1
|
Jiang F, Xu C, Fan X, Yang S, Fan W, Li M, Song J, Wei W, Chen H, Zhong D, Li G. MyD88 Inhibition Attenuates Cerebral Ischemia-reperfusion Injury by Regulating the Inflammatory Response and Reducing Blood-brain Barrier Damage. Neuroscience 2024; 549:121-137. [PMID: 38754722 DOI: 10.1016/j.neuroscience.2024.05.010] [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: 12/30/2023] [Revised: 05/01/2024] [Accepted: 05/09/2024] [Indexed: 05/18/2024]
Abstract
Myeloid differentiation primary response gene 88 (MyD88), a downstream molecule directly linked to Toll-like receptor (TLRs) and IL1 receptor, has been implicated in ischemia-reperfusion injury across various organs. However, its role in cerebral ischemia-reperfusion injury (CIRI) remains unclear. Five transient middle cerebral artery occlusion (tMCAO) microarray datasets were obtained from the Gene Expression Omnibus (GEO) database. We screened these datasets for differentially expressed genes (DEGs) using the GSE35338 and GSE58720 datasets and performed weighted gene co-expression network analysis (WGCNA) using the GSE30655, GSE28731, and GSE32529 datasets to identify the core module related to tMCAO. A protein-protein interaction (PPI) network was constructed using the intersecting DEGs and genes in the core module. Finally, we identified Myd88 was the core gene. In addition, Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Set Enrichment Analysis (GSEA) validated that TNFα, IL17, and MyD88 signaling pathways were significantly enriched in tMCAO. Subsequently, we investigated the mechanistic role of MyD88 in the tMCAO model using male C57BL/6 mice. MyD88 expression increased significantly 24 h after reperfusion. After intraperitoneal administration of TJ-M2010-5, a MyD88-specific inhibitor, during reperfusion, the infarction volumes in the mice were ameliorated. TJ-M2010-5 inhibits the activation of microglia and astrocytes. Moreover, it attenuates the upregulation of inflammatory cytokines TNFα, IL17, and MMP9 while preserving the expression level of ZO1 after tMCAO, thereby safeguarding against blood-brain barrier (BBB) disruption. Finally, our findings suggest that MyD88 regulates the IRAK4/IRF5 signaling pathway associated with microglial activation. MyD88 participates in CIRI by regulating the inflammatory response and BBB damage following tMCAO.
Collapse
Affiliation(s)
- Fangchao Jiang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chen Xu
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuehui Fan
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuai Yang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Fan
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Meng Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jihe Song
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wan Wei
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongping Chen
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Di Zhong
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Guozhong Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Neurology, Heilongjiang Provincial Hospital, Harbin, China.
| |
Collapse
|
2
|
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) is an all-encompassing term that describes cognitive impairment due to cerebrovascular origins. With the advancement of imaging and pathological studies, we now understand that VCID is often comorbid with Alzheimer disease. While researchers in the Alzheimer disease field have been working for years to establish and test blood-based biomarkers for Alzheimer disease diagnosis, prognosis, clinical therapy discovery, and early detection, blood-based biomarkers for VCID are in their infancy and also face challenges. VCID is heterogeneous, comprising many different pathological entities (ischemic, or hemorrhagic), and spatial and temporal differences (acute or chronic). This review highlights pathways that are aiding the search for sensitive and specific blood-based cerebrovascular dysfunction markers, describes promising candidates, and explains ongoing initiatives to discover blood-based VCID biomarkers.
Collapse
Affiliation(s)
- Kate E. Foley
- Stark Neurosciences Research Institute, Indiana University, Indianapolis IN, USA
- Department of Neurology, School of Medicine, Indiana University, Indianapolis IN, USA
| | - Donna M. Wilcock
- Stark Neurosciences Research Institute, Indiana University, Indianapolis IN, USA
- Department of Neurology, School of Medicine, Indiana University, Indianapolis IN, USA
| |
Collapse
|
3
|
Anttila JE, Mattila OS, Liew HK, Mätlik K, Mervaala E, Lindholm P, Lindahl M, Lindsberg PJ, Tseng KY, Airavaara M. MANF protein expression is upregulated in immune cells in the ischemic human brain and systemic recombinant MANF delivery in rat ischemic stroke model demonstrates anti-inflammatory effects. Acta Neuropathol Commun 2024; 12:10. [PMID: 38229173 DOI: 10.1186/s40478-023-01701-y] [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: 09/26/2023] [Accepted: 12/03/2023] [Indexed: 01/18/2024] Open
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF) has cytoprotective effects on various injuries, including cerebral ischemia, and it can promote recovery even when delivered intracranially several days after ischemic stroke. In the uninjured rodent brain, MANF protein is expressed almost exclusively in neurons, but post-ischemic MANF expression has not been characterized. We aimed to investigate how endogenous cerebral MANF protein expression evolves in infarcted human brains and rodent ischemic stroke models. During infarct progression, the cerebral MANF expression pattern both in human and rat brains shifted drastically from neurons to expression in inflammatory cells. Intense MANF immunoreactivity took place in phagocytic microglia/macrophages in the ischemic territory, peaking at two weeks post-stroke in human and one-week post-stroke in rat ischemic cortex. Using double immunofluorescence and mice lacking MANF gene and protein from neuronal stem cells, neurons, astrocytes, and oligodendrocytes, we verified that MANF expression was induced in microglia/macrophage cells in the ischemic hemisphere. Embarking on the drastic expression transition towards inflammatory cells and the impact of blood-borne inflammation in stroke, we hypothesized that exogenously delivered MANF protein can modulate tissue recovery processes. In an attempt to enhance recovery, we designed a set of proof-of-concept studies using systemic delivery of recombinant MANF in a rat model of cortical ischemic stroke. Intranasal recombinant MANF treatment decreased infarct volume and reduced the severity of neurological deficits. Intravenous recombinant MANF treatment decreased the levels of pro-inflammatory cytokines and increased the levels of anti-inflammatory cytokine IL-10 in the infarcted cortex one-day post-stroke. In conclusion, MANF protein expression is induced in activated microglia/macrophage cells in infarcted human and rodent brains, and this could implicate MANF's involvement in the regulation of post-stroke inflammation in patients and experimental animals. Moreover, systemic delivery of recombinant MANF shows promising immunomodulatory effects and therapeutic potential in experimental ischemic stroke.
Collapse
Affiliation(s)
- Jenni E Anttila
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Olli S Mattila
- Department of Neurology, Helsinki University Hospital and Clinical Neurosciences, University of Helsinki, 00290, Helsinki, Finland
| | - Hock-Kean Liew
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien County, Hualien, 970, Taiwan
| | - Kert Mätlik
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Eero Mervaala
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Päivi Lindholm
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Maria Lindahl
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Perttu J Lindsberg
- Department of Neurology, Helsinki University Hospital and Clinical Neurosciences, University of Helsinki, 00290, Helsinki, Finland
| | - Kuan-Yin Tseng
- Department of Neurological Surgery, Tri-Service General Hospital and National Defense Medical Center, Taipei, 114, Taiwan.
| | - Mikko Airavaara
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, P.O. Box 56, 00014, Helsinki, Finland.
- Neuroscience Center, University of Helsinki, 00014, Helsinki, Finland.
| |
Collapse
|
4
|
Abdel-Hameed SS, El-Daly M, Ahmed ASF, Bekhit AA, Heeba GH. Dapoxetine prevents neuronal damage and improves functional outcomes in a model of ischemic stroke through the modulation of inflammation and oxidative stress. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:253-266. [PMID: 37417988 PMCID: PMC10771602 DOI: 10.1007/s00210-023-02601-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 06/23/2023] [Indexed: 07/08/2023]
Abstract
Stroke is a medical emergency that is associated with substantial mortality and functional disability in adults. The most popular class of antidepressants, selective serotonin reuptake inhibitors SSRIs, have recently been shown in studies to have positive effects on post-stroke motor and cognitive function. Thus, we hypothesized that dapoxetine (DAP), a short-acting SSRI, would be effective against cerebral ischemia/reperfusion injury. Adult male Wister rats (200-250 g) were subjected to a sham operation or bilateral common carotid artery occlusion (BCCAO) for 30 min followed by 24 h of reperfusion to induce global cerebral ischemia/reperfusion (I/R) injury. Rats were treated with vehicle or DAP (30 or 60 mg/kg, i.p.) 1 h before BCCAO. The neurobehavioral performance of rats was assessed. The infarct volume, histopathological changes, oxidative stress parameters, and apoptotic and inflammatory mediators were determined in the brain tissues of euthanized rats. Our results confirmed that DAP significantly ameliorated cerebral I/R-induced neurobehavioral deficits, reduced cerebral infarct volume, and histopathological damage. Moreover, DAP pretreatment reduced lipid peroxidation, caspase-3, and inflammatory mediators (TNF-α and iNOS) compared to I/R-injured rats. Thus, DAP pretreatment potentially improves neurological function, and cerebral damage in cerebral ischemic rats may be partly related to the reduction in the inflammatory response, preservation of oxidative balance, and suppression of cell apoptosis in brain tissues.
Collapse
Affiliation(s)
| | - Mahmoud El-Daly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Al-Shaimaa F Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Amany A Bekhit
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, Egypt.
| | - Gehan H Heeba
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt.
| |
Collapse
|
5
|
A. Gawad EA, El Madani M, Mostafa H, Abdel halim RM. Precision medicine as a predictive factor for risk of hospitalization of recurrent ischemic stroke patients treated with low dose aspirin. A pilot study. EGYPTIAN JOURNAL OF ANAESTHESIA 2023. [DOI: 10.1080/11101849.2023.2174833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Affiliation(s)
| | | | - Hanan Mostafa
- Department of Anesthesia, surgical intensive care medicine and pain, Cairo University, Egypt
| | | |
Collapse
|
6
|
Gu C, Li Y, Liu J, Liu S, Long J, Zhang Q, Duan W, Feng T, Huang J, Qiu Y, Ahmed W, Cai H, Hu Y, Wu Y, Chen L. Neural stem cell-derived exosomes-loaded adhesive hydrogel controlled-release promotes cerebral angiogenesis and neurological function in ischemic stroke. Exp Neurol 2023; 370:114547. [PMID: 37743000 DOI: 10.1016/j.expneurol.2023.114547] [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: 04/21/2023] [Revised: 08/31/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
OBJECTIVE Ischemic stroke has become one of the leading diseases for international death, which brings burden to the economy and society. Exosomes (Exos) derived following neural stem cells (NSCs) stimulation promote neurogenesis and migration of NSCs. However, Exos themselves are easily to be removed in vivo. Our study is to investigate whether adhesive hyaluronic acid (HAD) hydrogel loading NSCs-derived-Exo (HAD-Exo) would promote the recovery of ischemic stroke. METHODS A mouse model of middle cerebral artery occlusion (MCAO) was established. PBS, Exo, HAD, and HAD-Exo groups were independently stereotactically injected in mice, respectively. The modified neurological severity score scale and behaviour tests were used to evaluate neurological improvement. Neuroimagings were used to observe the improvement of cerebral infarct volume and vessels. Immunofluorescence staining was used to verify the expression of vascular and cell proliferation-related proteins. RESULTS The structural and mechanical property of HAD and HAD-Exo were detected. Behavioral results showed that HAD-Exo significantly improved neurological functions, especially motor function. Neuroimagings showed that HAD-Exo significantly promoted infarct volume and angiogenesis. Immunofluorescence staining showed that HAD-Exo significantly promoted the cerebral angiogenesis and anti-inflammation. CONCLUSION NSCs derived exosomes-loaded adhesive HAD hydrogel controlled-release could promote cerebral angiogenesis and neurological function for ischemic stroke.
