1
|
Peng J, Huang Y, He T, Zhan Y, Liu J. NLRX1 mediated impaired microglial phagocytosis of NETs in cerebral ischemia and reperfusion injury. Cell Death Differ 2025:10.1038/s41418-025-01526-3. [PMID: 40399533 DOI: 10.1038/s41418-025-01526-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 04/25/2025] [Accepted: 05/06/2025] [Indexed: 05/23/2025] Open
Abstract
Ischemic stroke is one of the common causes of disability and death, and subsequent pathological processes consequent to revascularization could promote secondary tissue damage leading to neuronal death, namely cerebral ischemia and reperfusion injury. Neutrophils could invade injured brain parenchyma after vascularization and exert neurotoxicity by forming neutrophil extracellular traps (NETs). However, unwanted NETs were accumulated in the infarcted core of transient middle cerebral artery occlusion (tMCAO) rats and the mechanism is unknown. Efficient microglial phagocytosis is crucial for the homeostasis of cerebral parenchyma after stroke, and dysfunction of microglial phagocytosis of NETs were observed in the infarcted core cortex at tMCAO 1 d and the accumulation of NETs persisted to 7 d, which exerting deleterious neuronal damage after stroke. However, the detailed mechanisms underlying the dysfunction of microglial phagocytosis of NETs remained unclear. Our results further demonstrated that NLRX1 was mainly enhanced in the microglial cells in the infarcted core cortex at tMCAO 1 d and promoted galectin-3 expression on the lysosomes, facilitating the lysosomal dysfunction and impaired microglial phagocytosis via mTOR/TFEB signaling. NLRX1-silencing was able to suppress the galectin-3 intensity, inhibit the phosphorylation of mTOR and facilitate the nuclear localization of TFEB, ameliorating the lysosomal dysfunction and microglial phagocytosis of NETs. Our results uncovered the regulation of NLRX1 in the dysfunctional microglial phagocytosis of NETs and provided insights into the therapeutic potential for targeting at microglial lysosomal function in cerebral ischemia and reperfusion injury.
Collapse
Affiliation(s)
- Jialing Peng
- Department of Neurology and Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Yuxin Huang
- The Second Clinical School, Guangzhou Medical University, Guangzhou, 511495, China
| | - Tengjing He
- The Second Clinical School, Guangzhou Medical University, Guangzhou, 511495, China
| | - Yang Zhan
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Jun Liu
- Department of Neurology and Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China.
- Institute of Neurosciences, Department of Neurology and National Core Cognitive Center, The Second Affiliated Hospital of Guangzhou Medical University, No. 250 Changgang East Road, Haizhu District, Guangzhou, 510261, China.
| |
Collapse
|
2
|
Ye C, Mo Y, Su T, Huang G, Lu J, Tang S, Huang Q, Li Q, Jiang Q, Guo F, Wu P, Zhang G, Yan J. Cross-sectional study on the association between neutrophil-percentage-to-albumin ratio (NPAR) and prevalence of stroke among US adults: NHANES 1999-2018. Front Neurol 2025; 16:1520298. [PMID: 39935611 PMCID: PMC11812522 DOI: 10.3389/fneur.2025.1520298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/14/2025] [Indexed: 02/13/2025] Open
Abstract
The neutrophil-to-albumin ratio (NPAR) is a relatively novel composite biomarker of inflammation, which has been used for prognostication in cardiovascular diseases and may also be associated with stroke. A cross-sectional analysis was conducted using data from the National Health and Nutrition Examination Survey (NHANES) 1999-2018, including 48,734 individuals with complete NPAR and stroke data. The association between stroke prevalence and NPAR values was assessed through multivariate regression analysis. The relationship between these variables was further visualized using restricted cubic splines (RCS). Additionally, potential factors influencing this relationship were explored through subgroup analysis. The regression model revealed a significant association between NPAR and stroke prevalence, even after adjusting for other covariates [1.06 (1.04, 1.08)]. Stroke prevalence was 62% higher in the highest NPAR group compared to the lowest [1.62 (1.40, 1.89)]. The RCS analysis further confirmed this positive correlation. Subgroup analysis showed that this association was not significantly influenced by other factors. This study establishes a strong association between NPAR and stroke prevalence. However, further studies are needed to clarify the underlying mechanisms and establish a direct causal link.
Collapse
Affiliation(s)
- Chenglin Ye
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yong Mo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Tiansheng Su
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Guangxiang Huang
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jiachao Lu
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Shuling Tang
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qianrong Huang
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qiuyun Li
- Department of Breast Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qian Jiang
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Fangzhou Guo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Pinghua Wu
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Guozhong Zhang
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Jun Yan
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
3
|
Shen L, Xie H, Li G, Zhang Y, Ouyang Y, Li A, Lu C, Qu J. A commentary on 'Association of follow-up neutrophil-to-lymphocyte ratio and systemic inflammation response index with stroke-associated pneumonia and functional outcomes in cerebral hemorrhage patients: a case controlled study'. Int J Surg 2024; 110:5278-5279. [PMID: 38729167 PMCID: PMC11325948 DOI: 10.1097/js9.0000000000001565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 05/12/2024]
Affiliation(s)
- Lan Shen
- Emergency Center, Southern Central Hospital of Yunnan Province (The First People's Hospital of Honghe State), Yunnan, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Li B, Xu L, Wang Z, Shi Q, Cui Y, Fan W, Wu Q, Tong X, Yan H. Neutrophil Extracellular Traps Regulate Surgical Brain Injury by Activating the cGAS-STING Pathway. Cell Mol Neurobiol 2024; 44:36. [PMID: 38637346 PMCID: PMC11026279 DOI: 10.1007/s10571-024-01470-9] [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: 10/25/2023] [Accepted: 03/15/2024] [Indexed: 04/20/2024]
Abstract
Surgical brain injury (SBI), induced by neurosurgical procedures or instruments, has not attracted adequate attention. The pathophysiological process of SBI remains sparse compared to that of other central nervous system diseases thus far. Therefore, novel and effective therapies for SBI are urgently needed. In this study, we found that neutrophil extracellular traps (NETs) were present in the circulation and brain tissues of rats after SBI, which promoted neuroinflammation, cerebral edema, neuronal cell death, and aggravated neurological dysfunction. Inhibition of NETs formation by peptidylarginine deiminase (PAD) inhibitor or disruption of NETs with deoxyribonuclease I (DNase I) attenuated SBI-induced damages and improved the recovery of neurological function. We show that SBI triggered the activation of cyclic guanosine monophosphate-adenosine monophosphate synthase stimulator of interferon genes (cGAS-STING), and that inhibition of the cGAS-STING pathway could be beneficial. It is worth noting that DNase I markedly suppressed the activation of cGAS-STING, which was reversed by the cGAS product cyclic guanosine monophosphate-adenosine monophosphate (cGMP-AMP, cGAMP). Furthermore, the neuroprotective effect of DNase I in SBI was also abolished by cGAMP. NETs may participate in the pathophysiological regulation of SBI by acting through the cGAS-STING pathway. We also found that high-dose vitamin C administration could effectively inhibit the formation of NETs post-SBI. Thus, targeting NETs may provide a novel therapeutic strategy for SBI treatment, and high-dose vitamin C intervention may be a promising translational therapy with an excellent safety profile and low cost.
Collapse
Affiliation(s)
- Bingbing Li
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
| | - Lixia Xu
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Zhengang Wang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
| | - Qi Shi
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
| | - Yang Cui
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
| | - Weijia Fan
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Qiaoli Wu
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Xiaoguang Tong
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China.
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China.
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300350, China.
| | - Hua Yan
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China.
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China.
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300350, China.
| |
Collapse
|
5
|
Passino R, Finneran MC, Hafner H, Feng Q, Huffman LD, Zhao XF, Johnson CN, Kawaguchi R, Oses-Prieto JA, Burlingame AL, Geschwind DH, Benowitz LI, Giger RJ. Neutrophil-inflicted vasculature damage suppresses immune-mediated optic nerve regeneration. Cell Rep 2024; 43:113931. [PMID: 38492223 DOI: 10.1016/j.celrep.2024.113931] [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: 09/24/2023] [Revised: 01/03/2024] [Accepted: 02/21/2024] [Indexed: 03/18/2024] Open
Abstract
In adult mammals, injured retinal ganglion cells (RGCs) fail to spontaneously regrow severed axons, resulting in permanent visual deficits. Robust axon growth, however, is observed after intra-ocular injection of particulate β-glucan isolated from yeast. Blood-borne myeloid cells rapidly respond to β-glucan, releasing numerous pro-regenerative factors. Unfortunately, the pro-regenerative effects are undermined by retinal damage inflicted by an overactive immune system. Here, we demonstrate that protection of the inflamed vasculature promotes immune-mediated RGC regeneration. In the absence of microglia, leakiness of the blood-retina barrier increases, pro-inflammatory neutrophils are elevated, and RGC regeneration is reduced. Functional ablation of the complement receptor 3 (CD11b/integrin-αM), but not the complement components C1q-/- or C3-/-, reduces ocular inflammation, protects the blood-retina barrier, and enhances RGC regeneration. Selective targeting of neutrophils with anti-Ly6G does not increase axogenic neutrophils but protects the blood-retina barrier and enhances RGC regeneration. Together, these findings reveal that protection of the inflamed vasculature promotes neuronal regeneration.
Collapse
Affiliation(s)
- Ryan Passino
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Matthew C Finneran
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Hannah Hafner
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Qian Feng
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Lucas D Huffman
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Xiao-Feng Zhao
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Craig N Johnson
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Riki Kawaguchi
- Departments of Psychiatry and Neurology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Program in Neurogenetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Juan A Oses-Prieto
- University of California San Francisco, Department of Pharmaceutical Chemistry, San Francisco, CA 94158, USA
| | - Alma L Burlingame
- University of California San Francisco, Department of Pharmaceutical Chemistry, San Francisco, CA 94158, USA
| | - Daniel H Geschwind
- Departments of Psychiatry and Neurology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Program in Neurogenetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute of Precision Health, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Larry I Benowitz
- Departments of Neurosurgery and Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; Department of Neurosurgery, Boston Children's Hospital, Boston MA 02115, USA; Departmant of Ophthalmology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Roman J Giger
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
6
|
Koecke MHM, Strecker J, Straeten FA, Beuker C, Minnerup J, Schmidt‐Pogoda A, Börsch A. Inhibition of leukocyte migration after ischemic stroke by VE-cadherin mutation in a mouse model leads to reduced infarct volumes and improved motor skills. Brain Behav 2024; 14:e3449. [PMID: 38468566 PMCID: PMC10928452 DOI: 10.1002/brb3.3449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 01/28/2024] [Accepted: 02/04/2024] [Indexed: 03/13/2024] Open
Abstract
AIMS To distinguish between the genuine cellular impact of the ischemic cascade by leukocytes and unspecific effects of edema and humoral components, two knock-in mouse lines were utilized. Mouse lines Y731F and Y685F possess point mutations in VE-cadherin, which lead to a selective inhibition of transendothelial leukocyte migration or impaired vascular permeability. METHODS Ischemic stroke was induced by a model of middle cerebral artery occlusion. Analysis contained structural outcomes (infarct volume and extent of brain edema), functional outcomes (survival analysis, rotarod test, and neuroscore), and the extent and spatial distribution of leukocyte migration (heatmaps and fluorescence-activated cell sorting (FACS) analysis). RESULTS Inhibition of transendothelial leukocyte migration as in Y731F mice leads to smaller infarct volumes (52.33 ± 4719 vs. 70.43 ± 6483 mm3 , p = .0252) and improved motor skills (rotarod test: 85.52 ± 13.24 s vs. 43.06 ± 15.32 s, p = .0285). An impaired vascular permeability as in Y685F mice showed no effect on structural or functional outcomes. Both VE-cadherin mutations did not influence the total immune cell count or spatial distribution in ischemic brain parenchyma. CONCLUSION Selective inhibition of transendothelial leukocyte migration by VE-cadherin mutation after ischemic stroke in a mouse model leads to smaller infarct volumes and improved motor skills.
Collapse
Affiliation(s)
| | - Jan‐Kolja Strecker
- Department of Neurology with Institute of Translational NeurologyUniversity of MünsterMünsterGermany
| | - Frederike Anne Straeten
- Department of Neurology with Institute of Translational NeurologyUniversity of MünsterMünsterGermany
| | - Carolin Beuker
- Department of Neurology with Institute of Translational NeurologyUniversity of MünsterMünsterGermany
| | - Jens Minnerup
- Department of Neurology with Institute of Translational NeurologyUniversity of MünsterMünsterGermany
| | - Antje Schmidt‐Pogoda
- Department of Neurology with Institute of Translational NeurologyUniversity of MünsterMünsterGermany
| | - Anna‐Lena Börsch
- Department of Neurology with Institute of Translational NeurologyUniversity of MünsterMünsterGermany
| |
Collapse
|
7
|
Zawiah M, Khan AH, Farha RA, Usman A, Al-Ashwal FY, Akkaif MA. Assessing the predictive value of neutrophil percentage to albumin ratio for ICU admission in ischemic stroke patients. Front Neurol 2024; 15:1322971. [PMID: 38361641 PMCID: PMC10868651 DOI: 10.3389/fneur.2024.1322971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/09/2024] [Indexed: 02/17/2024] Open
Abstract
Background Acute ischemic stroke (AIS) remains a substantial global health challenge, contributing to increased morbidity, disability, and mortality. This study aimed at investigating the predictive value of the neutrophil percentage to albumin ratio (NPAR) in determining intensive care unit (ICU) admission among AIS patients. Methods A retrospective observational study was conducted, involving AIS cases admitted to a tertiary hospital in Jordan between 2015 and 2020. Lab data were collected upon admission, and the primary outcome was ICU admission during hospitalization. Descriptive and inferential analyses were performed using SPSS version 29. Results In this study involving 364 AIS patients, a subset of 77 (21.2%) required admission to the ICU during their hospital stay, most frequently within the first week of admission. Univariable analysis revealed significantly higher NPAR levels in ICU-admitted ischemic stroke patients compared to those who were not admitted (23.3 vs. 15.7, p < 0.001), and multivariable regression models confirmed that higher NPAR (≥19.107) independently predicted ICU admission in ischemic stroke patients (adjusted odds ratio [aOR] = 4.85, 95% CI: 1.83-12.83). Additionally, lower GCS scores and higher neutrophil-to-lymphocyte ratio (NLR) were also associated with increased likelihood of ICU admission. In terms of predictive performance, NPAR showed the highest accuracy with an AUC of 0.885, sensitivity of 0.805, and specificity of 0.854, using a cutoff value of 19.107. NPAR exhibits an AUC of 0.058, significantly outperforming NLR (Z = 2.782, p = 0.005). Conclusion NPAR emerged as a robust independent predictor of ICU admission in ischemic stroke patients, surpassing the predictive performance of the NLR.
