1
|
Miao Y, Yan T, Liu J, Zhang C, Yan J, Xu L, Zhang N, Zhang X. Meta-analysis of the association between interleukin-17 and ischemic cardiovascular disease. BMC Cardiovasc Disord 2024; 24:252. [PMID: 38750443 PMCID: PMC11097571 DOI: 10.1186/s12872-024-03897-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 04/22/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Interleukin-17 (IL-17) has been hypothesized to be involved in ischemic cardiovascular disease (ICVD). However, the association of IL-17 with ICVD remained unclear. The aim of this study was to systematically analyze the available evidence regarding the association between IL-17 and ICVD. METHODS We searched the PubMed, Web of Science, Cochrane Library, and Embase databases up to October 2023 to identify publications on the association between IL-17 and ICVD. The merged results were analyzed using a random effects model for meta-analysis and subgroup analysis. RESULTS A total of 955 publications were initially identified in our search and screened; six studies were eventually included in the analysis. The average age of study participants was 60.3 ± 12.6 years and 65.5% were men. There was a high degree of heterogeneity among studies. The results showed that IL-17 level were higher in the case group than those in the control group (standardized mean difference, SMD = 1.60, 95% confidence interval (95% CI): 0.53-2.66, P = 0.003). In sensitivity analysis, the merged results showed good robustness. Additionally, subgroup analysis showed that race and ethnicity, sample size, and detection methods were significant factors influencing heterogeneity in the published studies. CONCLUSION Our finding revealed that increased IL-17 level contributed to the development of ICVD, suggesting IL-17 as a potential risk marker. Further research is needed to establish IL-17 as a therapeutic biomarker of ICVD.
Collapse
Affiliation(s)
- Yu Miao
- Department of Health Statistics, Public Health College, Inner Mongolia Medical University, Hohhot, Inner Mongolia Province, 010000, China
| | - Tao Yan
- Department of Health Statistics, Public Health College, Inner Mongolia Medical University, Hohhot, Inner Mongolia Province, 010000, China
| | - Jia Liu
- Department of Health Statistics, Public Health College, Inner Mongolia Medical University, Hohhot, Inner Mongolia Province, 010000, China
| | - Chunfa Zhang
- Department of Health Statistics, Public Health College, Inner Mongolia Medical University, Hohhot, Inner Mongolia Province, 010000, China
| | - Jinli Yan
- Department of Health Statistics, Public Health College, Inner Mongolia Medical University, Hohhot, Inner Mongolia Province, 010000, China
| | - Lei Xu
- Department of Health Statistics, Public Health College, Inner Mongolia Medical University, Hohhot, Inner Mongolia Province, 010000, China
| | - Nan Zhang
- Department of Health Statistics, Public Health College, Inner Mongolia Medical University, Hohhot, Inner Mongolia Province, 010000, China.
| | - Xingguang Zhang
- Department of Health Statistics, Public Health College, Inner Mongolia Medical University, Hohhot, Inner Mongolia Province, 010000, China.
| |
Collapse
|
2
|
Hu J, Wang Z, Gong B, Feng L, Song Y, Zhang S, Wang L, Qu Y, Li G, Zhang L, Zheng C, Du F, Li P, Wang Y. IFN-γ promotes radioresistant Nestin-expressing progenitor regeneration in the developing cerebellum by augmenting Shh ligand production. CNS Neurosci Ther 2024; 30:e14485. [PMID: 37789668 PMCID: PMC10805445 DOI: 10.1111/cns.14485] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 09/05/2023] [Accepted: 09/18/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND Patients with brain tumors, especially pediatric brain tumors such as cerebellar medulloblastoma, always suffer from the severe side effects of radiotherapy. Regeneration of neural cells in irradiation-induced cerebellar injury has been reported, but the underlying mechanism remains elusive. METHODS We established an irradiation-induced developing cerebellum injury model in neonatal mice. Microarray, KEGG analysis and semi in vivo slice culture were performed for mechanistic study. RESULTS Nestin-expressing progenitors (NEPs) but not granule neuron precursors (GNPs) were resistant to irradiation and able to regenerate after irradiation. NEPs underwent less apoptosis but similar DNA damage following irradiation compared with GNPs. Subsequently, they started to proliferate and contributed to granule neurons regeneration dependent on the sonic hedgehog (Shh) pathway. In addition, irradiation increased Shh ligand provided by Purkinje cells. And microglia accumulated in the irradiated cerebellum producing more IFN-γ, which augmented Shh ligand production to promote NEP proliferation. CONCLUSIONS NEP was radioresistant and regenerative. IFN-γ was increased post irradiation to upregulate Shh ligand, contributing to NEP regeneration. Our study provides insight into the mechanisms of neural cell regeneration in irradiation injury of the developing cerebellum and will help to develop new therapeutic targets for minimizing the side effects of radiotherapy for brain tumors.
Collapse
Affiliation(s)
- Jian Hu
- Pediatric Cancer Center, Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical SciencesSoochow UniversitySuzhouChina
| | - Zixuan Wang
- Pediatric Cancer Center, Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical SciencesSoochow UniversitySuzhouChina
| | - Biao Gong
- Pediatric Cancer Center, Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical SciencesSoochow UniversitySuzhouChina
| | - Liyuan Feng
- Department of Pharmacognosy and Traditional Chinese Pharmacology, College of PharmacyArmy Medical UniversityChongqingChina
| | - Yan Song
- Pediatric Cancer Center, Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical SciencesSoochow UniversitySuzhouChina
| | - Shuo Zhang
- Pediatric Cancer Center, Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical SciencesSoochow UniversitySuzhouChina
| | - Lin Wang
- Pediatric Cancer Center, Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical SciencesSoochow UniversitySuzhouChina
| | - Yanghui Qu
- Pediatric Cancer Center, Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical SciencesSoochow UniversitySuzhouChina
| | - Gen Li
- Pediatric Cancer Center, Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical SciencesSoochow UniversitySuzhouChina
| | - Li Zhang
- Pediatric Cancer Center, Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical SciencesSoochow UniversitySuzhouChina
| | - Chaonan Zheng
- Pediatric Cancer Center, Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical SciencesSoochow UniversitySuzhouChina
| | - Fang Du
- Pediatric Cancer Center, Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical SciencesSoochow UniversitySuzhouChina
| | - Peng Li
- Department of Pharmacognosy and Traditional Chinese Pharmacology, College of PharmacyArmy Medical UniversityChongqingChina
| | - Yuan Wang
- Pediatric Cancer Center, Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical SciencesSoochow UniversitySuzhouChina
| |
Collapse
|
3
|
Radpour M, Khoshkroodian B, Asgari T, Pourbadie HG, Sayyah M. Interleukin 4 Reduces Brain Hyperexcitability after Traumatic Injury by Downregulating TNF-α, Upregulating IL-10/TGF-β, and Potential Directing Macrophage/Microglia to the M2 Anti-inflammatory Phenotype. Inflammation 2023; 46:1810-1831. [PMID: 37259014 DOI: 10.1007/s10753-023-01843-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/14/2023] [Accepted: 05/16/2023] [Indexed: 06/02/2023]
Abstract
Macrophage/microglia are activated after Traumatic brain injury (TBI), transform to inflammatory phenotype (M1) and trigger neuroinflammation, which provokes epileptogenesis. Interleukin-4 (IL-4) is a well-known drive of macrophage/microglia to the anti-inflammatory phenotype (M2). We tested effect of IL-4 on speed of epileptogenesis, brain expression of inflammatory and anti-inflammatory cytokines, and lesion size in TBI-injured male rats. Rats underwent TBI by Controlled Cortical Impact. Then 100 ng IL-4 was injected into cerebral ventricles. One day after TBI, pentylenetetrazole (PTZ) kindling started and development of generalized seizures was recorded. The lesion size, cell survival rate, TNF-α, TGF-β, IL-10, and Arginase1 (Arg1) was measured in the brain 6 h, 12 h, 24 h, 48 h, and 5 days after TBI. Astrocytes and macrophage/microglia activation/polarization was assessed by GFAP/Arg1 and Iba1/Arg1 immunostaining. TBI-injured rats were kindled by 50% less PTZ injections than control and sham-operated rats. IL-4 did not change kindling rate in sham-operated rats but inhibited acceleration of kindling rate in the TBI-injured rats. IL-4 decreased damage volume and number of destroyed neurons. IL-4 stopped TNF-α whereas upregulated TGF-β, IL-10, and Arg1 expressions. Iba1/Arg1 positive macrophage/microglia was notably increased 48 h after IL-4 administration. IL-4 suppresses TBI-induced acceleration of epileptogenesis in rats by directing TBI neuroinflammation toward an anti-inflammatory tone and inhibition of cell death.
Collapse
Affiliation(s)
- Mozhdeh Radpour
- Department of Physiology and Pharmacology, Pasteur Institute of Iran , Tehran, Iran
| | - Bahar Khoshkroodian
- Department of Physiology and Pharmacology, Pasteur Institute of Iran , Tehran, Iran
| | - Tara Asgari
- Department of Physiology and Pharmacology, Pasteur Institute of Iran , Tehran, Iran
| | | | - Mohammad Sayyah
- Department of Physiology and Pharmacology, Pasteur Institute of Iran , Tehran, Iran.
| |
Collapse
|
4
|
Fukasawa M, Nishio K, Oikawa D, Itou T, Iinuma T, Asano M. Allograft inflammatory factor-1 released from the cerebral microglia affect several organs in the body. J Mol Histol 2023; 54:147-156. [PMID: 36877416 DOI: 10.1007/s10735-023-10116-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 02/08/2023] [Indexed: 03/07/2023]
Abstract
Allograft inflammatory factor-1 (AIF-1) is expressed in microglia. Unilateral common carotid artery occlusion (UCCAO) was conducted to elucidate mechanisms that regulate AIF-1 expression in C57BL/6 male mice. Immunohistochemical reactivity of microglia against anti-AIF-1 antibody was increased significantly in the brain of this model. The increased AIF-1 production was further confirmed by ELISA using brain homogenate. Real-time PCR demonstrated that the increased AIF-1 production was regulated at the transcriptional level. Serum AIF-1 levels were further examined by ELISA and marked increase was observed on Day 1 of UCCAO. To examine the influence of AIF-1, immunohistochemical staining was performed and revealed that the immunoreactivity against anti-Iba-1 antibody was significantly increased in various organs. Among them, the accumulation of Iba-1+ cells were observed prominently in the spleen. Intraperitoneal injection of minocycline, a potent microglia inhibitor, reduced the number of Iba-1+ cells suggesting microglia activation-dependent accumulation. Based on these results, AIF-1 expression was further examined in the murine microglia cell line MG6. AIF-1 mRNA expression and secretion were up-regulated when the cells were cultured under hypoxic condition. Importantly, stimulation of the cells with recombinant AIF-1 induced the expression of AIF-1 mRNA. These results may suggest that increased AIF-1 production by microglia in cerebral ischemia regulate the AIF-1 mRNA expression at least in part by an autocrine manner.
Collapse
Affiliation(s)
- Mai Fukasawa
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry, Tokyo, Japan.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Kensuke Nishio
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry, Tokyo, Japan.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Daichi Oikawa
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry, Tokyo, Japan.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Tomoka Itou
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry, Tokyo, Japan.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Toshimitsu Iinuma
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry, Tokyo, Japan.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Masatake Asano
- Department of Pathology, Nihon University School of Dentistry, Tokyo, Japan. .,Division of Immunology and Pathobiology, Nihon University School of Dentistry, Tokyo, Japan.
| |
Collapse
|
5
|
Exaggerated levels of some specific TLRs, cytokines and chemokines in Japanese encephalitis infected BV2 and neuro 2A cell lines associated with worst outcome. Virol J 2023; 20:16. [PMID: 36707891 PMCID: PMC9881527 DOI: 10.1186/s12985-023-01966-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 01/04/2023] [Indexed: 01/29/2023] Open
Abstract
Japanese encephalitis (JE) disease, a viral brain fever is caused by Japanese encephalitis virus (JEV). Despite the availability of effective vaccines against this deadly infection, JE is the leading cause of epidemic viral encephalitis in children in South-east Asia. There is no treatment available for the JE disease which might be due to incomplete understanding of the pathogenesis of JE virus. The JEV infections lead to permanent neurological deficits even in those who survive from the infection. Activated microglia may play a potentially detrimental role by eliciting the expression of pro-inflammatory cytokines such as interleukin (IL)-1β, IL-6, and tumor necrosis factor-α (TNF-α) influencing the surrounding brain tissue. Microglial activation, proinflammatory cytokine release and leukocytes trafficking are associated following JEV infection in central nervous system (CNS). How the pattern recognition receptors sense the viral nucleic acid and how the microglial and neuronal cells behaves following JEV infection is still unelucidated. There is scarcity of data on the expression levels of toll like receptors (TLRs), cytokines and chemokines in JEV infection in invitro model. To explore the molecular mechanisms of JEV infection of microglial cells and neuronal cells, we studied the expression profile of TLRs, cytokines and chemokines in JEV infected microglial cell line BV2 and Neuronal cell line Neuro 2A. For the present study, we developed the mouse model of encephalitis by intracerebral (IC) injection of JE virus for virus propagation, disease progression and damage study. Our results demonstrate the exaggerated release of some specific TLRs, cytokines and chemokines in invitro cell culture of microglial and Neuro 2A cell line, which are associated with bad outcome in invivo study.