Collapse
Affiliation(s)
- Chenyang Gu
- Department of Neurosurgery, Neuroscience Centre, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510310, PR China; Department of Neurology, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, PR China
| | - Yajing Li
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan 523059, PR China
| | - Jiale Liu
- Department of Neurosurgery, Neuroscience Centre, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510310, PR China
| | - Sitian Liu
- Guangdong Engineering Research Centre for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Jun Long
- Department of Neurosurgery, Neuroscience Centre, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510310, PR China
| | - Qiankun Zhang
- Department of Neurosurgery, Neuroscience Centre, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510310, PR China
| | - Wenjie Duan
- Department of Neurosurgery, Neuroscience Centre, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510310, PR China
| | - Tingle Feng
- Department of Neurosurgery, Neuroscience Centre, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510310, PR China
| | - Jiajun Huang
- Department of Neurosurgery, Neuroscience Centre, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510310, PR China
| | - Yunhui Qiu
- Department of Pathology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510310, PR China
| | - Waqas Ahmed
- Department of Neurology, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, PR China
| | - Hengsen Cai
- Department of Neurosurgery, The Second People's Hospital of Pingnan, Pingnan 537300, PR China
| | - Yong Hu
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hongkong 999077, PR China
| | - Yaobin Wu
- Guangdong Engineering Research Centre for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, PR China.
| | - Lukui Chen
- Department of Neurosurgery, Neuroscience Centre, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510310, PR China.
| |
Collapse
|
7
|
Jaime Garcia D, Chagnot A, Wardlaw JM, Montagne A. A Scoping Review on Biomarkers of Endothelial Dysfunction in Small Vessel Disease: Molecular Insights from Human Studies. Int J Mol Sci 2023; 24:13114. [PMID: 37685924 PMCID: PMC10488088 DOI: 10.3390/ijms241713114] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Small vessel disease (SVD) is a highly prevalent disorder of the brain's microvessels and a common cause of dementia as well as ischaemic and haemorrhagic strokes. Though much about the underlying pathophysiology of SVD remains poorly understood, a wealth of recently published evidence strongly suggests a key role of microvessel endothelial dysfunction and a compromised blood-brain barrier (BBB) in the development and progression of the disease. Understanding the causes and downstream consequences associated with endothelial dysfunction in this pathological context could aid in the development of effective diagnostic and prognostic tools and provide promising avenues for potential therapeutic interventions. In this scoping review, we aim to summarise the findings from clinical studies examining the role of the molecular mechanisms underlying endothelial dysfunction in SVD, focussing on biochemical markers of endothelial dysfunction detectable in biofluids, including cell adhesion molecules, BBB transporters, cytokines/chemokines, inflammatory markers, coagulation factors, growth factors, and markers involved in the nitric oxide cascade.
Collapse
Affiliation(s)
- Daniela Jaime Garcia
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; (D.J.G.); (J.M.W.)
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK;
| | - Audrey Chagnot
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK;
| | - Joanna M. Wardlaw
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; (D.J.G.); (J.M.W.)
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK;
| | - Axel Montagne
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; (D.J.G.); (J.M.W.)
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK;
| |
Collapse
|
8
|
Liu P, Chen Y, Zhang Z, Yuan Z, Sun JG, Xia S, Cao X, Chen J, Zhang CJ, Chen Y, Zhan H, Jin Y, Bao X, Gu Y, Zhang M, Xu Y. Noncanonical contribution of microglial transcription factor NR4A1 to post-stroke recovery through TNF mRNA destabilization. PLoS Biol 2023; 21:e3002199. [PMID: 37486903 PMCID: PMC10365314 DOI: 10.1371/journal.pbio.3002199] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/14/2023] [Indexed: 07/26/2023] Open
Abstract
Microglia-mediated neuroinflammation is involved in various neurological diseases, including ischemic stroke, but the endogenous mechanisms preventing unstrained inflammation is still unclear. The anti-inflammatory role of transcription factor nuclear receptor subfamily 4 group A member 1 (NR4A1) in macrophages and microglia has previously been identified. However, the endogenous mechanisms that how NR4A1 restricts unstrained inflammation remain elusive. Here, we observed that NR4A1 is up-regulated in the cytoplasm of activated microglia and localizes to processing bodies (P-bodies). In addition, we found that cytoplasmic NR4A1 functions as an RNA-binding protein (RBP) that directly binds and destabilizes Tnf mRNA in an N6-methyladenosine (m6A)-dependent manner. Remarkably, conditional microglial deletion of Nr4a1 elevates Tnf expression and worsens outcomes in a mouse model of ischemic stroke, in which case NR4A1 expression is significantly induced in the cytoplasm of microglia. Thus, our study illustrates a novel mechanism that NR4A1 posttranscriptionally regulates Tnf expression in microglia and determines stroke outcomes.
Collapse
Affiliation(s)
- Pinyi Liu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Yan Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Zhi Zhang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Zengqiang Yuan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
- Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Jian-Guang Sun
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
- Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Shengnan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Xiang Cao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Jian Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Cun-Jin Zhang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Yanting Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Hui Zhan
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Yuexinzi Jin
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Yue Gu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Meijuan Zhang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neurology Clinic Medical Center, Nanjing, China
- Institute of Brain Sciences, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
9
|
Wang ZY, Li MZ, Li WJ, Ouyang JF, Gou XJ, Huang Y. Mechanism of action of Daqinjiao decoction in treating cerebral small vessel disease explored using network pharmacology and molecular docking technology. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154538. [PMID: 36370638 DOI: 10.1016/j.phymed.2022.154538] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 10/14/2022] [Accepted: 11/01/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND AND PURPOSE Cerebral small vessel disease (CSVD) is a clinically commonly-seen slow-progressing cerebral vascular disease. As a classic Chinese formula for the treatment of stroke, Daqinjiao Decoction (DQJD) is now used to treat CSVD with desirable effect. Since the mechanism of action is still unclear, this article will explore the therapeutic effect and mechanism of action of the formula using network pharmacology technology. METHODS The major chemical components and potential target genes of DQJD were screened by bioinformatics. The key targets in CSVD were identified based on network modules. Gene Ontology (GO) functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed. Pharmacodynamics of the decoction was evaluated by establishing a rat model with bilateral common carotid artery occlusion in the brain. Molecular docking, Western blot analysis and quantitative real-time polymerase chain reaction (QRT-PCR) were performed to confirm the effectiveness of targets in related pathways. RESULTS Network pharmacology showed that 16 targets and 30 pathways were involved in the DQJD-targeted pathway network. Results revealed that DQJD might play a role by targeting the key targets including Caspse3 and P53 and regulating the P53 signaling pathway. Cognitive function and neuronal cell changes of rats were evaluated using Morris water maze, open field test and HE staining. It was indicated that DQJD could keep the nerve cells intact and neatly arranged. The decoction could improve the memory and learning ability of rats compared with the model group. It decreased the protein and mRNA expression levels of Caspse3 and P53 significantly (p<0.01). CONCLUSION The study shows that baicalein, quercetin and wogonin, the effective components of DQJD, may regulate multiple signaling pathways by targeting the targets like Caspse3 and P53 and treat CSVD by reducing the damage to brain nerve cells.
Collapse
Affiliation(s)
- Zhuo-Yuan Wang
- Central Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai 201999, China
| | - Ming-Zhe Li
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Wen-Jie Li
- Experimental Research center, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Beijing 100700, China
| | - Jing-Feng Ouyang
- Experimental Research center, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Beijing 100700, China
| | - Xiao-Jun Gou
- Central Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai 201999, China.
| | - Ying Huang
- Experimental Research center, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Beijing 100700, China.
| |
Collapse
|
10
|
Wu QJ, Sun X, Teves L, Mayor D, Tymianski M. Mice and Rats Exhibit Striking Inter-species Differences in Gene Response to Acute Stroke. Cell Mol Neurobiol 2022; 42:2773-2789. [PMID: 34350530 PMCID: PMC11421588 DOI: 10.1007/s10571-021-01138-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 07/30/2021] [Indexed: 10/20/2022]
Abstract
Neuroprotection in acute stroke has not been successfully translated from animals to humans. Animal research on promising agents continues largely in rats and mice which are commonly available to researchers. However, controversies continue on the most suitable species to model the human situation. Generally, putative agents seem less effective in mice as compared with rats. We hypothesized that this may be due to inter-species differences in stroke response and that this might be manifest at a genetic level. Here we used whole-genome microarrays to examine the differential gene regulation in the ischemic penumbra of mice and rats at 2 and 6 h after permanent middle cerebral artery occlusion (pMCAO; Raw microarray CEL data files are available in the GEO database with an accession number GSE163654). Differentially expressed genes (adj. p ≤ 0.05) were organized by hierarchical clustering, correlation plots, Venn diagrams and pathway analyses in each species and at each time-point. Emphasis was placed on genes already known to be associated with stroke, including validation by RT-PCR. Gene expression patterns in the ischemic penumbra differed strikingly between the species at both 2 h and 6 h. Nearly 90% of significantly regulated genes and most pathways modulated by ischemia differed between mice and rats. These differences were evident globally, among stroke-associated genes, immediate early genes, genes implicated in stress response, inflammation, neuroprotection, ion channels, and signal transduction. The findings of this study may have significant implications for the choice of species for screening putative stroke therapies.
Collapse
Affiliation(s)
- Qiu Jing Wu
- Krembil Research Institute, University Health Network, 60 Leonard Ave., Toronto, ON, M5T0S8, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Xiujun Sun
- Krembil Research Institute, University Health Network, 60 Leonard Ave., Toronto, ON, M5T0S8, Canada
| | - Lucy Teves
- Krembil Research Institute, University Health Network, 60 Leonard Ave., Toronto, ON, M5T0S8, Canada
| | - Diana Mayor
- Krembil Research Institute, University Health Network, 60 Leonard Ave., Toronto, ON, M5T0S8, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Michael Tymianski
- Krembil Research Institute, University Health Network, 60 Leonard Ave., Toronto, ON, M5T0S8, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, Canada.
- Division of Neurosurgery, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
11
|
Wang X, Wang Q, Wang K, Ni Q, Li H, Su Z, Xu Y. Is Immune Suppression Involved in the Ischemic Stroke? A Study Based on Computational Biology. Front Aging Neurosci 2022; 14:830494. [PMID: 35250546 PMCID: PMC8896355 DOI: 10.3389/fnagi.2022.830494] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/20/2022] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE To identify the genetic mechanisms of immunosuppression-related genes implicated in ischemic stroke. BACKGROUND A better understanding of immune-related genes (IGs) involved in the pathophysiology of ischemic stroke may help identify drug targets beneficial for immunomodulatory approaches and reducing stroke-induced immunosuppression complications. METHODS Two datasets related to ischemic stroke were downloaded from the GEO database. Immunosuppression-associated genes were obtained from three databases (i.e., DisGeNET, HisgAtlas, and Drugbank). The CIBERSORT algorithm was used to calculate the mean proportions of 22 immune-infiltrating cells in the stroke samples. Differential gene expression analysis was performed to identify the differentially expressed genes (DEGs) involved in stroke. Immunosuppression-related crosstalk genes were identified as the overlapping genes between ischemic stroke-DEGs and IGs. Feature selection was performed using the Boruta algorithm and a classifier model was constructed to evaluate the prediction accuracy of the obtained immunosuppression-related crosstalk genes. Functional enrichment analysis, gene-transcriptional factor and gene-drug interaction networks were constructed. RESULTS Twenty two immune cell subsets were identified in stroke, where resting CD4 T memory cells were significantly downregulated while M0 macrophages were significantly upregulated. By overlapping the 54 crosstalk genes obtained by feature selection with ischemic stroke-related genes obtained from the DisGenet database, 17 potentially most valuable immunosuppression-related crosstalk genes were obtained, ARG1, CD36, FCN1, GRN, IL7R, JAK2, MAFB, MMP9, PTEN, STAT3, STAT5A, THBS1, TLR2, TLR4, TLR7, TNFSF10, and VASP. Regulatory transcriptional factors targeting key immunosuppression-related crosstalk genes in stroke included STAT3, SPI1, CEPBD, SP1, TP53, NFIL3, STAT1, HIF1A, and JUN. In addition, signaling pathways enriched by the crosstalk genes, including PD-L1 expression and PD-1 checkpoint pathway, NF-kappa B signaling, IL-17 signaling, TNF signaling, and NOD-like receptor signaling, were also identified. CONCLUSION Putative crosstalk genes that link immunosuppression and ischemic stroke were identified using bioinformatics analysis and machine learning approaches. These may be regarded as potential therapeutic targets for ischemic stroke.
Collapse
Affiliation(s)
- Xin Wang
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qian Wang
- Postdoctoral Workstation, Taian City Central Hospital, Taian, China
| | - Kun Wang
- Postdoctoral Workstation, Taian City Central Hospital, Taian, China
| | - Qingbin Ni
- Postdoctoral Workstation, Taian City Central Hospital, Taian, China
| | - Hu Li
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Zhiqiang Su
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuzhen Xu
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| |
Collapse
|
12
|
Bitencourt ACS, Timóteo RP, Bazan R, Silva MV, da Silveira Filho LG, Ratkevicius CMA, de Assunção TSF, de Oliveira APS, Luvizutto GJ. Association of Proinflammatory Cytokine Levels with Stroke Severity, Infarct Size, and Muscle Strength in the Acute Phase of Stroke. J Stroke Cerebrovasc Dis 2021; 31:106187. [PMID: 34749297 DOI: 10.1016/j.jstrokecerebrovasdis.2021.106187] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 10/19/2022] Open
Abstract
OBJECTIVE Patterns of cytokine levels and their association with stroke severity, infarct size, and muscle strength are obscure. We aimed to analyze the immune mediators linked to T helper (Th)1, Th2, Th17, and regulatory T cell patterns and their association with stroke severity, infarct size, and muscle strength. MATERIALS AND METHODS We included patients with acute stroke (n = 15) and healthy non-disabled individuals (n = 20) aged > 18 years. The dependent variables were stroke severity according to the National Institute of Health Stroke Scale (NIHSS), infarct size on computed tomography, handgrip strength by dynamometry, and global muscle strength according to the Medical Research Council (MRC) scale. The independent variables were the circulating cytokine levels. The cytokine levels were compared between the groups, and correlations between the clinical data were verified. RESULTS The stroke group had higher interleukin (IL)-6 (p < 0.0001) and IL-10 (p < 0.0001) levels, but lower tumor necrosis factor (TNF)-α (p = 0.036) levels than the control group. IL-10 and soluble tumor necrosis factor receptor (sTNF-RII) levels were correlated with each other (r = 0.533; p = 0.042) and infarct size (r = 0.653; p = 0.033 and r = 0.689; p = 0.018, respectively). MRC scores were positively and negatively correlated with handgrip strength of the affected side (r = 0.78; p = 0.001) and NIHSS scores (r = -0.87; p < 0.0001), respectively. CONCLUSIONS Plasma levels of some cytokines were associated with changes in the acute phase of stroke, and IL-10 and sTNF-RII levels are potential biomarkers of infarct size.