Collapse
Affiliation(s)
- Mohammed Zawiah
- Department of Clinical Pharmacy, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Amer Hayat Khan
- Discipline of Clinical Pharmacy, School of Pharmaceutical Sciences Universiti Sains Malaysia, Penang, Malaysia
| | - Rana Abu Farha
- Department of Clinical Pharmacy and Therapeutics, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - Abubakar Usman
- Department of Clinical Pharmacy and Practice, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Fahmi Y. Al-Ashwal
- Department of Clinical Pharmacy, College of Pharmacy, Al-Ayen Iraqi University, Thi-Qar, Iraq
| | - Mohammed Ahmed Akkaif
- Department of Cardiology, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
8
|
Bricio-Moreno L, Kurt-Jones EA, Sorensen EW, Luster AD, Michael BD. Using Multiphoton Intravital Microscopy to Study Neutrophil Transmigration and Blood-Brain Barrier Permeability in a Mouse Model of Herpes Simplex Virus Encephalitis. Methods Mol Biol 2024; 2828:45-55. [PMID: 39147969 DOI: 10.1007/978-1-0716-4023-4_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Multiphoton intravital microscopy (MP-IVM) is an imaging technique used for the observation of living organisms at a microscopic resolution. The tissue of interest is exposed through a window allowing imaging of cells in real time. Using MP-IVM, the temporospatial kinetics of leukocyte transendothelial migration can be visualized and quantitated using reporter mice and cell-specific fluorophore-conjugated monoclonal antibodies to track the leukocytes within and outside of vascular beds. Here we describe a method used to study neutrophil transendothelial migration and blood-brain barrier permeability in a mouse model of herpes simplex virus I (HSV) encephalitis.
Collapse
Affiliation(s)
- Laura Bricio-Moreno
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Evelyn A Kurt-Jones
- Department of Medicine, Division of Infectious Disease and Immunology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Elizabeth W Sorensen
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew D Luster
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Benedict D Michael
- Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, UK.
- The National Institute for Health Research Health Protection Research Unit for Emerging and Zoonotic Infections, University of Liverpool, Liverpool, UK.
- Department of Clinical Infection Microbiology and Immunology, Institute of Infection, Veterinary, and Ecological Sciences, University of Liverpool, Liverpool, UK.
| |
Collapse
|
9
|
Rana AK, Kumar R, Shukla DN, Singh D. Lithium co-administration with rutin improves post-stroke neurological outcomes via suppressing Gsk-3β activity in a rat model. Free Radic Biol Med 2023; 207:107-119. [PMID: 37414348 DOI: 10.1016/j.freeradbiomed.2023.07.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/24/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
Cerebral ischemic stroke is one of the leading causes of adult disability worldwide. Reperfusion is the only therapeutic option with a lot of side effects. In the current study, we investigated the efficacy of rutin and lithium co-treatment in improving post-stroke neurological outcomes in a transient global cerebral ischemia-reperfusion injury rat model. Middle-aged male rats were subjected to transient global cerebral ischemia-reperfusion. NORT and Y-maze were used to assess the cognitive processes. Lipid peroxidation, protein carbonylation, and nitric oxide assays were performed to study oxidative stress. The excitotoxicity index was calculated by HPLC. Real time-PCR and western blotting were performed to study gene and protein expressions. The co-administration of rutin and lithium improved the overall survival, recognition memory, spatial working memory, and neurological score following cerebral ischemia-reperfusion in rats. Further, a marked decrease in malonaldehyde, protein carbonyls, and nitric oxide levels was observed following combined treatment. The mRNA expression of antioxidant (Hmox1 and Nqo1) and pro-inflammatory (Il2, Il6, and Il1β) markers were significantly attenuated in the rutin and lithium co-administrated group. The treatment inhibited the Gsk-3β and maintained a normal pool of the downstream β-catenin and Nrf2 proteins. The results revealed that co-administration of rutin and lithium had a neuroprotective potential, suggesting it to be a viable treatment to overcome post-stroke deaths and neurological complications.
Collapse
Affiliation(s)
- Anil Kumar Rana
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Rajneesh Kumar
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Durgesh Nandan Shukla
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India
| | - Damanpreet Singh
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
10
|
Zawiah M, Khan AH, Abu Farha R, Usman A, AbuHammour K, Abdeen M, Albooz R. Predictors of stroke-associated pneumonia and the predictive value of neutrophil percentage-to-albumin ratio. Postgrad Med 2023; 135:681-689. [PMID: 37756038 DOI: 10.1080/00325481.2023.2261354] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/13/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND Early recognition of stroke-associated pneumonia (SAP) is critical to reducing morbidity and mortality associated with SAP. This study investigated the predictors of SAP, and the predictive value of the neutrophil percentage-to-albumin ratio (NPAR) for SAP. METHODS This retrospective cohort study was conducted among stroke patients admitted to Jordan University Hospital from January 2015 to May 2021. Multivariable logistic regression was used to identify independent predictors for SAP. The predictive performance was assessed using C-statistics, described as the area under the receiver-operating characteristic curve (AUC, ROC) with a 95% confidence interval. RESULTS Four hundred and six patients were included in the analysis, and the prevalence of SAP was 19.7%. Multivariable logistic analysis showed that males (Adjusted Odds Ratio (AOR): 5.74; 95% Confidence Interval (95%CI): 2.04-1 6.1)], dysphagia (AOR: 5.29; 95% CI: 1.80-15.5), hemiparesis (AOR: 3.27; 95% CI: 1.13-9.47), lower GCS score (AOR: 0.73; 95% CI: 0.58-0.91), higher levels of neutrophil-lymphocyte ratio (NLR) (AOR: 1.15; 95% CI: 1.07-1.24), monocyte-lymphocyte ratio (MLR) (AOR: 1.49; 95% CI: 1.13-1.96), and neutrophil percentage to albumin ratio (NPAR) (AOR: 1.53; 95% CI: 1.33-1.76) were independent predictors of SAP. The NPAR demonstrated a significantly higher AUC than both the NLR (0.939 versus 0.865, Z = 3.169, p = 0.002) and MLR (0.939 versus 0.842, Z = 3.940, p < 0.001). The AUCs of the NLR and MLR were comparable (0.865 versus 0.842, Z = 1.274, p = 0.203). CONCLUSION Male gender, dysphagia and hemiparesis were the strongest predictors of SAP, and NPAR has an excellent performance in predicting SAP which was better than high NLR and MLR.
Collapse
Affiliation(s)
- Mohammed Zawiah
- Discipline of Clinical Pharmacy, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang, Malaysia
| | - Amer Hayat Khan
- Discipline of Clinical Pharmacy, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang, Malaysia
| | - Rana Abu Farha
- Department of Clinical Pharmacy and Therapeutics, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - Abubakar Usman
- Discipline of Clinical Pharmacy, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang, Malaysia
- Department of Clinical Pharmacy and Practice, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Khawla AbuHammour
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Marwa Abdeen
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Rawand Albooz
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman, Jordan
| |
Collapse
|
11
|
Laaker C, Baenen C, Kovács KG, Sandor M, Fabry Z. Immune cells as messengers from the CNS to the periphery: the role of the meningeal lymphatic system in immune cell migration from the CNS. Front Immunol 2023; 14:1233908. [PMID: 37662908 PMCID: PMC10471710 DOI: 10.3389/fimmu.2023.1233908] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
In recent decades there has been a large focus on understanding the mechanisms of peripheral immune cell infiltration into the central nervous system (CNS) in neuroinflammatory diseases. This intense research led to several immunomodulatory therapies to attempt to regulate immune cell infiltration at the blood brain barrier (BBB), the choroid plexus (ChP) epithelium, and the glial barrier. The fate of these infiltrating immune cells depends on both the neuroinflammatory environment and their type-specific interactions with innate cells of the CNS. Although the fate of the majority of tissue infiltrating immune cells is death, a percentage of these cells could become tissue resident immune cells. Additionally, key populations of immune cells can possess the ability to "drain" out of the CNS and act as messengers reporting signals from the CNS toward peripheral lymphatics. Recent data supports that the meningeal lymphatic system is involved not just in fluid homeostatic functions in the CNS but also in facilitating immune cell migration, most notably dendritic cell migration from the CNS to the meningeal borders and to the draining cervical lymph nodes. Similar to the peripheral sites, draining immune cells from the CNS during neuroinflammation have the potential to coordinate immunity in the lymph nodes and thus influence disease. Here in this review, we will evaluate evidence of immune cell drainage from the brain via the meningeal lymphatics and establish the importance of this in animal models and humans. We will discuss how targeting immune cells at sites like the meningeal lymphatics could provide a new mechanism to better provide treatment for a variety of neurological conditions.
Collapse
Affiliation(s)
- Collin Laaker
- Neuroscience Training Program, University of Wisconsin Madison, Madison, WI, United States
| | - Cameron Baenen
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, WI, United States
| | - Kristóf G. Kovács
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, WI, United States
| | - Matyas Sandor
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, WI, United States
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, WI, United States
| |
Collapse
|
12
|
Tariq MB, Lee J, McCullough LD. Sex differences in the inflammatory response to stroke. Semin Immunopathol 2023; 45:295-313. [PMID: 36355204 PMCID: PMC10924671 DOI: 10.1007/s00281-022-00969-x] [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: 07/29/2022] [Accepted: 10/18/2022] [Indexed: 11/11/2022]
Abstract
Ischemic stroke is a leading cause of morbidity and mortality and disproportionally affects women, in part due to their higher longevity. Older women have poorer outcomes after stroke with high rates of cognitive deficits, depression, and reduced quality of life. Post-stroke inflammatory responses are also sexually dimorphic and drive differences in infarct size and recovery. Factors that influence sex-specific immune responses can be both intrinsic and extrinsic. Differences in gonadal hormone exposure, sex chromosome compliment, and environmental/social factors can drive changes in transcriptional and metabolic profiles. In addition, how these variables interact, changes across the lifespan. After the onset of ischemic injury, necrosis and apoptosis occur, which activate microglia and other glial cells within the central nervous system, promoting the release of cytokines and chemokines and neuroinflammation. Cells involved in innate and adaptive immune responses also have dual functions after stroke as they can enhance inflammation acutely, but also contribute to suppression of the inflammatory cascade and later repair. In this review, we provide an overview of the current literature on sex-specific inflammatory responses to ischemic stroke. Understanding these differences is critical to identifying therapeutic options for both men and women.
Collapse
Affiliation(s)
- Muhammad Bilal Tariq
- Memorial Hermann Hospital-Texas Medical Center, Houston, TX, 77030, USA
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin St, MSB7044B, Houston, TX, 77030, USA
| | - Juneyoung Lee
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin St, MSB7044B, Houston, TX, 77030, USA
| | - Louise D McCullough
- Memorial Hermann Hospital-Texas Medical Center, Houston, TX, 77030, USA.
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin St, MSB7044B, Houston, TX, 77030, USA.
| |
Collapse
|
13
|
Bernis ME, Zweyer M, Maes E, Schleehuber Y, Sabir H. Neutrophil Extracellular Traps Release following Hypoxic-Ischemic Brain Injury in Newborn Rats Treated with Therapeutic Hypothermia. Int J Mol Sci 2023; 24:3598. [PMID: 36835009 PMCID: PMC9966013 DOI: 10.3390/ijms24043598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/01/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023] Open
Abstract
The peripheral immune system plays a critical role in neuroinflammation of the central nervous system after an insult. Hypoxic-ischemic encephalopathy (HIE) induces a strong neuroinflammatory response in neonates, which is often associated with exacerbated outcomes. In adult models of ischemic stroke, neutrophils infiltrate injured brain tissue immediately after an ischemic insult and aggravate inflammation via various mechanisms, including neutrophil extracellular trap (NETs) formation. In this study, we used a neonatal model of experimental hypoxic-ischemic (HI) brain injury and demonstrated that circulating neutrophils were rapidly activated in neonatal blood. We observed an increased infiltration of neutrophils in the brain after exposure to HI. After treatment with either normothermia (NT) or therapeutic hypothermia (TH), we observed a significantly enhanced expression level of the NETosis marker Citrullinated H3 (Cit-H3), which was significantly more pronounced in animals treated with TH than in those treated with NT. NETs and NLR family pyrin domain containing 3 (NLRP-3) inflammasome assembly are closely linked in adult models of ischemic brain injury. In this study, we observed an increase in the activation of the NLRP-3 inflammasome at the time points analyzed, particularly immediately after TH, when we observed a significant increase in NETs structures in the brain. Together, these results suggest the important pathological functions of early arriving neutrophils and NETosis following neonatal HI, particularly after TH treatment, which is a promising starting point for the development of potential new therapeutic targets for neonatal HIE.
