliation(s)
- Chao Xu
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China
| | - Yongxin Dai
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China
| | - Jing Bai
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China; School Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Bo Ren
- School Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Jing Xu
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China; School Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Fujia Gao
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China
| | - Lu Wang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China
| | - Wenli Zhang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China
| | - Ruimin Wang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China; School Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, 063210, China.
| |
Collapse
|
48
|
Hernández IH, Villa-González M, Martín G, Soto M, Pérez-Álvarez MJ. Glial Cells as Therapeutic Approaches in Brain Ischemia-Reperfusion Injury. Cells 2021; 10:1639. [PMID: 34208834 PMCID: PMC8305833 DOI: 10.3390/cells10071639] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/24/2021] [Accepted: 06/26/2021] [Indexed: 02/07/2023] Open
Abstract
Ischemic stroke is the second cause of mortality and the first cause of long-term disability constituting a serious socioeconomic burden worldwide. Approved treatments include thrombectomy and rtPA intravenous administration, which, despite their efficacy in some cases, are not suitable for a great proportion of patients. Glial cell-related therapies are progressively overcoming inefficient neuron-centered approaches in the preclinical phase. Exploiting the ability of microglia to naturally switch between detrimental and protective phenotypes represents a promising therapeutic treatment, in a similar way to what happens with astrocytes. However, the duality present in many of the roles of these cells upon ischemia poses a notorious difficulty in disentangling the precise pathways to target. Still, promoting M2/A2 microglia/astrocyte protective phenotypes and inhibiting M1/A1 neurotoxic profiles is globally rendering promising results in different in vivo models of stroke. On the other hand, described oligodendrogenesis after brain ischemia seems to be strictly beneficial, although these cells are the less studied players in the stroke paradigm and negative effects could be described for oligodendrocytes in the next years. Here, we review recent advances in understanding the precise role of mentioned glial cell types in the main pathological events of ischemic stroke, including inflammation, blood brain barrier integrity, excitotoxicity, reactive oxygen species management, metabolic support, and neurogenesis, among others, with a special attention to tested therapeutic approaches.
Collapse
Affiliation(s)
- Ivó H Hernández
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Mario Villa-González
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Gerardo Martín
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Manuel Soto
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - María José Pérez-Álvarez
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
49
|
Pan Z, Ma G, Kong L, Du G. Hypoxia-inducible factor-1: Regulatory mechanisms and drug development in stroke. Pharmacol Res 2021; 170:105742. [PMID: 34182129 DOI: 10.1016/j.phrs.2021.105742] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/13/2021] [Accepted: 06/23/2021] [Indexed: 12/14/2022]
Abstract
Stroke is an acute cerebrovascular disease caused by sudden rupture of blood vessels in the brain or blockage of blood vessels, which has now become one of the main causes of adult death. During stroke, hypoxia-inducible factor-1 (HIF-1), as an important regulator under hypoxia conditions, is involved in the pathological process of stroke by regulating multi-pathways, such as glucose metabolism, angiogenesis, erythropoiesis, cell survival. However, the roles of HIF-1 in stroke are still controversial, which are related with ischemic time and degree of ischemia. The regulatory mechanisms of HIF-1 in stroke include inflammation, autophagy, oxidative stress, apoptosis and energy metabolism. The potential drugs targeting HIF-1 have attracted more attention, such as HIF-1 inhibitors, HIF-1 stabilizers and natural products. Based on the role of HIF-1 in stroke, HIF-1 is expected to be a potential target for stroke treatment. Resolving when and what interventions for HIF-1 to take during stroke will provide novel strategies for stroke treatment.
Collapse
Affiliation(s)
- Zirong Pan
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Guodong Ma
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Linglei Kong
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| | - Guanhua Du
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| |
Collapse
|
50
|
Zhang Y, Lu X, Tai B, Li W, Li T. Ferroptosis and Its Multifaceted Roles in Cerebral Stroke. Front Cell Neurosci 2021; 15:615372. [PMID: 34149358 PMCID: PMC8209298 DOI: 10.3389/fncel.2021.615372] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 04/09/2021] [Indexed: 12/11/2022] Open
Abstract
Ferroptosis is a unique regulated cell death defined by the intracellular iron overload and distinct biological features compared with other well-known programmed cell death. Ferroptosis can be triggered by many causes including decreased expression of glutathione (GSH), inhibition of the function of glutathione-dependent peroxidase 4 (GPX4), and system xc–, all of which finally lead to the over-accumulation of lipid peroxides in the cell. Ferroptosis has been reported to play an important role in the pathophysiological process of various cancers. In recent years, much evidence also proved that ferroptosis is involved in the progress of cerebral stroke. In this review, we summarized the characteristics of ferroptosis and the potential relationship between ferroptosis and ischemic and hemorrhagic stroke, to provide new targets and ideas for the therapy of stroke.
Collapse
Affiliation(s)
- Yongfa Zhang
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunhua Hospital, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Xiaoyang Lu
- Department of Neurosurgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China.,Translational Neurosurgery and Neurobiology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Bai Tai
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunhua Hospital, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Weijia Li
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunhua Hospital, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Tao Li
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunhua Hospital, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|