Collapse
Affiliation(s)
| | - Rodolfo Pessato Timóteo
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Rodrigo Bazan
- Department of Neurology Psychology and Psychiatry, Botucatu Medical School, Botucatu, São Paulo, Brazil
| | - Marcos Vinícius Silva
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | | | | | | | | | - Gustavo José Luvizutto
- Department of Applied Physical Therapy, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil.
| |
Collapse
|
13
|
Liu K, Tao X, Su J, Li F, Mu F, Zhao S, Lu X, Li J, Chen S, Dong T, Duan J, Wei P, Xi M. Network pharmacology and molecular docking reveal the effective substances and active mechanisms of Dalbergia Odoriferain protecting against ischemic stroke. PLoS One 2021; 16:e0255736. [PMID: 34582494 PMCID: PMC8478192 DOI: 10.1371/journal.pone.0255736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/22/2021] [Indexed: 11/29/2022] Open
Abstract
Dalbergia Odorifera (DO) has been widely used for the treatment of cardiovascular and cerebrovascular diseasesinclinical. However, the effective substances and possible mechanisms of DO are still unclear. In this study, network pharmacology and molecular docking were used toelucidate the effective substances and active mechanisms of DO in treating ischemic stroke (IS). 544 DO-related targets from 29 bioactive components and 344 IS-related targets were collected, among them, 71 overlapping common targets were got. Enrichment analysis showed that 12 components were the possible bioactive components in DO, which regulating 9 important signaling pathways in 3 biological processes including 'oxidative stress' (KEGG:04151, KEGG:04068, KEGG:04915), 'inflammatory response'(KEGG:04668, KEGG:04064) and 'vascular endothelial function regulation'(KEGG:04066, KEGG:04370). Among these, 5 bioactive components with degree≥20 among the 12 potential bioactive components were selected to be docked with the top5 core targets using AutodockVina software. According to the results of molecular docking, the binding sites of core target protein AKT1 and MOL002974, MOL002975, and MOL002914 were 9, 8, and 6, respectively, and they contained 2, 1, and 0 threonine residues, respectively. And some binding sites were consistent, which may be the reason for the similarities and differences between the docking results of the 3 core bioactive components. The results of in vitro experiments showed that OGD/R could inhibit cell survival and AKT phosphorylation which were reversed by the 3 core bioactive components. Among them, MOL002974 (butein) had a slightly better effect. Therefore, the protective effect of MOL002974 (butein) against cerebral ischemia was further evaluated in a rat model of middle cerebral artery occlusion (MCAO) by detecting neurological score, cerebral infarction volume and lactate dehydrogenase (LDH) level. The results indicated that MOL002974 (butein) could significantly improve the neurological score of rats, decrease cerebral infarction volume, and inhibit the level of LDH in the cerebral tissue and serum in a dose-dependent manner. In conclusion, network pharmacology and molecular docking predicate the possible effective substances and mechanisms of DO in treating IS. And the results are verified by the in vitro and in vivo experiments. This research reveals the possible effective substances from DO and its active mechanisms for treating IS and provides a new direction for the secondary development of DO for treating IS.
Collapse
Affiliation(s)
- Kedi Liu
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
- TANK Medicinal Biology Institute of Xi’an, Xi’an, Shaanxi, China
| | - Xingru Tao
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
- TANK Medicinal Biology Institute of Xi’an, Xi’an, Shaanxi, China
| | - Jing Su
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
- TANK Medicinal Biology Institute of Xi’an, Xi’an, Shaanxi, China
| | - Fei Li
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Fei Mu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Shi Zhao
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
- TANK Medicinal Biology Institute of Xi’an, Xi’an, Shaanxi, China
| | - Xinming Lu
- YouYi Clinical Laboratories of Shaanxi, Xi’an, Shaanxi, China
| | - Jing Li
- YouYi Clinical Laboratories of Shaanxi, Xi’an, Shaanxi, China
| | - Sha Chen
- YouYi Clinical Laboratories of Shaanxi, Xi’an, Shaanxi, China
| | - Taiwei Dong
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Jialin Duan
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Peifeng Wei
- National Drug Clinical Trial Institute, The Second Affiliated Hospital, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Miaomiao Xi
- TANK Medicinal Biology Institute of Xi’an, Xi’an, Shaanxi, China
- National Drug Clinical Trial Institute, The Second Affiliated Hospital, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| |
Collapse
|
14
|
Shademan B, Nourazarian A, Laghousi D, Karamad V, Nikanfar M. Exploring potential serum levels of Homocysteine, interleukin-1 beta, and apolipoprotein B 48 as new biomarkers for patients with ischemic stroke. J Clin Lab Anal 2021; 35:e23996. [PMID: 34492129 PMCID: PMC8551691 DOI: 10.1002/jcla.23996] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/25/2021] [Accepted: 08/30/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Stroke is the second leading cause of death worldwide with heterogeneous characteristics. The subtypes of stroke are due to different pathophysiological regulations and causes. This study aimed to investigate the correlation of serum levels of apolipoprotein B 48, interleukin-1β and Homocysteine with BMI in patients with ischemic stroke (IS). METHODS Over one hundred controls (120) and an equal number of IS patients, including 31 women and 89 men, were recruited to participate in the case-control study conducted at Imam Reza Hospital (Tabriz, Iran) from February 2019 to March 2020. We measured serum levels of apolipoprotein B 48, interleukin-1β, and Homocysteine. Receiver operating characteristic analysis (ROC) was performed to evaluate the diagnostic value of these indices in patients and control groups. RESULTS The mean serum levels of apolipoprotein B 48, interleukin-1β, and Homocysteine, were significantly increased in the experimental group compared to the control group with a p-value of 0.001. The ROC curve analysis showed that the area under the curve for apo B48, IL -1β, hs-CRP, and Homocysteine serum levels were 0.94, 0.98, 0.99, and 1, respectively. CONCLUSIONS The results of our current study show that the determination of serum levels of apolipoprotein B 48, interleukin-1β, and Homocysteine can potentially be used to monitor and diagnose IS patients. However, there was no statistically significant correlation between serum levels of apolipoprotein B 48, interleukin 1β and Homocysteine and BMI in the patient group. However, there was a statistically significant inverse correlation between serum levels of high-sensitivity C-reactive protein (hs-CRP) and BMI in the patient group.
Collapse
Affiliation(s)
- Behrouz Shademan
- Department of Medical BiologyFaculty of MedicineEGE UniversityIzmirTurkey
| | - Alireza Nourazarian
- Neurosciences Research Center (NSRC)Tabriz University of Medical SciencesTabrizIran
- Department of Biochemistry and Clinical LaboratoriesFaculty of MedicineTabriz University of Medical SciencesTabrizIran
| | - Delara Laghousi
- Social Determinants of Health Research CenterTabriz University of Medical SciencesTabrizIran
| | - Vahidreza Karamad
- Department of Medical BiologyFaculty of MedicineEGE UniversityIzmirTurkey
| | - Masoud Nikanfar
- Department of NeurologyFaculty of MedicineTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
15
|
Zhou Y, Liao J, Mei Z, Liu X, Ge J. Insight into Crosstalk between Ferroptosis and Necroptosis: Novel Therapeutics in Ischemic Stroke. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9991001. [PMID: 34257829 PMCID: PMC8257382 DOI: 10.1155/2021/9991001] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/30/2021] [Accepted: 06/14/2021] [Indexed: 12/16/2022]
Abstract
Ferroptosis is a nonapoptotic form of cell death characterized by iron-dependent accumulation of lipid hydroperoxides to lethal levels. Necroptosis, an alternative form of programmed necrosis, is regulated by receptor-interacting protein (RIP) 1 activation and by RIP3 and mixed-lineage kinase domain-like (MLKL) phosphorylation. Ferroptosis and necroptosis both play important roles in the pathological progress in ischemic stroke, which is a complex brain disease regulated by several cell death pathways. In the past few years, increasing evidence has suggested that the crosstalk occurs between necroptosis and ferroptosis in ischemic stroke. However, the potential links between ferroptosis and necroptosis in ischemic stroke have not been elucidated yet. Hence, in this review, we overview and analyze the mechanism underlying the crosstalk between necroptosis and ferroptosis in ischemic stroke. And we find that iron overload, one mechanism of ferroptosis, leads to mitochondrial permeability transition pore (MPTP) opening, which aggravates RIP1 phosphorylation and contributes to necroptosis. In addition, heat shock protein 90 (HSP90) induces necroptosis and ferroptosis by promoting RIP1 phosphorylation and suppressing glutathione peroxidase 4 (GPX4) activation. In this work, we try to deliver a new perspective in the exploration of novel therapeutic targets for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yue Zhou
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jun Liao
- Medical School, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Zhigang Mei
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei 443002, China
| | - Xun Liu
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jinwen Ge
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- School of Medicine, Shaoyang University, Shaoyang, Hunan 422000, China
| |
Collapse
|
16
|
Lin SY, Wang YY, Chang CY, Wu CC, Chen WY, Liao SL, Chen CJ. TNF-α Receptor Inhibitor Alleviates Metabolic and Inflammatory Changes in a Rat Model of Ischemic Stroke. Antioxidants (Basel) 2021; 10:851. [PMID: 34073455 PMCID: PMC8228519 DOI: 10.3390/antiox10060851] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/14/2021] [Accepted: 05/24/2021] [Indexed: 11/25/2022] Open
Abstract
Hyperglycemia and inflammation, with their augmented interplay, are involved in cases of stroke with poor outcomes. Interrupting this vicious cycle thus has the potential to prevent stroke disease progression. Tumor necrosis factor-α (TNF-α) is an emerging molecule, which has inflammatory and metabolic roles. Studies have shown that TNF-α receptor inhibitor R-7050 possesses neuroprotective, antihyperglycemic, and anti-inflammatory effects. Using a rat model of permanent cerebral ischemia, pretreatment with R-7050 offered protection against poststroke neurological deficits, brain infarction, edema, oxidative stress, and caspase 3 activation. In the injured cortical tissues, R-7050 reversed the activation of TNF receptor-I (TNFRI), NF-κB, and interleukin-6 (IL-6), as well as the reduction of zonula occludens-1 (ZO-1). In the in vitro study on bEnd.3 endothelial cells, R-7050 reduced the decline of ZO-1 levels after TNF-α-exposure. R-7050 also reduced the metabolic alterations occurring after ischemic stroke, such as hyperglycemia and increased plasma corticosterone, free fatty acids, C reactive protein, and fibroblast growth factor-15 concentrations. In the gastrocnemius muscles of rats with stroke, R-7050 improved activated TNFRI/NF-κB, oxidative stress, and IL-6 pathways, as well as impaired insulin signaling. Overall, our findings highlight a feasible way to combat stroke disease based on an anti-TNF therapy that involves anti-inflammatory and metabolic mechanisms.
Collapse
Affiliation(s)
- Shih-Yi Lin
- Center for Geriatrics and Gerontology, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei City 112, Taiwan;
| | - Ya-Yu Wang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei City 112, Taiwan;
- Department of Family Medicine, Taichung Veterans General Hospital, Taichung City 407, Taiwan
| | - Cheng-Yi Chang
- Department of Surgery, Feng Yuan Hospital, Taichung City 420, Taiwan;
| | - Chih-Cheng Wu
- Department of Anesthesiology, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
- Department of Financial Engineering, Providence University, Taichung City 433, Taiwan
- Department of Data Science and Big Data Analytics, Providence University, Taichung City 433, Taiwan
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung-Hsing University, Taichung City 402, Taiwan;
| | - Su-Lan Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung City 404, Taiwan
| |
Collapse
|
17
|
Gao P, Tang S, Chen H, Zhou X, Ou Y, Shen R, He Y. Preconditioning increases brain resistance against acute brain injury via neuroinflammation modulation. Exp Neurol 2021; 341:113712. [PMID: 33819449 DOI: 10.1016/j.expneurol.2021.113712] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/16/2021] [Accepted: 03/26/2021] [Indexed: 01/10/2023]
Abstract
Acute brain injury (ABI) is a broad concept mainly comprised of sudden parenchymal brain injury. Acute brain injury outcomes are dependent not only on the severity of the primary injury, but the delayed secondary injury that subsequently follows as well. These are both taken into consideration when determining the patient's prognosis. Growing clinical and experimental evidence demonstrates that "preconditioning," a prophylactic approach in which the brain is exposed to various pre-injury stressors, can induce varying degrees of "tolerance" against the impact of the ABI by modulating neuroinflammation. In this review, we will summarize the pathophysiology of ABI, and discuss the involved mechanisms of neuroinflammation in ABI, as well as existing experimental and clinical studies demonstrating the efficacy of preconditioning methods in various types of ABI by modulating neuroinflammation.