Collapse
Affiliation(s)
- Maria E. Bernis
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany
- Deutsche Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Margit Zweyer
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany
- Deutsche Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Elke Maes
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany
- Deutsche Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Yvonne Schleehuber
- Deutsche Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Hemmen Sabir
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany
- Deutsche Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| |
Collapse
|
14
|
Clemente-Moragón A, Oliver E, Calle D, Cussó L, Gómez M, Pradillo JM, Castejón R, Rallón N, Benito JM, Fernández-Ferro JC, Carneado-Ruíz J, Moro MA, Sánchez-González J, Fuster V, Cortés-Canteli M, Desco M, Ibáñez B. Neutrophil β 1 adrenoceptor blockade blunts stroke-associated neuroinflammation. Br J Pharmacol 2023; 180:459-478. [PMID: 36181002 PMCID: PMC10100149 DOI: 10.1111/bph.15963] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 04/10/2022] [Accepted: 04/28/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Reperfusion therapy is the standard of care for ischaemic stroke; however, there is a need to identify new therapeutic targets able to ameliorate cerebral damage. Neutrophil β1 adrenoceptors (β1AR) have been linked to neutrophil migration during exacerbated inflammation. Given the central role of neutrophils in cerebral damage during stroke, we hypothesize that β1AR blockade will improve stroke outcomes. EXPERIMENTAL APPROACH Rats were subjected to middle cerebral artery occlusion-reperfusion to evaluate the effect on stroke of the selective β1AR blocker metoprolol (12.5 mg·kg-1 ) when injected i.v. 10 min before reperfusion. KEY RESULTS Magnetic resonance imaging and histopathology analysis showed that pre-reperfusion i.v. metoprolol reduced infarct size. This effect was accompanied by reduced cytotoxic oedema at 24 h and vasogenic oedema at 7 days. Metoprolol-treated rats showed reduced brain neutrophil infiltration and those which infiltrated displayed a high proportion of anti-inflammatory phenotype (N2, YM1+ ). Additional inflammatory models demonstrated that metoprolol specifically blocked neutrophil migration via β1AR and excluded a significant effect on the glia compartment. Consistently, metoprolol did not protect the brain in neutrophil-depleted rats upon stroke. In patients suffering an ischaemic stroke, β1AR blockade by metoprolol reduced circulating neutrophil-platelet co-aggregates. CONCLUSIONS AND IMPLICATIONS Our findings describe that β1AR blockade ameliorates cerebral damage by targeting neutrophils, identifying a novel therapeutic target to improve outcomes in patients with stroke. This therapeutic strategy is in the earliest stages of the translational pathway and should be further explored.
Collapse
Affiliation(s)
- Agustín Clemente-Moragón
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Eduardo Oliver
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Daniel Calle
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Lorena Cussó
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Madrid, Spain.,Centro de Investigación Biomédica en Salud Mental (CIBERSAM), Madrid, Spain
| | - Mónica Gómez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jesús M Pradillo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Madrid, Spain
| | - Raquel Castejón
- Internal Medicine Department, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Norma Rallón
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain.,Hospital Universitario Rey Juan Carlos, Madrid, Spain
| | - José M Benito
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain.,Department of Neurology, Hospital Universitario Rey Juan Carlos, Madrid, Spain
| | - José C Fernández-Ferro
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Department of Neurology, Hospital Universitario Rey Juan Carlos, Madrid, Spain
| | | | - María A Moro
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Cardiovascular Risk Factors and Brain Function program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Valentín Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Marta Cortés-Canteli
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Cardiovascular Risk Factors and Brain Function program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Manuel Desco
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Madrid, Spain.,Centro de Investigación Biomédica en Salud Mental (CIBERSAM), Madrid, Spain
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Cardiology Department, IIS-Fundación Jiménez Díaz Hospital, Madrid, Spain
| |
Collapse
|
15
|
Shi Z, Jiang X, Geng Y, Yue X, Gao J, Cheng X, Zhao M, Zhu L. Expression profile of cytokines and chemokines in a mouse high-altitude cerebral edema model. Int J Immunopathol Pharmacol 2023; 37:3946320231177189. [PMID: 37188519 DOI: 10.1177/03946320231177189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023] Open
Abstract
INTRODUCTION High-altitude cerebral edema (HACE) is considered to be the end-stage of acute mountain sickness (AMS); however, its pathophysiological mechanism remains unknown. Increasing evidences support that inflammation is an important risk factor for the occurrence of HACE. Including our published papers, previous studies demonstrated that the levels of IL-6, IL-1β, and TNF-α in both serum and hippocampus were increased in the mouse HACE model induced by LPS stimulation combined with hypobaric hypoxia exposure; however, the expression profile of other cytokines and chemokines remains unknown. OBJECTIVE This study was to analyze the expression profile of cytokines and chemokines in the HACE model. METHODS The mouse HACE model was established by LPS stimulation combined with hypobaric hypoxia exposure (LH). The mice were divided into the normoxic group, LH-6 h group, LH-1 d group, and LH-7 d group. Brain water content (BWC) was determined using the wet/dry weight ratio. The levels of 30 cytokines and chemokines in the serum and hippocampal tissue were detected using LiquiChip. The mRNA expression of cytokines and chemokines in hippocampal tissue were determined by q-PCR. RESULTS In the current study, we found that the brain water content was increased after the combinational treatment of LPS and hypobaric hypoxia. The results of LiquiChip showed that, in the serum and hippocampal tissue, most factors in all 30 cytokines and chemokines were dramatically upregulated at 6 h, and then declined at the 1st d and 7th d. Among these factors, G-CSF, M-CSF, MCP-1, KC, MIG, Eotaxin, Rantes, IP10, IL-6, MIP-2, and MIP-1β were all increased in both serum and hippocampal tissue at 6 h. In addition, the results of q-PCR showed the mRNA levels of G-CSF, MCP-1, KC, MIG, Eotaxin, Rantes, IP10, IL-6, MIP-2, and MIP-1β in hippocampal tissue were dramatically upregulated at 6 h. CONCLUSION This study showed that the dynamic expression profile of 30 cytokines and chemokines in a mouse HACE model induced by LPS plus hypobaric hypoxia. The levels of G-CSF, MCP-1, KC, MIG, Eotaxin, Rantes, IP10, IL-6, MIP-2, and MIP-1β in both serum and hippocampus were significantly increased at 6 h, which may be involved in the occurrence and development of HACE.
Collapse
Affiliation(s)
- Zibi Shi
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Xiufang Jiang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yanan Geng
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Xiangpei Yue
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jiayue Gao
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Xiang Cheng
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Ming Zhao
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Lingling Zhu
- Beijing Institute of Basic Medical Sciences, Beijing, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- School of Pharmaceutical Sciences, University of South China, Hengyang, China
| |
Collapse
|
16
|
Luo H, Guo H, Zhou Y, Fang R, Zhang W, Mei Z. Neutrophil Extracellular Traps in Cerebral Ischemia/Reperfusion Injury: Friend and Foe. Curr Neuropharmacol 2023; 21:2079-2096. [PMID: 36892020 PMCID: PMC10556361 DOI: 10.2174/1570159x21666230308090351] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/19/2022] [Accepted: 12/26/2022] [Indexed: 03/10/2023] Open
Abstract
Cerebral ischemic injury, one of the leading causes of morbidity and mortality worldwide, triggers various central nervous system (CNS) diseases, including acute ischemic stroke (AIS) and chronic ischemia-induced Alzheimer's disease (AD). Currently, targeted therapies are urgently needed to address neurological disorders caused by cerebral ischemia/reperfusion injury (CI/RI), and the emergence of neutrophil extracellular traps (NETs) may be able to relieve the pressure. Neutrophils are precursors to brain injury following ischemic stroke and exert complicated functions. NETs extracellularly release reticular complexes of neutrophils, i.e., double-stranded DNA (dsDNA), histones, and granulins. Paradoxically, NETs play a dual role, friend and foe, under different conditions, for example, physiological circumstances, infection, neurodegeneration, and ischemia/reperfusion. Increasing evidence indicates that NETs exert anti-inflammatory effects by degrading cytokines and chemokines through protease at a relatively stable and moderate level under physiological conditions, while excessive amounts of NETs release (NETosis) irritated by CI/RI exacerbate the inflammatory response and aggravate thrombosis, disrupt the blood-brain barrier (BBB), and initiates sequential neuron injury and tissue damage. This review provides a comprehensive overview of the machinery of NETs formation and the role of an abnormal cascade of NETs in CI/RI, as well as other ischemia-induced neurological diseases. Herein, we highlight the potential of NETs as a therapeutic target against ischemic stroke that may inspire translational research and innovative clinical approaches.
Collapse
Affiliation(s)
- Haoyue Luo
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Hanjing Guo
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Yue Zhou
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Rui Fang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Wenli Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China
| |
Collapse
|
17
|
Bagatella S, Haghayegh Jahromi N, Monney C, Polidori M, Gall FM, Marchionatti E, Serra F, Riedl R, Engelhardt B, Oevermann A. Bovine neutrophil chemotaxis to Listeria monocytogenes in neurolisteriosis depends on microglia-released rather than bacterial factors. J Neuroinflammation 2022; 19:304. [PMID: 36527076 PMCID: PMC9758797 DOI: 10.1186/s12974-022-02653-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Listeria monocytogenes (Lm) is a bacterial pathogen of major concern for humans and ruminants due to its neuroinvasive potential and its ability to cause deadly encephalitis (neurolisteriosis). On one hand, polymorphonuclear neutrophils (PMN) are key players in the defense against Lm, but on the other hand intracerebral infiltration with PMN is associated with significant neural tissue damage. Lm-PMN interactions in neurolisteriosis are poorly investigated, and factors inducing PMN chemotaxis to infectious foci containing Lm in the central nervous system (CNS) remain unidentified. METHODS In this study, we assessed bovine PMN chemotaxis towards Lm and supernatants of infected endogenous brain cell populations in ex vivo chemotaxis assays, to identify chemotactic stimuli for PMN chemotaxis towards Lm in the brain. In addition, microglial secretion of IL-8 was assessed both ex vivo and in situ. RESULTS Our data show that neither Lm cell wall components nor intact bacteria elicit chemotaxis of bovine PMN ex vivo. Moreover, astrocytes and neural cells fail to induce bovine PMN chemotaxis upon infection. In contrast, supernatant from Lm infected microglia readily induced chemotaxis of bovine PMN. Microglial expression and secretion of IL-8 was identified during early Lm infection in vitro and in situ, although IL-8 blocking with a specific antibody could not abrogate PMN chemotaxis towards Lm infected microglial supernatant. CONCLUSIONS These data provide evidence that host-derived rather than bacterial factors trigger PMN chemotaxis to bacterial foci in the CNS, that microglia have a primary role as initiators of bovine PMN chemotaxis into the brain during neurolisteriosis and that blockade of these factors could be a therapeutic target to limit intrathecal PMN chemotaxis and PMN associated damage in neurolisteriosis.
Collapse
Affiliation(s)
- Stefano Bagatella
- grid.5734.50000 0001 0726 5157Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, CH-3012 Bern, Switzerland ,grid.5734.50000 0001 0726 5157Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Neda Haghayegh Jahromi
- grid.5734.50000 0001 0726 5157Theodor Kocher Institute (TKI), University of Bern, Bern, Switzerland
| | - Camille Monney
- grid.5734.50000 0001 0726 5157Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, CH-3012 Bern, Switzerland
| | - Margherita Polidori
- grid.5734.50000 0001 0726 5157Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, CH-3012 Bern, Switzerland ,grid.5734.50000 0001 0726 5157Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Flavio Max Gall
- grid.19739.350000000122291644Institute of Chemistry and Biotechnology, Competence Center for Drug Discovery, Zurich University of Applied Sciences (ZHAW), Wädenswil, Switzerland
| | - Emma Marchionatti
- grid.5734.50000 0001 0726 5157Clinic for Ruminants, Department of Clinical Veterinary Medicine, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | | | - Rainer Riedl
- grid.19739.350000000122291644Institute of Chemistry and Biotechnology, Competence Center for Drug Discovery, Zurich University of Applied Sciences (ZHAW), Wädenswil, Switzerland
| | - Britta Engelhardt
- grid.5734.50000 0001 0726 5157Theodor Kocher Institute (TKI), University of Bern, Bern, Switzerland
| | - Anna Oevermann
- grid.5734.50000 0001 0726 5157Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, CH-3012 Bern, Switzerland
| |
Collapse
|
18
|
Chen W, Zhang Y, Zhai X, Xie L, Guo Y, Chen C, Li Y, Wang F, Zhu Z, Zheng L, Wan J, Li P. Microglial phagocytosis and regulatory mechanisms after stroke. J Cereb Blood Flow Metab 2022; 42:1579-1596. [PMID: 35491825 PMCID: PMC9441720 DOI: 10.1177/0271678x221098841] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Stroke, including ischemic stroke and hemorrhagic stroke can cause massive neuronal death and disruption of brain structure, which is followed by secondary inflammatory injury initiated by pro-inflammatory molecules and cellular debris. Phagocytic clearance of cellular debris by microglia, the brain's scavenger cells, is pivotal for neuroinflammation resolution and neurorestoration. However, microglia can also exacerbate neuronal loss by phagocytosing stressed-but-viable neurons in the penumbra, thereby expanding the injury area and hindering neurofunctional recovery. Microglia constantly patrol the central nervous system using their processes to scour the cellular environment and start or cease the phagocytosis progress depending on the "eat me" or "don't eat me'' signals on cellular surface. An optimal immune response requires a delicate balance between different phenotypic states to regulate neuro-inflammation and facilitate reconstruction after stroke. Here, we examine the literature and discuss the molecular mechanisms and cellular pathways regulating microglial phagocytosis, their resulting effects in brain injury and neural regeneration, as well as the potential therapeutic targets that might modulate microglial phagocytic activity to improve neurological function after stroke.
