251
|
Abstract
The treatment of acute ischemic stroke continues to advance. The mainstay of treatment remains intravenous thrombolysis with alteplase. Recent studies demonstrated that later treatment with alteplase is beneficial in patients selected with advanced imaging techniques. Tenecteplase has been evaluated as an alternative thrombolytic drug and evidence suggests that it is as least as effective as alteplase and may lyse large vessel clots more effectively. Endovascular therapy with mechanical thrombectomy has now been shown to be beneficial up to 24 hours after stroke onset in carefully selected patients with proximal, large vessel occlusions. Ongoing studies are evaluating the effectiveness of thrombectomy in patients with more distal vessel occlusions and patients with proximal large vessel occlusions with larger ischemic core volumes and also in patients with milder neurological deficits. Cytoprotection is another potential acute stroke therapy that has not demonstrated efficacy in prior clinical trials. It should be reconsidered as an adjunct to reperfusion and a variety of new clinical trials can be envisioned to evaluate the potential benefits of cytoprotection in patients before and after reperfusion.
Collapse
Affiliation(s)
- Yunyun Xiong
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (Y.X.).,Chinese Institute of Brain Research (Y.X.)
| | - Ajay K Wakhloo
- Department of Neurointerventional Radiology Beth Israel Lahey Health Medical Center, Tufts University School of Medicine, Burlington' MA (A.K.W.)
| | - Marc Fisher
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School' Boston' MA (M.F.)
| |
Collapse
|
252
|
Emerging Roles for the Orphan GPCRs, GPR37 and GPR37 L1, in Stroke Pathophysiology. Int J Mol Sci 2022; 23:ijms23074028. [PMID: 35409385 PMCID: PMC9000135 DOI: 10.3390/ijms23074028] [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: 03/02/2022] [Revised: 03/31/2022] [Accepted: 04/02/2022] [Indexed: 11/17/2022] Open
Abstract
Recent studies have shed light on the diverse and complex roles of G-protein coupled receptors (GPCRs) in the pathophysiology of stroke. These receptors constitute a large family of seven transmembrane-spanning proteins that play an intricate role in cellular communication mechanisms which drive both tissue injury and repair following ischemic stroke. Orphan GPCRs represent a unique sub-class of GPCRs for which no natural ligands have been found. Interestingly, the majority of these receptors are expressed within the central nervous system where they represent a largely untapped resource for the treatment of neurological diseases. The focus of this review will thus be on the emerging roles of two brain-expressed orphan GPCRs, GPR37 and GPR37 L1, in regulating various cellular and molecular processes underlying ischemic stroke.
Collapse
|
253
|
Wei H, Peng Z, Chen Y, Guo J, Chen L, Shao K. cPKCγ ameliorates ischemic injury in cultured neurons exposed to oxygen glucose deprivation/reoxygenation by inhibiting ferroptosis. Neurosci Res 2022; 181:95-104. [DOI: 10.1016/j.neures.2022.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 03/17/2022] [Accepted: 04/06/2022] [Indexed: 12/27/2022]
|
254
|
Hetman M, Slomnicki L, Hodges E, Ohri SS, Whittemore SR. Role of circadian rhythms in pathogenesis of acute CNS injuries: Insights from experimental studies. Exp Neurol 2022; 353:114080. [DOI: 10.1016/j.expneurol.2022.114080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/28/2022] [Accepted: 04/05/2022] [Indexed: 11/16/2022]
|
255
|
Kim H, Kim MJ, Kwon YW, Jeon S, Lee S, Kim C, Choi BT, Shin Y, Hong SW, Shin HK. Benefits of a Skull-Interfaced Flexible and Implantable Multilight Emitting Diode Array for Photobiomodulation in Ischemic Stroke. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104629. [PMID: 35076161 PMCID: PMC9008794 DOI: 10.1002/advs.202104629] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/08/2022] [Indexed: 05/05/2023]
Abstract
Photobiomodulation (PBM) has received attention due to its potential for improving tissue function and enhancing regeneration in stroke. A lightweight, compact, and simple system of miniaturized electronic devices consisting of packaged light-emitting diodes (LEDs) that incorporates a flexible substrate for in vivo brain PBM in a mouse model is developed. Using this device platform, the preventive and therapeutic effects of PBM affixed to the exposed skull of mice in the photothrombosis and middle cerebral artery occlusion stroke model are evaluated. Among the wavelength range of 630, 850, and 940 nm LED array, the PBM with 630-nm LED array is proved to be the most effective for reducing the infarction volume and neurological impairment after ischemic stroke. Moreover, the PBM with 630 nm LED array remarkably improves the capability of spatial learning and memory in the chronic poststroke phase, attenuates AIM2 inflammasome activation and inflammasome-mediated pyroptosis, and modulates microglial polarization in the hippocampus and cortex 7 days following ischemic stroke. Thus, PBM may prevent tissue and functional damage in acute ischemic injury, thereby attenuating the development of cognitive impairment after stroke.