Collapse
|
6
|
An J, Li H, Xia D, Xu B, Wang J, Qiu H, He J. The role of interleukin-17 in epilepsy. Epilepsy Res 2022; 186:107001. [PMID: 35994860 DOI: 10.1016/j.eplepsyres.2022.107001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/26/2022] [Accepted: 08/11/2022] [Indexed: 11/30/2022]
Abstract
Epilepsy is a common neurological disorder that seriously affects human health. It is a chronic central nervous system dysfunction caused by abnormal discharges of neurons. About 50 million patients worldwide are affected by epilepsy. Although epileptic symptoms of most patients are controllable, some patients with refractory epilepsy have no response to antiseizure medications. It is necessary to investigate the pathogenesis of epilepsy and identify new therapeutic targets for refractory epilepsy. Epileptic disorders often accompany cerebral inflammatory reactions. Recently, the role of inflammation in the onset of epilepsy has increasingly attracted attention. The activation of both innate and adaptive immunity plays a significant role in refractory epilepsy. According to several clinical studies, interleukin-17, an essential inflammatory mediator linking innate and adaptive immunity, increased significantly in the body liquid and epileptic focus of patients with epilepsy. Experimental studies also indicated that interleukin-17 participated in epileptogenesis through various mechanisms. This review summarized the current studies about interleukin-17 in epilepsy and aimed at finding new therapeutic targets for refractory epilepsy.
Collapse
Affiliation(s)
- Jiayin An
- Emergency Department, Naval Hospital of Eastern Theater, Zhejiang, China.
| | - He Li
- Emergency Department, Naval Hospital of Eastern Theater, Zhejiang, China.
| | - Demeng Xia
- Emergency Department, Naval Hospital of Eastern Theater, Zhejiang, China; Luodian Clinical Drug Research Center, Shanghai Baoshan Luodian Hospital, Shanghai University, Shanghai, China.
| | - Bin Xu
- Emergency Department, Naval Hospital of Eastern Theater, Zhejiang, China.
| | - Jiayan Wang
- Emergency Department, Naval Hospital of Eastern Theater, Zhejiang, China.
| | - Huahui Qiu
- Zhoushan Hospital, Zhejiang University, Zhoushan, Zhejiang, China.
| | - Jiaojiang He
- Department of Neurosurgery, West China Hospital of Sichuan University, Sichuan, China.
| |
Collapse
|
7
|
Stanzione R, Forte M, Cotugno M, Bianchi F, Marchitti S, Rubattu S. Role of DAMPs and of Leukocytes Infiltration in Ischemic Stroke: Insights from Animal Models and Translation to the Human Disease. Cell Mol Neurobiol 2022; 42:545-556. [PMID: 32996044 PMCID: PMC11441194 DOI: 10.1007/s10571-020-00966-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/22/2020] [Indexed: 02/08/2023]
Abstract
Stroke is a leading cause of death and disability worldwide. Several mechanisms are involved in the pathogenesis of ischemic stroke (IS). The contributory role of the inflammatory and immunity processes was demonstrated both in vitro and in animal models, and was confirmed in humans. IS evokes an immediate inflammatory response that involves complex cellular and molecular mechanisms. All components of the innate and adaptive immunity systems are involved in several steps of the ischemic cascade. In the early phase, inflammatory and immune mechanisms contribute to the brain tissue damage, whereas, in the late phase, they participate to the tissue repair processes. In particular, damage-associated molecular patterns (DAMPs) appear critical for the promotion of altered blood brain barrier permeability, leukocytes infiltration, tissue edema and brain injury. Conversely, the activation of regulatory T lymphocytes (Tregs) plays protective effects. The identification of specific cellular/molecular elements belonging to the inflammatory and immune responses, contributing to the brain ischemic injury and tissue remodeling, offers the advantage to design adequate therapeutic strategies. In this article, we will present an overview of the knowledge on inflammatory and immunity processes in IS, with a particular focus on the role of DAMPs and leukocytes infiltration. We will discuss evidence obtained in preclinical models of IS and in humans. The main molecular mechanisms useful for the development of novel therapeutic approaches will be highlighted. The translation of experimental findings to the human disease is still a difficult step to pursue. Further investigations are required to fill up the existing gaps.
Collapse
Affiliation(s)
| | | | | | | | | | - Speranza Rubattu
- IRCCS Neuromed, Pozzilli, IS, Italy.
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
8
|
Ryan E, Kelly L, Stacey C, Huggard D, Duff E, McCollum D, Leonard A, Boran G, Doherty DR, Bolger T, Molloy EJ. Mild-to-severe traumatic brain injury in children: altered cytokines reflect severity. J Neuroinflammation 2022; 19:36. [PMID: 35130911 PMCID: PMC8822689 DOI: 10.1186/s12974-022-02390-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 01/14/2022] [Indexed: 11/10/2022] Open
Abstract
Background Paediatric traumatic brain injury (TBI) is recognised to have significant longer-term neurocognitive effects. Childhood is a time of high risk for head injury. Functional recovery is variable with a combination of any or all of physical, cognitive and emotional impairment. Immune activation and alteration in cytokine levels are present following TBI which may differ from adults. Methods Pro- and anti-inflammatory cytokines including Interleukin (IL)-2, IL-4, IL-6, IL-8, IL-10, IL-17A, Tumor Necrosis Factor (TNF)-α and Interferon (IFN)-γ were examined at baseline and following in vitro treatment with endotoxin of whole blood, in the following children: severe TBI (sTBI: initial Glasgow coma scale(GCS) ≤ 8), mild TBI (mTBI; GCS 14/15) at 0-4d and at 10-14d post-TBI and compared to healthy age-matched controls. Results The study enrolled 208 children, including 110 with TBI cohort (n = 104 mild; 6 severe) and controls (n = 98). At baseline all children with TBI had increased IL-6. The mTBI group had significantly increased IFN-γ versus controls. In sTBI at baseline, IFN-γ was decreased compared to controls. At baseline IL-8, IL-10, IL-17A, and TNF-α were decreased in mTBI compared to controls. This persisted at 2 week post-mTBI. The AUC for detecting mTBI was 0.801 CI (0.73–086) using IL6/IL10 ratio. mTBI showed a greater fold change in IL-8 and TNF-α in response to endotoxin stimulation, a response that persisted at 2 weeks. Children with sTBI did not have a significant IL-6 response to endotoxin, but did show an increase in IL-17A. Conclusion Children with all TBI including mTBI show altered cytokine profiles and altered endotoxin responses. Although cytokines increased in sTBI especially in response to endotoxin, suppressed responses were found in mTBI coupled with persistent immune dysfunction post-injury. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02390-5.
Collapse
Affiliation(s)
- Emer Ryan
- Department of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, Tallaght University Hospital, Dublin 24, Ireland.,Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Trinity Translational Medicine Institute, St James Hospital, Dublin 8, Ireland.,Paediatric Emergency Medicine, Children's Hospital Ireland (CHI) at Tallaght, Dublin 24, Ireland
| | - Lynne Kelly
- Department of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, Tallaght University Hospital, Dublin 24, Ireland.,Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Trinity Translational Medicine Institute, St James Hospital, Dublin 8, Ireland
| | - Catherine Stacey
- Department of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, Tallaght University Hospital, Dublin 24, Ireland.,Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Trinity Translational Medicine Institute, St James Hospital, Dublin 8, Ireland
| | - Dean Huggard
- Department of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, Tallaght University Hospital, Dublin 24, Ireland.,Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Trinity Translational Medicine Institute, St James Hospital, Dublin 8, Ireland.,Department of Paediatrics, Waterford Regional Hospital, Waterford, Ireland
| | - Eimear Duff
- Department of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, Tallaght University Hospital, Dublin 24, Ireland.,Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Trinity Translational Medicine Institute, St James Hospital, Dublin 8, Ireland
| | - Danielle McCollum
- Department of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, Tallaght University Hospital, Dublin 24, Ireland.,Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Trinity Translational Medicine Institute, St James Hospital, Dublin 8, Ireland.,Paediatric Emergency Medicine, Children's Hospital Ireland (CHI) at Tallaght, Dublin 24, Ireland.,Department of Paediatric Emergency Medicine, Royal Children's Hospital, Melbourne, Australia
| | - Ann Leonard
- Department of Biochemistry, Tallaght University Hospital, Dublin 24, Ireland
| | - Gerard Boran
- Department of Biochemistry, Tallaght University Hospital, Dublin 24, Ireland
| | - Dermot R Doherty
- Critical Care Medicine, Children's Health Ireland (CHI) at Temple Street, Dublin 1, Ireland
| | - Turlough Bolger
- Department of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Paediatric Emergency Medicine, Children's Hospital Ireland (CHI) at Tallaght, Dublin 24, Ireland
| | - Eleanor J Molloy
- Department of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland. .,Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, Tallaght University Hospital, Dublin 24, Ireland. .,Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Trinity Translational Medicine Institute, St James Hospital, Dublin 8, Ireland. .,Paediatric Emergency Medicine, Children's Hospital Ireland (CHI) at Tallaght, Dublin 24, Ireland. .,Department of Neonatology, CHI at Crumlin, Dublin, Ireland. .,Department of Neonatology, Coombe Women and Infants University Hospital Dublin, Dublin, Ireland.
| |
Collapse
|
9
|
Foschi M, Padroni M, Abu-Rumeileh S, Abdelhak A, Russo M, D'Anna L, Guarino M. Diagnostic and Prognostic Blood Biomarkers in Transient Ischemic Attack and Minor Ischemic Stroke: An Up-To-Date Narrative Review. J Stroke Cerebrovasc Dis 2022; 31:106292. [PMID: 35026496 DOI: 10.1016/j.jstrokecerebrovasdis.2021.106292] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Early diagnosis and correct risk stratification in patients with transient ischemic attack (TIA) and minor ischemic stroke (MIS) is crucial for the high rate of subsequent disabling stroke. Although highly improved, diagnosis and prognostication of TIA/MIS patients remain still based on clinical and neuroimaging findings, with some inter-rater variability even among trained neurologists. OBJECTIVES To provide an up-to-date overview of diagnostic and prognostic blood biomarkers in TIA and MIS patients. MATERIAL AND METHODS We performed a bibliographic search on PubMed database with last access on July 10th 2021. More than 680 articles were screened and we finally included only primary studies on blood biomarkers. RESULTS In a narrative fashion, we discussed about blood biomarkers investigated in TIA/MIS patients, including inflammatory, thrombosis, neuronal injury and cardiac analytes, antibodies and microRNAs. Other soluble molecules have been demonstrated to predict the risk of recurrent cerebrovascular events or treatment response in these patients. A rapid point of care assay, combining the determination of different biomarkers, has been developed to improve triage recognition of acute cerebrovascular accidents. CONCLUSIONS The implementation of blood biomarkers in the clinical management of TIA/MIS could ameliorate urgent identification, risk stratification and individual treatment choice. Large prospective and longitudinal studies, adopting standardized sampling and analytic procedures, are needed to clarify blood biomarkers kinetic and their relationship with TIA and minor stroke etiology.