Collapse
Affiliation(s)
- Weijie Chen
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yueman Zhang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaozhu Zhai
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lv Xie
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunlu Guo
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Chen
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Li
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fajun Wang
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Ziyu Zhu
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Zheng
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieqing Wan
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Yu F, Wang Y, Stetler AR, Leak RK, Hu X, Chen J. Phagocytic microglia and macrophages in brain injury and repair. CNS Neurosci Ther 2022; 28:1279-1293. [PMID: 35751629 PMCID: PMC9344092 DOI: 10.1111/cns.13899] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/31/2022] [Accepted: 06/04/2022] [Indexed: 12/21/2022] Open
Abstract
AIMS Phagocytosis is the cellular digestion of extracellular particles, such as pathogens and dying cells, and is a key element in the evolution of central nervous system (CNS) disorders. Microglia and macrophages are the professional phagocytes of the CNS. By clearing toxic cellular debris and reshaping the extracellular matrix, microglia/macrophages help pilot the brain repair and functional recovery process. However, CNS resident and invading immune cells can also magnify tissue damage by igniting runaway inflammation and phagocytosing stressed-but viable-neurons. DISCUSSION Microglia/macrophages help mediate intercellular communication and react quickly to the "find-me" signals expressed by dead/dying neurons. The activated microglia/macrophages then migrate to the injury site to initiate the phagocytic process upon encountering "eat-me" signals on the surfaces of endangered cells. Thus, healthy cells attempt to avoid inappropriate engulfment by expressing "do not-eat-me" signals. Microglia/macrophages also have the capacity to phagocytose immune cells that invade the injured brain (e.g., neutrophils) and to regulate their pro-inflammatory properties. During brain recovery, microglia/macrophages engulf myelin debris, initiate synaptogenesis and neurogenesis, and sculpt a favorable extracellular matrix to support network rewiring, among other favorable roles. Here, we review the multilayered nature of phagocytotic microglia/macrophages, including the molecular and cellular mechanisms that govern microglia/macrophage-induced phagocytosis in acute brain injury, and discuss strategies that tap into the therapeutic potential of this engulfment process. CONCLUSION Identification of biological targets that can temper neuroinflammation after brain injury without hindering the essential phagocytic functions of microglia/macrophages will expedite better medical management of the stroke recovery stage.
Collapse
Affiliation(s)
- Fang Yu
- Geriatric Research, Education and Clinical CenterVeterans Affairs Pittsburgh Health Care SystemPittsburghPennsylvaniaUSA
- Pittsburgh Institute of Brain Disorders & Recovery and Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Yangfan Wang
- Geriatric Research, Education and Clinical CenterVeterans Affairs Pittsburgh Health Care SystemPittsburghPennsylvaniaUSA
- Pittsburgh Institute of Brain Disorders & Recovery and Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Anne R. Stetler
- Geriatric Research, Education and Clinical CenterVeterans Affairs Pittsburgh Health Care SystemPittsburghPennsylvaniaUSA
- Pittsburgh Institute of Brain Disorders & Recovery and Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Rehana K. Leak
- Graduate School of Pharmaceutical SciencesSchool of Pharmacy, Duquesne UniversityPittsburghPennsylvaniaUSA
| | - Xiaoming Hu
- Geriatric Research, Education and Clinical CenterVeterans Affairs Pittsburgh Health Care SystemPittsburghPennsylvaniaUSA
- Pittsburgh Institute of Brain Disorders & Recovery and Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Jun Chen
- Geriatric Research, Education and Clinical CenterVeterans Affairs Pittsburgh Health Care SystemPittsburghPennsylvaniaUSA
- Pittsburgh Institute of Brain Disorders & Recovery and Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
20
|
Wang N, Yang Y, Qiu B, Gao Y, Wang A, Xu Q, Meng X, Xu Y, Song B, Wang Y, Wang Y. Correlation of the systemic immune-inflammation index with short- and long-term prognosis after acute ischemic stroke. Aging (Albany NY) 2022; 14:6567-6578. [PMID: 35985678 PMCID: PMC9467411 DOI: 10.18632/aging.204228] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 06/01/2022] [Indexed: 01/05/2023]
Abstract
Immune and inflammatory responses after stroke are important pathophysiological processes. This study explored the relationship between the systemic immune-inflammation index (SII) and stroke prognosis. Patients from the China National Stroke Registry III were investigated. SII was defined as neutrophils × platelets/lymphocytes, and the patients were divided into four groups according to quartiles based on SII values. The primary outcome was poor functional outcome, assessed by the modified Rankin Scale (mRS), defined as an mRS score of ≥3. The secondary outcome was the incidence of all-cause death and recurrent stroke. Data were analyzed using either the logistic regression or Cox regression models. As the SII quartile increased, the percentage of patients with poor functional outcomes increased: 178 (7.8%), 223 (9.8%), 292 (12.8%), and 417 (18.3%) (P < 0.0001) at the 90-day follow-up and 172 (7.6%), 203 (8.9%), 266 (11.7%), and 386 (17.0%) (P < 0.0001) at the 1-year follow-up. Compared to patients in the quartile (Q)1 group, those in the Q4 group had a higher risk for adverse events, especially all-cause death at the 90-day follow-up (adjusted hazard ratio [HR], 2.409; 95% confidence interval [CI], 1.273–4.559, P = 0.0069) and at the 1-year follow-up visits (adjusted HR, 2.209; 95% CI, 1.474–3.311, P = 0.0001). The SII was closely related to the short- and long-term prognosis of patients with acute ischemic stroke, and patients with higher SIIs were more likely to have poor outcomes.
Collapse
Affiliation(s)
- Nan Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan, China.,Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yingying Yang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Centre for Neurological Diseases, Beijing, China
| | - Baoshan Qiu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Centre for Neurological Diseases, Beijing, China
| | - Ying Gao
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Centre for Neurological Diseases, Beijing, China
| | - Anxin Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Centre for Neurological Diseases, Beijing, China
| | - Qin Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Centre for Neurological Diseases, Beijing, China
| | - Xia Meng
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Centre for Neurological Diseases, Beijing, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Bo Song
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Yongjun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Centre for Neurological Diseases, Beijing, China
| | - Yilong Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Centre for Neurological Diseases, Beijing, China
| |
Collapse
|
21
|
Hikosaka M, Kawano T, Wada Y, Maeda T, Sakurai T, Ohtsuki G. Immune-Triggered Forms of Plasticity Across Brain Regions. Front Cell Neurosci 2022; 16:925493. [PMID: 35978857 PMCID: PMC9376917 DOI: 10.3389/fncel.2022.925493] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/16/2022] [Indexed: 01/03/2023] Open
Abstract
Immune cells play numerous roles in the host defense against the invasion of microorganisms and pathogens, which induces the release of inflammatory mediators (e.g., cytokines and chemokines). In the CNS, microglia is the major resident immune cell. Recent efforts have revealed the diversity of the cell types and the heterogeneity of their functions. The refinement of the synapse structure was a hallmark feature of the microglia, while they are also involved in the myelination and capillary dynamics. Another promising feature is the modulation of the synaptic transmission as synaptic plasticity and the intrinsic excitability of neurons as non-synaptic plasticity. Those modulations of physiological properties of neurons are considered induced by both transient and chronic exposures to inflammatory mediators, which cause behavioral disorders seen in mental illness. It is plausible for astrocytes and pericytes other than microglia and macrophage to induce the immune-triggered plasticity of neurons. However, current understanding has yet achieved to unveil what inflammatory mediators from what immune cells or glia induce a form of plasticity modulating pre-, post-synaptic functions and intrinsic excitability of neurons. It is still unclear what ion channels and intracellular signaling of what types of neurons in which brain regions of the CNS are involved. In this review, we introduce the ubiquitous modulation of the synaptic efficacy and the intrinsic excitability across the brain by immune cells and related inflammatory cytokines with the mechanism for induction. Specifically, we compare neuro-modulation mechanisms by microglia of the intrinsic excitability of cerebellar Purkinje neurons with cerebral pyramidal neurons, stressing the inverted directionality of the plasticity. We also discuss the suppression and augmentation of the extent of plasticity by inflammatory mediators, as the meta-plasticity by immunity. Lastly, we sum up forms of immune-triggered plasticity in the different brain regions with disease relevance. Together, brain immunity influences our cognition, sense, memory, and behavior via immune-triggered plasticity.
Collapse
Affiliation(s)
| | | | | | | | | | - Gen Ohtsuki
- Department of Drug Discovery Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
22
|
Zang X, Chen S, Zhu J, Ma J, Zhai Y. The Emerging Role of Central and Peripheral Immune Systems in Neurodegenerative Diseases. Front Aging Neurosci 2022; 14:872134. [PMID: 35547626 PMCID: PMC9082639 DOI: 10.3389/fnagi.2022.872134] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/25/2022] [Indexed: 12/31/2022] Open
Abstract
For decades, it has been widely believed that the blood-brain barrier (BBB) provides an immune privileged environment in the central nervous system (CNS) by blocking peripheral immune cells and humoral immune factors. This view has been revised in recent years, with increasing evidence revealing that the peripheral immune system plays a critical role in regulating CNS homeostasis and disease. Neurodegenerative diseases are characterized by progressive dysfunction and the loss of neurons in the CNS. An increasing number of studies have focused on the role of the connection between the peripheral immune system and the CNS in neurodegenerative diseases. On the one hand, peripherally released cytokines can cross the BBB, cause direct neurotoxicity and contribute to the activation of microglia and astrocytes. On the other hand, peripheral immune cells can also infiltrate the brain and participate in the progression of neuroinflammatory and neurodegenerative diseases. Neurodegenerative diseases have a high morbidity and disability rate, yet there are no effective therapies to stop or reverse their progression. In recent years, neuroinflammation has received much attention as a therapeutic target for many neurodegenerative diseases. In this review, we highlight the emerging role of the peripheral and central immune systems in neurodegenerative diseases, as well as their interactions. A better understanding of the emerging role of the immune systems may improve therapeutic strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Xin Zang
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Si Chen
- Department of Neurology, the Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - JunYao Zhu
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Junwen Ma
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yongzhen Zhai
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
23
|
Wen SW, Shim R, Hall P, Bedo J, Wilson JL, Nicholls AJ, Hickey MJ, Wong CHY. Lung Imaging Reveals Stroke-Induced Impairment in Pulmonary Intravascular Neutrophil Function, a Response Exacerbated with Aging. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2019-2028. [PMID: 35365565 DOI: 10.4049/jimmunol.2100997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/05/2022] [Indexed: 06/14/2023]
Abstract
In stroke patients, infection is a significant contributor to morbidity and mortality. Moreover, older stroke patients show an increased risk of developing stroke-associated infection, although the mechanisms underlying this increased susceptibility to infection are unknown. In this study, using an experimental mouse model of ischemic stroke, we showed that older (12-15 mo of age) mice had elevated lung bacterial infection and inflammatory damage after stroke when compared with young (8-10 wk of age) counterparts, despite undergoing the same degree of brain injury. Intravital microscopy of the lung microvasculature revealed that in younger mice, stroke promoted neutrophil arrest in pulmonary microvessels, but this response was not seen in older poststroke mice. In addition, bacterial phagocytosis by neutrophils in the lung microvasculature was reduced by both aging and stroke, such that neutrophils in aged poststroke mice showed the greatest impairment in this function. Analysis of neutrophil migration in vitro and in the cremaster muscle demonstrated that stroke alone did not negatively impact neutrophil migration, but that the combination of increased age and stroke led to reduced effectiveness of neutrophil chemotaxis. Transcriptomic analysis of pulmonary neutrophils using RNA sequencing identified 79 genes that were selectively altered in the context of combined aging and stroke, and they were associated with pathways that control neutrophil chemotaxis. Taken together, the findings of this study show that stroke in older animals results in worsening of neutrophil antibacterial responses and changes in neutrophil gene expression that have the potential to underpin elevated risk of stroke-associated infection in the context of increased age.
Collapse
Affiliation(s)
- Shu Wen Wen
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Raymond Shim
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Pam Hall
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Justin Bedo
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; and
- School of Computing and Information Systems, The University of Melbourne, Parkville, Victoria, Australia
| | - Jenny L Wilson
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Alyce J Nicholls
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Michael J Hickey
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Connie H Y Wong
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia;
| |
Collapse
|
24
|
Sienel RI, Kataoka H, Kim SW, Seker FB, Plesnila N. Adhesion of Leukocytes to Cerebral Venules Precedes Neuronal Cell Death and Is Sufficient to Trigger Tissue Damage After Cerebral Ischemia. Front Neurol 2022; 12:807658. [PMID: 35140676 PMCID: PMC8818753 DOI: 10.3389/fneur.2021.807658] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/20/2021] [Indexed: 12/18/2022] Open
Abstract
Background Leukocytes contribute to tissue damage after cerebral ischemia; however, the mechanisms underlying this process are still unclear. This study investigates the temporal and spatial relationship between vascular leukocyte recruitment and tissue damage and aims to uncover which step of the leukocyte recruitment cascade is involved in ischemic brain injury. Methods Male wild-type, ICAM-1-deficient, anti-CD18 antibody treated, or selectin-deficient [fucusyltransferase (FucT IV/VII−/−)] mice were subjected to 60 min of middle cerebral artery occlusion (MCAo). The interaction between leukocytes and the cerebrovascular endothelium was quantified by in vivo fluorescence microscopy up to 15 h thereafter. Temporal dynamics of neuronal cell death and leukocyte migration were assessed at the same time points and in the same tissue volume by histology. Results In wild-type mice, leukocytes started to firmly adhere to the wall of pial postcapillary venules two hours after reperfusion. Three hours later, neuronal loss started and 13 h later, leukocytes transmigrated into brain tissue. Loss of selectin function did not influence this process. Application of an anti-CD18 antibody or genetic deletion of ICAM-1, however, significantly reduced tight adhesion of leukocytes to the cerebrovascular endothelium (-60%; p < 0.01) and increased the number of viable neurons in the ischemic penumbra by 5-fold (p < 0.01); the number of intraparenchymal leukocytes was not affected. Conclusions Our findings suggest that ischemia triggers only a transient adhesion of leukocytes to the venous endothelium and that inhibition of this process is sufficient to partly prevent ischemic tissue damage.