Collapse
Affiliation(s)
- Hyunha Kim
- Department of Korean Medical ScienceGraduate Training Program of Korean Medical Therapeutics for Healthy‐AgingSchool of Korean MedicinePusan National UniversityYangsan50612Republic of Korea
- Department of Pharmacology and NeuroscienceCreighton University School of MedicineOmahaNE68178USA
| | - Min Jae Kim
- Department of Korean Medical ScienceGraduate Training Program of Korean Medical Therapeutics for Healthy‐AgingSchool of Korean MedicinePusan National UniversityYangsan50612Republic of Korea
| | - Young Woo Kwon
- Department of Nano‐Fusion TechnologyCollege of Nanoscience & NanotechnologyPusan National UniversityBusan46241Republic of Korea
| | - Sangheon Jeon
- Department of Congo‐Mechatronics EngineeringDepartment of Optics and Mechatronics EngineeringCollege of Nanoscience & NanotechnologyPusan National UniversityBusan46241Republic of Korea
| | - Seo‐Yeon Lee
- Department of PharmacologyWonkwang University School of MedicineIksan54538Republic of Korea
| | - Chang‐Seok Kim
- Department of Congo‐Mechatronics EngineeringDepartment of Optics and Mechatronics EngineeringCollege of Nanoscience & NanotechnologyPusan National UniversityBusan46241Republic of Korea
| | - Byung Tae Choi
- Department of Korean Medical ScienceGraduate Training Program of Korean Medical Therapeutics for Healthy‐AgingSchool of Korean MedicinePusan National UniversityYangsan50612Republic of Korea
| | - Yong‐Il Shin
- Department of Rehabilitation MedicineSchool of MedicinePusan National UniversityYangsan50612Republic of Korea
| | - Suck Won Hong
- Department of Congo‐Mechatronics EngineeringDepartment of Optics and Mechatronics EngineeringCollege of Nanoscience & NanotechnologyPusan National UniversityBusan46241Republic of Korea
| | - Hwa Kyoung Shin
- Department of Korean Medical ScienceGraduate Training Program of Korean Medical Therapeutics for Healthy‐AgingSchool of Korean MedicinePusan National UniversityYangsan50612Republic of Korea
| |
Collapse
|
256
|
An J, Zhao L, Duan R, Sun K, Lu W, Yang J, Liang Y, Liu J, Zhang Z, Li L, Shi J. Potential nanotherapeutic strategies for perioperative stroke. CNS Neurosci Ther 2022; 28:510-520. [PMID: 35243774 PMCID: PMC8928924 DOI: 10.1111/cns.13819] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 01/24/2022] [Accepted: 02/04/2022] [Indexed: 12/12/2022] Open
Abstract
AIMS Based on the complex pathological environment of perioperative stroke, the development of targeted therapeutic strategies is important to control the development of perioperative stroke. DISCUSSIONS Recently, great progress has been made in nanotechnology, and nanodrug delivery systems have been developed for the treatment of ischemic stroke. CONCLUSION In this review, the pathological processes and mechanisms of ischemic stroke during perioperative stroke onset were systematically sorted. As a potential treatment strategy for perioperative stroke, the review also summarizes the multifunctional nanodelivery systems based on ischemic stroke, thus providing insight into the nanotherapeutic strategies for perioperative stroke.