Collapse
Affiliation(s)
- Pan Gao
- Department of Translational Neurodegeneration, German Centre for Neurodegenerative Diseases (DZNE), Munich 81377, Germany.
| | - Sicheng Tang
- Medical Clinic and Polyclinic IV, Ludwig-Maximilians University Munich (LMU), Munich 80336, Germany
| | - Hanmin Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Xiangyue Zhou
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Yibo Ou
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Ronghua Shen
- Department of Psychological Rehabilitation, Hankou Hospital, Wuhan, Hubei 430010, PR China.
| | - Yue He
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China.
| |
Collapse
|
18
|
Huang D, Zhou J, Li W, Zhang L, Wang X, Liu Q. Casticin protected against neuronal injury and inhibited the TLR4/NF-κB pathway after middle cerebral artery occlusion in rats. Pharmacol Res Perspect 2021; 9:e00752. [PMID: 33704926 PMCID: PMC7948701 DOI: 10.1002/prp2.752] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/12/2021] [Accepted: 02/13/2021] [Indexed: 12/18/2022] Open
Abstract
Although stroke is a major human neurological disease, there is a paucity of effective neuroprotectants that can improve its treatment. Casticin is a natural monomer drug with many biological effects such as anti-inflammatory and anti-tumor actions. However, it is not clear whether it has a neuroprotective effect in ischemic stroke. In this study, the neuroprotective effect of casticin in a rat middle cerebral artery occlusion (MCAO) model was investigated. Results showed that casticin reduced the volume of the cerebral infarction, mNSS scores, swimming distance, time to find the submerged platform, and serum concentrations of TNF-α, TGF-β, IL-6 in MCAO rats. Moreover, casticin also decreased the expression of TLR4, NF-κB p65, and NF-κB p50 proteins and reversed the reduced expression of IκB protein in the brain tissue of MCAO rats. The in vitro study revealed that casticin decreased apoptosis of OGD/R-PC12 cells, reduced the expression of TLR4, NF-κB p65, and NF-κB p50, while increased IκB protein expression. In conclusion, casticin improved the neurological functions of MCAO rats via inhibiting the TLR4/NF-κB pathway and might have the potential to be developed into a neuroprotective agent for stroke patients.
Collapse
Affiliation(s)
- Dan Huang
- Department of Neurology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jiafan Zhou
- Department of Neurology, Qionghai People's Hospital, Qionghai, China
| | - Wenning Li
- Department of Neurology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Li Zhang
- Department of Pharmacology, Hainan Medical University, Haikou, China
| | - Xiaomeng Wang
- Department of Pharmacology, Hainan Medical University, Haikou, China
| | - Qiang Liu
- Department of Pharmacology, Hainan Medical University, Haikou, China
| |
Collapse
|
19
|
Eidson LN, Gao Q, Qu H, Kikuchi DS, Campos ACP, Faidley EA, Sun YY, Kuan CY, Pagano RL, Lassègue B, Tansey MG, Griendling KK, Hernandes MS. Poldip2 controls leukocyte infiltration into the ischemic brain by regulating focal adhesion kinase-mediated VCAM-1 induction. Sci Rep 2021; 11:5533. [PMID: 33692398 PMCID: PMC7970934 DOI: 10.1038/s41598-021-84987-z] [Citation(s) in RCA: 10] [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: 06/10/2020] [Accepted: 02/22/2021] [Indexed: 11/29/2022] Open
Abstract
Stroke is a multiphasic process involving a direct ischemic brain injury which is then exacerbated by the influx of immune cells into the brain tissue. Activation of brain endothelial cells leads to the expression of adhesion molecules such vascular cell adhesion molecule 1 (VCAM-1) on endothelial cells, further increasing leukocyte recruitment. Polymerase δ-interacting protein 2 (Poldip2) promotes brain vascular inflammation and leukocyte recruitment via unknown mechanisms. This study aimed to define the role of Poldip2 in mediating vascular inflammation and leukocyte recruitment following cerebral ischemia. Cerebral ischemia was induced in Poldip2+/+ and Poldip2+/- mice and brains were isolated and processed for flow cytometry or RT-PCR. Cultured rat brain microvascular endothelial cells were used to investigate the effect of Poldip2 depletion on focal adhesion kinase (FAK)-mediated VCAM-1 induction. Poldip2 depletion in vivo attenuated the infiltration of myeloid cells, inflammatory monocytes/macrophages and decreased the induction of adhesion molecules. Focusing on VCAM-1, we demonstrated mechanistically that FAK activation was a critical intermediary in Poldip2-mediated VCAM-1 induction. In conclusion, Poldip2 is an important mediator of endothelial dysfunction and leukocyte recruitment. Thus, Poldip2 could be a therapeutic target to improve morbidity following ischemic stroke.
Collapse
Affiliation(s)
- Lori N Eidson
- Department of Physiology, Emory University, Atlanta, GA, 30322, USA
| | - Qingzeng Gao
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308-C WMB, Atlanta, GA, 30322, USA
| | - Hongyan Qu
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308-C WMB, Atlanta, GA, 30322, USA
| | - Daniel S Kikuchi
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308-C WMB, Atlanta, GA, 30322, USA
| | - Ana Carolina P Campos
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308-C WMB, Atlanta, GA, 30322, USA
- Department of Neuroscience, Hospital Sírio-Libanês, São Paulo, Brazil
| | - Elizabeth A Faidley
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308-C WMB, Atlanta, GA, 30322, USA
| | - Yu-Yo Sun
- Department of Neuroscience, University of Virginia, Charlottesville, VA, 22904, USA
| | - Chia-Yi Kuan
- Department of Neuroscience, University of Virginia, Charlottesville, VA, 22904, USA
| | - Rosana L Pagano
- Department of Neuroscience, Hospital Sírio-Libanês, São Paulo, Brazil
| | - Bernard Lassègue
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308-C WMB, Atlanta, GA, 30322, USA
| | - Malú G Tansey
- Department of Physiology, Emory University, Atlanta, GA, 30322, USA
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, College of Medicine, Normal Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, 32610, USA
- Department of Neurology, Center for Translational Research in Neurodegenerative Disease, College of Medicine, Normal Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, 32610, USA
| | - Kathy K Griendling
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308-C WMB, Atlanta, GA, 30322, USA
| | - Marina S Hernandes
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308-C WMB, Atlanta, GA, 30322, USA.
| |
Collapse
|
20
|
Chen RH, Du WD, Wang Q, Li ZF, Wang DX, Yang SL, Feng YL. Effects of Acanthopanax senticosus (Rupr. & Maxim.) Harms on cerebral ischemia-reperfusion injury revealed by metabolomics and transcriptomics. JOURNAL OF ETHNOPHARMACOLOGY 2021; 264:113212. [PMID: 32768643 DOI: 10.1016/j.jep.2020.113212] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cerebral ischemia-reperfusion (CIR) injury is one of the main diseases leading to death and disability. Acanthopanax senticosus (Rupr. & Maxim.) Harms (AS), also known as Panax ginseng, has neuroprotective effects on anti-CIR injury. However, the underlying molecular mechanism of its therapeutic effects is not clear. AIM OF THE STUDY To systematically study and explore the mechanism of Acanthopanax senticosus (Rupr. & Maxim.) Harms extract (ASE) in the treatment of CIR injury based on metabolomics and transcriptomics. MATERIALS AND METHODS The pharmacological basis of ASE in the treatment of CIR was evaluated, and samples were used in plasma metabolomics and brain tissue transcriptomics to reveal potential biomarkers. Finally, according to online database, we analyzed biomarkers identified by the two technologies, explained reasons for the therapeutic effect of ASE, and identify therapeutic targets. RESULTS A total of 53 differential metabolites (DMs) were identified in plasma and 3138 differentially expressed genes (DEGs) were identified in brain tissue from three groups of rats, including sham, ischemia-reperfusion (I/R), and ASE groups. Enrichment analysis showed that Nme6, Tk1, and Pold1 that are involved in the production of deoxycytidine and thymine were significantly up-regulated and Dck was significantly down-regulated by the intervention with ASE. These findings indicated that ASE participates in the pyrimidine metabolism by significantly regulating the balance between dCTP and dTTP. In addition, ASE repaired and promoted the lipid metabolism in rats, which might be due to the significant expression of Dgkz, Chat, and Gpcpd1. CONCLUSIONS The findings of this study suggest that ASE regulates the significant changes in gene expression in metabolites pyrimidine, and lipid metabolism in CIR rats and plays an active role in the treatment of CIR injury through multiple targets and pathways.
Collapse
Affiliation(s)
- Ren-Hao Chen
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330002, China
| | - Wei-Dong Du
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330002, China
| | - Qi Wang
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang, 330006, China
| | - Zhi-Feng Li
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330002, China; Nanchang Key Laboratory of Active Ingredients of Traditional Chinese Medicine and Natural Medicine, Nanchang, 330006, China.
| | - Dong-Xu Wang
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330002, China
| | - Shi-Lin Yang
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang, 330006, China
| | - Yu-Lin Feng
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang, 330006, China.
| |
Collapse
|
21
|
Clausen BH, Wirenfeldt M, Høgedal SS, Frich LH, Nielsen HH, Schrøder HD, Østergaard K, Finsen B, Kristensen BW, Lambertsen KL. Characterization of the TNF and IL-1 systems in human brain and blood after ischemic stroke. Acta Neuropathol Commun 2020; 8:81. [PMID: 32503645 PMCID: PMC7273684 DOI: 10.1186/s40478-020-00957-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/27/2020] [Indexed: 01/17/2023] Open
Abstract
Preclinical and clinical proof-of-concept studies have suggested the effectiveness of pharmacological modulation of inflammatory cytokines in ischemic stroke. Experimental evidence shows that targeting tumor necrosis factor (TNF) and interleukin (IL)-1 holds promise, and these cytokines are considered prime targets in the development of new stroke therapies. So far, however, information on the cellular expression of TNF and IL-1 in the human ischemic brain is sparse.We studied 14 cases of human post-mortem ischemic stroke, representing 21 specimens of infarcts aged 1 to > 8 days. We characterized glial and leukocyte reactions in the infarct/peri-infarct (I/PI) and normal-appearing tissue (NAT) and the cellular location of TNF, TNF receptor (TNFR)1 and TNFR2, IL-1α, IL-1β, and IL-1 receptor antagonist (IL-1Ra). The immunohistochemically stained tissue sections received a score reflecting the number of immunoreactive cells and the intensity of the immunoreactivity (IR) in individual cells where 0 = no immunoreactive cells, 1 = many intermediately to strongly immunoreactive cells, and 2 = numerous and intensively immunoreactive cells. Additionally, we measured blood TNF, TNFR, and IL-1 levels in surviving ischemic stroke patients within the first 8 h and again at 72 h after symptom onset and compared levels to healthy controls.We observed IL-1α and IL-1β IR in neurons, glia, and macrophages in all specimens. IL-1Ra IR was found in glia, in addition to macrophages. TNF IR was initially found in neurons located in I/PI and NAT but increased in glia in older infarcts. TNF IR increased in macrophages in all specimens. TNFR1 IR was found in neurons and glia and macrophages, while TNFR2 was expressed only by glia in I/PI and NAT, and by macrophages in I/PI. Our results suggest that TNF and IL-1 are expressed by subsets of cells and that TNFR2 is expressed in areas with increased astrocytic reactivity. In ischemic stroke patients, we demonstrate that plasma TNFR1 and TNFR2 levels increased in the acute phase after symptom onset compared to healthy controls, whereas TNF, IL-1α, IL-1β, and IL-1Ra did not change.Our findings of increased brain cytokines and plasma TNFR1 and TNFR2 support the hypothesis that targeting post-stroke inflammation could be a promising add-on therapy in ischemic stroke patients.