Collapse
Affiliation(s)
- Rebecca Isabella Sienel
- Laboratory of Experimental Stroke Research, Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center, Munich, Germany
- Munich Cluster of Systems Neurology (Synergy), Munich, Germany
| | - Hiroharu Kataoka
- Department of Neurosurgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Seong-Woong Kim
- Department of Neurosurgery, University of Giessen, Giessen, Germany
| | - Fatma Burcu Seker
- Laboratory of Experimental Stroke Research, Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center, Munich, Germany
- Munich Cluster of Systems Neurology (Synergy), Munich, Germany
| | - Nikolaus Plesnila
- Laboratory of Experimental Stroke Research, Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center, Munich, Germany
- Munich Cluster of Systems Neurology (Synergy), Munich, Germany
- *Correspondence: Nikolaus Plesnila
| |
Collapse
|
25
|
Yuan J, Li L, Yang Q, Ran H, Wang J, Hu K, Pu W, Huang J, Wen L, Zhou L, Jiang Y, Xiong X, Zhang J, Zhou Z. Targeted Treatment of Ischemic Stroke by Bioactive Nanoparticle-Derived Reactive Oxygen Species Responsive and Inflammation-Resolving Nanotherapies. ACS NANO 2021; 15:16076-16094. [PMID: 34606239 DOI: 10.1021/acsnano.1c04753] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Stroke is a primary cause of death and disability worldwide, while effective and safe drugs remain to be developed for its clinical treatment. Herein, we report bioactive nanoparticle-derived multifunctional nanotherapies for ischemic stroke, which are engineered from a pharmacologically active oligosaccharide material (termed as TPCD) prepared by covalently conjugating a radical-scavenging compound (Tempol) and a hydrogen-peroxide-eliminating moiety of phenylboronic acid pinacol ester (PBAP) on β-cyclodextrin. Of note, combined functional moieties of Tempol and PBAP on β-cyclodextrin contribute to antioxidative and anti-inflammatory activities of TPCD. Cellularly, TPCD nanoparticles (i.e., TPCD NPs) reduced oxygen-glucose deprivation-induced overproduction of oxidative mediators, increased antioxidant enzyme expression, and suppressed microglial-mediated inflammation, thereby inhibiting neuronal apoptosis. After intravenous (i.v.) delivery, TPCD NPs could efficiently accumulate at the cerebral ischemic injury site of mice with middle cerebral artery occlusion (MCAO), showing considerable distribution in cells relevant to the pathogenesis of stroke. Therapeutically, TPCD NPs significantly decreased infarct volume and accelerated recovery of neurological function in MCAO mice. Mechanistically, efficacy of TPCD NPs is achieved by its antioxidative, anti-inflammatory, and antiapoptotic effects. Furthermore, TPCD NPs can function as a reactive oxygen species labile nanovehicle to efficiently load and triggerably release an inflammation-resolving peptide Ac2-26, giving rise to an inflammation-resolving nanotherapy (i.e., ATPCD NP). Compared to TPCD NP, ATPCD NP demonstrated notably enhanced in vivo efficacies, largely resulting from its additional inflammation-resolving activity. Consequently, TPCD NP-derived nanomedicines can be further developed as promising targeted therapies for stroke and other inflammation-associated cerebrovascular diseases.
Collapse
Affiliation(s)
- Jichao Yuan
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lanlan Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qinghua Yang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hong Ran
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jie Wang
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Kaiyao Hu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wendan Pu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jialu Huang
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lan Wen
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Linke Zhou
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ying Jiang
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zhenhua Zhou
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
26
|
Levochkina M, McQuillan L, Awan N, Barton D, Maczuzak J, Bianchine C, Trombley S, Kotes E, Wiener J, Wagner A, Calcagno J, Maza A, Nierstedt R, Ferimer S, Wagner A. Neutrophil-to-Lymphocyte Ratios and Infections after Traumatic Brain Injury: Associations with Hospital Resource Utilization and Long-Term Outcome. J Clin Med 2021; 10:jcm10194365. [PMID: 34640381 PMCID: PMC8509449 DOI: 10.3390/jcm10194365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/20/2021] [Accepted: 09/24/2021] [Indexed: 12/30/2022] Open
Abstract
Traumatic brain injury (TBI) induces immune dysfunction that can be captured clinically by an increase in the neutrophil-to-lymphocyte ratio (NLR). However, few studies have characterized the temporal dynamics of NLR post-TBI and its relationship with hospital-acquired infections (HAI), resource utilization, or outcome. We assessed NLR and HAI over the first 21 days post-injury in adults with moderate-to-severe TBI (n = 196) using group-based trajectory (TRAJ), changepoint, and mixed-effects multivariable regression analysis to characterize temporal dynamics. We identified two groups with unique NLR profiles: a high (n = 67) versus a low (n = 129) TRAJ group. High NLR TRAJ had higher rates (76.12% vs. 55.04%, p = 0.004) and earlier time to infection (p = 0.003). In changepoint-derived day 0–5 and 6–20 epochs, low lymphocyte TRAJ, early in recovery, resulted in more frequent HAIs (p = 0.042), subsequently increasing later NLR levels (p ≤ 0.0001). Both high NLR TRAJ and HAIs increased hospital length of stay (LOS) and days on ventilation (p ≤ 0.05 all), while only high NLR TRAJ significantly increased odds of unfavorable six-month outcome as measured by the Glasgow Outcome Scale (GOS) (p = 0.046) in multivariable regression. These findings provide insight into the temporal dynamics and interrelatedness of immune factors which collectively impact susceptibility to infection and greater hospital resource utilization, as well as influence recovery.
Collapse
Affiliation(s)
- Marina Levochkina
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
- Department of Infectious Diseases & Microbiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Leah McQuillan
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Nabil Awan
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - David Barton
- Department of Emergency Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - John Maczuzak
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Claudia Bianchine
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Shannon Trombley
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Emma Kotes
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Joshua Wiener
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Audrey Wagner
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Jason Calcagno
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Andrew Maza
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Ryan Nierstedt
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Stephanie Ferimer
- Division of Pediatric Rehabilitation Medicine, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA;
| | - Amy Wagner
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Correspondence:
| |
Collapse
|
27
|
Strinitz M, Pham M, März AG, Feick J, Weidner F, Vogt ML, Essig F, Neugebauer H, Stoll G, Schuhmann MK, Kollikowski AM. Immune Cells Invade the Collateral Circulation during Human Stroke: Prospective Replication and Extension. Int J Mol Sci 2021; 22:9161. [PMID: 34502070 PMCID: PMC8430889 DOI: 10.3390/ijms22179161] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/17/2021] [Accepted: 08/24/2021] [Indexed: 01/09/2023] Open
Abstract
It remains unclear if principal components of the local cerebral stroke immune response can be reliably and reproducibly observed in patients with acute large-vessel-occlusion (LVO) stroke. We prospectively studied a large independent cohort of n = 318 consecutive LVO stroke patients undergoing mechanical thrombectomy during which cerebral blood samples from within the occluded anterior circulation and systemic control samples from the ipsilateral cervical internal carotid artery were obtained. An extensive protocol was applied to homogenize the patient cohort and to standardize the procedural steps of endovascular sample collection, sample processing, and laboratory analyses. N = 58 patients met all inclusion criteria. (1) Mean total leukocyte counts were significantly higher within the occluded ischemic cerebral vasculature (I) vs. intraindividual systemic controls (S): +9.6%, I: 8114/µL ± 529 vs. S: 7406/µL ± 468, p = 0.0125. (2) This increase was driven by neutrophils: +12.1%, I: 7197/µL ± 510 vs. S: 6420/µL ± 438, p = 0.0022. Leukocyte influx was associated with (3) reduced retrograde collateral flow (R2 = 0.09696, p = 0.0373) and (4) greater infarct extent (R2 = 0.08382, p = 0.032). Despite LVO, leukocytes invade the occluded territory via retrograde collateral pathways early during ischemia, likely compromising cerebral hemodynamics and tissue integrity. This inflammatory response can be reliably observed in human stroke by harvesting immune cells from the occluded cerebral vascular compartment.
Collapse
Affiliation(s)
- Marc Strinitz
- Department of Neuroradiology, University Hospital of Würzburg, 97080 Würzburg, Germany; (M.S.); (M.P.); (A.G.M.); (J.F.); (F.W.); (M.L.V.)
| | - Mirko Pham
- Department of Neuroradiology, University Hospital of Würzburg, 97080 Würzburg, Germany; (M.S.); (M.P.); (A.G.M.); (J.F.); (F.W.); (M.L.V.)
| | - Alexander G. März
- Department of Neuroradiology, University Hospital of Würzburg, 97080 Würzburg, Germany; (M.S.); (M.P.); (A.G.M.); (J.F.); (F.W.); (M.L.V.)
| | - Jörn Feick
- Department of Neuroradiology, University Hospital of Würzburg, 97080 Würzburg, Germany; (M.S.); (M.P.); (A.G.M.); (J.F.); (F.W.); (M.L.V.)
| | - Franziska Weidner
- Department of Neuroradiology, University Hospital of Würzburg, 97080 Würzburg, Germany; (M.S.); (M.P.); (A.G.M.); (J.F.); (F.W.); (M.L.V.)
| | - Marius L. Vogt
- Department of Neuroradiology, University Hospital of Würzburg, 97080 Würzburg, Germany; (M.S.); (M.P.); (A.G.M.); (J.F.); (F.W.); (M.L.V.)
| | - Fabian Essig
- Department of Neurology, University Hospital of Würzburg, 97080 Würzburg, Germany; (F.E.); (H.N.); (G.S.); (M.K.S.)
| | - Hermann Neugebauer
- Department of Neurology, University Hospital of Würzburg, 97080 Würzburg, Germany; (F.E.); (H.N.); (G.S.); (M.K.S.)
| | - Guido Stoll
- Department of Neurology, University Hospital of Würzburg, 97080 Würzburg, Germany; (F.E.); (H.N.); (G.S.); (M.K.S.)
| | - Michael K. Schuhmann
- Department of Neurology, University Hospital of Würzburg, 97080 Würzburg, Germany; (F.E.); (H.N.); (G.S.); (M.K.S.)
| | - Alexander M. Kollikowski
- Department of Neuroradiology, University Hospital of Würzburg, 97080 Würzburg, Germany; (M.S.); (M.P.); (A.G.M.); (J.F.); (F.W.); (M.L.V.)
| |
Collapse
|
28
|
Wang H, Zhou J, Bi H, Yang X, Chen W, Jiang K, Yao Y, Ni W. Bioactive Ingredients from Nitraria tangutorun Bobr. Protect Against Cerebral Ischemia/Reperfusion Injury Through Attenuation of Oxidative Stress and the Inflammatory Response. J Med Food 2021; 24:686-696. [PMID: 34280030 DOI: 10.1089/jmf.2020.4848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Nitraria tangutorun Bobr. has been used for thousands of years as a native folk medicine to alleviate dizziness and neurasthenia due to oxygen. In our previous study, natural antioxidant components (namely, NJBE) were isolated from industrial N. tangutorun Bobr. juice byproducts (NJBE) from the Qinghai-Tibet plateau. The current investigation assessed the effects of NJBE on ischemic stroke in mice and the potential mechanisms. C57BL/6 mice received NJBE (25, 50, or 100 mg/Kg) by gavage for 14 days and then stroke was induced by the middle cerebral artery occlusion (MCAO) model, followed by reperfusion for 72 h. The evaluation of brain infarct size, behavioral tests, and functional assessments was conducted to assess the effects of NJBE after MCAO. Our results suggested that NJBE significantly decreases infarct size, improves neurological deficits, as well as reduces the number of GFAP+ and Iba-1+ cells after MCAO. NJBE inhibited nitric oxide and malondialdehyde production in the ischemic brain. Meanwhile, it attenuated the expressions of superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx). Also, NJBE significantly attenuated the expression levels of proinflammatory indicators, including TNF-α, IL-1β, IL-6, and IL-12. This process was accompanied by the downregulation of TLR4, TRAF6, pIκB/pIκB, and MMP9 expression and the upregulation of claudin-5 expression. NJBE induced improvements in brain injury. The neuroprotective effect of NJBE provides evidence for its potential application in stroke treatment.
Collapse
Affiliation(s)
- Hailiang Wang
- The Second Hospital of Jilin University, Changchun, China
| | - Jianhong Zhou
- College of Basic Medical Science of Jilin University, Changchun, China
| | - Hongtao Bi
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China
| | - Xiaoyu Yang
- College of Basic Medical Science of Jilin University, Changchun, China
| | - Wenlong Chen
- The Second Hospital of Jilin University, Changchun, China
| | - Kuijun Jiang
- The Second Hospital of Jilin University, Changchun, China
| | - Yang Yao
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Weihua Ni
- College of Basic Medical Science of Jilin University, Changchun, China
| |
Collapse
|
29
|
The Role of Neutrophil Extracellular Traps in Central Nervous System Diseases and Prospects for Clinical Application. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9931742. [PMID: 34336122 PMCID: PMC8294981 DOI: 10.1155/2021/9931742] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 07/01/2021] [Indexed: 12/13/2022]
Abstract
Neutrophil extracellular traps (NETs) are complexes of decondensed DNA fibers and antimicrobial peptides that are released by neutrophils and play important roles in many noninfectious diseases, such as cystic fibrosis, systemic lupus erythematosus, diabetes, and cancer. Recently, the formation of NETs has been detected in many central nervous system diseases and is thought to play different roles in the occurrence and development of these diseases. Researchers have detected NETs in acute ischemic stroke thrombi, and these NETs are thought to promote coagulation and thrombosis. NETs in ischemic brain parenchyma were identified as the cause of secondary nerve damage. High levels of NETs were also detected in grade IV glioma tissues, where NETs were involved in the proliferation and invasion of glioma cells by activating a signaling pathway. Extracellular web-like structures have also recently been observed in mice with traumatic brain injury (TBI), and it was hypothesized that NETs contribute to the development of edema after TBI. This article reviews the effect of NETs on multiple diseases that affect the CNS and explores their clinical application prospects.