Collapse
Affiliation(s)
- Matteo Foschi
- Department of Neuroscience, Neurology Unit, S. Maria delle Croci Hospital of Ravenna, AUSL Romagna, Ravenna, Italy; Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy.
| | - Marina Padroni
- Neurology Unit, Azienda Ospedaliero-Universitaria di Ferrara, Cona, Ferrara, Italy
| | - Samir Abu-Rumeileh
- Department of Neurology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Ahmed Abdelhak
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, USA; Department of Neurology, Ulm University Hospital, Ulm, Germany
| | - Michele Russo
- Department of Cardiovascular Diseases, Division of Cardiology - S. Maria delle Croci Hospital, AUSL Romagna, Ravenna, Italy
| | - Lucio D'Anna
- Department of Stroke and Neuroscience, Charing Cross Hospital, Imperial College London, NHS Healthcare Trust, London, United Kingdom; Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Maria Guarino
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| |
Collapse
|
10
|
Nakajima S, Tanaka R, Yamashiro K, Chiba A, Noto D, Inaba T, Kurita N, Miyamoto N, Kuroki T, Shimura H, Ueno Y, Urabe T, Miyake S, Hattori N. Mucosal-Associated Invariant T Cells Are Involved in Acute Ischemic Stroke by Regulating Neuroinflammation. J Am Heart Assoc 2021; 10:e018803. [PMID: 33733818 PMCID: PMC8174378 DOI: 10.1161/jaha.120.018803] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 02/08/2021] [Indexed: 12/22/2022]
Abstract
Background Mucosal-associated invariant T (MAIT) cells have been associated with inflammation in several autoimmune diseases. However, their relation to ischemic stroke remains unclear. This study attempted to elucidate the role of MAIT cells in acute ischemic stroke in mice. Methods and Results We used MR1 knockout C57BL/6 (MR1-/-) mice and wild-type littermates (MR1+/+). After performing a transient middle cerebral artery occlusion (tMCAO), we evaluated the association with inflammation and prognosis in the acute cerebral ischemia. Furthermore, we analyzed the tMCAO C57BL/6 mice administered with the suppressive MR1 ligand and the vehicle control. We also evaluated the infiltration of MAIT cells into the ischemic brain by flow cytometry. Results showed a reduction of infarct volume and an improvement of neurological impairment in MR1-/- mice (n=8). There was a reduction in the number of infiltrating microglia/macrophages (n=3-5) and in their activation (n=5) in the peri-infarct area of MR1-/- mice. The cytokine levels of interleukin-6 and interleukin-17 at 24 hours after tMCAO (n=3-5), and for interleukin-17 at 72 hours after tMCAO (n=5), were lower in the MR1-/- mice. The administration of the suppressive MR1 ligand reduced the infarct volume and improved functional impairment (n=5). Flow cytometric analysis demonstrated there was a reduction of MAIT cells infiltrating into the ischemic brain at 24 hours after tMCAO (n=17). Conclusions Our results showed that MAIT cells play an important role in neuroinflammation after focal cerebral ischemia and the use of MAIT cell regulation has a potential role as a novel neuroprotectant for the treatment of acute ischemic stroke.
Collapse
Affiliation(s)
- Sho Nakajima
- Department of NeurologyJuntendo University Faculty of MedicineTokyoJapan
| | - Ryota Tanaka
- Department of NeurologyJuntendo University Faculty of MedicineTokyoJapan
- Stroke Center and Division of NeurologyDepartment of MedicineJichi Medical UniversityTochigiJapan
| | - Kazuo Yamashiro
- Department of NeurologyJuntendo University Urayasu HospitalChibaJapan
| | - Asako Chiba
- Department of ImmunologyJuntendo University Faculty of MedicineTokyoJapan
| | - Daisuke Noto
- Department of ImmunologyJuntendo University Faculty of MedicineTokyoJapan
| | - Toshiki Inaba
- Department of NeurologyJuntendo University Faculty of MedicineTokyoJapan
| | - Naohide Kurita
- Department of NeurologyJuntendo University Urayasu HospitalChibaJapan
| | - Nobukazu Miyamoto
- Department of NeurologyJuntendo University Faculty of MedicineTokyoJapan
| | - Takuma Kuroki
- Department of NeurologyJuntendo University Faculty of MedicineTokyoJapan
| | - Hideki Shimura
- Department of NeurologyJuntendo University Urayasu HospitalChibaJapan
| | - Yuji Ueno
- Department of NeurologyJuntendo University Faculty of MedicineTokyoJapan
| | - Takao Urabe
- Department of NeurologyJuntendo University Urayasu HospitalChibaJapan
| | - Sachiko Miyake
- Department of ImmunologyJuntendo University Faculty of MedicineTokyoJapan
| | - Nobutaka Hattori
- Department of NeurologyJuntendo University Faculty of MedicineTokyoJapan
| |
Collapse
|
11
|
Prakash R, Mishra RK, Ahmad A, Khan MA, Khan R, Raza SS. Sivelestat-loaded nanostructured lipid carriers modulate oxidative and inflammatory stress in human dental pulp and mesenchymal stem cells subjected to oxygen-glucose deprivation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 120:111700. [PMID: 33545859 DOI: 10.1016/j.msec.2020.111700] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/21/2020] [Accepted: 11/02/2020] [Indexed: 12/16/2022]
Abstract
Stroke remains the leading cause of morbidity and mortality. Stem cell-based therapy offers promising hope for survivors and their families. Despite the clinical translation of stem cell-based therapies in stroke patients for almost two decades, results of these randomized controlled trials are not very optimistic. In these lines, an amalgamation of nanocarriers based drug delivery with stem cells holds great promise in enhancing stroke recovery. In the present study, we treated oxygen-glucose deprivation (OGD) exposed dental pulp stem cells (DPSCs) and mesenchymal stem cells (MSCs) with sivelestat-loaded nanostructured lipid carriers (NLCs). Various physicochemical limitations associated with sivelestat drug applications and its recent inefficacy in the clinical trials necessitates the development of novel delivery approaches for sivelestat. Therefore, to improve its efficacy on the survival of DPSCs and MSCs cell types under OGD insult, the current NLCs were formulated and characterized. Resulting NLCs exhibited a hydrodynamic diameter of 160-180 nm by DLS technique and possessed good PDI values of 0.2-0.3. Their shape, size and surface morphology were corroborated with microscopic techniques like TEM, SEM, and AFM. FTIR and UV-Vis techniques confirmed nanocarrier's loading capacity, encapsulation efficiency of sivelestat, and drug release profile. Oxidative stress in DPSCs and MSCs was assessed by DHE and DCFDA staining, and cell viability was assessed by Trypan blue exclusion test and MTT assay. Results indicated that sivelestat-loaded NLCs protected the loss of cell membrane integrity and restored cell morphology. Furthermore, NLCs successfully defended human DPSCs and MSCs against OGD-induced oxidative and inflammatory stress. In conclusion, modulation of oxidative and inflammatory stress by treatment with sivelestat-loaded NLCs in DPSCs and MSCs provides a novel strategy to rescue stem cells during ischemic stroke.
Collapse
Affiliation(s)
- Ravi Prakash
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Sarfarazganj, Lucknow 226003, Uttar Pradesh, India
| | - Rakesh Kumar Mishra
- Department of Nano-Therapeutics, Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India
| | - Anas Ahmad
- Department of Nano-Therapeutics, Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India
| | | | - Rehan Khan
- Department of Nano-Therapeutics, Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India.
| | - Syed Shadab Raza
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Sarfarazganj, Lucknow 226003, Uttar Pradesh, India; Department of Stem Cell Biology and Regenerative Medicine, Era University, Sarfarazganj, Lucknow 226003, Uttar Pradesh, India.
| |
Collapse
|
12
|
Kim E, Cho S. CNS and peripheral immunity in cerebral ischemia: partition and interaction. Exp Neurol 2021; 335:113508. [PMID: 33065078 PMCID: PMC7750306 DOI: 10.1016/j.expneurol.2020.113508] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/28/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023]
Abstract
Stroke elicits excessive immune activation in the injured brain tissue. This well-recognized neural inflammation in the brain is not just an intrinsic organ response but also a result of additional intricate interactions between infiltrating peripheral immune cells and the resident immune cells in the affected areas. Given that there is a finite number of immune cells in the organism at the time of stroke, the partitioned immune systems of the central nervous system (CNS) and periphery must appropriately distribute the limited pool of immune cells between the two domains, mounting a necessary post-stroke inflammatory response by supplying a sufficient number of immune cells into the brain while maintaining peripheral immunity. Stroke pathophysiology has mainly been neurocentric in focus, but understanding the distinct roles of the CNS and peripheral immunity in their concerted action against ischemic insults is crucial. This review will discuss stroke-induced influences of the peripheral immune system on CNS injury/repair and of neural inflammation on peripheral immunity, and how comorbidity influences each.
Collapse
Affiliation(s)
- Eunhee Kim
- Vivian L. Smith Department of Neurosurgery at University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Sunghee Cho
- Burke Neurological Institute, White Plains, NY, United States of America; Feil Brain Mind Research Institute, Weill Cornell Medicine, New York, NY, United States of America.
| |
Collapse
|
13
|
Zhang Q, Liao Y, Liu Z, Dai Y, Li Y, Li Y, Tang Y. Interleukin-17 and ischaemic stroke. Immunology 2020; 162:179-193. [PMID: 32935861 DOI: 10.1111/imm.13265] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/22/2020] [Accepted: 08/26/2020] [Indexed: 12/26/2022] Open
Abstract
Interleukin-17 (IL-17) is a cytokine family that includes 6 members, IL-17A through IL-17F, most of them are reported to have pro-inflammatory role. Through binding to their receptors (IL-17Rs), IL-17 activates the intracellular signalling pathways to play an important role in autoimmune diseases, including rheumatoid arthritis (RA) and multiple sclerosis (MS). Ischaemic stroke is a complex pathophysiological process mainly caused by regional cerebral ischaemia. Inflammatory factors contribute to the physiological process of stroke that leads to poor prognosis. IL-17 plays a crucial role in promoting inflammatory response and inducing secondary injury in post-stroke. Though immune cells and inflammatory factors have been reported to be involved in the damage of stroke, the functions of IL-17 in this process need to be elucidated. This review focuses on the pathological modulation and the mechanism of IL-17 family in ischaemic stroke and seeking to provide new insights for future therapies.
Collapse
Affiliation(s)
- Qiaohui Zhang
- Chinese Medical Institute, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Liao
- Chinese Medical Institute, Beijing University of Chinese Medicine, Beijing, China
| | - Zhenquan Liu
- School of Chinese Materia Medical, Beijing University of Chinese Medicine, Beijing, China
| | - Yajie Dai
- Chinese Medical Institute, Beijing University of Chinese Medicine, Beijing, China
| | - Yunxin Li
- Chinese Medical Institute, Beijing University of Chinese Medicine, Beijing, China
| | - Yue Li
- School of Chinese Materia Medical, Beijing University of Chinese Medicine, Beijing, China
| | - Yibo Tang
- Chinese Medical Institute, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
14
|
Neuroprotective and Anti-inflammatory Effect of Tangeretin Against Cerebral Ischemia-Reperfusion Injury in Rats. Inflammation 2020; 43:2332-2343. [DOI: 10.1007/s10753-020-01303-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
15
|
Nagy EE, Frigy A, Szász JA, Horváth E. Neuroinflammation and microglia/macrophage phenotype modulate the molecular background of post-stroke depression: A literature review. Exp Ther Med 2020; 20:2510-2523. [PMID: 32765743 PMCID: PMC7401670 DOI: 10.3892/etm.2020.8933] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/05/2020] [Indexed: 12/16/2022] Open
Abstract
Increasing evidence hints to the central role of neuroinflammation in the development of post-stroke depression. Danger signals released in the acute phase of ischemia trigger microglial activation, along with the infiltration of neutrophils and macrophages. The increased secretion of proinflammatory cytokines interleukin (IL)-1β, IL-6, IL-8, and tumor necrosis factor α (TNFα) provokes neuronal degeneration and apoptosis, whereas IL-6, interferon γ (IFNγ), and TNFα induce aberrant tryptophane degradation with the accumulation of the end-product quinolinic acid in resident glial cells. This promotes glutamate excitotoxicity via hyperexcitation of N-methyl-D-aspartate receptors and antagonizes 5-hydroxy-tryptamine, reducing synaptic plasticity and neuronal survival, thus favoring depression. In the post-stroke period, CX3CL1 and the CD200-CD200R interaction mediates the activation of glial cells, whereas CCL-2 attracts infiltrating macrophages. CD206 positive cells grant the removal of excessive danger signals; the high number of regulatory T cells, IL-4, IL-10, transforming growth factor β (TGFβ), and intracellular signaling via cAMP response element-binding protein (CREB) support the M2 type differentiation. In favorable conditions, these cells may exert efficient clearance, mediate tissue repair, and might be essential players in the downregulation of molecular pathways that promote post-stroke depression.