Collapse
Affiliation(s)
- Jingyi An
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, China.,Key Laboratories of the Ministry of Education, Zhengzhou University, Zhengzhou, China
| | - Ling Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ranran Duan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ke Sun
- Department of Urinary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenxin Lu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jiali Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yan Liang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Junjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, China.,Key Laboratories of the Ministry of Education, Zhengzhou University, Zhengzhou, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, China.,Key Laboratories of the Ministry of Education, Zhengzhou University, Zhengzhou, China
| | - Li Li
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, China.,Key Laboratories of the Ministry of Education, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
257
|
Inflammatory Response and Secondary White Matter Damage to the Corpus Callosum after Focal Striatal Stroke in Rats. Int J Mol Sci 2022; 23:ijms23063179. [PMID: 35328600 PMCID: PMC8955860 DOI: 10.3390/ijms23063179] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/28/2022] [Accepted: 01/30/2022] [Indexed: 02/06/2023] Open
Abstract
Stroke is one of the leading causes of death and long-term disabilities worldwide, resulting in a debilitating condition occasioned by disturbances in the cerebral vasculature. Primary damage due to metabolic collapse is a quick outcome following stroke, but a multitude of secondary events, including excitotoxicity, inflammatory response, and oxidative stress cause further cell death and functional impairment. In the present work, we investigated whether a primary ischemic damage into the dorsal striatum may cause secondary damage in the circumjacent corpus callosum (CC). Animals were injected with endothelin-1 and perfused at 3, 7, 14, and 30 post-lesion days (PLD). Sections were stained with Cresyl violet for basic histopathology and immunolabeled by antibodies against astrocytes (anti-GFAP), macrophages/microglia (anti-IBA1/anti MHC-II), oligodendrocytes (anti-TAU) and myelin (anti-MBP), and Anti-Nogo. There were conspicuous microgliosis and astrocytosis in the CC, followed by later oligodendrocyte death and myelin impairment. Our results suggest that secondary white matter damage in the CC follows a primary focal striatal ischemia in adult rats.
Collapse
|
258
|
Astrocyte-secreted chordin-like 1 regulates spine density after ischemic injury. Sci Rep 2022; 12:4176. [PMID: 35264691 PMCID: PMC8907233 DOI: 10.1038/s41598-022-08031-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 03/01/2022] [Indexed: 11/09/2022] Open
Abstract
Ischemic injury occurs when the brain is deprived of blood flow, preventing cells from receiving essential nutrients. The injury core is the brain region directly deprived and is surrounded by the peri-infarct area, the region with recovery potential. In the peri-infarct area neurons undergo acute loss of dendritic spines, which modifies synaptic plasticity and determines neuronal survival. Astrocytes can be protective or detrimental to the ischemic injury response depending on the specific stage, yet we lack clear understanding of the underlying mechanisms. Chordin-like 1 (Chrdl1) is an astrocyte-secreted protein that promotes synaptic maturation and limits experience-dependent plasticity in the mouse visual cortex. Given this plasticity-limiting function we asked if Chrdl1 regulates the response to ischemic injury, modelled using photothrombosis (PT). We find that Chrdl1 mRNA is upregulated in astrocytes in the peri-infarct area in both acute and sub-acute phases post-PT. To determine the impact of increased Chrdl1 on the response to PT we analyzed Chrdl1 knock-out mice. We find that absence of Chrdl1 prevents ischemia-induced spine loss in the peri-infarct area and reduces cell death in the core, without impacting gliosis. These findings highlight the important role of astrocyte-secreted proteins in regulating structural plasticity in response to brain ischemic injuries.
Collapse
|
259
|
Bhattacharya R, Zekavat SM, Haessler J, Fornage M, Raffield L, Uddin MM, Bick AG, Niroula A, Yu B, Gibson C, Griffin G, Morrison AC, Psaty BM, Longstreth WT, Bis JC, Rich SS, Rotter JI, Tracy RP, Correa A, Seshadri S, Johnson A, Collins MPH JM, Hayden KM, Madsen TE, Ballantyne CM, Jaiswal S, Ebert BL, Kooperberg C, Manson JE, Whitsel EA, Natarajan P, Reiner AP. Clonal Hematopoiesis Is Associated With Higher Risk of Stroke. Stroke 2022; 53:788-797. [PMID: 34743536 PMCID: PMC8885769 DOI: 10.1161/strokeaha.121.037388] [Citation(s) in RCA: 98] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 10/25/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND PURPOSE Clonal hematopoiesis of indeterminate potential (CHIP) is a novel age-related risk factor for cardiovascular disease-related morbidity and mortality. The association of CHIP with risk of incident ischemic stroke was reported previously in an exploratory analysis including a small number of incident stroke cases without replication and lack of stroke subphenotyping. The purpose of this study was to discover whether CHIP is a risk factor for ischemic or hemorrhagic stroke. METHODS We utilized plasma genome sequence data of blood DNA to identify CHIP in 78 752 individuals from 8 prospective cohorts and biobanks. We then assessed the association of CHIP and commonly mutated individual CHIP driver genes (DNMT3A, TET2, and ASXL1) with any stroke, ischemic stroke, and hemorrhagic stroke. RESULTS CHIP was associated with an increased risk of total stroke (hazard ratio, 1.14 [95% CI, 1.03-1.27]; P=0.01) after adjustment for age, sex, and race. We observed associations with CHIP with risk of hemorrhagic stroke (hazard ratio, 1.24 [95% CI, 1.01-1.51]; P=0.04) and with small vessel ischemic stroke subtypes. In gene-specific association results, TET2 showed the strongest association with total stroke and ischemic stroke, whereas DMNT3A and TET2 were each associated with increased risk of hemorrhagic stroke. CONCLUSIONS CHIP is associated with an increased risk of stroke, particularly with hemorrhagic and small vessel ischemic stroke. Future studies clarifying the relationship between CHIP and subtypes of stroke are needed.