Collapse
|
30
|
Cross-Talk of Atherosclerosis and Ischemic Stroke: Dramatic Role of Neutrophils. ARCHIVES OF NEUROSCIENCE 2021. [DOI: 10.5812/ans.104433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Context: Current investigations illustrate the increasing prevalence of atherosclerosis (AS) through the aggravating role of inappropriate lifestyle patterns. Atherosclerosis is the cause of important vascular-related diseases such as ischemic stroke (IS). Understanding AS pathophysiology can help reduce the incidence of AS-mediated diseases like ischemic stroke. Evidence Acquisition: For this narrative review article, we used the five mega databases of PubMed, Google Scholar, Scopus, Springer, and Science Direct. We searched from 2010 Jan to 2020 Dec and based on keywords and inclusion criteria, 77 articles were enrolled. Results: Based on prior articles on atherosclerosis and ischemic stroke pathophysiology, local and systemic inflammation is a vigorous factor in both diseasesIndeed, the fundamental inflammatory pathway involved atherosclerosis, and ischemic stroke is associated with the toll-like receptor 4/myeloid differentiation primary response 88/nuclear factor-kappa B (TLR4/ Myd88/ NF-κB) cascade. The functional paw of these intricate mechanisms are pro-inflammatory mediators, such as interleukin-1 beta (IL-1β), tumor necrosis factor (TNF-α), and interleukin-18 (IL-18) incite inflammation. Besides, the essential structures termed inflammasomes (multi proteins components), and multiplicity of immune and non-immune cells (i.e., neutrophils, monocytes, platelets, and macrophages) are beneficial in the induction of inflammatory microenvironment. Conclusions: Neutrophils could be the most effective cells in the inflammation-based mechanism in IS and AS. It is clarified that neutrophils with the recruitment of own vesicles and granules can afford to amplify inflammatory conditions and be a key cell in AS and IS cross-talk. Therefore, utilizing methods to control neutrophils-mediated mechanisms could be an effective method for the prevention of AS and IS.
Collapse
|
31
|
Four Decades of Ischemic Penumbra and Its Implication for Ischemic Stroke. Transl Stroke Res 2021; 12:937-945. [PMID: 34224106 DOI: 10.1007/s12975-021-00916-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 12/15/2022]
Abstract
The ischemic penumbra defined four decades ago has been the main battleground of ischemic stroke. The evolving ischemic penumbra concept has been providing insight for the development of vascular and cellular approaches as well as diagnostic tools for the treatment of ischemic stroke. rt-PA thrombolytic therapy to prevent the transition of ischemic penumbra to core has been approved for acute ischemic stroke within 3 h and was later recommended to extend to 4.5 h after symptom onset. Mechanical thrombectomy was introduced for the treatment of acute ischemic stroke with a therapeutic window of up to 24 h after stroke onset. Multiple modalities brain imaging techniques have been developed that provide guidance to define ischemic penumbra for reperfusion therapy in clinical practice. Cellular and molecular dissection of ischemic penumbra has been providing targets for the development of neuroprotective therapy for ischemic stroke. However, the dynamic nature of ischemic penumbra implicates that infarct core eventually expands into penumbra over time without reperfusion, dictating relative short therapeutic windows and limiting the impact of current reperfusion intervention. Entering the 5th decade since the introduction, ischemic penumbra remains the main focus of ischemic stroke research and clinical practice. In this review, we summarized the evolving ischemic penumbra concept and its implication in the development of vascular and cellular interventions as well as diagnostic tools for acute ischemic stroke. In addition, we discussed future perspectives on expansion of the campaign beyond ischemic penumbra to develop treatment for ischemic stroke.
Collapse
|
32
|
Liu Q, Sorooshyari SK. Quantitative and Correlational Analysis of Brain and Spleen Immune Cellular Responses Following Cerebral Ischemia. Front Immunol 2021; 12:617032. [PMID: 34194419 PMCID: PMC8238006 DOI: 10.3389/fimmu.2021.617032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 04/15/2021] [Indexed: 11/18/2022] Open
Abstract
Stroke is a multiphasic process, and the initial ischemic phase of neuronal damage is followed by secondary innate and adaptive responses that unfold over days after stroke, offer a longer time frame of intervention, and represent a novel therapeutic target. Therefore, revealing the distinct functions of immune cells in both brain and periphery is important for identification of immunotherapeutic targets for stroke to extend the treatment time window. In this paper an examination of the cellular dynamics of the immune response in the central nervous system (CNS) and periphery provoked by cerebral ischemia is provided. New data is presented for the number of immune cells in brain and spleen of mice during the 7 days following middle cerebral artery occlusion (MCAO). A novel analysis of the correlation among various cell types in the brain and spleen following stroke is presented. It is found that the infiltrated macrophages in the ischemic hemisphere positively correlate with neutrophils which implies their synergic effect in migrating into the brain after stroke onset. It is noted that during infiltration of adaptive immune cells, the number of neutrophils correlate positively with T cells, which suggests neutrophils contribute to T cell infiltration in the stroked brain. Furthermore, the correlation among neurological deficit and various immune cells suggests that microglia and splenic adaptive immune cells (T and B cells) are protective while infiltrating peripheral myeloid cells (macrophage and neutrophils) worsen stroke outcome. Comprehension of such immune responses post cerebral ischemia is crucial for differentiating the drivers of outcomes and also predicting the stroke outcome.
Collapse
Affiliation(s)
- Qingkun Liu
- Department of Neurology, School of Medicine, Stanford, CA, United States
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Siamak K. Sorooshyari
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, United States
| |
Collapse
|
33
|
Decoding the Transcriptional Response to Ischemic Stroke in Young and Aged Mouse Brain. Cell Rep 2021; 31:107777. [PMID: 32553170 DOI: 10.1016/j.celrep.2020.107777] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 03/25/2020] [Accepted: 05/26/2020] [Indexed: 12/17/2022] Open
Abstract
Ischemic stroke is a well-recognized disease of aging, yet it is unclear how the age-dependent vulnerability occurs and what are the underlying mechanisms. To address these issues, we perform a comprehensive RNA-seq analysis of aging, ischemic stroke, and their interaction in 3- and 18-month-old mice. We assess differential gene expression across injury status and age, estimate cell type proportion changes, assay the results against a range of transcriptional signatures from the literature, and perform unsupervised co-expression analysis, identifying modules of genes with varying response to injury. We uncover downregulation of axonal and synaptic maintenance genetic program, and increased activation of type I interferon (IFN-I) signaling following stroke in aged mice. Together, these results paint a picture of ischemic stroke as a complex age-related disease and provide insights into interaction of aging and stroke on cellular and molecular level.
Collapse
|
34
|
Jover-Mengual T, Hwang JY, Byun HR, Court-Vazquez BL, Centeno JM, Burguete MC, Zukin RS. The Role of NF-κB Triggered Inflammation in Cerebral Ischemia. Front Cell Neurosci 2021; 15:633610. [PMID: 34040505 PMCID: PMC8141555 DOI: 10.3389/fncel.2021.633610] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
Cerebral ischemia is a devastating disease that affects many people worldwide every year. The neurodegenerative damage as a consequence of oxygen and energy deprivation, to date, has no known effective treatment. The ischemic insult is followed by an inflammatory response that involves a complex interaction between inflammatory cells and molecules which play a role in the progression towards cell death. However, there is presently a matter of controversy over whether inflammation could either be involved in brain damage or be a necessary part of brain repair. The inflammatory response is triggered by inflammasomes, key multiprotein complexes that promote secretion of pro-inflammatory cytokines. An early event in post-ischemic brain tissue is the release of certain molecules and reactive oxygen species (ROS) from injured neurons which induce the expression of the nuclear factor-kappaB (NF-κB), a transcription factor involved in the activation of the inflammasome. There are conflicting observations related to the role of NF-κB. While some observe that NF-κB plays a damaging role, others suggest it to be neuroprotective in the context of cerebral ischemia, indicating the need for additional investigation. Here we discuss the dual role of the major inflammatory signaling pathways and provide a review of the latest research aiming to clarify the relationship between NF-κB mediated inflammation and neuronal death in cerebral ischemia.
Collapse
Affiliation(s)
- Teresa Jover-Mengual
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States.,Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe-Universidad de Valencia, Valencia, Spain.,Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - Jee-Yeon Hwang
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States.,Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, United States
| | - Hyae-Ran Byun
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| | - Brenda L Court-Vazquez
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| | - José M Centeno
- Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - María C Burguete
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe-Universidad de Valencia, Valencia, Spain.,Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - R Suzanne Zukin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
35
|
Delvasto-Nuñez L, Jongerius I, Zeerleder S. It takes two to thrombosis: Hemolysis and complement. Blood Rev 2021; 50:100834. [PMID: 33985796 DOI: 10.1016/j.blre.2021.100834] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 01/12/2023]
Abstract
Thromboembolic events represent the most common complication of hemolytic anemias characterized by complement-mediated hemolysis such as paroxysmal nocturnal hemoglobinuria and autoimmune hemolytic anemia. Similarly, atypical hemolytic uremic syndrome is characterized by hemolysis and thrombotic abnormalities. The main player in the development of thrombosis in hemolytic diseases is suggested to be the complement system. However, the release of extracellular hemoglobin and heme by hemolysis itself can also drive procoagulant responses. Both, complement activation and hemolysis promote the activation of neutrophils resulting in the formation of neutrophil extracellular traps and induce inflammation and vascular damage which all together might (synergistically) lead to hypercoagulability. In this review we aim to summarize the current knowledge on the role of complement activation and hemolysis in the onset of thrombosis in hemolytic diseases. This review will discuss the interplay between different biological systems and neutrophil activation contributing to the pathogenesis of thrombosis. Finally, we will combine this fundamental knowledge and address the pathophysiology of hemolysis in prototypical complement-driven diseases.
Collapse
Affiliation(s)
- Laura Delvasto-Nuñez
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Department of Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Ilse Jongerius
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Department of Pediatric Immunology, Amsterdam UMC, University of Amsterdam, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam, the Netherlands
| | - Sacha Zeerleder
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Switzerland; Department for BioMedical Research, University of Bern, Switzerland.
| |
Collapse
|
36
|
Chavda V, Madhwani K, Chaurasia B. Stroke and immunotherapy: Potential mechanisms and its implications as immune-therapeutics. Eur J Neurosci 2021; 54:4338-4357. [PMID: 33829590 DOI: 10.1111/ejn.15224] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 12/19/2022]
Abstract
Ischemia or brain injuries are mostly associated with emergency admissions and huge mortality rates. Stroke is a fatal cerebrovascular malady and second top root of disability and death in both developing and developed countries with a projected rise of 24.9% (from 2010) by 2030. It's the most frequent cause of morbidities and systemic permanent morbidities due to its multi-organ systemic pathology. Brain edema or active immune response cause disturbed or abnormal systemic affects causing inflammatory damage leading to secondary infection and secondary immune response which leads to activation like pneumonia or urine tract infections. There are a variety of post stroke treatments available which claims their usefulness in reducing or inhibiting post stroke and recurrent stroke damage followed by heavy inflammatory actions. Stroke does change the quality of life and also ensures daily chronic rapid neurodegeneration and cognitive decline. The only approved therapies for stroke are alteplase and thrombectomy which is associated with adverse outcomes and are not a total cure for ischemic stroke. Stroke and immune response are reciprocal to the pathology and time of event and it progresses till untreated. The immune reaction during ischemia opens new doors for advanced targeted therapeutics. Nowadays stem cell therapy has shown better results in stroke-prone individuals. Few monoclonal antibodies like natalizumab have shown great impact on pre-clinical and clinical stroke trial studies. In this current review, we have explored an immunology of stroke, current therapeutic scenario and future potential targets as immunotherapeutic agents in stroke therapeutics.
Collapse
Affiliation(s)
- Vishal Chavda
- Division of Anesthesia, Sardar Women's Hospital, Ahmadabad, Gujarat, India
| | - Kajal Madhwani
- Department of Microbiology, Nirma University, Ahmadabad, Gujarat, India
| | | |
Collapse
|
37
|
Ansari J, Gavins FNE. The impact of thrombo-inflammation on the cerebral microcirculation. Microcirculation 2021; 28:e12689. [PMID: 33638262 DOI: 10.1111/micc.12689] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/12/2021] [Accepted: 02/22/2021] [Indexed: 12/16/2022]
Abstract
The intertwined processes of thrombosis and inflammation (termed "thrombo-inflammation") are significant drivers of cerebrovascular diseases, and as such, they represent prime targets for drug discovery programs focusing on treatment and management of cerebrovascular diseases. Most cerebrovascular events result from chronic systemic microcirculatory dysfunction due to underlying conditions, for example, hypertension, diabetes mellitus, coronary artery disease, dyslipidemia, and sickle cell disease. Immune cells especially neutrophils play a critical role in the onset and maintenance of neuroinflammatory responses in the microcirculation. Neutrophils have the ability to drive both inflammatory and anti-inflammatory/pro-resolution effects depending on the underlying vascular state (physiological vs. pathological). In this article, we highlight the pathophysiological role of neutrophils in stroke and discuss ongoing pharmacotherapeutic strategies that are focused on identifying potential therapeutic targets for enhancing neuroprotection, mitigating inflammatory pathways, and enabling resolution.