Collapse
Affiliation(s)
- Bettina H. Clausen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21, st, DK-5000 Odense C, Denmark
- BRIDGE, Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark
| | - Martin Wirenfeldt
- BRIDGE, Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark
- Department of Pathology, Odense University Hospital, Odense, J.B. Winsloewsvej 15, DK-5000 Odense C, Denmark
| | - Sofie S. Høgedal
- Department of Neurology, Odense University Hospital, J.B. Winsloewsvej 4, DK-5000 Odense C, Denmark
| | - Lars H. Frich
- Orthopedic Research Unit, University of Southern Denmark, DK-5000 Odense C, Denmark
- OPEN, Open Patient data Explorative Network, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, J.B. Winsloewsvej 9a, DK-5000 Odense, Denmark
| | - Helle H. Nielsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21, st, DK-5000 Odense C, Denmark
- BRIDGE, Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark
- Department of Neurology, Odense University Hospital, J.B. Winsloewsvej 4, DK-5000 Odense C, Denmark
| | - Henrik D. Schrøder
- Department of Pathology, Odense University Hospital, Odense, J.B. Winsloewsvej 15, DK-5000 Odense C, Denmark
| | - Kamilla Østergaard
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21, st, DK-5000 Odense C, Denmark
| | - Bente Finsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21, st, DK-5000 Odense C, Denmark
- BRIDGE, Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark
| | - Bjarne W. Kristensen
- BRIDGE, Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark
- Department of Pathology, Odense University Hospital, Odense, J.B. Winsloewsvej 15, DK-5000 Odense C, Denmark
| | - Kate L. Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21, st, DK-5000 Odense C, Denmark
- BRIDGE, Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark
- Department of Neurology, Odense University Hospital, J.B. Winsloewsvej 4, DK-5000 Odense C, Denmark
- OPEN, Open Patient data Explorative Network, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, J.B. Winsloewsvej 9a, DK-5000 Odense, Denmark
| |
Collapse
|
22
|
Manolis TA, Manolis AA, Apostolopoulos EJ, Melita H, Manolis AS. Atrial Fibrillation and Cognitive Impairment: An Associated Burden or Burden by Association? Angiology 2020; 71:498-519. [DOI: 10.1177/0003319720910669] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Growing evidence suggests that atrial fibrillation (AF), in addition to its thromboembolic risk, is a risk factor for cognitive impairment (CI) via several pathways and mechanisms, further contributing to morbidity/mortality. Prior stroke is a contributor to CI, but AF is also associated with CI independently from prior stroke. Silent brain infarctions, microemboli and microbleeds, brain atrophy, cerebral hypoperfusion from widely fluctuating ventricular rates, altered hemostatic function, vascular oxidative stress, and inflammation may all exacerbate CI, particularly in patients with persistent/permanent rather than paroxysmal AF and with increased duration/burden of the arrhythmia. Brain magnetic resonance imaging is an important screening tool in eliciting and monitoring vascular and nonvascular lesions contributing to CI. Evidence is also emerging about the role of genetics in CI development. Anticoagulation and rhythm/rate control strategies may protect against CI preventing or slowing its progression or conversion to dementia, particularly at the early stages when CI may still be a treatable condition. Importantly, AF and CI share many common risk factors. Thus, screening for these 2 conditions and searching for and managing modifiable risk factors and potentially reversible causes for both AF and CI remains an important step toward prevention or amelioration of the impact incurred by these 2 conditions.
Collapse
Affiliation(s)
| | | | | | | | - Antonis S. Manolis
- First and Third Department of Cardiology, Athens University School of Medicine, Athens, Greece
| |
Collapse
|
23
|
Zhao Q, Yan T, Chopp M, Venkat P, Chen J. Brain-kidney interaction: Renal dysfunction following ischemic stroke. J Cereb Blood Flow Metab 2020; 40:246-262. [PMID: 31766979 PMCID: PMC7370616 DOI: 10.1177/0271678x19890931] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Stroke is a leading cause of mortality and morbidity, with long-term debilitating effects. Accumulating evidence from experimental studies as well as observational studies in patients suggests a cross talk between the brain and kidney after stroke. Stroke may lead to kidney dysfunction which can adversely impact patient outcome. In this review article, we discuss the epidemiology and mechanisms of brain–kidney interaction following ischemic stroke. Specifically, we discuss the role of the central autonomic network, autoregulation, inflammatory and immune responses, the role of extracellular vesicles and their cargo microRNA, in mediating brain–kidney interaction following stroke. Understanding the bidirectional nature of interaction between the brain and kidney after cerebral injury would have clinical implications for the treatment of stroke and overall patient outcome.
Collapse
Affiliation(s)
- Qiang Zhao
- Tianjin Neurological Institute, Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Tao Yan
- Tianjin Neurological Institute, Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.,Department of Physics, Oakland University, Rochester, MI, USA
| | - Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
24
|
Li L, Dong L, Xiao Z, He W, Zhao J, Pan H, Chu B, Cheng J, Wang H. Integrated analysis of the proteome and transcriptome in a MCAO mouse model revealed the molecular landscape during stroke progression. J Adv Res 2020; 24:13-27. [PMID: 32181013 PMCID: PMC7063112 DOI: 10.1016/j.jare.2020.01.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/10/2020] [Accepted: 01/10/2020] [Indexed: 12/24/2022] Open
Abstract
DIA proteomics was applied to MCAO mice detection for the first time. Proteomics and bioinformatics revealed relationship between stroke process and immunity, especially inflammation. C3, Apoa4 and S100a9 were highlighted as a marker or drug targets for stroke.
Strokes usually results in long-term disability and death, and they occur worldwide. Recently, increased research on both on the physiopathological mechanisms and the transcriptome during stroke progression, have highlighted the relationship between stroke progression and immunity, with a special focus on inflammation. Here, we applied proteome analysis to a middle carotid artery occlusion (MCAO) mouse model at 0 h, 6 h, 12 h and 24 h, in which proteome profiling was performed with 23 samples, and 41 differentially expressed proteins (DEPs) were identified. Bioinformatics studies on our data revealed the importance of the immune response and particularly identified the inflammatory response, cytokine- cytokine receptor interactions, the innate immune response and reactive oxygen species (ROS) during stroke progression. In addition, we compared our data with multiple gene expression omnibus (GEO) datasets with and without a time series, in which similar pathways were identified, and three proteins, C3, Apoa4 and S100a9, were highlighted as markers or drug targets for stroke; these three proteins were significantly upregulated in the MCAO model, both in our proteomic data and in the GEO database.
Collapse
Affiliation(s)
- Litao Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang 050051, Hebei, China
| | - Lipeng Dong
- Department of Neurology, Hebei General Hospital, Shijiazhuang 050051, Hebei, China
| | - Zhen Xiao
- College of Life Sciences, Shanghai Normal University, Shanghai 200234, China
| | - Weiliang He
- Department of Neurology, Hebei General Hospital, Shijiazhuang 050051, Hebei, China
| | - Jingru Zhao
- Department of Neurology, Hebei General Hospital, Shijiazhuang 050051, Hebei, China
| | - Henan Pan
- Department of Neurology, Hebei General Hospital, Shijiazhuang 050051, Hebei, China.,North China University of Science and Technology, Tangshan 063210, Hebei, China
| | - Bao Chu
- Department of Neurology, Hebei General Hospital, Shijiazhuang 050051, Hebei, China
| | - Jinming Cheng
- Department of Neurology, Hebei General Hospital, Shijiazhuang 050051, Hebei, China
| | - Hebo Wang
- Department of Neurology, Hebei General Hospital, Shijiazhuang 050051, Hebei, China
| |
Collapse
|
25
|
Wang D, Wang Q, Chen R, Yang S, Li Z, Feng Y. Exploring the effects of Gastrodia elata Blume on the treatment of cerebral ischemia-reperfusion injury using UPLC-Q/TOF-MS-based plasma metabolomics. Food Funct 2019; 10:7204-7215. [PMID: 31609374 DOI: 10.1039/c9fo01729a] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Gastrodia elata Blume (Orchidaceae, GEB) is a medicinal plant that has been widely used in the treatment of cerebrovascular disease. This study explored the protective effects of GEB against cerebral ischemia-reperfusion using Information-Dependent Acquisition (IDA)-mediated UPLC-Q/TOF-MS-based plasma metabolomics. Cerebral ischemia-reperfusion (IR) injury was induced in male Wistar rats using the Zea Longa method. Biochemical and histological assays were performed to evaluate the therapeutic effects of GEB on IR rats. We found that the neurobehavioral scores and infarction areas of GEB and nimodipine treated groups were dramatically lower than those of the IR groups. Hematoxylin and Eosin (HE) staining and TdT-mediated dUTP Nick-End Labeling (TUNEL) showed that GEB significantly improved neuronal injury and prevented neuronal apoptosis. Biochemical analysis revealed that GEB prevented cerebral ischemia-reperfusion injury by regulating inflammation and oxidative injury. Through ultra-high-performance liquid chromatography-quadrupole time-of-flight mass spectrometry-metabolomics-based approaches, 43 plasma metabolites related to GEB treatment were detected, 6 of which significantly differed (p < 0.05) between the model and GEB groups. The levels of l-histidine, sphinganine, thymine, spermidine and deoxycytidine in the IR group were significantly higher than those in the sham group, but decreased following GEB treatment. Arachidonic acid levels were lower in the IR group, but dramatically increased in response to GEB. Pharmacodynamics and metabolomics confirmed that the mechanism of GEB in the treatment of cerebral ischemia was not only related to the reduction of inflammation, oxidation, neurotoxicity, and apoptosis, but also mediated through arachidonic acid metabolism, histidine metabolism, pyrimidine metabolism, arginine and proline metabolism, sphingolipid metabolism, and glycerophospholipid metabolism in vivo.
Collapse
Affiliation(s)
- Dongxu Wang
- Jiangxi University of Traditional Chinese Medicine, Nanchang 330002, China.
| | - Qi Wang
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang 330006, China.
| | - Renhao Chen
- Jiangxi University of Traditional Chinese Medicine, Nanchang 330002, China.
| | - Shinlin Yang
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang 330006, China.
| | - Zhifeng Li
- Jiangxi University of Traditional Chinese Medicine, Nanchang 330002, China.
| | - Yulin Feng
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang 330006, China.
| |
Collapse
|
26
|
Ma S, Liu X, Cheng B, Jia Z, Hua H, Xin Y. Chemical characterization of polysaccharides isolated from scrophularia ningpoensis and its protective effect on the cerebral ischemia/reperfusin injury in rat model. Int J Biol Macromol 2019; 139:955-966. [DOI: 10.1016/j.ijbiomac.2019.08.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 04/12/2019] [Accepted: 08/05/2019] [Indexed: 12/22/2022]
|
27
|
Dukay B, Csoboz B, Tóth ME. Heat-Shock Proteins in Neuroinflammation. Front Pharmacol 2019; 10:920. [PMID: 31507418 PMCID: PMC6718606 DOI: 10.3389/fphar.2019.00920] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/22/2019] [Indexed: 01/01/2023] Open
Abstract
The heat-shock response, one of the main pro-survival mechanisms of a living organism, has evolved as the biochemical response of cells to cope with heat stress. The most well-characterized aspect of the heat-shock response is the accumulation of a conserved set of proteins termed heat-shock proteins (HSPs). HSPs are key players in protein homeostasis acting as chaperones by aiding the folding and assembly of nascent proteins and protecting against protein aggregation. HSPs have been associated with neurological diseases in the context of their chaperone activity, as they were found to suppress the aggregation of misfolded toxic proteins. In recent times, HSPs have proven to have functions apart from the classical molecular chaperoning in that they play a role in a wider scale of neurological disorders by modulating neuronal survival, inflammation, and disease-specific signaling processes. HSPs are gaining importance based on their ability to fine-tune inflammation and act as immune modulators in various bodily fluids. However, their effect on neuroinflammation processes is not yet fully understood. In this review, we summarize the role of neuroinflammation in acute and chronic pathological conditions affecting the brain. Moreover, we seek to explore the existing literature on HSP-mediated inflammatory function within the central nervous system and compare the function of these proteins when they are localized intracellularly compared to being present in the extracellular milieu.
Collapse
Affiliation(s)
- Brigitta Dukay
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.,Doctoral School in Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Bálint Csoboz
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Melinda E Tóth
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| |
Collapse
|
28
|
Jayaraj RL, Azimullah S, Beiram R, Jalal FY, Rosenberg GA. Neuroinflammation: friend and foe for ischemic stroke. J Neuroinflammation 2019; 16:142. [PMID: 31291966 PMCID: PMC6617684 DOI: 10.1186/s12974-019-1516-2] [Citation(s) in RCA: 892] [Impact Index Per Article: 148.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/10/2019] [Indexed: 12/13/2022] Open
Abstract
Stroke, the third leading cause of death and disability worldwide, is undergoing a change in perspective with the emergence of new ideas on neurodegeneration. The concept that stroke is a disorder solely of blood vessels has been expanded to include the effects of a detrimental interaction between glia, neurons, vascular cells, and matrix components, which is collectively referred to as the neurovascular unit. Following the acute stroke, the majority of which are ischemic, there is secondary neuroinflammation that both promotes further injury, resulting in cell death, but conversely plays a beneficial role, by promoting recovery. The proinflammatory signals from immune mediators rapidly activate resident cells and influence infiltration of a wide range of inflammatory cells (neutrophils, monocytes/macrophages, different subtypes of T cells, and other inflammatory cells) into the ischemic region exacerbating brain damage. In this review, we discuss how neuroinflammation has both beneficial as well as detrimental roles and recent therapeutic strategies to combat pathological responses. Here, we also focus on time-dependent entry of immune cells to the ischemic area and the impact of other pathological mediators, including oxidative stress, excitotoxicity, matrix metalloproteinases (MMPs), high-mobility group box 1 (HMGB1), arachidonic acid metabolites, mitogen-activated protein kinase (MAPK), and post-translational modifications that could potentially perpetuate ischemic brain damage after the acute injury. Understanding the time-dependent role of inflammatory factors could help in developing new diagnostic, prognostic, and therapeutic neuroprotective strategies for post-stroke inflammation.