Collapse
Affiliation(s)
- Előd Ernő Nagy
- Department of Biochemistry and Environmental Chemistry, 'George Emil Palade' University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
| | - Attila Frigy
- Department of Internal Medicine IV, 'George Emil Palade' University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540103 Targu Mures, Romania
| | - József Attila Szász
- Neurology Clinic II, 'George Emil Palade' University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540136 Targu Mures, Romania
| | - Emőke Horváth
- Department of Pathology, 'George Emil Palade' University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
| |
Collapse
|
16
|
Milovanovic J, Arsenijevic A, Stojanovic B, Kanjevac T, Arsenijevic D, Radosavljevic G, Milovanovic M, Arsenijevic N. Interleukin-17 in Chronic Inflammatory Neurological Diseases. Front Immunol 2020; 11:947. [PMID: 32582147 PMCID: PMC7283538 DOI: 10.3389/fimmu.2020.00947] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/22/2020] [Indexed: 12/15/2022] Open
Abstract
A critical role for IL-17, a cytokine produced by T helper 17 (Th17) cells, has been indicated in the pathogenesis of chronic inflammatory and autoimmune diseases. A positive effect of blockade of IL-17 secreted by autoreactive T cells has been shown in various inflammatory diseases. Several cytokines, whose production is affected by environmental factors, control Th17 differentiation and its maintenance in tissues during chronic inflammation. The roles of IL-17 in the pathogenesis of chronic neuroinflammatory conditions, multiple sclerosis (MS), experimental autoimmune encephalomyelitis (EAE), Alzheimer's disease, and ischemic brain injury are reviewed here. The role of environmental stimuli in Th17 differentiation is also summarized, highlighting the role of viral infection in the regulation of pathogenic T helper cells in EAE.
Collapse
Affiliation(s)
- Jelena Milovanovic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
- Department of Histology and Embriology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Aleksandar Arsenijevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | - Bojana Stojanovic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Tatjana Kanjevac
- Department of Dentistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Dragana Arsenijevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Gordana Radosavljevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | - Marija Milovanovic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | - Nebojsa Arsenijevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
17
|
Janach GMS, Reetz O, Döhne N, Stadler K, Grosser S, Byvaltcev E, Bräuer AU, Strauss U. Interferon-γ acutely augments inhibition of neocortical layer 5 pyramidal neurons. J Neuroinflammation 2020; 17:69. [PMID: 32087716 PMCID: PMC7035745 DOI: 10.1186/s12974-020-1722-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 01/20/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Interferon-γ (IFN-γ, a type II IFN) is present in the central nervous system (CNS) under various conditions. Evidence is emerging that, in addition to its immunological role, IFN-γ modulates neuronal morphology, function, and development in several brain regions. Previously, we have shown that raising levels of IFN-β (a type I IFN) lead to increased neuronal excitability of neocortical layer 5 pyramidal neurons. Because of shared non-canonical signaling pathways of both cytokines, we hypothesized a similar neocortical role of acutely applied IFN-γ. METHODS We used semi-quantitative RT-PCR, immunoblotting, and immunohistochemistry to analyze neuronal expression of IFN-γ receptors and performed whole-cell patch-clamp recordings in layer 5 pyramidal neurons to investigate sub- and suprathreshold excitability, properties of hyperpolarization-activated cyclic nucleotide-gated current (Ih), and inhibitory neurotransmission under the influence of acutely applied IFN-γ. RESULTS We show that IFN-γ receptors are present in the membrane of rat's neocortical layer 5 pyramidal neurons. As expected from this and the putative overlap in IFN type I and II alternative signaling pathways, IFN-γ diminished Ih, mirroring the effect of type I IFNs, suggesting a likewise activation of protein kinase C (PKC). In contrast, IFN-γ did neither alter subthreshold nor suprathreshold neuronal excitability, pointing to augmented inhibitory transmission by IFN-γ. Indeed, IFN-γ increased electrically evoked inhibitory postsynaptic currents (IPSCs) on neocortical layer 5 pyramidal neurons. Furthermore, amplitudes of spontaneous IPSCs and miniature IPSCs were elevated by IFN-γ, whereas their frequency remained unchanged. CONCLUSIONS The expression of IFN-γ receptors on layer 5 neocortical pyramidal neurons together with the acute augmentation of inhibition in the neocortex by direct application of IFN-γ highlights an additional interaction between the CNS and immune system. Our results strengthen our understanding of the role of IFN-γ in neocortical neurotransmission and emphasize its impact beyond its immunological properties, particularly in the pathogenesis of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Gabriel M S Janach
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Cell Biology & Neurobiology, Charitéplatz 1, 10117, Berlin, Germany
| | - Olivia Reetz
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Cell Biology & Neurobiology, Charitéplatz 1, 10117, Berlin, Germany
| | - Noah Döhne
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Cell Biology & Neurobiology, Charitéplatz 1, 10117, Berlin, Germany
| | - Konstantin Stadler
- Industrial Ecology Programme, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Sabine Grosser
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Integrative Neuroanatomy, Berlin, Germany
| | - Egor Byvaltcev
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Cell Biology & Neurobiology, Charitéplatz 1, 10117, Berlin, Germany
| | - Anja U Bräuer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Cell Biology & Neurobiology, Charitéplatz 1, 10117, Berlin, Germany.,Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.,Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Ulf Strauss
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Cell Biology & Neurobiology, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
18
|
Wang Y, Xiao G, He S, Liu X, Zhu L, Yang X, Zhang Y, Orgah J, Feng Y, Wang X, Zhang B, Zhu Y. Protection against acute cerebral ischemia/reperfusion injury by QiShenYiQi via neuroinflammatory network mobilization. Biomed Pharmacother 2020; 125:109945. [PMID: 32028240 DOI: 10.1016/j.biopha.2020.109945] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/14/2022] Open
Abstract
Cerebral ischemia/reperfusion injury (CI/RI) is a common feature of ischemic stroke, involving a period of impaired blood supply to the brain, followed by the restoration of cerebral perfusion through medical intervention. Although ischemia and reperfusion brain damage is a complex pathological process with an unclear physiological mechanism, more attention is currently focused on the neuroinflammatory response of an ischemia/reperfusion origin, and anti-inflammatory appears to be a potential therapeutic strategy following ischemic stroke. QiShenYiQi (QSYQ), a component-based Chinese medicine with Qi-tonifying and blood-activating property, has pharmacological actions of anti-inflammatory, antioxidant, mitochondrial protectant, anti-apoptosis, and antiplatelet aggregation. We have previously reported that the cardioprotective effect of QSYQ against ischemia/reperfusion injury is via improvement of mitochondrial functional integrity. In this research work, we aimed to investigate the possible mechanism involved in the neuroprotection of QSYQ in mice model of cerebral ischemia/reperfusion injury based on the inflammatory pathway. The cerebral protection was evaluated in the stroke mice after 24 h reperfusion by assessing the neurological deficit, cerebral infarction, brain edema, BBB functionality, and via histopathological assessment. TCM-based network pharmacology method was performed to establish and analyze compound-target-disease & function-pathway network so as to find the possible mechanism linking to the role of QSYQ in CI/RI. In addition, RT-qPCR was used to verify the accuracy of predicted signaling gene expression. As a result, improvement of neurological outcome, reduction of infarct volume and brain edema, a decrease in BBB disruption, and amelioration of histopathological alteration were observed in mice pretreated with QSYQ after experimental stroke surgery. Network pharmacology analysis revealed neuroinflammatory response was associated with the action of QSYQ in CI/RI. RT-qPCR data showed that the mice pretreated with QSYQ could significantly decrease IFNG-γ, IL-6, TNF-α, NF-κB p65, and TLR-4 mRNA levels and increase TGF-β1 mRNA level in the brain compared to the untreated mice after CI/RI (p < 0.05). In conclusion, our study indicated the cerebral protective effect of pretreatment with QSYQ against CI/RI, which may be partly related to its potential to the reduction of neuroinflammatory response in a stroke subject.
Collapse
Affiliation(s)
- Yule Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Guangxu Xiao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Shuang He
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Xinyan Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Lin Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Xinyue Yang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Yiqian Zhang
- State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin Tasly Holding Group Co., Ltd., Tianjin, China
| | - John Orgah
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Yuxin Feng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Xiaoying Wang
- Neuroscience Program, Neuroprotection Research Laboratory, Department of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Boli Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China.
| |
Collapse
|
19
|
Gorlé N, Vandenbroucke RE. Interferons: A molecular switch between damage and repair in ageing and Alzheimer's disease. Mech Ageing Dev 2019; 183:111148. [PMID: 31541624 DOI: 10.1016/j.mad.2019.111148] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 09/13/2019] [Accepted: 09/17/2019] [Indexed: 10/26/2022]
Abstract
Alzheimer's disease was first described over 100 years ago, yet it remains incurable and affects 44 million people worldwide. Traditionally, research has largely focused on the amyloid cascade hypothesis, but interest in the importance of inflammation in the progression of the disease has recently been increasing. Interferons, a large family of cytokines that trigger the immune system, are believed to play a crucial role in the pathology of Alzheimer's disease. This review focuses on how interferons affect the brain during ageing and whether they could be candidate therapeutic targets for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- N Gorlé
- VIB Center for Inflammation Research, VIB, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
| | - R E Vandenbroucke
- VIB Center for Inflammation Research, VIB, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium.
| |
Collapse
|
20
|
Singh D, Reeta K, Sharma U, Jagannathan N, Dinda A, Gupta Y. Neuro-protective effect of monomethyl fumarate on ischemia reperfusion injury in rats: Role of Nrf2/HO1 pathway in peri-infarct region. Neurochem Int 2019; 126:96-108. [DOI: 10.1016/j.neuint.2019.03.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 02/28/2019] [Accepted: 03/11/2019] [Indexed: 12/30/2022]
|
21
|
Ahmad A, Fauzia E, Kumar M, Mishra RK, Kumar A, Khan MA, Raza SS, Khan R. Gelatin-Coated Polycaprolactone Nanoparticle-Mediated Naringenin Delivery Rescue Human Mesenchymal Stem Cells from Oxygen Glucose Deprivation-Induced Inflammatory Stress. ACS Biomater Sci Eng 2018; 5:683-695. [PMID: 33405831 DOI: 10.1021/acsbiomaterials.8b01081] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Ischemic stroke involves pro-inflammatory species, which implicates inflammation in the disease mechanism. Recent studies indicate that the prevalence of therapeutic choice such as stem cell transplantation has seen an upsurge in ischemic stroke. However, after transplantation the fate of transplanted cells is largely unknown. Human mesenchymal stem cells (MSCs), due to their robust survival rate upon transplantation in brain tissue, are being widely employed to treat ischemic stroke. In the present study, we have evaluated naringenin-loaded gelatin-coated polycaprolactone nanoparticles (nar-gel-c-PCL NPs) to rescue MSCs against oxygen glucose deprived insult. Naringenin, due to its strong anti-inflammatory effects, remains a therapeutic choice in neurological disorders. Though, the low solubility and inefficient delivery remain challenges in using naringenin as a therapeutic drug. The present study showed that inflammation occurred in MSCs during their treatment with oxygen glucose deprivation (OGD) and was well overturned by treatment with nar-gel-c-PCL NPs. In brief, the results indicated that nar-gel-c-PCL NPs were able to protect the loss of cell membrane integrity and restored neuronal morphology. Then nar-gel-c-PCL NPs successfully protected the human MSCs against OGD-induced inflammation as evident by reduced level of pro-inflammatory cytokine (TNF-α, IFN-γ, and IL-1β) and other inflammatory biomarkers (COX2, iNOS, and MPO activity). Therefore, the modulation of inflammation by treatment with nar-gel-c-PCL NPs in MSCs could provide a novel strategy to improve MSC-based therapy, and thus, our nanoformulation may find a wide therapeutic application in ischemic stroke and other neuro-inflammatory diseases.