Collapse
Affiliation(s)
- Junaid Ansari
- Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Felicity N E Gavins
- Department of Life Sciences, The Centre for Inflammation Research and Translational Medicine (CIRTM), Brunel University London, Uxbridge, Middlesex, UK
| |
Collapse
|
38
|
Cellular and Molecular Mechanisms of R/S-Roscovitine and CDKs Related Inhibition under Both Focal and Global Cerebral Ischemia: A Focus on Neurovascular Unit and Immune Cells. Cells 2021; 10:cells10010104. [PMID: 33429982 PMCID: PMC7827530 DOI: 10.3390/cells10010104] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/29/2020] [Accepted: 01/05/2021] [Indexed: 12/29/2022] Open
Abstract
Ischemic stroke is the second leading cause of death worldwide. Following ischemic stroke, Neurovascular Unit (NVU) inflammation and peripheral leucocytes infiltration are major contributors to the extension of brain lesions. For a long time restricted to neurons, the 10 past years have shown the emergence of an increasing number of studies focusing on the role of Cyclin-Dependent Kinases (CDKs) on the other cells of NVU, as well as on the leucocytes. The most widely used CDKs inhibitor, (R)-roscovitine, and its (S) isomer both decreased brain lesions in models of global and focal cerebral ischemia. We previously showed that (S)-roscovitine acted, at least, by modulating NVU response to ischemia. Interestingly, roscovitine was shown to decrease leucocytes-mediated inflammation in several inflammatory models. Specific inhibition of roscovitine majors target CDK 1, 2, 5, 7, and 9 showed that these CDKs played key roles in inflammatory processes of NVU cells and leucocytes after brain lesions, including ischemic stroke. The data summarized here support the investigation of roscovitine as a potential therapeutic agent for the treatment of ischemic stroke, and provide an overview of CDK 1, 2, 5, 7, and 9 functions in brain cells and leucocytes during cerebral ischemia.
Collapse
|
39
|
Li T, Chen Z, Zhu X, Tang X, Pan S, Gong F, Xu L, Wang M, Zhang H, Guo Y, Zhang J, Qin B, Zhang Z, Liu Y, Fei Z, Pan W, Yu X, Liu D. Neutrophil Count, Intracranial Atherosclerotic Stenosis, and Prognosis of Ischemic Stroke After Endovascular Treatment: A Mediation Analysis. Front Neurol 2021; 11:605852. [PMID: 33391168 PMCID: PMC7775673 DOI: 10.3389/fneur.2020.605852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 11/05/2020] [Indexed: 11/26/2022] Open
Abstract
Background and Purpose: Data on the relationship among neutrophil count, intracranial atherosclerotic stenosis (ICAS), and functional outcomes after endovascular thrombectomy (EVT) for ischemic stroke patients remains unclear. We aimed to evaluate the association between neutrophil count and prognosis of EVT patients and to determine whether the association was mediated by ICAS. Methods: We retrospectively analyzed consecutive patients who underwent EVT at two comprehensive stroke centers between June 2016 and December 2019. A remaining stenosis >70%, or a lesser degree of stenosis with a tendency toward re-occlusion or flow impairment during the procedure, was classified as ICAS. A poor outcome was defined as a 90-day modified Rankin Scale score of 3–6. Results: Of the 221 patients (mean age, 65.9 years; males, 61.1%) included in this study, 81 (36.3%) had ICAS, and 120 (54.3%) experienced a poor outcome at 90 days, respectively. In the multivariate adjustment for potential confounders, neutrophil count (odds ratio [OR], 1.19; 95% confidence interval [CI], 1.04–1.36; P = 0.012) and presence of ICAS (OR, 2.65; 95CI%, 1.28–5.45; P = 0.008) were risk factors of poor outcomes. Furthermore, mediation analysis indicated that total ICAS mediated the association between increased neutrophil count and worse functional outcome after EVT (the regression coefficient was changed by 11.7% for poor outcome, and 17.1% for modified Rankin Scale score, respectively). Conclusions: Our study demonstrated that a higher neutrophil count might increase the risk of a poor outcome among ischemic stroke patients who underwent EVT, which was partially mediated by ICAS.
Collapse
Affiliation(s)
- Tingting Li
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhonglun Chen
- Department of Neurology, MianYang Central Hospital, Mianyang, China
| | - Xuyin Zhu
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xianbiao Tang
- Department of Rehabilitation Medicine, Puer Hospital of Traditional Chinese Medicine, Pu'er, China
| | - Song Pan
- Medical Imaging Department of Shanxi Medical University, Taiyuan, China
| | - Fan Gong
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Leyi Xu
- Department of Neurosurgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingzhe Wang
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongzhi Zhang
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yongmei Guo
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingsi Zhang
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Baofeng Qin
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zongqi Zhang
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yun Liu
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhimin Fei
- Department of Neurosurgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weidong Pan
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaofei Yu
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dezhi Liu
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
40
|
Cheng W, Chen L, Yu H, Lu D, Yu R, Chen J. Value of Combining of the NLR and the Fibrinogen Level for Predicting Stroke-Associated Pneumonia. Neuropsychiatr Dis Treat 2021; 17:1697-1705. [PMID: 34093013 PMCID: PMC8169056 DOI: 10.2147/ndt.s311036] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 05/14/2021] [Indexed: 12/21/2022] Open
Abstract
PURPOSE To evaluate the value of the NLR (neutrophil-to-lymphocyte ratio) and the fibrinogen level in predicting stroke-associated pneumonia (SAP) in acute ischemic stroke (AIS) patients. PATIENTS AND METHODS In total, we enrolled 734 medical-ward patients with AIS in this retrospective study. Patients were divided into SAP (n=52) and non-SAP (n=682) groups according to the diagnostic criteria of SAP. Binary logistic regression analysis was used to analyze the relationship between the NLR, serum fibrinogen concentration and SAP. Receiver operating characteristic (ROC) curves were generated to identify the optimal cutoff points and assess the diagnostic value of the NLR, serum fibrinogen and the combination of NLR and fibrinogen in predicting SAP. RESULTS SAP occurred in 52 (7.08%) patients among the enrolled AIS patients. Binary logistic regression analysis showed that the NLR (odds ratio [OR]: 2.802, 95% confidence interval [CI]: 1.302-6.032, P=0.008) and serum fibrinogen concentration (OR: 7.850, 95% CI: 3.636-16.949, P=0.000) were independently associated with a higher risk of SAP incidence after adjusting for age, sex, ASPECT score, atrial fibrillation, nasogastric tube feeding, LDL-C and TC, temperature at admission and mechanical ventilation. The optimal cutoff points of the NLR and serum fibrinogen to distinguish SAP among AIS patients were 3.603 (AUC, 0.690; NPV, 95.78; PPV, 19.01) and 4.595 (AUC, 0.727; NPV, 95.60; PPV, 24.49), respectively. When the combination of NLR and fibrinogen was used to predict SAP, the optimal cutoff points were >2.436 for NLR and >3.24 for fibrinogen (AUC, 0.758; NPV, 98.50; PPV, 11.80). CONCLUSION The NLR and serum fibrinogen might have greater negative diagnostic value in predicting SAP among AIS patients. Combining the NLR and serum fibrinogen showed an increased AUC for predicting SAP among AIS patients.
Collapse
Affiliation(s)
- Wei Cheng
- Department of Respiratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Lichang Chen
- Department of Respiratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Huapeng Yu
- Department of Respiratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Dongzhu Lu
- Department of Respiratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Rong Yu
- Department of Respiratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Jian Chen
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
41
|
Deng J, Zhao F, Zhang Y, Zhou Y, Xu X, Zhang X, Zhao Y. Neutrophil extracellular traps increased by hyperglycemia exacerbate ischemic brain damage. Neurosci Lett 2020; 738:135383. [PMID: 32937190 DOI: 10.1016/j.neulet.2020.135383] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/29/2020] [Accepted: 09/09/2020] [Indexed: 12/13/2022]
Abstract
Hyperglycemia is common and associated with poor outcome in acute ischemic stroke patients. Neutrophil extracellular traps (NETs) are increased either in diabetes or ischemic stroke. We aimed to determine the role of NETs in acute ischemic stroke with hyperglycemia as it has not been clarified. NETs were observed in thrombi retrieved from ischemic stroke patients undergoing endovascular treatment. Citrullinated histone H3 was much more abundant in thrombi from hyperglycemic patients than in those from normoglycemic patients. BKS-db/db and wild-type mice injected with glucose were used to establish the permanent middle cerebral artery occlusion model with hyperglycemia. NETs induced by hyperglycemia were detected in the peri-ischemic brain tissue. Inhibition of NET formation decreased the infarction volume both in db/db and wild-type mice with hyperglycemia. Neurological function deficits were alleviated by blocking NET formation. These findings suggest that NETs increased by hyperglycemia play a role in exacerbation of ischemic brain damage.
Collapse
Affiliation(s)
- Jiangshan Deng
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Fei Zhao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yunlong Zhang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yajun Zhou
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Xiaofeng Xu
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Xiaojie Zhang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yuwu Zhao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| |
Collapse
|
42
|
Ischemic Stroke among the Symptoms Caused by the COVID-19 Infection. J Clin Med 2020; 9:jcm9092688. [PMID: 32825182 PMCID: PMC7565891 DOI: 10.3390/jcm9092688] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/11/2020] [Accepted: 08/17/2020] [Indexed: 12/22/2022] Open
Abstract
The 2019 global pandemic of coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been declared a public health emergency of international concern by the World Health Organization (WHO). The WHO recognized the spread of COVID-19 as a pandemic on 11 March 2020. Based on statistics from 10 August 2020, more than 20.2 million cases of COVID-19 have been reported resulting in more than 738,000 deaths. This completely new coronavirus has spread worldwide in a short period, causing economic crises and healthcare system failures worldwide. Initially, it was thought that the main health threat was associated with respiratory system failures, but since then, SARS-CoV-2 has been linked to a broad spectrum of symptoms indicating neurological manifestations, including ischemic stroke. Current knowledge about SARS-CoV-2 and its complications is very limited because of its rapidly evolving character. However, further research is undoubtedly necessary to understand the causes of neurological abnormalities, including acute cerebrovascular disease. The viral infection is inextricably associated with the activation of the immune system and the release of pro-inflammatory factors, that can stimulate the host organism to defend itself. However, the body’s immune response is a double-edged sword that on one hand, destroys the virus but also disrupts the homeostasis leading to serious complications, including thrombosis. Numerous studies have linked coagulopathies with COVID-19, however, there is great uncertainty regarding it functions on the molecular level. In this review, a detailed insight into the biological processes associated with ischemic stroke in COVID-19 patients and suggest a possible explanation for this phenomenon is provided.
Collapse
|
43
|
Sabouri E, Majdi A, Jangjui P, Rahigh Aghsan S, Naseri Alavi SA. Neutrophil-to-Lymphocyte Ratio and Traumatic Brain Injury: A Review Study. World Neurosurg 2020; 140:142-147. [DOI: 10.1016/j.wneu.2020.04.185] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 11/28/2022]
|
44
|
Abstract
Only a handful of published reports exist today that describe neurological complications following smoke inhalation injury. In this study, we characterize acute pathophysiological changes in the brain of sheep exposed to smoke inhalation, with- and without third-degree skin burn that models the injuries sustained by human victims of fire accidents. Blood-brain barrier integrity and hemorrhage were analyzed throughout the brain using specific histological stains: Hematoxylin & Eosin, Luxol fast blue, Periodic acid-Schiff (PAS), and Martius, Scarlet and Blue (MSB). Our data show that, following smoke inhalation injury, alone and in combination with third-degree skin burn, there was a significant increase in the number of congested and dilated blood vessels in the frontal cortex, basal ganglia, amygdala, hippocampus, pons, cerebellum, and pituitary gland as compared to sham-injured controls. Positive PAS staining confirmed damage to the basement membrane of congested and dilated blood vessels throughout the brain. Severe rupturing of blood vessels, microvascular hemorrhaging and bleeding throughout the brain was also observed in the injured groups. No significant changes in hemodynamics and PaO2 were observed. Our data demonstrate for the first time that acute smoke inhalation alone results in diffuse blood-brain barrier dysfunction and massive bleeding in the brain in the absence of hypoxia and changes in hemodynamics. These findings provide critical information and prompt further mechanistic and interventional studies necessary to develop effective and novel treatments aimed at alleviating CNS dysfunction in patients with smoke and burn injuries.
Collapse
|
45
|
Kollikowski AM, Schuhmann MK, Nieswandt B, Müllges W, Stoll G, Pham M. Local Leukocyte Invasion during Hyperacute Human Ischemic Stroke. Ann Neurol 2020; 87:466-479. [PMID: 31899551 DOI: 10.1002/ana.25665] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 12/27/2019] [Accepted: 12/30/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Bridging the gap between experimental stroke and patients by ischemic blood probing during the hyperacute stage of vascular occlusion is crucial to assess the role of inflammation in human stroke and for the development of adjunct treatments beyond recanalization. METHODS We prospectively observed 151 consecutive ischemic stroke patients with embolic large vessel occlusion of the anterior circulation who underwent mechanical thrombectomy. In all these patients, we attempted microcatheter aspiration of 3 different arterial blood samples: (1) within the core of the occluded vascular compartment and controlled by (2) carotid and (3) femoral samples obtained under physiological flow conditions. Subsequent laboratory analyses comprised leukocyte counting and differentiation, platelet counting, and the quantification of 13 proinflammatory human chemokines/cytokines. RESULTS Forty patients meeting all clinical, imaging, interventional, and laboratory inclusion criteria could be analyzed, showing that the total number of leukocytes significantly increased under the occlusion condition. This increase was predominantly driven by neutrophils. Significant increases were also apparent for lymphocytes and monocytes, accompanied by locally elevated plasma levels of the T-cell chemoattractant CXCL-11. Finally, we found evidence that short-term clinical outcome (National Institute of Health Stroke Scale at 72 hours) was negatively associated with neutrophil accumulation. INTERPRETATION We provide the first direct human evidence that neutrophils, lymphocytes, and monocytes, accompanied by specific chemokine upregulation, accumulate in the ischemic vasculature during hyperacute stroke and may affect outcome. These findings strongly support experimental evidence that immune cells contribute to acute ischemic brain damage and indicate that ischemic inflammation initiates already during vascular occlusion. Ann Neurol 2020;87:466-479.