Collapse
Affiliation(s)
- Richard L. Jayaraj
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Sheikh Azimullah
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Rami Beiram
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Fakhreya Y. Jalal
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Gary A. Rosenberg
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131 USA
| |
Collapse
|
29
|
Safflower Yellow B Protects Brain against Cerebral Ischemia Reperfusion Injury through AMPK/NF-kB Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:7219740. [PMID: 30854014 PMCID: PMC6378026 DOI: 10.1155/2019/7219740] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 01/15/2019] [Indexed: 11/24/2022]
Abstract
Inflammation had showed its important role in the pathogenesis of cerebral ischemia and secondary damage. Safflower yellow B (SYB) had neuroprotective effects against oxidative stress-induced brain injuries, but the mechanisms were still largely unknown to us. In this study, we tried to investigate the anti-inflammation effects of SYB and the possible roles of AMPK/NF-κB signaling pathway on these protective effects. In vivo, brain ischemia/reperfusion (I/R) was induced by transient middle cerebral artery occlusion for 2 h and reperfusion for 20 h. Neurofunctional evaluation, infarction area, and brain water contents were measured. Brain injury markers and inflammatory cytokines levels were measured by ELISA kits. In vitro, cell viability, apoptosis, and LDH leakage were measured after I/R in PC12 cells. The expression and phosphorylation levels of AMPK, NF-κB p65, and P-IκB-α in cytoplasm and nuclear were measured by Western blotting. SiRNA experiment was performed to certify the role of AMPK. The results showed SYB reduced infarct size, improved neurological outcomes, and inhibited brain injury after I/R. In vitro test, SYB treatment alleviated PC12 cells injury and apoptosis and inhibited the inflammatory cytokines (IL-1, IL-6, TNF-α, and COX-2) in a dose-dependent manner. SYB treatment induced AMPK phosphorylation and inhibited NF-κB p65 nuclear translocation both in brain and in PC12 cells. Further studies also showed that the inhibition of NF-κB activity of SYB was through AMPK. In conclusion, SYB protected brain I/R injury through reducing expression of inflammatory cytokines and this effect might be partly due to the inhibition of NF-κB mediated by AMPK.
Collapse
|
30
|
Flurbiprofen axetil attenuates cerebral ischemia/reperfusion injury by reducing inflammation in a rat model of transient global cerebral ischemia/reperfusion. Biosci Rep 2018. [PMID: 29540536 PMCID: PMC6435563 DOI: 10.1042/bsr20171562] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Ischemic stroke has been ranked as the second cause of death in patients worldwide. Inflammation which is activated during cerebral ischemia/reperfusion (I/R) is an important mechanism leading to brain injury. The present study aimed to investigate the effect of flurbiprofen axetil on cerebral I/R injury and the role of inflammation in this process. Rats were subjected to sham operation or global cerebral I/R with or without flurbiprofen axetil (5 or 10 mg/kg). Global cerebral ischemia was achieved by occlusion of bilateral common carotid arteries combined with hypotension for 20 min followed by reperfusion for 72 h. Then the neurological deficit score, hippocampal cell apoptosis, levels of aquaporin (AQP) 4, AQP9, intercellular cell adhesion molecule-1 (ICAM-1), nuclear factor-κB (NF-κB), tumor necrosis factor (TNF-α), interleukin-1 β (IL-1β), thromboxane B2 (TXB2), and 6-keto-PGI1α were assessed. After reperfusion, neurological deficit score was significantly increased accompanied by severe neuronal damage (exacerbated morphological deficit, increased terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay (TUNEL)-positive cells and cleaved caspase-3 protein expression in hippocampal CA1 region). Cerebral I/R injury also enhanced expressions of TNF-α, IL-1β, NF-κB, AQP4 and AQP9 as well as TXB2 and TXB2/6-keto-PGI1α. All these changes were reversed by pretreatment with flurbiprofen axetil. Flurbiprofen axetil protects the brain from cerebral I/R injury through reducing inflammation and brain edema.
Collapse
|
31
|
Effects of Angiotensin-Converting Enzyme Inhibition on Circulating Endothelial Progenitor Cells in Patients with Acute Ischemic Stroke. Stem Cells Int 2018; 2018:2827580. [PMID: 29853909 PMCID: PMC5944289 DOI: 10.1155/2018/2827580] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/03/2018] [Accepted: 03/29/2018] [Indexed: 12/25/2022] Open
Abstract
Background Therapeutic neovascularization might represent an important strategy to salvage tissue after ischemia. Circulating bone marrow-derived endothelial progenitor cells (EPCs) were previously shown to augment the neovascularization of ischemic tissue. Angiotensin-converting enzyme inhibitors (ACEIs) might modulate EPC mobilization. We evaluated populations of circulating stem cells and early EPCs in acute ischemic stroke (AIS) patients and the effect of ACEI on circulating EPCs in these patients with respect to aspects of stroke pathogenesis. Methods We studied 43 AIS patients (group I), comprising 33 treated with ACEI (group Ia) and 10 untreated (group Ib). Risk factor controls (group II) included 22 subjects. EPCs were measured by flow cytometry. Results In AIS patients, the number of circulating stem cells and early EPCs upon admission was similar to that in control group individuals. There were no significant differences in the numbers of stem cells and early EPCs over subsequent days after AIS. There were also no significant differences in stem cell and early EPC numbers over the first 3 days between group Ia and group Ib. However, on day 7, these numbers were significantly higher in group Ib than in group Ia (p < 0.05). In AIS patients chronically treated with ACEI, there was a negative correlation between CD133+ cell number and neurological deficit on the first, third, and seventh days (p < 0.005). Conclusions An increased number of circulating stem cells and early EPCs were not observed in stroke patients chronically treated with ACEI. In patients chronically treated with ACEI, a significant correlation was observed between decreased neurological deficit and higher levels of CD133+ cells; this could be due to the positive influence of these cells on the regeneration of the endothelium and improved circulation in the ischemic penumbra.
Collapse
|
32
|
De Doncker W, Dantzer R, Ormstad H, Kuppuswamy A. Mechanisms of poststroke fatigue. J Neurol Neurosurg Psychiatry 2018; 89:287-293. [PMID: 28939684 DOI: 10.1136/jnnp-2017-316007] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/05/2017] [Accepted: 09/07/2017] [Indexed: 11/04/2022]
Abstract
Poststroke fatigue is a debilitating symptom and is poorly understood. Here we summarise molecular, behavioural and neurophysiological changes related to poststroke fatigue and put forward potential theories for mechanistic understanding of poststroke fatigue.
Collapse
Affiliation(s)
| | - Robert Dantzer
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Heidi Ormstad
- Faculty of Health and Social Sciences, University of South West Norway, Oslo, Norway
| | | |
Collapse
|
33
|
Hernandes MS, Lassègue B, Hilenski LL, Adams J, Gao N, Kuan CY, Sun YY, Cheng L, Kikuchi DS, Yepes M, Griendling KK. Polymerase delta-interacting protein 2 deficiency protects against blood-brain barrier permeability in the ischemic brain. J Neuroinflammation 2018; 15:45. [PMID: 29452577 PMCID: PMC5816395 DOI: 10.1186/s12974-017-1032-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 12/11/2017] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Polymerase δ-interacting protein 2 (Poldip2) is a multifunctional protein that regulates vascular extracellular matrix composition and matrix metalloproteinase (MMP) activity. The blood-brain barrier (BBB) is a dynamic system assembled by endothelial cells, basal lamina, and perivascular astrocytes, raising the possibility that Poldip2 may be involved in maintaining its structure. We investigated the role of Poldip2 in the late BBB permeability induced by cerebral ischemia. METHODS Transient middle cerebral artery occlusion (tMCAO) was induced in Poldip2+/+ and Poldip2+/- mice. The volume of the ischemic lesion was measured in triphenyltetrazolium chloride-stained sections. BBB breakdown was evaluated by Evans blue dye extravasation. Poldip2 protein expression was evaluated by western blotting. RT-PCR, zymography, and ELISAs were used to measure mRNA levels, activity, and protein levels of cytokines and MMPs. Cultured astrocytes were transfected with Poldip2 siRNA, and mRNA levels of cytokines were evaluated as well as IκBα protein degradation. RESULTS Cerebral ischemia induced the expression of Poldip2. Compared to Poldip2+/+ mice, Poldip2+/- animals exhibited decreased Evans blue dye extravasation and improved survival 24 h following stroke. Poldip2 expression was upregulated in astrocytes exposed to oxygen and glucose deprivation (OGD) and siRNA-mediated downregulation of Poldip2 abrogated OGD-induced IL-6 and TNF-α expression. In addition, siRNA against Poldip2 inhibited TNF-α-induced IκBα degradation. TNF-α, IL-6, MCP-1, VEGF, and MMP expression induced by cerebral ischemia was abrogated in Poldip2+/- mice. The protective effect of Poldip2 depletion on the increased permeability of the BBB was partially reversed by systemic administration of TNF-α. CONCLUSIONS Poldip2 is upregulated following ischemic stroke and mediates the breakdown of the BBB by increasing cerebral cytokine production and MMP activation. Therefore, Poldip2 appears to be a promising novel target for the development of therapeutic strategies to prevent the development of cerebral edema in the ischemic brain.
Collapse
Affiliation(s)
- Marina S Hernandes
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308 WMB, Atlanta, GA, 30322, USA
| | - Bernard Lassègue
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308 WMB, Atlanta, GA, 30322, USA
| | - Lula L Hilenski
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308 WMB, Atlanta, GA, 30322, USA
| | - Jonathan Adams
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Ning Gao
- Division of Neurology, Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Chia-Yi Kuan
- Division of Neurology, Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Yu-Yo Sun
- Division of Neurology, Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Lihong Cheng
- Department of Neurology, Emory University, Atlanta, GA, 30322, USA
| | - Daniel S Kikuchi
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308 WMB, Atlanta, GA, 30322, USA
| | - Manuel Yepes
- Department of Neurology, Emory University, Atlanta, GA, 30322, USA
- Division of Neuroscience, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Kathy K Griendling
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308 WMB, Atlanta, GA, 30322, USA.
| |
Collapse
|
34
|
Muñoz R, Santamaría E, Rubio I, Ausín K, Ostolaza A, Labarga A, Roldán M, Zandio B, Mayor S, Bermejo R, Mendigaña M, Herrera M, Aymerich N, Olier J, Gállego J, Mendioroz M, Fernández-Irigoyen J. Mass Spectrometry-Based Proteomic Profiling of Thrombotic Material Obtained by Endovascular Thrombectomy in Patients with Ischemic Stroke. Int J Mol Sci 2018; 19:ijms19020498. [PMID: 29414888 PMCID: PMC5855720 DOI: 10.3390/ijms19020498] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/01/2018] [Accepted: 02/02/2018] [Indexed: 12/25/2022] Open
Abstract
Thrombotic material retrieved from acute ischemic stroke (AIS) patients represents a valuable source of biological information. In this study, we have developed a clinical proteomics workflow to characterize the protein cargo of thrombi derived from AIS patients. To analyze the thrombus proteome in a large-scale format, we developed a workflow that combines the isolation of thrombus by endovascular thrombectomy and peptide chromatographic fractionation coupled to mass-spectrometry. Using this workflow, we have characterized a specific proteomic expression profile derived from four AIS patients included in this study. Around 1600 protein species were unambiguously identified in the analyzed material. Functional bioinformatics analyses were performed, emphasizing a clustering of proteins with immunological functions as well as cardiopathy-related proteins with blood-cell dependent functions and peripheral vascular processes. In addition, we established a reference proteomic fingerprint of 341 proteins commonly detected in all patients. Protein interactome network of this subproteome revealed protein clusters involved in the interaction of fibronectin with 14-3-3 proteins, TGFβ signaling, and TCP complex network. Taken together, our data contributes to the repertoire of the human thrombus proteome, serving as a reference library to increase our knowledge about the molecular basis of thrombus derived from AIS patients, paving the way toward the establishment of a quantitative approach necessary to detect and characterize potential novel biomarkers in the stroke field.
Collapse
Affiliation(s)
- Roberto Muñoz
- Department of Neurology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Enrique Santamaría
- Clinical Neuroproteomics Laboratory, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, IDISNA, Navarra Institute for Health Research, Pamplona 31008, Spain.
- Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, IDISNA, Navarra Institute for Health Research, Pamplona 31008, Spain.
| | - Idoya Rubio
- Department of Neurology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Karina Ausín
- Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, IDISNA, Navarra Institute for Health Research, Pamplona 31008, Spain.
| | - Aiora Ostolaza
- Department of Neurology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Alberto Labarga
- Bioinformatics Laboratory, Navarrabiomed-Departamento de Salud, Universidad Pública de Navarra, IDISNA, Navarra Institute for Health Research, Pamplona 31008, Spain.
| | - Miren Roldán
- Neuroepigenetics Laboratory, Navarrabiomed-Departamento de Salud, Universidad Pública de Navarra, IDISNA, Navarra Institute for Health Research, Pamplona 31008, Spain.
| | - Beatriz Zandio
- Department of Neurology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Sergio Mayor
- Department of Neurology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Rebeca Bermejo
- Department of Interventional Neuroradiology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Mónica Mendigaña
- Department of Interventional Neuroradiology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - María Herrera
- Department of Neurology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Nuria Aymerich
- Department of Neurology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Jorge Olier
- Department of Interventional Neuroradiology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Jaime Gállego
- Department of Neurology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Maite Mendioroz
- Department of Neurology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
- Neuroepigenetics Laboratory, Navarrabiomed-Departamento de Salud, Universidad Pública de Navarra, IDISNA, Navarra Institute for Health Research, Pamplona 31008, Spain.
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Laboratory, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, IDISNA, Navarra Institute for Health Research, Pamplona 31008, Spain.
- Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, IDISNA, Navarra Institute for Health Research, Pamplona 31008, Spain.
| |
Collapse
|
35
|
Vijayan M, Reddy PH. Stroke, Vascular Dementia, and Alzheimer's Disease: Molecular Links. J Alzheimers Dis 2018; 54:427-43. [PMID: 27567871 DOI: 10.3233/jad-160527] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Stroke is a brain disease that occurs when blood flow stops, resulting in reduced oxygen supply to neurons. Stroke occurs at any time and at any age, but increases after the age of 55. It is the second leading cause of death and the third leading cause of disability-adjusted, life-years. The pathophysiology of ischemic stroke is complex and recent molecular, cellular, and animal models and postmortem brain studies have revealed that multiple cellular changes have been implicated, including oxidative stress/mitochondrial dysfunction, inflammatory responses, micro RNA alterations, and marked changes in brain proteins. These cellular changes provide new information for developing therapeutic strategies for ischemic stroke treatment. Research also revealed that stroke increases with a number of modifiable factors and most strokes can be prevented and/or controlled through pharmacological or surgical interventions and lifestyle changes. Ischemic stroke is the major risk factor for vascular dementia and Alzheimer's disease. This review summarizes the latest research findings on stroke, including causal factors and molecular links between stroke and vascular disease/Alzheimer's disease.
Collapse
Affiliation(s)
- Murali Vijayan
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Neuroscience and Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Speech, Language and Hearing Sciences Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
36
|
Huang L, Shang E, Fan W, Li X, Li B, He S, Fu Y, Zhang Y, Li Y, Fang W. S-oxiracetam protect against ischemic stroke via alleviating blood brain barrier dysfunction in rats. Eur J Pharm Sci 2017; 109:40-47. [DOI: 10.1016/j.ejps.2017.07.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 06/20/2017] [Accepted: 07/21/2017] [Indexed: 12/14/2022]
|
37
|
Ramakrishnan A, Vijayakumar N. Urea cycle pathway targeted therapeutic action of naringin against ammonium chloride induced hyperammonemic rats. Biomed Pharmacother 2017; 94:1028-1037. [DOI: 10.1016/j.biopha.2017.08.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 08/04/2017] [Accepted: 08/04/2017] [Indexed: 12/25/2022] Open
|
38
|
Dietzel J, Haeusler KG, Endres M. Does atrial fibrillation cause cognitive decline and dementia? Europace 2017; 20:408-419. [DOI: 10.1093/europace/eux031] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 02/07/2017] [Indexed: 12/12/2022] Open
Affiliation(s)
- Joanna Dietzel
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Germany
| | - Karl Georg Haeusler
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Germany
- Department of Neurology, Charité - Universitätsmedizin Berlin, Germany
| | - Matthias Endres
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Germany
- Department of Neurology, Charité - Universitätsmedizin Berlin, Germany
- German Center for Cardiovascular Research (DZHK)
- German Center for Neurodegenerative Disease (DZNE)
- ExcellenceCluster NeuroCure, Charité- Universitätsmedizin Berlin, Germany
| |
Collapse
|
39
|
Zhang X, Xue X, Xian L, Guo Z, Ito Y, Sun W. Potential neuroprotection of protodioscin against cerebral ischemia-reperfusion injury in rats through intervening inflammation and apoptosis. Steroids 2016; 113:52-63. [PMID: 27343977 PMCID: PMC8725987 DOI: 10.1016/j.steroids.2016.06.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 06/09/2016] [Accepted: 06/15/2016] [Indexed: 11/16/2022]
Abstract
The aim of the current research is to investigate the cerebral-protection of protodioscin on a transient cerebral ischemia-reperfusion (I/R) model and to explore its possible underlying mechanisms. The rats were preconditioned with protodioscin at the doses of 25 and 50mgkg(-1) prior to surgery. Then the animals were subjected to right middle cerebral artery occlusion (MCAO) using an intraluminal method by inserting a thread (90min surgery). After the blood flow was restored in 24h via withdrawing the thread, some representative indicators for the cerebral injury were evaluated by various methods including TTC-staining, TUNEL, immunohistochemistry, and Western blotting. As compared with the operated rats without drug intervening, treatment with protodioscin apparently lowered the death rate and improved motor coordination abilities through reducing the deficit scores and cerebral infarct volume. What's more, an apparent decrease in neuron apoptosis detected in hippocampus CA1 and cortex of the ipsilateral hemisphere might attribute to alleviate the increase in Caspase-3 and Bax/Bcl-2 ratio. Meanwhile, concentrations of several main pro-inflammatory cytokines (TNF-α, IL-1β and IL-6) in the serum were also significantly suppressed. Finally, the NF-κB and IκBa protein expressions in the cytoplasm of right injured brain were remarkably up-regulated, while NF-κB in nucleus was down-regulated. Therefore, these observed findings demonstrated that protodioscin appeared to reveal potential neuroprotection against the I/R injury due to its anti-inflammatory and anti-apoptosis properties. This therapeutic effect was probably mediated by the inactivation of NF-κB signal pathways.
Collapse
Affiliation(s)
- Xinxin Zhang
- College of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xuanji Xue
- College of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Liang Xian
- College of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Zengjun Guo
- College of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Yoichiro Ito
- Laboratory of Bioseparation Technology, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Wenji Sun
- Biomedicine Key Laboratory of Shaanxi Province, Northwest University, Xi'an 710069, China.
| |
Collapse
|
40
|
Madsen PM, Clausen BH, Degn M, Thyssen S, Kristensen LK, Svensson M, Ditzel N, Finsen B, Deierborg T, Brambilla R, Lambertsen KL. Genetic ablation of soluble tumor necrosis factor with preservation of membrane tumor necrosis factor is associated with neuroprotection after focal cerebral ischemia. J Cereb Blood Flow Metab 2016; 36:1553-69. [PMID: 26661199 PMCID: PMC5012516 DOI: 10.1177/0271678x15610339] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 09/07/2015] [Indexed: 11/16/2022]
Abstract
Microglia respond to focal cerebral ischemia by increasing their production of the neuromodulatory cytokine tumor necrosis factor, which exists both as membrane-anchored tumor necrosis factor and as cleaved soluble tumor necrosis factor forms. We previously demonstrated that tumor necrosis factor knockout mice display increased lesion volume after focal cerebral ischemia, suggesting that tumor necrosis factor is neuroprotective in experimental stroke. Here, we extend our studies to show that mice with intact membrane-anchored tumor necrosis factor, but no soluble tumor necrosis factor, display reduced infarct volumes at one and five days after stroke. This was associated with improved functional outcome after experimental stroke. No changes were found in the mRNA levels of tumor necrosis factor and tumor necrosis factor-related genes (TNFR1, TNFR2, TACE), pro-inflammatory cytokines (IL-1β, IL-6) or chemokines (CXCL1, CXCL10, CCL2); however, protein expression of TNF, IL-1β, IL-6 and CXCL1 was reduced in membrane-anchored tumor necrosis factor(Δ/Δ) compared to membrane-anchored tumor necrosis factor(wt/wt) mice one day after experimental stroke. This was paralleled by reduced MHCII expression and a reduction in macrophage infiltration in the ipsilateral cortex of membrane-anchored tumor necrosis factor(Δ/Δ) mice. Collectively, these findings indicate that membrane-anchored tumor necrosis factor mediates the protective effects of tumor necrosis factor signaling in experimental stroke, and therapeutic strategies specifically targeting soluble tumor necrosis factor could be beneficial in clinical stroke therapy.
Collapse
Affiliation(s)
- Pernille M Madsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA
| | - Bettina H Clausen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Matilda Degn
- Molecular Sleep Lab, Department of Diagnostics, Glostrup Hospital, Glostrup, Denmark
| | - Stine Thyssen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Lotte K Kristensen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Martina Svensson
- Department of Experimental Medical Sciences, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Nicholas Ditzel
- KMEB, Molecular Endocrinology, Odense University Hospital, Odense, Denmark
| | - Bente Finsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Tomas Deierborg
- Department of Experimental Medical Sciences, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Roberta Brambilla
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA
| | - Kate L Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
41
|
Kawabori M, Yenari MA. Inflammatory responses in brain ischemia. Curr Med Chem 2016; 22:1258-77. [PMID: 25666795 DOI: 10.2174/0929867322666150209154036] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/02/2014] [Accepted: 02/02/2015] [Indexed: 12/20/2022]
Abstract
Brain infarction causes tissue death by ischemia due to occlusion of the cerebral vessels and recent work has shown that post stroke inflammation contributes significantly to the development of ischemic pathology. Because secondary damage by brain inflammation may have a longer therapeutic time window compared to the rescue of primary damage following arterial occlusion, controlling inflammation would be an obvious therapeutic target. A substantial amount of experimentall progress in this area has been made in recent years. However, it is difficult to elucidate the precise mechanisms of the inflammatory responses following ischemic stroke because inflammation is a complex series of interactions between inflammatory cells and molecules, all of which could be either detrimental or beneficial. We review recent advances in neuroinflammation and the modulation of inflammatory signaling pathways in brain ischemia. Potential targets for treatment of ischemic stroke will also be covered. The roles of the immune system and brain damage versus repair will help to clarify how immune modulation may treat stroke.
Collapse
Affiliation(s)
| | - Midori A Yenari
- Dept. of Neurology, University of California, San Francisco and the San Francisco Veterans Affairs Medical Center, 4150 Clement Street, San Francisco, CA 94121, USA.
| |
Collapse
|
42
|
Xiang Y, Zhao H, Wang J, Zhang L, Liu A, Chen Y. Inflammatory mechanisms involved in brain injury following cardiac arrest and cardiopulmonary resuscitation. Biomed Rep 2016; 5:11-17. [PMID: 27330748 DOI: 10.3892/br.2016.677] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/25/2016] [Indexed: 12/24/2022] Open
Abstract
Cardiac arrest (CA) is a leading cause of fatality and long-term disability worldwide. Recent advances in cardiopulmonary resuscitation (CPR) have improved survival rates; however, the survivors are prone to severe neurological injury subsequent to successful CPR following CA. Effective therapeutic options to protect the brain from CA remain limited, due to the complexities of the injury cascades caused by global cerebral ischemia/reperfusion (I/R). Although the precise mechanisms of neurological impairment following CA-initiated I/R injury require further clarification, evidence supports that one of the key cellular pathways of cerebral injury is inflammation. The inflammatory response is orchestrated by activated glial cells in response to I/R injury. Increased release of danger-associated molecular pattern molecules and cellular dysfunction in activated microglia and astrocytes contribute to ischemia-induced cytotoxic and pro-inflammatory cytokines generation, and ultimately to delayed death of neurons. Furthermore, cytokines and adhesion molecules generated within activated microglia, as well as astrocytes, are involved in the innate immune response; modulate influx of peripheral immune and inflammatory cells into the brain, resulting in neurological injury. The present review discusses the molecular aspects of immune and inflammatory mechanisms in global cerebral I/R injury following CA and CPR, and the potential therapeutic strategies that target neuroinflammation and the innate immune system.