Collapse
Affiliation(s)
- Anas Ahmad
- Department of Nano-Therapeutics, Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India
| | - Eram Fauzia
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College Hospital, Sarfarazganj, Lucknow-226003, India
| | - Manish Kumar
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College Hospital, Sarfarazganj, Lucknow-226003, India
| | - Rakesh Kumar Mishra
- Department of Nano-Therapeutics, Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India
| | - Ajay Kumar
- Department of Nano-Therapeutics, Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India
| | - Mohsin Ali Khan
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College Hospital, Sarfarazganj, Lucknow-226003, India
| | - Syed Shadab Raza
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College Hospital, Sarfarazganj, Lucknow-226003, India.,Department of Stem Cell Biology and Regenerative Medicine, Era's Lucknow Medical College Hospital, Sarfarazganj, Lucknow-226003, India
| | - Rehan Khan
- Department of Nano-Therapeutics, Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India
| |
Collapse
|
22
|
Lively S, Schlichter LC. Microglia Responses to Pro-inflammatory Stimuli (LPS, IFNγ+TNFα) and Reprogramming by Resolving Cytokines (IL-4, IL-10). Front Cell Neurosci 2018; 12:215. [PMID: 30087595 PMCID: PMC6066613 DOI: 10.3389/fncel.2018.00215] [Citation(s) in RCA: 245] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/28/2018] [Indexed: 12/11/2022] Open
Abstract
Microglia respond to CNS injuries and diseases with complex reactions, often called "activation." A pro-inflammatory phenotype (also called classical or M1 activation) lies at one extreme of the reactivity spectrum. There were several motivations for this study. First, bacterial endotoxin (lipopolysaccharide, LPS) is the most commonly used pro-inflammatory stimulus for microglia, both in vitro and in vivo; however, pro-inflammatory cytokines (e.g., IFNγ, TNFα) rather than LPS will be encountered with sterile CNS damage and disease. We lack direct comparisons of responses between LPS and such cytokines. Second, while transcriptional profiling is providing substantial data on microglial responses to LPS, these studies mainly use mouse cells and models, and there is increasing evidence that responses of rat microglia can differ. Third, the cytokine milieu is dynamic after acute CNS damage, and an important question in microglial biology is: How malleable are their responses? There are very few studies of effects of resolving cytokines, particularly for rat microglia, and much of the work has focused on pro-inflammatory outcomes. Here, we first exposed primary rat microglia to LPS or to IFNγ+TNFα (I+T) and compared hallmark functional (nitric oxide production, migration) and molecular responses (almost 100 genes), including surface receptors that can be considered part of the sensome. Protein changes for exemplary molecules were also quantified: ARG1, CD206/MRC1, COX-2, iNOS, and PYK2. Despite some similarities, there were notable differences in responses to LPS and I+T. For instance, LPS often evoked higher pro-inflammatory gene expression and also increased several anti-inflammatory genes. Second, we compared the ability of two anti-inflammatory, resolving cytokines (IL-4, IL-10), to counteract responses to LPS and I+T. IL-4 was more effective after I+T than after LPS, and IL-10 was surprisingly ineffective after either stimulus. These results should prove useful in modeling microglial reactivity in vitro; and comparing transcriptional responses to sterile CNS inflammation in vivo.
Collapse
Affiliation(s)
- Starlee Lively
- Division of Genetics & Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Lyanne C Schlichter
- Division of Genetics & Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
23
|
Shekhar S, Cunningham MW, Pabbidi MR, Wang S, Booz GW, Fan F. Targeting vascular inflammation in ischemic stroke: Recent developments on novel immunomodulatory approaches. Eur J Pharmacol 2018; 833:531-544. [PMID: 29935175 DOI: 10.1016/j.ejphar.2018.06.028] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/02/2018] [Accepted: 06/19/2018] [Indexed: 10/28/2022]
Abstract
Ischemic stroke is a devastating and debilitating medical condition with limited therapeutic options. However, accumulating evidence indicates a central role of inflammation in all aspects of stroke including its initiation, the progression of injury, and recovery or wound healing. A central target of inflammation is disruption of the blood brain barrier or neurovascular unit. Here we discuss recent developments in identifying potential molecular targets and immunomodulatory approaches to preserve or protect barrier function and limit infarct damage and functional impairment. These include blocking harmful inflammatory signaling in endothelial cells, microglia/macrophages, or Th17/γδ T cells with biologics, third generation epoxyeicosatrienoic acid (EET) analogs with extended half-life, and miRNA antagomirs. Complementary beneficial pathways may be enhanced by miRNA mimetics or hyperbaric oxygenation. These immunomodulatory approaches could be used to greatly expand the therapeutic window for thrombolytic treatment with tissue plasminogen activator (t-PA). Moreover, nanoparticle technology allows for the selective targeting of endothelial cells for delivery of DNA/RNA oligonucleotides and neuroprotective drugs. In addition, although likely detrimental to the progression of ischemic stroke by inducing inflammation, oxidative stress, and neuronal cell death, 20-HETE may also reduce susceptibility of onset of ischemic stroke by maintaining autoregulation of cerebral blood flow. Although the interaction between inflammation and stroke is multifaceted, a better understanding of the mechanisms behind the pro-inflammatory state at all stages will hopefully help in developing novel immunomodulatory approaches to improve mortality and functional outcome of those inflicted with ischemic stroke.
Collapse
Affiliation(s)
- Shashank Shekhar
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, USA; Institute of Clinical Medicine, University of Turku, Turku, Finland
| | - Mark W Cunningham
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Mallikarjuna R Pabbidi
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Shaoxun Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - George W Booz
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
24
|
Abstract
Stroke-induced endothelial cell injury leads to destruction of cerebral microvasculature and significant damage to the brain tissue. A subacute phase of cerebral ischemia is associated with regeneration involving the activation of vascular remodeling, neuroplasticity, neurogenesis, and neuroinflammation processes. Effective restoration and improvement of blood supply to the damaged brain tissue offers a potential therapy for stroke. microRNAs (miRNAs) are recently identified small RNA molecules that regulate gene expression and significantly influence the essential cellular processes associated with brain repair following stroke. A number of specific miRNAs are implicated in regulating the development and propagation of the ischemic tissue damage as well as in mediating post-stroke regeneration. In this review, I discuss the functions of the miRNA miR-155 and the effect of its in vivo inhibition on brain recovery following experimental cerebral ischemia. The article introduces new and unexplored approach to cerebral regeneration: regulation of brain tissue repair through a direct modulation of specific miRNA activity.
Collapse
Affiliation(s)
- Tamara Roitbak
- Department of Neurosurgery, Health Sciences Center, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
25
|
Zhang G, Guo X, Chen L, Li B, Gu B, Wang H, Wu G, Kong J, Chen W, Yu Y. Interferon-γ Promotes Neuronal Repair by Transplanted Neural Stem Cells in Ischemic Rats. Stem Cells Dev 2018; 27:355-366. [PMID: 29298609 DOI: 10.1089/scd.2017.0225] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Ischemic stroke represents the leading cause of adult neurological disability, with no effective therapeutic strategy. Stem cell transplantation promises a new promising for treating stroke, through cell replacement and cytokine paracrine. However, due to the effect of hostile immune microenvironment, the survival and differentiation of stem cells are limited in vivo. Furthermore, the delayed inflammatory response to stroke induced secondary neurological injury. IFN-γ as pro-inflammatory cytokine has the potential to protect stem cell population during inflammatory response, as well as stimulates neurogenesis of stem cells. The purpose of this study was to investigate whether co-injection of neural stem cells and IFN-γ can improve therapeutic outcomes in ischemic stroke model. In this study, we found that IFN-γ did not interfere with the proliferation of neural stem cells (NSCs) in vitro and induced levels of subsequent neuronal differentiation significantly superior to those of other four cytokines BDNF, VEGF, TGF-β, and IGF-1. Co-delivery of IFN-γ (concentration: 50 ng) enhanced the effectiveness of NSC transplantation therapy in ischemic rats. And combined IFN-γ treatment significantly increased neurogenesis in vivo, with more BrdU/DCX dual-positive cells found in ischemic areas. Moreover, co-treatment with IFN-γ and NSCs exerted additional neurological benefits compared with NSC transplantation alone. In conclusion, low concentration of IFN-γ can promote the functions of transplanted NSCs and facilitate their ability of neurological repair. Thus, our findings suggest that co-delivery of NSCs and IFN-γ without genetic modification may be an effective, simple, and novel approach for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Guilong Zhang
- Department of Neurosurgery, Zhongda Hospital, School of Medicine, Southeast University , Nanjing, China
| | - Xiaoyuan Guo
- Department of Neurosurgery, Zhongda Hospital, School of Medicine, Southeast University , Nanjing, China
| | - Lukui Chen
- Department of Neurosurgery, Zhongda Hospital, School of Medicine, Southeast University , Nanjing, China
| | - Bingqian Li
- Department of Neurosurgery, Zhongda Hospital, School of Medicine, Southeast University , Nanjing, China
| | - Bin Gu
- Department of Neurosurgery, Zhongda Hospital, School of Medicine, Southeast University , Nanjing, China
| | - Hong Wang
- Department of Neurosurgery, Zhongda Hospital, School of Medicine, Southeast University , Nanjing, China
| | - Guojian Wu
- Department of Neurosurgery, Zhongda Hospital, School of Medicine, Southeast University , Nanjing, China
| | - Jun Kong
- Department of Neurosurgery, Zhongda Hospital, School of Medicine, Southeast University , Nanjing, China
| | - Wanghao Chen
- Department of Neurosurgery, Zhongda Hospital, School of Medicine, Southeast University , Nanjing, China
| | - Yongbo Yu
- Department of Neurosurgery, Zhongda Hospital, School of Medicine, Southeast University , Nanjing, China
| |
Collapse
|
26
|
Cruz Y, García EE, Gálvez JV, Arias-Santiago SV, Carvajal HG, Silva-García R, Bonilla-Jaime H, Rojas-Castañeda J, Ibarra A. Release of interleukin-10 and neurotrophic factors in the choroid plexus: possible inductors of neurogenesis following copolymer-1 immunization after cerebral ischemia. Neural Regen Res 2018; 13:1743-1752. [PMID: 30136689 PMCID: PMC6128049 DOI: 10.4103/1673-5374.238615] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Copolymer-1 (Cop-1) is a peptide with immunomodulatory properties, approved by the Food and Drug Administration of United States in the treatment of multiple sclerosis. Cop-1 has been shown to exert neuroprotective effects and induce neurogenesis in cerebral ischemia models. Nevertheless, the mechanism involved in the neurogenic action of this compound remains unknown. The choroid plexus (CP) is a network of cells that constitute the interphase between the immune and central nervous systems, with the ability to mediate neurogenesis through the release of cytokines and growth factors. Therefore, the CP could play a role in Cop-1-induced neurogenesis. In order to determine the participation of the CP in the induction of neurogenesis after Cop-1 immunization, we evaluated the gene expression of various growth factors (brain-derived neurotrophic factor, insulin-like growth factor 1, neurotrophin-3) and cytokines (tumor necrosis factor alpha, interferon-gamma, interleukin-4 (IL-4), IL-10 and IL-17), in the CP at 14 days after ischemia. Furthermore, we analyzed the correlation between the expression of these genes and neurogenesis. Our results showed that Cop-1 was capable of stimulating an upregulation in the expression of the genes encoding for brain-derived neurotrophic factor, insulin-like growth factor 1, neurotrophin-3 and IL-10 in the CP, which correlated with an increase in neurogenesis in the subventricular and subgranular zone. As well, we observed a downregulation of IL-17 gene expression. This study demonstrates the effect of Cop-1 on the expression of growth factors and IL-10 in the CP, in the same way, presents a possible mechanism involved in the neurogenic effect of Cop-1.
Collapse
Affiliation(s)
- Yolanda Cruz
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Norte, Huixquilucan, Estado de México; Lab. De Biología de la reproducción, UAMI. Ciudad de México; Doctorado en Ciencias Biológicas, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa. Ciudad de México, México
| | - Edna E García
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Norte, Huixquilucan, Estado de México, México
| | - Jessica V Gálvez
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Norte, Huixquilucan, Estado de México, México
| | - Stella V Arias-Santiago
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Norte, Huixquilucan, Estado de México, México
| | - Horacio G Carvajal
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Norte, Huixquilucan, Estado de México, México
| | | | | | - Julio Rojas-Castañeda
- Subdirección de Medicina Experimental, Instituto Nacional de Pediatría. Ciudad de México, México
| | - Antonio Ibarra
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Norte, Huixquilucan, Estado de México, México
| |
Collapse
|
27
|
Torres MD, Garcia O, Tang C, Busciglio J. Dendritic spine pathology and thrombospondin-1 deficits in Down syndrome. Free Radic Biol Med 2018; 114:10-14. [PMID: 28965914 PMCID: PMC7185223 DOI: 10.1016/j.freeradbiomed.2017.09.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/25/2017] [Accepted: 09/26/2017] [Indexed: 11/27/2022]
Abstract
Abnormal dendritic spine structure and function is one of the most prominent features associated with neurodevelopmental disorders including Down syndrome (DS). Defects in both spine morphology and spine density may underlie alterations in neuronal and synaptic plasticity, ultimately affecting cognitive ability. Here we briefly examine the role of astrocytes in spine alterations and more specifically the involvement of astrocyte-secreted thrombospondin 1 (TSP-1) deficits in spine and synaptic pathology in DS.