Collapse
Affiliation(s)
- Romit Bhattacharya
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Seyedeh M. Zekavat
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA
- Yale School of Medicine, New Haven, CT
| | - Jeffrey Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Laura Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Md Mesbah Uddin
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA
| | - Alexander G. Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Abhishek Niroula
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA
- Department of Laboratory Medicine, Lund University, Lund, Sweden
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Bing Yu
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Christopher Gibson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Gabriel Griffin
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
- Epigenomics Program, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Alanna C. Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Services, University of Washington, Seattle, WA
| | | | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA USA
| | - Russell P. Tracy
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington
| | - Adolfo Correa
- Jackson Heart Study, University of Mississippi Medical Center, Jackson, MS, USA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX 78229
- Boston University and the NHLBI’s Framingham Heart Study, Boston, MA, 02215, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Andrew Johnson
- Boston University and the NHLBI’s Framingham Heart Study, Boston, MA, 02215, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, Framingham, MA 01702
| | - Jason M. Collins MPH
- Department of Epidemiology, University of North Carolina, Gillings School of Global Public Health, Chapel Hill, NC
| | - Kathleen M. Hayden
- Department of Social Sciences and Health Policy, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Tracy E. Madsen
- Departments of Emergency Medicine and Epidemiology, Brown University
| | | | - Siddhartha Jaiswal
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Benjamin L. Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Howard Hughes Medical Institute, Boston, MA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - JoAnn E. Manson
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Eric A. Whitsel
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, Framingham, MA 01702
- Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC
| | | | - Pradeep Natarajan
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Alexander P. Reiner
- Department of Epidemiology, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
260
|
Takahashi S. Metabolic Contribution and Cerebral Blood Flow Regulation by Astrocytes in the Neurovascular Unit. Cells 2022; 11:cells11050813. [PMID: 35269435 PMCID: PMC8909328 DOI: 10.3390/cells11050813] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/17/2022] [Accepted: 02/24/2022] [Indexed: 12/10/2022] Open
Abstract
The neurovascular unit (NVU) is a conceptual framework that has been proposed to better explain the relationships between the neural cells and blood vessels in the human brain, focused mainly on the brain gray matter. The major components of the NVU are the neurons, astrocytes (astroglia), microvessels, pericytes, and microglia. In addition, we believe that oligodendrocytes should also be included as an indispensable component of the NVU in the white matter. Of all these components, astrocytes in particular have attracted the interest of researchers because of their unique anatomical location; these cells are interposed between the neurons and the microvessels of the brain. Their location suggests that astrocytes might regulate the cerebral blood flow (CBF) in response to neuronal activity, so as to ensure an adequate supply of glucose and oxygen to meet the metabolic demands of the neurons. In fact, the adult human brain, which accounts for only 2% of the entire body weight, consumes approximately 20–25% of the total amount of glucose and oxygen consumed by the whole body. The brain needs a continuous supply of these essential energy sources through the CBF, because there are practically no stores of glucose or oxygen in the brain; both acute and chronic cessation of CBF can adversely affect brain functions. In addition, another important putative function of the NVU is the elimination of heat and waste materials produced by neuronal activity. Recent evidence suggests that astrocytes play pivotal roles not only in supplying glucose, but also fatty acids and amino acids to neurons. Loss of astrocytic support can be expected to lead to malfunction of the NVU as a whole, which underlies numerous neurological disorders. In this review, we shall focus on historical and recent findings with regard to the metabolic contributions of astrocytes in the NVU.