Collapse
Affiliation(s)
- Yanxiao Xiang
- Department of Clinical Pharmacy, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China; Department of Emergency, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Hua Zhao
- Department of Orthopedics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jiali Wang
- Chest Pain Center, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China; Institute of Emergency and Critical Care Medicine, Shandong University, Jinan, Shandong 250012, P.R. China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Luetao Zhang
- Chest Pain Center, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China; Institute of Emergency and Critical Care Medicine, Shandong University, Jinan, Shandong 250012, P.R. China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Anchang Liu
- Department of Clinical Pharmacy, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yuguo Chen
- Department of Emergency, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China; Chest Pain Center, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China; Institute of Emergency and Critical Care Medicine, Shandong University, Jinan, Shandong 250012, P.R. China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
43
|
Pettigrew LC, Kryscio RJ, Norris CM. The TNFα-Transgenic Rat: Hippocampal Synaptic Integrity, Cognition, Function, and Post-Ischemic Cell Loss. PLoS One 2016; 11:e0154721. [PMID: 27144978 PMCID: PMC4856338 DOI: 10.1371/journal.pone.0154721] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 04/18/2016] [Indexed: 11/18/2022] Open
Abstract
The cytokine, tumor necrosis factor α (TNFα), is a key regulator of neuroinflammation linked to numerous neurodegenerative conditions and diseases. The present study used transgenic rats that overexpress a murine TNFα gene, under the control of its own promoter, to investigate the impact of chronically elevated TNFα on hippocampal synaptic function. Neuronal viability and cognitive recovery in TNFα Tg rats were also determined following an ischemic insult arising from reversible middle cerebral artery occlusion (MCAO). Basal CA3-CA1 synaptic strength, recorded in acute brain slices, was not significantly different between eight-week-old TNFα Tg rats and non-Tg rats. In contrast, slices from TNFα Tg rats showed significantly greater levels of long-term potentiation (LTP) in response to 100 Hz stimulation, suggesting that synaptic networks may be hyperexcitable in the context of elevated TNFα. Cognitive and motor deficits (assessed on the Morris Water Maze and Rotarod task, respectively) were present in TNFα Tg rats in the absence of significant differences in the loss of cortical and hippocampal neurons. TNF overexpression exacerbated MCAO-dependent deficits on the rotarod, but ameliorated cortical neuron loss in response to MCAO.
Collapse
Affiliation(s)
- L. Creed Pettigrew
- Paul G. Blazer, Jr. Stroke Research Laboratory, University of Kentucky, Lexington, Kentucky, United States of America
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Neurology, University of Kentucky, Lexington, Kentucky, United States of America
- Veterans Administration (VA) Medical Center, Lexington, Kentucky, United States of America
- * E-mail:
| | - Richard J. Kryscio
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Statistics and School of Public Health, University of Kentucky, Lexington, Kentucky, United States of America
| | - Christopher M. Norris
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, United States of America
| |
Collapse
|
44
|
Xing C, Lo EH. Help-me signaling: Non-cell autonomous mechanisms of neuroprotection and neurorecovery. Prog Neurobiol 2016; 152:181-199. [PMID: 27079786 DOI: 10.1016/j.pneurobio.2016.04.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 04/06/2016] [Accepted: 04/09/2016] [Indexed: 12/11/2022]
Abstract
Self-preservation is required for life. At the cellular level, this fundamental principle is expressed in the form of molecular mechanisms for preconditioning and tolerance. When the cell is threatened, internal cascades of survival signaling become triggered to protect against cell death and defend against future insults. Recently, however, emerging findings suggest that this principle of self-preservation may involve not only intracellular signals; the release of extracellular signals may provide a way to recruit adjacent cells into an amplified protective program. In the central nervous system where multiple cell types co-exist, this mechanism would allow threatened neurons to "ask for help" from glial and vascular compartments. In this review, we describe this new concept of help-me signaling, wherein damaged or diseased neurons release signals that may shift glial and vascular cells into potentially beneficial phenotypes, and help remodel the neurovascular unit. Understanding and dissecting these non-cell autonomous mechanisms of self-preservation in the CNS may lead to novel opportunities for neuroprotection and neurorecovery.
Collapse
Affiliation(s)
- Changhong Xing
- Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA.
| | - Eng H Lo
- Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA.
| |
Collapse
|
45
|
Rainer TH, Leung LY, Chan CPY, Leung YK, Abrigo JM, Wang D, Graham CA. Plasma miR-124-3p and miR-16 concentrations as prognostic markers in acute stroke. Clin Biochem 2016; 49:663-668. [PMID: 26968104 DOI: 10.1016/j.clinbiochem.2016.02.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/19/2016] [Accepted: 02/24/2016] [Indexed: 11/29/2022]
Abstract
OBJECTIVES This study aimed to investigate plasma concentrations of miR-124-3p and miR-16 as prognostic markers in emergency department patients with acute stroke. DESIGN AND METHODS Plasma concentrations of miR-124-3p and miR-16 of 84 stroke patients (presenting to the emergency department within 24h from onset of symptoms) were determined by RT-qPCR. The primary outcome measure was 3-month mortality and the secondary outcome measure was post-stroke modified Rankin Score (mRS). RESULTS Twelve patients (14.3%) died within 3months of hospital admission and forty-one (48.8%) patients as achieved a 3-month mRS>2. Median plasma miR-124-3p concentrations were elevated in patients who died compared to patients who survived (p=0.0052), and its levels were found to be higher in patients with a 3-month mRS>2 compared with patients with mRS≤2 (p=0.0312). Higher plasma miR-16 concentrations were observed in patients who survived than in patients who died (p=0.0394), while its concentrations were lower in patients achieving mRS>2 than in patients with mRS≤2 (p=0.0124). For a subgroup of cases presenting to the emergency department within 6h from time of symptom onset (n=36), plasma miR-124-3p concentrations predicted 3-month mortality with an area under the ROC curve of 0.87 (95%CI: 0.72-0.96). CONCLUSIONS Plasma miR-124-3p and miR-16 are molecular markers which could be useful for the early prediction of mortality and mRS.
Collapse
Affiliation(s)
- Timothy Hudson Rainer
- Accident and Emergency Medicine Academic Unit, The Chinese University of Hong Kong, Hong Kong
| | - Ling Yan Leung
- Accident and Emergency Medicine Academic Unit, The Chinese University of Hong Kong, Hong Kong
| | - Cangel Pui Yee Chan
- Accident and Emergency Medicine Academic Unit, The Chinese University of Hong Kong, Hong Kong
| | - Yuk Ki Leung
- Accident and Emergency Medicine Academic Unit, The Chinese University of Hong Kong, Hong Kong
| | - Jill Morales Abrigo
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong
| | - Defeng Wang
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong
| | - Colin A Graham
- Accident and Emergency Medicine Academic Unit, The Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|
46
|
Zhang Q, Li CS, Wang S, Gu W. Effects of Chinese medicine shen-fu injection on the expression of inflammatory cytokines and complements during post-resuscitation immune dysfunction in a porcine model. Chin J Integr Med 2016; 22:101-9. [PMID: 25253551 DOI: 10.1007/s11655-014-1857-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Indexed: 01/03/2023]
Abstract
OBJECTIVE To investigate the action of Shen-Fu Injection (SFI) in regulating the expression of the serum complements and inflammatory cytokines synthesized and released in response to the stress of global ischemia accompanying cardiac arrest (CA) and resuscitation. METHODS Thirty pigs were randomly divided into the sham (n=6) and 3 returns of spontaneous circulation (ROSC) groups (n=24). After 8-min untreated ventricular fibrillation and 2-min basic life support, 24 pigs of the ROSC groups were randomized into three groups (n=8 per group), which received central venous injection of SFI (SFI group), epinephrine (EP group), or saline (SA group). Hemodynamic status and blood samples were obtained at 0, 0.5, 1, 2, 4, 6, 12, and 24 h after ROSC. RESULTS Serum concentrations of specific activation markers of the complement system C3, C4 and C5b-9 were increased during cardiopulmonary resuscitation through 24 h after ROSC. There were intense changes of various pro-inflammatory cytokines and anti-inflammatory cytokines as early as 0.5 h after CA. Compared with the EP and SA groups, SFI treatment reduced the proinflammatory cytokines levels of interleukin (IL)-6, IL-8 and tumor necrosis factor α (TNF-α, P<0.05), and increased the anti-inflammatory cytokine levels of IL-4 and IL-10 (P<0.05). Further, SFI treatment decreased the values of C3, C4 and C5b-9 compared with the EP and SA groups. CONCLUSIONS SFI, derived from the ancient Chinese medicine, has significant effects in attenuating post-resuscitation immune dysfunction by modulating the expression of complements and cytokines levels. The current study provided an experimental basis for the clinical application of a potential pharmacologic target for post resuscitation immune dysfunction.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Chun-sheng Li
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Shuo Wang
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Wei Gu
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| |
Collapse
|
47
|
Sutherland BA, Neuhaus AA, Couch Y, Balami JS, DeLuca GC, Hadley G, Harris SL, Grey AN, Buchan AM. The transient intraluminal filament middle cerebral artery occlusion model as a model of endovascular thrombectomy in stroke. J Cereb Blood Flow Metab 2016; 36:363-9. [PMID: 26661175 PMCID: PMC4759672 DOI: 10.1177/0271678x15606722] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/19/2015] [Indexed: 01/10/2023]
Abstract
The clinical relevance of the transient intraluminal filament model of middle cerebral artery occlusion (tMCAO) has been questioned due to distinct cerebral blood flow profiles upon reperfusion between tMCAO (abrupt reperfusion) and alteplase treatment (gradual reperfusion), resulting in differing pathophysiologies. Positive results from recent endovascular thrombectomy trials, where the occluding clot is mechanically removed, could revolutionize stroke treatment. The rapid cerebral blood flow restoration in both tMCAO and endovascular thrombectomy provides clinical relevance for this pre-clinical model. Any future clinical trials of neuroprotective agents as adjuncts to endovascular thrombectomy should consider tMCAO as the model of choice to determine pre-clinical efficacy.
Collapse
Affiliation(s)
- Brad A Sutherland
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Ain A Neuhaus
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Yvonne Couch
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Joyce S Balami
- Centre for Evidence Based Medicine, University of Oxford, Oxford, UK Norfolk and Norwich University Teaching Hospital NHS Trust, Norwich, UK
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Gina Hadley
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Scarlett L Harris
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Adam N Grey
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Alastair M Buchan
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
48
|
Yang Z, Weian C, Susu H, Hanmin W. Protective effects of mangiferin on cerebral ischemia–reperfusion injury and its mechanisms. Eur J Pharmacol 2016; 771:145-51. [DOI: 10.1016/j.ejphar.2015.12.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 11/23/2015] [Accepted: 12/01/2015] [Indexed: 01/16/2023]
|
49
|
Han D, Fang W, Zhang R, Wei J, Kodithuwakku ND, Sha L, Ma W, Liu L, Li F, Li Y. Clematichinenoside protects blood brain barrier against ischemic stroke superimposed on systemic inflammatory challenges through up-regulating A20. Brain Behav Immun 2016; 51:56-69. [PMID: 26231971 DOI: 10.1016/j.bbi.2015.07.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 07/16/2015] [Accepted: 07/27/2015] [Indexed: 12/20/2022] Open
Abstract
Suppression of excessive inflammation can ameliorate blood brain barrier (BBB) injury, which shows therapeutic potential for clinical treatment of brain injury induced by stroke superimposed on systemic inflammatory diseases. In this study, we investigated whether and how clematichinenoside (AR), an anti-inflammatory triterpene saponin, protects brain injury from stroke superimposed on systemic inflammation. Lipopolysaccharide (LPS) was intraperitoneally injected immediately after middle cerebral artery occlusion (MCAO) in rats. Rat microvessel endothelial cells (rBMECs) were exposed to hypoxia/reoxygenation (H/R) coexisting with LPS. The results revealed that AR suppressed the excessive inflammation, restored BBB dysfunction, alleviated brain edema, decreased neutrophil infiltration, lessened neurological dysfunction, and decreased infarct rate. Further study demonstrated that the expression of nucleus nuclear factor kappa B (NF-κB), inducible nitric oxide synthase (iNOS), intercellular adhesion molecule-1 (ICAM-1), tumor necrosis factor-α (TNF-α) and interlukin-1β (IL-1β) were suppressed by AR via zinc finger protein A20. Besides, AR increased in vitro BBB integrity through A20. In conclusion, AR alleviated cerebral inflammatory injury through A20-NF-κB signal pathway, offering an alternative medication for stroke associated with systemic inflammatory diseases.
Collapse
Affiliation(s)
- Dan Han
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Rui Zhang
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jie Wei
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Nandani Darshika Kodithuwakku
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Lan Sha
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Wenhuan Ma
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Lifang Liu
- Department of Pharmacognosy and the Key Laboratory of Modern Chinese Medicines, Ministry of Education, China Pharmaceutical University, 210009, PR China
| | - Fengwen Li
- Department of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yunman Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
50
|
Targeting of Tumor Necrosis Factor Alpha Receptors as a Therapeutic Strategy for Neurodegenerative Disorders. Antibodies (Basel) 2015. [DOI: 10.3390/antib4040369] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|