Collapse
Affiliation(s)
- Maria D Torres
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), and Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, CA 92697, United States
| | - Octavio Garcia
- Facultad de Psicología, Universidad Nacional Autónoma de México, 04510 Coyoacán, Ciudad de México, México
| | - Cindy Tang
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), and Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, CA 92697, United States
| | - Jorge Busciglio
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), and Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, CA 92697, United States.
| |
Collapse
|
28
|
Stokum JA, Kwon MS, Woo SK, Tsymbalyuk O, Vennekens R, Gerzanich V, Simard JM. SUR1-TRPM4 and AQP4 form a heteromultimeric complex that amplifies ion/water osmotic coupling and drives astrocyte swelling. Glia 2017; 66:108-125. [PMID: 28906027 DOI: 10.1002/glia.23231] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 07/23/2017] [Accepted: 08/23/2017] [Indexed: 12/17/2022]
Abstract
Astrocyte swelling occurs after central nervous system injury and contributes to brain swelling, which can increase mortality. Mechanisms proffered to explain astrocyte swelling emphasize the importance of either aquaporin-4 (AQP4), an astrocyte water channel, or of Na+ -permeable channels, which mediate cellular osmolyte influx. However, the spatio-temporal functional interactions between AQP4 and Na+ -permeable channels that drive swelling are poorly understood. We hypothesized that astrocyte swelling after injury is linked to an interaction between AQP4 and Na+ -permeable channels that are newly upregulated. Here, using co-immunoprecipitation and Förster resonance energy transfer, we report that AQP4 physically co-assembles with the sulfonylurea receptor 1-transient receptor potential melastatin 4 (SUR1-TRPM4) monovalent cation channel to form a novel heteromultimeric water/ion channel complex. In vitro cell-swelling studies using calcein fluorescence imaging of COS-7 cells expressing various combinations of AQP4, SUR1, and TRPM4 showed that the full tripartite complex, comprised of SUR1-TRPM4-AQP4, was required for fast, high-capacity transmembrane water transport that drives cell swelling, with these findings corroborated in cultured primary astrocytes. In a murine model of brain edema involving cold-injury to the cerebellum, we found that astrocytes newly upregulate SUR1-TRPM4, that AQP4 co-associates with SUR1-TRPM4, and that genetic inactivation of the solute pore of the SUR1-TRPM4-AQP4 complex blocked in vivo astrocyte swelling measured by diolistic labeling, thereby corroborating our in vitro functional studies. Together, these findings demonstrate a novel molecular mechanism involving the SUR1-TRPM4-AQP4 complex to account for bulk water influx during astrocyte swelling. These findings have broad implications for the understanding and treatment of AQP4-mediated pathological conditions.
Collapse
Affiliation(s)
- Jesse A Stokum
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, 21201-1595
| | - Min S Kwon
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, 21201-1595
| | - Seung K Woo
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, 21201-1595
| | - Orest Tsymbalyuk
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, 21201-1595
| | - Rudi Vennekens
- Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, Katholieke Universiteit Leuven, Leuven, 3000, Belgium
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, 21201-1595
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, 21201-1595.,Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, 21201-1595.,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, 21201-1595
| |
Collapse
|
29
|
Zhang B, Yang N, Mo ZM, Lin SP, Zhang F. IL-17A Enhances Microglial Response to OGD by Regulating p53 and PI3K/Akt Pathways with Involvement of ROS/HMGB1. Front Mol Neurosci 2017; 10:271. [PMID: 28912678 PMCID: PMC5583146 DOI: 10.3389/fnmol.2017.00271] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 08/11/2017] [Indexed: 01/01/2023] Open
Abstract
Cerebral ischemia-reperfusion injury (IRI) has a complex pathogenesis, and interleukin-17 (IL-17) is a newly identified class of the cytokine family that plays an important role in ischemic inflammation. An oxygen-glucose deprivation (OGD) model showed that IL-17A expression was significantly up-regulated in microglial cells. After IL-17A siRNA transfection, the inhibition of proliferation, and the increased apoptosis in microglial cells, induced by OGD/reperfusion, was improved, and the elevation of Caspase-3, Caspase-8, Caspase-9, and poly ADP ribose polymerase (PARP) activities was inhibited. Mass spectrometry demonstrated that IL-17A functioned through a series of factors associated with oxidative stress and apoptosis and regulated Caspase-3 activity and apoptosis in microglial cells via the p53 and PI3K/Akt signaling pathways. IL-17A, HMGB1, and ROS were regulated mutually to exhibit a synergistic effect in the OGD model of microglial cells, but the down-regulation of IL-17A or HMGB1 expression did not completely inhibit the production of ROS. These findings demonstrated that ROS might be located upstream of IL-17A and HMGB1 so that ROS can regulate HMGB1/IL-17A expression to affect the p53 and PI3K/Akt signaling pathways and therefore promote the occurrence of apoptosis in microglial cells. These findings provide a novel evidence for the role of IL-17A in ischemic cerebral diseases.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Neurology, the Fifth Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Ning Yang
- Department of Neurology, the Fifth Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Zhi-Ming Mo
- Department of Neurology, the Fifth Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Shao-Peng Lin
- Department of Emergency, the Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Feng Zhang
- Department of Neurosurgery, the Fourth Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| |
Collapse
|
30
|
Pena-Philippides JC, Caballero-Garrido E, Lordkipanidze T, Roitbak T. In vivo inhibition of miR-155 significantly alters post-stroke inflammatory response. J Neuroinflammation 2016; 13:287. [PMID: 27829437 PMCID: PMC5103429 DOI: 10.1186/s12974-016-0753-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/27/2016] [Indexed: 12/12/2022] Open
Abstract
Background MicroRNA miR-155 is implicated in modulation of the inflammatory processes in various pathological conditions. In our previous studies, we demonstrated that in vivo inhibition of miR-155 promotes functional recovery after mouse experimental stroke. In the present study, we explored if this beneficial effect is associated with miR-155 inhibition-induced alterations in post-stroke inflammatory response. Methods Intravenous injections of a specific miR-155 inhibitor were initiated at 48 h after mouse distal middle cerebral artery occlusion (dMCAO). Temporal changes in the expression of cytokines and key molecules associated with cytokine signaling were assessed at 7, 14, and 21 days after dMCAO, using mouse cytokine gene and protein arrays and Western blot analyses. Electron and immunofluorescence confocal microscopy techniques were used to evaluate the ultrastructural changes, as well as altered expression of specific phenotypic markers, at different time points after dMCAO. Results In the inhibitor-injected mice (inhibitor group), there was a significant decrease in CCL12 and CXCL3 cytokine expression at 7 days and significantly increased levels of major cytokines IL-10, IL-4, IL-6, MIP-1α, IL-5, and IL-17 at 14 days after dMCAO. These temporal changes correlated with altered expression of miR-155 target proteins SOCS-1, SHIP-1, and C/EBP-β and phosphorylation levels of cytokine signaling regulator STAT-3. Electron microscopy showed decreased number of phagocytically active peri-vascular microglia/macrophages in the inhibitor samples. Immunofluorescence and Western blot of these samples demonstrated that expression of leukocyte/ macrophage marker CD45 and phagocytosis marker CD68 was reduced at 7 days, and in contrast, significantly increased at 14 days after dMCAO, as compared to controls. Conclusions Based on our findings, we propose that in vivo miR-155 inhibition following mouse stroke significantly alters the time course of the expression of major cytokines and inflammation-associated molecules, which could influence inflammation process and tissue repair after experimental cerebral ischemia. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0753-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Juan Carlos Pena-Philippides
- Department of Neurosurgery, University of New Mexico Health Sciences Center, 1101 Yale Blvd, Albuquerque, NM, 87106-3834, USA
| | - Ernesto Caballero-Garrido
- Department of Neurosurgery, University of New Mexico Health Sciences Center, 1101 Yale Blvd, Albuquerque, NM, 87106-3834, USA
| | | | - Tamara Roitbak
- Department of Neurosurgery, University of New Mexico Health Sciences Center, 1101 Yale Blvd, Albuquerque, NM, 87106-3834, USA.
| |
Collapse
|
31
|
Minhas G, Sharma J, Khan N. Cellular Stress Response and Immune Signaling in Retinal Ischemia-Reperfusion Injury. Front Immunol 2016; 7:444. [PMID: 27822213 PMCID: PMC5075763 DOI: 10.3389/fimmu.2016.00444] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/07/2016] [Indexed: 01/04/2023] Open
Abstract
Ischemia–reperfusion injury is a well-known pathological hallmark associated with diabetic retinopathy, glaucoma, and other related retinopathies that ultimately can lead to visual impairment and vision loss. Retinal ischemia pathogenesis involves a cascade of detrimental events that include energy failure, excitotoxic damage, calcium imbalance, oxidative stress, and eventually cell death. Retina for a long time has been known to be an immune privileged site; however, recent investigations reveal that retina, as well as the central nervous system, elicits immunological responses during various stress cues. Stress condition, such as reperfusion of blood supply post-ischemia results in the sequestration of different immune cells, inflammatory mediators including cytokines, chemokines, etc., to the ischemic region, which in turn facilitates induction of inflammatory conditions in these tissues. The immunological activation during injury or stress per se is beneficial for repair and maintenance of cellular homeostasis, but whether the associated inflammation is good or bad, during ischemia–reperfusion injury, hitherto remains to be explored. Keeping all these notions in mind, the current review tries to address the immune response and host stress response mechanisms involved in ischemia–reperfusion injury with the focus on the retina.
Collapse
Affiliation(s)
- Gillipsie Minhas
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad , Hyderabad , Telangana, India
| | - Jyoti Sharma
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad , Hyderabad , Telangana, India
| | - Nooruddin Khan
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad , Hyderabad , Telangana, India
| |
Collapse
|
32
|
Qian L, Yuanshao L, Wensi H, Yulei Z, Xiaoli C, Brian W, Wanli Z, Zhengyi C, Jie X, Wenhui Z, Tieer Y, Hong W, Jincai H, Kunlin J, Bei S. Serum IL-33 Is a Novel Diagnostic and Prognostic Biomarker in Acute Ischemic Stroke. Aging Dis 2016; 7:614-622. [PMID: 27699084 PMCID: PMC5036956 DOI: 10.14336/ad.2016.0207] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 02/07/2016] [Indexed: 01/12/2023] Open
Abstract
Interleukin-33 (IL-33), a newly recognized IL-1 family member, is expressed in various tissues and cells, and involved in pathogenesis of many human diseases. For example, IL-33 plays a protective role in cardiovascular diseases. However, the role of IL-33 in acute ischemic stroke (AIS) remains unclear. This study aims to investigate whether IL-33 level in AIS patient serum can be used as a potential diagnostic and prognostic marker. The study included two hundred and six patients with first-ever ischemic stroke, who were admitted within 72 hours after stroke onset. The serum level of IL-33 was measured with ELISA and the severity of AIS patients on admission was evaluated based on the National Institutes of Health Stroke Scale (NIHSS) score. The functional outcome at 3 months was determined using the Barthel index (BI). We found that serum IL-33 was significantly higher (P < 0.001) in patients with AIS [57.68 ng/L (IQR, 44.95-76.73)] compared with healthy controls [47.48 ng/L (IQR, 38.67-53.78)]. IL-33 was an independent diagnostic biomarker for AIS with an OR of 1.051 (95%Cl, 1.018-1.085; P=0.002). Serum IL-33 was higher (P < 0.05) in the stroke patients with small cerebral infarction volume compared to AIS patients with large cerebral infarction. In addition, serum IL-33 was also significantly higher (P = 0.001) in the patients with mild stroke, compared to the patients with severe stroke. Furthermore, serum IL-33 level in AIS patients with a worse outcome was higher (P < 0.001) compared to AIS patients with a better outcome. IL-33 was also an independent predictor for the functional outcome with an adjusted OR of 0.932 (95% CI, 0.882-0.986). Our results suggest that the lower level of serum IL-33 is associated with large infarction volume and greater stroke severity in AIS patients. Thus, IL-33 can be used as a novel and independent diagnostic and predicting prognostic marker in AIS.