Collapse
Affiliation(s)
| | | | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Wolfgang Müllges
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Guido Stoll
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Mirko Pham
- Department of Neuroradiology, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
46
|
Cirillo C, Brihmat N, Castel-Lacanal E, Le Friec A, Barbieux-Guillot M, Raposo N, Pariente J, Viguier A, Simonetta-Moreau M, Albucher JF, Olivot JM, Desmoulin F, Marque P, Chollet F, Loubinoux I. Post-stroke remodeling processes in animal models and humans. J Cereb Blood Flow Metab 2020; 40:3-22. [PMID: 31645178 PMCID: PMC6928555 DOI: 10.1177/0271678x19882788] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 08/28/2019] [Accepted: 09/05/2019] [Indexed: 01/05/2023]
Abstract
After cerebral ischemia, events like neural plasticity and tissue reorganization intervene in lesioned and non-lesioned areas of the brain. These processes are tightly related to functional improvement and successful rehabilitation in patients. Plastic remodeling in the brain is associated with limited spontaneous functional recovery in patients. Improvement depends on the initial deficit, size, nature and localization of the infarction, together with the sex and age of the patient, all of them affecting the favorable outcome of reorganization and repair of damaged areas. A better understanding of cerebral plasticity is pivotal to design effective therapeutic strategies. Experimental models and clinical studies have fueled the current understanding of the cellular and molecular processes responsible for plastic remodeling. In this review, we describe the known mechanisms, in patients and animal models, underlying cerebral reorganization and contributing to functional recovery after ischemic stroke. We also discuss the manipulations and therapies that can stimulate neural plasticity. We finally explore a new topic in the field of ischemic stroke pathophysiology, namely the brain-gut axis.
Collapse
Affiliation(s)
- Carla Cirillo
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Nabila Brihmat
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Evelyne Castel-Lacanal
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Alice Le Friec
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | | | - Nicolas Raposo
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Jérémie Pariente
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Alain Viguier
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Marion Simonetta-Moreau
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Jean-François Albucher
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Jean-Marc Olivot
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Franck Desmoulin
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Philippe Marque
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - François Chollet
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Isabelle Loubinoux
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| |
Collapse
|
47
|
The Brain Entangled: The Contribution of Neutrophil Extracellular Traps to the Diseases of the Central Nervous System. Cells 2019; 8:cells8121477. [PMID: 31766346 PMCID: PMC6953104 DOI: 10.3390/cells8121477] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/17/2019] [Accepted: 11/18/2019] [Indexed: 12/24/2022] Open
Abstract
Under normal conditions, neutrophils are restricted from trafficking into the brain parenchyma and cerebrospinal fluid by the presence of the brain–blood barrier (BBB). Yet, infiltration of the central nervous system (CNS) by neutrophils is a well-known phenomenon in the course of different pathological conditions, e.g., infection, trauma or neurodegeneration. Different studies have shown that neutrophil products, i.e., free oxygen radicals and proteolytic enzymes, play an important role in the pathogenesis of BBB damage. It was recently observed that accumulating granulocytes may release neutrophil extracellular traps (NETs), which damage the BBB and directly injure surrounding neurons. In this review, we discuss the emerging role of NETs in various pathological conditions affecting the CNS.
Collapse
|
48
|
Bieber M, Schuhmann MK, Volz J, Kumar GJ, Vaidya JR, Nieswandt B, Pham M, Stoll G, Kleinschnitz C, Kraft P. Description of a Novel Phosphodiesterase (PDE)-3 Inhibitor Protecting Mice From Ischemic Stroke Independent From Platelet Function. Stroke 2019; 50:478-486. [PMID: 30566040 PMCID: PMC6358218 DOI: 10.1161/strokeaha.118.023664] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Supplemental Digital Content is available in the text. Background and Purpose— Acetylsalicylic acid and clopidogrel are the 2 main antithrombotic drugs for secondary prevention in patients with ischemic stroke (IS) without indication for anticoagulation. Because of their limited efficacy and potential side effects, novel antiplatelet agents are urgently needed. Cilostazol, a specific phosphodiesterase (PDE)-3 inhibitor, protected from IS in clinical studies comprising mainly Asian populations. Nevertheless, the detailed mechanistic role of PDE-3 inhibitors in IS pathophysiology is hardly understood. In this project, we analyzed the efficacy and pathophysiologic mechanisms of a novel and only recently described PDE-3 inhibitor (substance V) in a mouse model of focal cerebral ischemia. Methods— Focal cerebral ischemia was induced by transient middle cerebral artery occlusion in 6- to 8-week-old male C57Bl/6 wild-type mice receiving substance V or vehicle 1 hour after ischemia induction. Infarct volumes and functional outcomes were assessed between day 1 and day 7, and findings were validated by magnetic resonance imaging. Blood-brain barrier damage, as well as the extent of local inflammatory response and cell death, was determined. Results— Inhibition of PDE-3 by pharmacological blockade with substance V significantly reduced infarct volumes and improved neurological outcome on day 1 and 7 after experimental cerebral ischemia. Reduced blood-brain barrier damage, attenuated brain tissue inflammation, and decreased local cell death could be identified as potential mechanisms. PDE-3 inhibitor treatment did neither increase the number of intracerebral hemorrhages nor affect platelet function. Conclusions— The novel PDE-3 inhibitor substance V protected mice from IS independent from platelet function. Pharmaceutical inactivation of PDE-3 might become a promising therapeutic approach to combat IS via inhibition of thromboinflammatory mechanisms and stabilization of the blood-brain barrier.
Collapse
Affiliation(s)
- Michael Bieber
- From the Department of Neurology (M.B., M.K.S., G.S., C.K., P.K.), University Hospital Würzburg, Germany
| | - Michael K Schuhmann
- From the Department of Neurology (M.B., M.K.S., G.S., C.K., P.K.), University Hospital Würzburg, Germany
| | - Julia Volz
- Rudolf Virchow Center, DFG Research Center for Experimental Biomedicine, University of Würzburg, Germany (J.V., B.N.)
| | - Gangasani Jagadeesh Kumar
- Fluro Agro Chemicals (Organic Chemistry II) Division (G.J.K., J.R.V.) and AcSIR (G.J.K., J.R.V.), CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India
| | - Jayathirtha Rao Vaidya
- Fluro Agro Chemicals (Organic Chemistry II) Division (G.J.K., J.R.V.) and AcSIR (G.J.K., J.R.V.), CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India
| | - Bernhard Nieswandt
- Rudolf Virchow Center, DFG Research Center for Experimental Biomedicine, University of Würzburg, Germany (J.V., B.N.)
| | - Mirko Pham
- Department of Neuroradiology (M.P.), University Hospital Würzburg, Germany
| | - Guido Stoll
- From the Department of Neurology (M.B., M.K.S., G.S., C.K., P.K.), University Hospital Würzburg, Germany
| | - Christoph Kleinschnitz
- From the Department of Neurology (M.B., M.K.S., G.S., C.K., P.K.), University Hospital Würzburg, Germany.,Department of Neurology, University Hospital Essen, Germany (C.K.)
| | - Peter Kraft
- From the Department of Neurology (M.B., M.K.S., G.S., C.K., P.K.), University Hospital Würzburg, Germany.,Department of Neurology, Klinikum Main-Spessart, Lohr, Germany (P.K.)
| |
Collapse
|
49
|
Yang CS, Guo A, Li Y, Shi K, Shi FD, Li M. Dl-3-n-butylphthalide Reduces Neurovascular Inflammation and Ischemic Brain Injury in Mice. Aging Dis 2019; 10:964-976. [PMID: 31595195 PMCID: PMC6764730 DOI: 10.14336/ad.2019.0608] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/08/2019] [Indexed: 12/13/2022] Open
Abstract
Dl-3-n-butylphthalide (NBP) is a synthetic compound that has been approved for the treatment of ischemic stroke in China. The mechanisms underlying the treatment efficacy of NBP have been reported in multiple studies and remain controversial. Here, we show that NBP treatment attenuated ischemic brain injury in mice subjected to transient middle cerebral artery occlusion or photothrombosis-induced permanent cerebral ischemia. NBP induced downregulation of intercellular adhesion molecule 1 and protease-activated receptor 1 in cerebrovascular endothelial cells after cerebral ischemia and reperfusion. This effect was associated with the reduced brain infiltration of myeloid cells and improved cerebral blood flow after reperfusion. The beneficial effects of NBP were diminished in mice subjected to the depletion of Gr1+ myeloid cells before brain ischemia. Therefore, the restriction of neurovascular inflammation is a key mode of action for NBP in ischemic stroke.
Collapse
Affiliation(s)
- Chun-Sheng Yang
- 1Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ai Guo
- 1Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yulin Li
- 1Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Kaibin Shi
- 2Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Fu-Dong Shi
- 1Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Minshu Li
- 1Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
50
|
Torres-Aguila NP, Carrera C, Muiño E, Cullell N, Cárcel-Márquez J, Gallego-Fabrega C, González-Sánchez J, Bustamante A, Delgado P, Ibañez L, Heitsch L, Krupinski J, Montaner J, Martí-Fàbregas J, Cruchaga C, Lee JM, Fernandez-Cadenas I. Clinical Variables and Genetic Risk Factors Associated with the Acute Outcome of Ischemic Stroke: A Systematic Review. J Stroke 2019; 21:276-289. [PMID: 31590472 PMCID: PMC6780022 DOI: 10.5853/jos.2019.01522] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 08/28/2019] [Indexed: 12/19/2022] Open
Abstract
Stroke is a complex disease and one of the main causes of morbidity and mortality among the adult population. A huge variety of factors is known to influence patient outcome, including demographic variables, comorbidities or genetics. In this review, we expound what is known about the influence of clinical variables and related genetic risk factors on ischemic stroke outcome, focusing on acute and subacute outcome (within 24 to 48 hours after stroke and until day 10, respectively), as they are the first indicators of stroke damage. We searched the PubMed data base for articles that investigated the interaction between clinical variables or genetic factors and acute or subacute stroke outcome. A total of 61 studies were finally included in this review. Regarding the data collected, the variables consistently associated with acute stroke outcome are: glucose levels, blood pressure, presence of atrial fibrillation, prior statin treatment, stroke severity, type of acute treatment performed, severe neurological complications, leukocyte levels, and genetic risk factors. Further research and international efforts are required in this field, which should include genome-wide association studies.
Collapse
Affiliation(s)
- Nuria P Torres-Aguila
- Stroke Pharmacogenomics and Genetics Laboratory, Sant Pau Research Institute, Barcelona, Spain.,Neurovascular Research Laboratory, Vall d'Hebron Research Institute (VHIR), Autonomous University of Barcelona, Barcelona, Spain
| | - Caty Carrera
- Stroke Pharmacogenomics and Genetics Laboratory, Sant Pau Research Institute, Barcelona, Spain.,Neurovascular Research Laboratory, Vall d'Hebron Research Institute (VHIR), Autonomous University of Barcelona, Barcelona, Spain
| | - Elena Muiño
- Stroke Pharmacogenomics and Genetics Laboratory, Sant Pau Research Institute, Barcelona, Spain
| | - Natalia Cullell
- Stroke Pharmacogenomics and Genetics Laboratory, Mutua Terrasa Foundation of Teaching and Research, Mutua Terrassa Hospital, Terrassa, Spain
| | - Jara Cárcel-Márquez
- Stroke Pharmacogenomics and Genetics Laboratory, Sant Pau Research Institute, Barcelona, Spain
| | - Cristina Gallego-Fabrega
- Stroke Pharmacogenomics and Genetics Laboratory, Sant Pau Research Institute, Barcelona, Spain.,Stroke Pharmacogenomics and Genetics Laboratory, Mutua Terrasa Foundation of Teaching and Research, Mutua Terrassa Hospital, Terrassa, Spain
| | - Jonathan González-Sánchez
- Stroke Pharmacogenomics and Genetics Laboratory, Sant Pau Research Institute, Barcelona, Spain.,Stroke Pharmacogenomics and Genetics Laboratory, Mutua Terrasa Foundation of Teaching and Research, Mutua Terrassa Hospital, Terrassa, Spain.,Health Care Science Department, The Manchester Metropolitan University of All Saints, Manchester, UK
| | - Alejandro Bustamante
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute (VHIR), Autonomous University of Barcelona, Barcelona, Spain
| | - Pilar Delgado
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute (VHIR), Autonomous University of Barcelona, Barcelona, Spain
| | - Laura Ibañez
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Laura Heitsch
- Division of Emergency Medicine, Washington University School of Medicine, St. Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jerzy Krupinski
- Stroke Pharmacogenomics and Genetics Laboratory, Mutua Terrasa Foundation of Teaching and Research, Mutua Terrassa Hospital, Terrassa, Spain.,Health Care Science Department, The Manchester Metropolitan University of All Saints, Manchester, UK
| | - Joan Montaner
- Department of Neurology, Virgin Rocío and Macarena Hospitals, Institute of Biomedicine of Seville (IBiS), Seville, Spain
| | - Joan Martí-Fàbregas
- Stroke Unit, Department of Neurology, Saint Cross and Saint Pau Hospital, Barcelona, Spain
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Jin-Moo Lee
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | | | -
- Stroke Pharmacogenomics and Genetics Laboratory, Sant Pau Research Institute, Barcelona, Spain
| |
Collapse
|