Collapse
Affiliation(s)
- Shinichi Takahashi
- Department of Neurology and Stroke, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka-shi 350-1298, Japan; ; Tel.: +81-42-984-4111 (ext. 7412) or +81-3-3353-1211 (ext. 62613); Fax: +81-42-984-0664 or +81-3-3357-5445
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
261
|
Stroke induces disease-specific myeloid cells in the brain parenchyma and pia. Nat Commun 2022; 13:945. [PMID: 35177618 PMCID: PMC8854573 DOI: 10.1038/s41467-022-28593-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 01/18/2022] [Indexed: 11/09/2022] Open
Abstract
Inflammation triggers secondary brain damage after stroke. The meninges and other CNS border compartments serve as invasion sites for leukocyte influx into the brain thus promoting tissue damage after stroke. However, the post-ischemic immune response of border compartments compared to brain parenchyma remains poorly characterized. Here, we deeply characterize tissue-resident leukocytes in meninges and brain parenchyma and discover that leukocytes respond differently to stroke depending on their site of residence. We thereby discover a unique phenotype of myeloid cells exclusive to the brain after stroke. These stroke-associated myeloid cells partially resemble neurodegenerative disease-associated microglia. They are mainly of resident microglial origin, partially conserved in humans and exhibit a lipid-phagocytosing phenotype. Blocking markers specific for these cells partially ameliorates stroke outcome thus providing a potential therapeutic target. The injury-response of myeloid cells in the CNS is thus compartmentalized, adjusted to the type of injury and may represent a therapeutic target.
Collapse
|
262
|
Li TT, Wan Q, Zhang X, Xiao Y, Sun LY, Zhang YR, Liu XN, Yang WC. Stellate ganglion block reduces inflammation and improves neurological function in diabetic rats during ischemic stroke. Neural Regen Res 2022; 17:1991-1997. [PMID: 35142688 PMCID: PMC8848600 DOI: 10.4103/1673-5374.335162] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Diabetes mellitus is an independent risk factor for ischemic stroke. Both diabetes mellitus and stroke are linked to systemic inflammation that aggravates patient outcomes. Stellate ganglion block can effectively regulate the inflammatory response. Therefore, it is hypothesized that stellate ganglion block could be a potential therapy for ischemic stroke in diabetic subjects. In this study, we induced diabetes mellitus in rats by feeding them a high-fat diet for 4 successive weeks. The left middle cerebral artery was occluded to establish models of ischemic stroke in diabetic rats. Subsequently, we performed left stellate ganglion block with 1% lidocaine using the percutaneous posterior approach 15 minutes before reperfusion and again 20 and 44 hours after reperfusion. Our results showed that stellate ganglion block did not decrease the blood glucose level in diabetic rats with diabetes mellitus but did reduce the cerebral infarct volume and the cerebral water content. It also improved the recovery of neurological function, increased 28-day survival rate, inhibited Toll like receptor 4/nuclear factor kappa B signaling pathway and reduced inflammatory response in the plasma of rats. However, injection of Toll like receptor 4 agonist lipopolysaccharide 5 minutes before stellate ganglion block inhibited the effect of stellate ganglion block, whereas injection of Toll like receptor 4 inhibitor TAK242 had no such effect. We also found that stellate ganglion block performed at night had no positive effect on diabetic ischemic stroke. These findings suggest that stellate ganglion block is a potential therapy for diabetic ischemic stroke and that it may be mediated through the Toll like receptor 4/nuclear factor kappa B signaling pathway. We also found that the therapeutic effect of stellate ganglion block is affected by circadian rhythm.
Collapse
Affiliation(s)
- Ting-Ting Li
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Qiang Wan
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xin Zhang
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yuan Xiao
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Li-Ying Sun
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yu-Rong Zhang
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xiang-Nan Liu
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wan-Chao Yang
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
263
|
Tao W, Zhang X, Ding J, Yu S, Ge P, Han J, Luo X, Cui W, Chen J. The effect of propofol on hypoxia- and TNF-α-mediated BDNF/TrkB pathway dysregulation in primary rat hippocampal neurons. CNS Neurosci Ther 2022; 28:761-774. [PMID: 35112804 PMCID: PMC8981449 DOI: 10.1111/cns.13809] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/12/2022] [Accepted: 01/26/2022] [Indexed: 12/17/2022] Open
Abstract
AIMS Hypoxia and inflammation may lead to BDNF/TrkB dysregulation and neurological disorders. Propofol is an anesthetic with neuroprotective properties. We wondered whether and how propofol affected BDNF/TrkB pathway in hippocampal neurons and astrocytes. METHODS Primary rat hippocampal neurons and astrocytes were cultured and exposed to propofol followed by hypoxia or TNF-α treatment. The expression of BDNF and the expression/truncation/phosphorylation of TrkB were measured. The underlying mechanisms were investigated. RESULTS Hypoxia and TNF-α reduced the expression of BDNF, which was reversed by pretreatment of 25 μM propofol in hippocampal neurons. Furthermore, hypoxia and TNF-α increased the phosphorylation of ERK and phosphorylation of CREB at Ser142, while reduced the phosphorylation of CREB at Ser133, which were all reversed by 25 μM propofol and 10 μM ERK inhibitor. In addition, hypoxia or TNF-α did not affect TrkB expression, truncation, or phosphorylation in hippocampal neurons and astrocytes. However, in hippocampal neurons, 50 μM propofol induced TrkB phosphorylation, which may be mediated by p35 expression and Cdk5 activation, as suggested by the data showing that blockade of p35 or Cdk5 expression mitigated propofol-induced TrkB phosphorylation. CONCLUSIONS Propofol modulated BDNF/TrkB pathway in hippocampal neurons via ERK/CREB and p35/Cdk5 under the condition of hypoxia or TNF-α exposure.