Collapse
Affiliation(s)
- Li Qian
- 1Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research
| | - Lin Yuanshao
- 1Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research
| | - Huang Wensi
- 1Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research
| | - Zhou Yulei
- 1Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research
| | - Chen Xiaoli
- 1Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research
| | - Wang Brian
- 3Institute for Healthy Aging, Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, Texas 76107, USA
| | - Zhang Wanli
- 1Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research
| | - Cai Zhengyi
- 1Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research
| | - Xue Jie
- 1Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research
| | - Zhang Wenhui
- 2Department of Clinical Laboratory Medicine, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 35000, China
| | - Yu Tieer
- 1Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research
| | - Wang Hong
- 1Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research
| | - He Jincai
- 1Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research
| | - Jin Kunlin
- 1Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research,; 3Institute for Healthy Aging, Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, Texas 76107, USA
| | - Shao Bei
- 1Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research
| |
Collapse
|
33
|
Jansson D, Scotter EL, Rustenhoven J, Coppieters N, Smyth LCD, Oldfield RL, Bergin PS, Mee EW, Graham ES, Faull RLM, Dragunow M. Interferon-γ blocks signalling through PDGFRβ in human brain pericytes. J Neuroinflammation 2016; 13:249. [PMID: 27654972 PMCID: PMC5031293 DOI: 10.1186/s12974-016-0722-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/13/2016] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Neuroinflammation and blood-brain barrier (BBB) disruption are common features of many brain disorders, including Alzheimer's disease, epilepsy, and motor neuron disease. Inflammation is thought to be a driver of BBB breakdown, but the underlying mechanisms for this are unclear. Brain pericytes are critical cells for maintaining the BBB and are immunologically active. We sought to test the hypothesis that inflammation regulates the BBB by altering pericyte biology. METHODS We exposed primary adult human brain pericytes to chronic interferon-gamma (IFNγ) for 4 days and measured associated functional aspects of pericyte biology. Specifically, we examined the influence of inflammation on platelet-derived growth factor receptor-beta (PDGFRβ) expression and signalling, as well as pericyte proliferation and migration by qRT-PCR, immunocytochemistry, flow cytometry, and western blotting. RESULTS Chronic IFNγ treatment had marked effects on pericyte biology most notably through the PDGFRβ, by enhancing agonist (PDGF-BB)-induced receptor phosphorylation, internalization, and subsequent degradation. Functionally, chronic IFNγ prevented PDGF-BB-mediated pericyte proliferation and migration. CONCLUSIONS Because PDGFRβ is critical for pericyte function and its removal leads to BBB leakage, our results pinpoint a mechanism linking chronic brain inflammation to BBB dysfunction.
Collapse
Affiliation(s)
- Deidre Jansson
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Gravida National Centre for Growth and Development, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Emma L Scotter
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Justin Rustenhoven
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Natacha Coppieters
- Department of Anatomy and Medical Imaging, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Leon C D Smyth
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | | | - Peter S Bergin
- Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand.,Auckland City Hospital, 1023, Auckland, New Zealand
| | - Edward W Mee
- Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand.,Auckland City Hospital, 1023, Auckland, New Zealand
| | - E Scott Graham
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Richard L M Faull
- Department of Anatomy and Medical Imaging, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Mike Dragunow
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand. .,Gravida National Centre for Growth and Development, The University of Auckland, 1023, Auckland, New Zealand. .,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand. .,Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, 1142, Auckland, New Zealand.
| |
Collapse
|
34
|
Interleukin 6 promoter 174 G/C polymorphisms in acute ischemic stroke: G allele is protective but not associated with IL-6 levels or stroke outcome. J Neuroimmunol 2016; 293:22-27. [DOI: 10.1016/j.jneuroim.2016.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 01/27/2016] [Accepted: 02/02/2016] [Indexed: 02/06/2023]
|
35
|
Matsui T, Yoshida Y. Reduction of the expression and production of adhesion molecules and chemokines by brain endothelial cells in response to tumor necrosis factor-α and interleukin-17 in hypothermia. ACTA ACUST UNITED AC 2016. [DOI: 10.1111/cen3.12298] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Tomohiro Matsui
- Department of Laboratory Sciences; Yamaguchi University Graduate School of Medicine; Ube Japan
| | - Yusuke Yoshida
- Research and Development Department; ACEL; Sagamihara Japan
| |
Collapse
|
36
|
The role of IL-17 in CNS diseases. Acta Neuropathol 2015; 129:625-37. [PMID: 25716179 DOI: 10.1007/s00401-015-1402-7] [Citation(s) in RCA: 244] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/18/2015] [Accepted: 02/19/2015] [Indexed: 12/19/2022]
Abstract
Cytokines of the IL-17 family are uniquely placed on the border between immune cells and tissue. Although IL-17 was originally found to induce the activation and mobilization of neutrophils to sites of inflammation, its tissue-specific function is not yet fully understood. The best-studied IL-17 family members, IL-17A and IL-17F, are both typically produced by immune cells such as Th17, γδ T cells and innate lymphoid cells group 3. However, the cells that respond to these cytokines are mostly found in inflamed tissue. As seen in psoriatic skin lesions or in joints of rheumatoid arthritis patients, high levels of IL-17 have been detected in the central nervous system (CNS) during inflammatory responses. Here, we provide a general review of the molecular function of IL-17 and its role in the CNS in particular. Of the different inflammatory conditions of the CNS, we found multiple sclerosis (MS) to be the one most associated with the presence of Th17 cells and IL-17. In particular, many studies using the murine model for MS, experimental autoimmune encephalomyelitis, found a clear association of Th17 and IL-17 with disease severity and progression. We summarize the recent advances made in correlating the presence of IL-17 with impaired blood-brain barrier integrity as well as the activation of astrocytes and microglia and the consequences for disease progression. There is also evidence that IL-17 plays a pathogenic role in the post-ischemic phase of stroke as well as its experimental model. We review the limited but promising data on the sources of post-stroke IL-17 production and its effects on CNS-resident target cells. In addition to MS and stroke, there is also evidence linking high levels of IL-17 to depression, as a frequent comorbidity of several inflammatory diseases, as well as to different types of infections of the CNS. The evidence we supply here suggests that inhibiting the function of the IL-17 cytokine family could have a beneficial effect on pathogenic conditions in the CNS.
Collapse
|
37
|
Zheng Y, Zhong D, Chen H, Ma S, Sun Y, Wang M, Liu Q, Li G. Pivotal role of cerebral interleukin-23 during immunologic injury in delayed cerebral ischemia in mice. Neuroscience 2015; 290:321-31. [PMID: 25637493 DOI: 10.1016/j.neuroscience.2015.01.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/18/2014] [Accepted: 01/06/2015] [Indexed: 10/24/2022]
Abstract
BACKGROUND Interleukin-23 (IL-23) is required for T helper 17 (Th17) cell responses and IL-17 production in ischemic stroke. We previously showed that the IL-23/IL-17 axis aggravates immune injury after cerebral infarction in mice. However, IL-23 might activate other cytokines and transcription factor forkhead box P3 (Foxp3) production in cerebral ischemia. We aimed to determine whether IL-23p19 knockdown prevents cerebral ischemic injury by reducing ischemic-induced inflammation. METHODS Ischemic stroke models were established by permanent middle cerebral arterial occlusion (pMCAO) in male C57BL/6 mice. In vivo gene knockdown was achieved by intravenous delivery of lentiviral vectors (LVs) encoding IL-23p19 short hairpin RNA (LV-IL-23p19 shRNA). Enzyme-linked immunoassay (ELISA) and quantitative real-time polymerase chain reaction (qRT-PCR) confirmed inhibitory efficiency. Behavioral deficits were evaluated by adhesive-removal somatic-sensory test. Brain infarct volume was measured at day 5 after pMCAO by 2,3,5-triphenyltetrazolium chloride (TTC) staining. Expression of IL-17, IL-4, interferon (IFN)-γ and Foxp3 in ischemic brain tissues were detected by qRT-PCR and Western blotting, respectively. Additionally, immunohistochemical staining located cytokines in ischemic brain tissues. RESULTS RNA interference knockdown of IL-23p19 resulted in improved neurological function and reduced infarct volume. IL-23p19 knockdown suppressed IL-17 gene and protein expression. Moreover, IL-23p19 deficiency enhanced IFN-γ and Foxp3 expressions in delayed cerebral ischemic mice, and did not impact IL-4 expression. Immunohistochemical staining showed that IL-17, IL-4, IFN-γ and Foxp3-positive cells were located around ischemic lesions of the ipsilateral hemisphere. CONCLUSIONS IL-23p19 knockdown prevents delayed cerebral ischemic injury by dampening the ischemia-induced inflammation, and is a promising approach for clinically managing ischemic stroke.
Collapse
Affiliation(s)
- Y Zheng
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - D Zhong
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - H Chen
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - S Ma
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - Y Sun
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - M Wang
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - Q Liu
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - G Li
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China.
| |
Collapse
|
38
|
Interleukin-17 inhibits adult hippocampal neurogenesis. Sci Rep 2014; 4:7554. [PMID: 25523081 PMCID: PMC4271266 DOI: 10.1038/srep07554] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 10/16/2014] [Indexed: 12/20/2022] Open
Abstract
Interleukin 17(A) (IL-17) is a potent pro-inflammatory cytokine that acts as a central regulator of inflammatory response within the brain, but its physiological roles under non-inflammatory conditions remain elusive. Here we report that endogenous IL-17 ablates neurogenesis in the adult dentate gyrus (DG) of hippocampus. Genetic deletion of IL-17 increased the number of adult-born neurons in the DG. Further, we found that IL-17 deletion altered cytokine network, facilitated basal excitatory synaptic transmission, enhanced intrinsic neuronal excitability, and increased expression of proneuronal genes in neuronal progenitor cells (NPCs). Our findings suggest a profound role of IL-17 in the negative regulation of adult hippocampal neurogenesis under physiology conditions.
Collapse
|
39
|
Hypothermia Reduces but Hyperthermia Augments T Cell-Derived Release of Interleukin-17 and Granzyme B that Mediate Neuronal Cell Death. Neurocrit Care 2014; 23:116-26. [DOI: 10.1007/s12028-014-0094-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
40
|
Doll D, Barr TL, Simpkins JW. Cytokines: their role in stroke and potential use as biomarkers and therapeutic targets. Aging Dis 2014; 5:294-306. [PMID: 25276489 DOI: 10.14336/ad.2014.0500294] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 05/16/2014] [Accepted: 05/27/2014] [Indexed: 12/30/2022] Open
Abstract
Inflammatory mechanisms both in the periphery and in the CNS are important in the pathophysiologic processes occurring after the onset of ischemic stroke (IS). Cytokines are key players in the inflammatory mechanism and contribute to the progression of ischemic damage. This literature review focuses on the effects of inflammation on ischemic stroke, and the role pro-inflammatory and anti-inflammatory cytokines play on deleterious or beneficial stroke outcome. The discovery of biomarkers and novel therapeutics for stroke has been the focus of extensive research recently; thus, understanding the roles of pro-inflammatory and anti-inflammatory cytokines that are up-regulated during stroke will help us further understand how inflammation contributes to the progression of ischemic damage and provide potential targets for novel therapeutics and biomarkers for diagnosis and prognosis of stroke.
Collapse
Affiliation(s)
| | - Taura L Barr
- School of Nursing, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA ; Center for Basic and Translational Stroke Research, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA
| | - James W Simpkins
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA ; Center for Basic and Translational Stroke Research, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA
| |
Collapse
|
41
|
Dotson AL, Zhu W, Libal N, Alkayed NJ, Offner H. Different immunological mechanisms govern protection from experimental stroke in young and older mice with recombinant TCR ligand therapy. Front Cell Neurosci 2014; 8:284. [PMID: 25309326 PMCID: PMC4174768 DOI: 10.3389/fncel.2014.00284] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 08/26/2014] [Indexed: 12/02/2022] Open
Abstract
Stroke is a leading cause of death and disability in the United States. The lack of clinical success in stroke therapies can be attributed, in part, to inadequate basic research on aging rodents. The current study demonstrates that recombinant TCR ligand therapy uses different immunological mechanisms to protect young and older mice from experimental stroke. In young mice, RTL1000 therapy inhibited splenocyte efflux while reducing frequency of T cells and macrophages in the spleen. Older mice treated with RTL1000 exhibited a significant reduction in inflammatory cells in the brain and inhibition of splenic atrophy. Our data suggest age specific differences in immune response to stroke that allow unique targeting of stroke immunotherapies.
Collapse
Affiliation(s)
- Abby L Dotson
- Neuroimmunology Research, VA Medical Center Portland, OR, USA ; Department of Neurology, Oregon Health and Science University Portland, OR, USA
| | - Wenbin Zhu
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University Portland, OR, USA
| | - Nicole Libal
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University Portland, OR, USA
| | - Nabil J Alkayed
- Department of Neurology, Oregon Health and Science University Portland, OR, USA ; Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University Portland, OR, USA ; Knight Cardiovascular Institute, Oregon Health and Science University Portland, OR, USA
| | - Halina Offner
- Neuroimmunology Research, VA Medical Center Portland, OR, USA ; Department of Neurology, Oregon Health and Science University Portland, OR, USA ; Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University Portland, OR, USA
| |
Collapse
|
42
|
Deng G, Carter J, Traystman RJ, Wagner DH, Herson PS. Pro-inflammatory T-lymphocytes rapidly infiltrate into the brain and contribute to neuronal injury following cardiac arrest and cardiopulmonary resuscitation. J Neuroimmunol 2014; 274:132-40. [PMID: 25084739 DOI: 10.1016/j.jneuroim.2014.07.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 07/03/2014] [Accepted: 07/15/2014] [Indexed: 01/11/2023]
Abstract
Although inflammatory mechanisms have been linked to neuronal injury following global cerebral ischemia, the presence of infiltrating peripheral immune cells remains understudied. We performed flow cytometry of single cell suspensions obtained from the brains of mice at varying time points after global cerebral ischemia induced by cardiac arrest and cardiopulmonary resuscitation (CA/CPR) to characterize the influx of lymphocytes into the injured brain. We observed that CA/CPR caused a large influx of lymphocytes within 3h of resuscitation that was maintained for the 3day duration of our experiments. Using cell staining flow cytometry we observed that the large majority of infiltrating lymphocytes were CD4(+) T cells. Intracellular stains revealed a large proportion of pro-inflammatory T cells expressing either TNFα or INFγ. Importantly, the lack of functional T cells in TCRα knockout mice reduced neuronal injury following CA/CPR, implicating pro-inflammatory T cells in the progression of ischemic neuronal injury. Finally, we made the remarkable observation that the novel CD4(+)CD40(+) (Th40) population of pro-inflammatory T cells that are strongly associated with autoimmunity are present in large numbers in the injured brain. These data indicate that studies investigating the neuro-immune response after global cerebral ischemia should consider the role of infiltrating T cells in orchestrating the acute and sustained immune response.
Collapse
Affiliation(s)
- Guiying Deng
- Department of Pharmacology, University of Colorado School of Medicine, 12800 E. 19th Ave., Aurora, CO 80045, USA
| | - Jessica Carter
- Webb Waring Center, University of Colorado School of Medicine, 12850 E. Montview Blvd., Aurora, CO 80045, USA
| | - Richard J Traystman
- Department of Pharmacology, University of Colorado School of Medicine, 12800 E. 19th Ave., Aurora, CO 80045, USA; Department of Anesthesiology, University of Colorado School of Medicine, 12800 E. 19th Ave., Aurora, CO 80045, USA
| | - David H Wagner
- Webb Waring Center, University of Colorado School of Medicine, 12850 E. Montview Blvd., Aurora, CO 80045, USA
| | - Paco S Herson
- Department of Pharmacology, University of Colorado School of Medicine, 12800 E. 19th Ave., Aurora, CO 80045, USA; Department of Anesthesiology, University of Colorado School of Medicine, 12800 E. 19th Ave., Aurora, CO 80045, USA.
| |
Collapse
|
43
|
Seifert HA, Pennypacker KR. Molecular and cellular immune responses to ischemic brain injury. Transl Stroke Res 2014; 5:543-53. [PMID: 24895236 DOI: 10.1007/s12975-014-0349-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 05/19/2014] [Accepted: 05/21/2014] [Indexed: 12/21/2022]
Abstract
Despite extensive research into stroke pathology, there have not been any major recent advancements in stroke therapeutics. Animal models of cerebral ischemia and clinical data have been used to investigate the progressive neural injury that occurs after an initial ischemic insult. This has lead researchers to focus more on the peripheral immune response that is generated as a result of cerebral ischemia. The therapies that have been developed as a result of this research thus far have proven ineffective in clinical trials. The failure of these therapeutics in clinical trials is thought to be due to the broad immunosuppression elicited as a result of the treatments and the cerebral ischemia itself. Emerging evidence indicates a more selective modulation of the immune system following stroke could be beneficial. The spleen has been shown to exacerbate neural injury following experimental stroke and would provide a strong therapeutic target. Selecting facets of the immune system to target would allow the protective and regenerative properties of the immune response to remain intact while blunting the pro-inflammatory response generated towards the injured brain.
Collapse
Affiliation(s)
- Hilary A Seifert
- Department of Molecular Pharmacology and Physiology, School of Basic Biomedical Sciences, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd., MDC Box 8, Tampa, FL, 33612, USA
| | | |
Collapse
|
44
|
LEÓN R, PAVÓN N, PENTÓN G, ALMAGUER W, MARÍN J, CRUZ A, LORIGADOS L, BLANCO L, ESTUPIÑÁN B, MERCERON D, MACÍAS L, BERGADO J. Experimental Model of Cerebral Hypoperfusion Produced Memory-learning Deficits, and Modifications in Gene Expression. ACTA BIOLÓGICA COLOMBIANA 2014. [DOI: 10.15446/abc.v20n1.40976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
45
|
Zhang Y, Gao Z, Wang D, Zhang T, Sun B, Mu L, Wang J, Liu Y, Kong Q, Liu X, Zhang Y, Zhang H, He J, Li H, Wang G. Accumulation of natural killer cells in ischemic brain tissues and the chemotactic effect of IP-10. J Neuroinflammation 2014; 11:79. [PMID: 24742325 PMCID: PMC4039314 DOI: 10.1186/1742-2094-11-79] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 04/05/2014] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Stroke is accompanied by a distinguished inflammatory reaction that is initiated by the infiltration of immunocytes, expression of cytokines, and other inflammatory mediators. As natural killer cells (NK cells) are a type of cytotoxic lymphocyte critical to the innate immune system, we investigated the mechanism of NK cells-induced brain injuries after cerebral ischemia and the chemotactic effect of IP-10 simultaneously. METHODS NK cells infiltration, interferon-gamma (IFN-γ) and IP-10 expression were detected by immunohistochemistry, immunofluorescence, PCR and flow cytometry in human and C57/BL6 wild type mouse ischemic brain tissues. The ischemia area was detected via 2,3,5-triphenyltetrazolium chloride staining. CXCR3 mean fluorescence intensity of isolated NK cells was measured by flow cytometry. The neuronal injury made by NK cells was examined via apoptosis experiment. The chemotactic of IP-10 was detected by migration and permeability assays. RESULTS In human ischemic brain tissue, infiltrations of NK cells were observed and reached a peak at 2 to 5 days. In a permanent middle cerebral artery occlusion (pMCAO) model, infiltration of NK cells into the ischemic infarct region reached their highest levels 12 hours after ischemia. IFN-γ-positive NK cells and levels of the chemokine IP-10 were also detected within the ischemic region, from 6 hours up to 4 days after pMCAO was performed, and IFN-γ levels decreased after NK cells depletion in vivo. Co-culture experiments of neural cells with NK cells also showed that neural necrosis was induced via IFN-γ. In parallel experiments with IP-10, the presence of CXCR3 indicates that NK cells were affected by IP-10 via CXCR3, and the effect was dose-dependent. After IP-10 depletion in vivo, NK cells decreased. In migration assays and permeability experiments, disintegration of the blood-brain barrier (BBB) was observed following the addition of NK cells. Moreover, in the presence of IP-10 this injury was aggravated. CONCLUSIONS All findings support the hypothesis that NK cells participate in cerebral ischemia and promote neural cells necrosis via IFN-γ. Moreover, IP-10 intensifies injury to the BBB by NK cells via CXCR3.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Hulun Li
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, Harbin Medical University, Xuefu Road, 150081, Heilongjiang, People's Republic of China.
| | | |
Collapse
|
46
|
IL-10 deficiency exacerbates the brain inflammatory response to permanent ischemia without preventing resolution of the lesion. J Cereb Blood Flow Metab 2013; 33:1955-66. [PMID: 24022622 PMCID: PMC3851905 DOI: 10.1038/jcbfm.2013.155] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 07/31/2013] [Accepted: 08/02/2013] [Indexed: 11/08/2022]
Abstract
Stroke induces inflammation that can aggravate brain damage. This work examines whether interleukin-10 (IL-10) deficiency exacerbates inflammation and worsens the outcome of permanent middle cerebral artery occlusion (pMCAO). Expression of IL-10 and IL-10 receptor (IL-10R) increased after ischemia. From day 4, reactive astrocytes showed strong IL-10R immunoreactivity. Interleukin-10 knockout (IL-10 KO) mice kept in conventional housing showed more mortality after pMCAO than the wild type (WT). This effect was associated with the presence of signs of colitis in the IL-10 KO mice, suggesting that ongoing systemic inflammation was a confounding factor. In a pathogen-free environment, IL-10 deficiency slightly increased infarct volume and neurologic deficits. Induction of proinflammatory molecules in the IL-10 KO brain was similar to that in the WT 6 hours after ischemia, but was higher at day 4, while differences decreased at day 7. Deficiency of IL-10 promoted the presence of more mature phagocytic cells in the ischemic tissue, and enhanced the expression of M2 markers and the T-cell inhibitory molecule CTLA-4. These findings agree with a role of IL-10 in attenuating local inflammatory reactions, but do not support an essential function of IL-10 in lesion resolution. Upregulation of alternative immunosuppressive molecules after brain ischemia can compensate, at least in part, the absence of IL-10.
Collapse
|
47
|
Seifert HA, Leonardo CC, Hall AA, Rowe DD, Collier LA, Benkovic SA, Willing AE, Pennypacker KR. The spleen contributes to stroke induced neurodegeneration through interferon gamma signaling. Metab Brain Dis 2012; 27:131-41. [PMID: 22354752 PMCID: PMC4739736 DOI: 10.1007/s11011-012-9283-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 02/02/2012] [Indexed: 01/01/2023]
Abstract
Delayed neuronal death associated with stroke has been increasingly linked to the immune response to the injury. Splenectomy prior to middle cerebral artery occlusion (MCAO) is neuroprotective and significantly reduces neuroinflammation. The present study investigated whether splenic signaling occurs through interferon gamma (IFNγ). IFNγ was elevated early in spleens but later in the brains of rats following MCAO. Splenectomy decreased the amount of IFNγ in the infarct post-MCAO. Systemic administration of recombinant IFNγ abolished the protective effects of splenectomy with a concurrent increase in INFγ expression in the brain. These results suggest a role for spleen-derived IFNγ in stroke pathology.
Collapse
Affiliation(s)
- Hilary A. Seifert
- Department of Molecular Pharmacology and Physiology, School of Basic Biomedical Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Christopher C. Leonardo
- Department of Molecular Pharmacology and Physiology, School of Basic Biomedical Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Aaron A. Hall
- Department of Molecular Pharmacology and Physiology, School of Basic Biomedical Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Derrick D. Rowe
- Department of Molecular Pharmacology and Physiology, School of Basic Biomedical Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Lisa A. Collier
- Department of Molecular Pharmacology and Physiology, School of Basic Biomedical Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | | | - Alison E. Willing
- Center for Excellence in Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Keith R. Pennypacker
- Department of Molecular Pharmacology and Physiology, School of Basic Biomedical Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
48
|
Prolonged elevation of cytokine levels after human acute ischaemic stroke with evidence of individual variability. J Neuroimmunol 2012; 246:78-84. [DOI: 10.1016/j.jneuroim.2012.02.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 02/22/2012] [Accepted: 02/23/2012] [Indexed: 01/16/2023]
|
49
|
The Splenic Response to Ischemic Stroke: Neuroinflammation, Immune Cell Migration, and Experimental Approaches to Defining Cellular Mechanisms. Transl Stroke Res 2012. [DOI: 10.1007/978-1-4419-9530-8_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
50
|
Yin Y, Li G. Hypoxia induces T Helper 17 cell upregulation in cultured peripheral blood mononuclear cells from chronic stage patients of severe cerebral infarction. Microbiol Immunol 2011; 55:130-4. [PMID: 21204953 DOI: 10.1111/j.1348-0421.2010.00301.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Studies have shown the IL-17A involvement in human ischemic stroke patients in vivo. Whether the IL-17A expression was originated from Th17 and could be stimulated by hypoxia remained unknown. Here we report the Th17 upregulation in anaerobic cultured PBMC from chronic stage patients of severe cerebral infarction. By using ELISA and FACS we examined IL-1β, IFN-γ, IL-23 and IL-17A protein levels in the supernatants and Th1/Th17 ratios in PBMC. Statistical significance of Th17 but not Th1 upregulation was proved in 6-hr anaerobic cultured patient groups (P < 0.001). Hence, Th17 might be essential in the autoimmune pathogenesis when hypoxia recurs in severe ischemic stroke patients.
Collapse
Affiliation(s)
- YanHong Yin
- Department of Neurology, Chinese People Liberation Army (PLA) General Hospital, 28 Fuxing Road, Haidian District, Beijing 100854, China.
| | | |
Collapse
|