Collapse
Affiliation(s)
- Weiping Tao
- Department of Anesthesiology, Jing'an District Central Hospital of Shanghai, Shanghai, China
| | - Xuesong Zhang
- Department of Anesthesiology, Shanghai Public Health Clinical Center, Shanghai, China
| | - Juan Ding
- Department of Anesthesiology, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shijian Yu
- Department of Anesthesiology, Jing'an District Central Hospital of Shanghai, Shanghai, China
| | - Peiqing Ge
- Department of Anesthesiology, Jing'an District Central Hospital of Shanghai, Shanghai, China
| | - Jingfeng Han
- Department of Anesthesiology, Jing'an District Central Hospital of Shanghai, Shanghai, China
| | - Xing Luo
- Department of Anesthesiology, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Cui
- Department of Anesthesiology, Jing'an District Central Hospital of Shanghai, Shanghai, China
| | - Jiawei Chen
- Department of Anesthesiology, Jing'an District Central Hospital of Shanghai, Shanghai, China
| |
Collapse
|
264
|
Exosomal microRNAs have great potential in the neurorestorative therapy for traumatic brain injury. Exp Neurol 2022; 352:114026. [DOI: 10.1016/j.expneurol.2022.114026] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/09/2022] [Accepted: 02/22/2022] [Indexed: 11/19/2022]
|
265
|
Biophoton imaging identification of delayed functional neural circuit injury after cerebral ischemia-reperfusion. J Neurosci Methods 2022; 367:109438. [PMID: 34896102 DOI: 10.1016/j.jneumeth.2021.109438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/30/2021] [Accepted: 12/06/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND The evaluation of structural changes after stroke has made great progress; however, it remains difficult to evaluate functional neural changes. NEW METHOD Here, we report a novel imaging technique that could monitor delayed functional neural circuit injury in an animal model of cerebral ischemia-reperfusion. The changes in 50 mM glutamate-induced biophotonic activities in functional neural circuits in rat brain slices after middle cerebral artery occlusion were investigated with an ultraweak biophoton imaging system. RESULTS Six hours after ischemia-reperfusion, the rats presented a significant decrease in motion ability together with a large part of the unstained 2,3,5-Triphenyltetrazolium chloride (TTC) area in the ischemia-reperfusion side, whereas the intensity of the biophoton emissions was consistent on both the ischemia-reperfusion and non-ischemic sides of brain slices. Twenty-four hours after reperfusion, the behavior evaluation and TTC staining recovered slightly, and the intensity of the biophoton emissions was weaker on the ischemia-reperfusion side than on the contralateral side. One week after reperfusion, the behavioral test and TTC staining recovered to normal levels; however, the intensity of the biophoton emissions was decreased significantly on both the ischemia-reperfusion and contralateral sides, and such changes were even distinguished in different brain areas, such as the sensory and motor coteries and striatum. CONCLUSION These findings suggest that delayed functional neural circuit injury induced by cerebral ischemia-reperfusion could be identified with biophoton imaging techniques, providing a novel functional evaluation method for animal models of cerebral ischemia-reperfusion.
Collapse
|
266
|
Yang Y, Li H, Yang C, Kong X, Wu X, Gong M, Li Y. The potent inhibitory role of suppressing TBK1 in RIPK1 associated cerebral ischemia-reperfusion injury. Brain Res 2022; 1781:147813. [PMID: 35120903 DOI: 10.1016/j.brainres.2022.147813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/19/2022] [Accepted: 01/28/2022] [Indexed: 12/12/2022]
Abstract
The pathological mechanism of cell death features in cerebral ischemia-reperfusion injury (CIRI) was complicated. The occurrence of various cell death pathways during the progression of ischemia/reperfusion injury promoted complex further neuroinflammation. RIPK1, receptor interacting protein kinase 1, was convinced to be involved in both necroptosis and apoptosis, which is a special RIPK1-dependent apoptosis. More evidences indicated the physiological role of RIPK1 in necroptosis, apoptosis and also autophagy. In this study, we elucidated the RIPK1 exhibited characterization in various cell death pathways in time-course dependent feature. The necroptosis occupied dominant neuron death within 24 hours after ischemia/reperfusion injury. However, the neuronal death feature seemed turned to apoptosis 24 hours after reperfusion. In this study, it was also found that TBK1 (TANK binding kinase 1) played as suppressor in the regulation of kinase activity of RIPK1. This result might provide a potential approach in mediating the kinase activity of RIPK1 in clinic.
Collapse
Affiliation(s)
- Yuting Yang
- Department of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Huijie Li
- Department of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Chaonan Yang
- Department of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xiaodong Kong
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, China
| | - Xiaohui Wu
- Department of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Min Gong
- Department of Pharmacy, Tianjin Medical University, Tianjin, China; Tianjin Neurological Institute, Tianjin Medical University General Hospital, China.
| | - Ying Li
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, China.
| |
Collapse
|
267
|
GSK-126 Protects CA1 Neurons from H3K27me3-Mediated Apoptosis in Cerebral Ischemia. Mol Neurobiol 2022; 59:2552-2562. [PMID: 35091962 PMCID: PMC9016005 DOI: 10.1007/s12035-021-02677-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/30/2021] [Indexed: 12/16/2022]
Abstract
Epigenetics, including histone modifications, play a significant role in central nervous system diseases, but the underlying mechanism remains to be elucidated. The aim of this study was to evaluate the role of H3K27me3 in regulating transcriptomic and pathogenic mechanisms following global ischemic stroke. Here, we found that in vivo ischemic/reperfusion (I/R) injury induced marked upregulation of H3K27me3 in the hippocampus. The administration of GSK-126 to rat brains decreased the levels of H3K27me3 in the hippocampus and reduced neuronal apoptosis after experimental stroke. Furthermore, ChIP-seq data demonstrated that the primary role of GSK-126 in the ischemic brain is to reduce H3K27me3 enrichment, mediating negative regulation of the execution phase of apoptosis and the MAPK signaling pathway. Further study suggested that the protective role of GSK-126 in ischemic rats was antagonized by U0126, an inhibitor of ERK1/2. Collectively, we demonstrated the potential of H3K27me3 as a novel stroke therapeutic target, and GSK-126 exerted a neuroprotective function in ischemic brain injury, which might be associated with activation of the MAPK/ERK pathway.
Collapse
|
268
|
The Quinazoline Otaplimastat (SP-8203) Reduces the Hemorrhagic Transformation and Mortality Aggravated after Delayed rtPA-Induced Thrombolysis in Cerebral Ischemia. Int J Mol Sci 2022; 23:ijms23031403. [PMID: 35163322 PMCID: PMC8835804 DOI: 10.3390/ijms23031403] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/24/2022] [Indexed: 02/01/2023] Open
Abstract
Acute ischemic stroke is the leading cause of morbidity and mortality worldwide. Recombinant tissue plasminogen activator (rtPA) is the only agent clinically approved by FDA for patients with acute ischemic stroke. However, delayed treatment of rtPA (e.g., more than 3 h after stroke onset) exacerbates ischemic brain damage by causing intracerebral hemorrhage and increasing neurotoxicity. In the present study, we investigated whether the neuroprotant otaplimastat reduced delayed rtPA treatment-evoked neurotoxicity in male Sprague Dawley rats subjected to embolic middle cerebral artery occlusion (eMCAO). Otaplimastat reduced cerebral infarct size and edema and improved neurobehavioral deficits. In particular, otaplimastat markedly reduced intracerebral hemorrhagic transformation and mortality triggered by delayed rtPA treatment, consequently extending the therapeutic time window of rtPA. We further found that ischemia-evoked extracellular matrix metalloproteases (MMPs) expression was closely correlated with cerebral hemorrhagic transformation and brain damage. In ischemic conditions, delayed rtPA treatment further increased brain injury via synergistic expression of MMPs in vascular endothelial cells. In oxygen-glucose-deprived endothelial cells, otaplimastat suppressed the activity rather than protein expression of MMPs by restoring the level of tissue inhibitor of metalloproteinase (TIMP) suppressed in ischemia, and consequently reduced vascular permeation. This paper shows that otaplimastat under clinical trials is a new drug which can inhibit stroke on its own and extend the therapeutic time window of rtPA, especially when administered in combination with rtPA.
Collapse
|
269
|
Pharmacological brain cytoprotection in acute ischaemic stroke — renewed hope in the reperfusion era. Nat Rev Neurol 2022; 18:193-202. [PMID: 35079135 PMCID: PMC8788909 DOI: 10.1038/s41582-021-00605-6] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2021] [Indexed: 11/08/2022]
Abstract
For over 40 years, attempts to develop treatments that protect neurons and other brain cells against the cellular and biochemical consequences of cerebral ischaemia in acute ischaemic stroke (AIS) have been unsuccessful. However, the advent of intravenous thrombolysis and endovascular thrombectomy has taken us into a new era of treatment for AIS in which highly effective reperfusion therapy is widely available. In this context, cytoprotective treatments should be revisited as adjunctive treatment to reperfusion therapy. Renewed efforts should focus on developing new drugs that target multiple aspects of the ischaemic cascade, and previously developed drugs should be reconsidered if they produced robust cytoprotective effects in preclinical models and their safety profiles were reasonable in previous clinical trials. Several development pathways for cytoprotection as an adjunct to reperfusion can be envisioned. In this Review, we outline the targets for cytoprotective therapy and discuss considerations for future drug development, highlighting the recent ESCAPE-NA1 trial of nerinetide, which produced the most promising results to date. We review new types of clinical trial to evaluate whether cytoprotective drugs can slow infarct growth prior to reperfusion and/or ameliorate the consequences of reperfusion, such as haemorrhagic transformation. We also highlight how advanced brain imaging can help to identify patients with salvageable ischaemic tissue who are likely to benefit from cytoprotective therapy. In this Review, Fisher and Savitz consider how the era of reperfusion therapy in ischaemic stroke provides new hope for the development of cytoprotective therapies to further improve outcomes, highlighting how promising recent findings can be built on to benefit patients. Highly successful reperfusion therapy with intravenous thrombolysis and endovascular thrombectomy is now widely available for the treatment of acute ischaemic stroke, making cytoprotective therapy a viable additional treatment approach. Previous attempts to develop cytoprotective therapy have been unsuccessful, but this approach should now be reconsidered as an adjunctive therapy to thrombolysis and thrombectomy. New cytoprotective drugs should be developed to target multiple aspects of the ischaemic cascade, and previously developed drugs should be reconsidered. Trials should be conducted to evaluate the effects of cytoprotective drugs when administered before or after reperfusion therapy or both. Advanced brain imaging should be used to select patients who are most likely to benefit from cytoprotective treatment for enrolment in new trials.
Collapse
|
270
|
Vespakinin-M, a natural peptide from Vespa magnifica, promotes functional recovery in stroke mice. Commun Biol 2022; 5:74. [PMID: 35058552 PMCID: PMC8776894 DOI: 10.1038/s42003-022-03024-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 12/28/2021] [Indexed: 02/06/2023] Open
Abstract
Acute ischemic stroke triggers complex systemic pathological responses for which the exploration of drug resources remains a challenge. Wasp venom extracted from Vespa magnifica (Smith, 1852) is most commonly used to treat rheumatoid arthritis as well as neurological disorders. Vespakinin-M (VK), a natural peptide from wasp venom, has remained largely unexplored for stroke. Herein, we first confirmed the structure, stability, toxicity and distribution of VK as well as its penetration into the blood–brain barrier. VK (150 and 300 µg/kg, i.p.) was administered to improve stroke constructed by middle cerebral artery occlusion in mice. Our results indicate that VK promote functional recovery in mice after ischemia stroke, including an improvement of neurological impairment, reduction of infarct volume, maintenance of blood-brain barrier integrity, and an obstruction of the inflammatory response and oxidative stress. In addition, VK treatment led to reduced neuroinflammation and apoptosis associated with the activation of PI3K–AKT and inhibition of IκBα–NF-κB signaling pathways. Simultaneously, we confirmed that VK can combine with bradykinin receptor 2 (B2R) as detected by molecular docking, the B2R antagonist HOE140 could counteract the neuro-protective effects of VK on stroke in mice. Overall, targeting the VK–B2R interaction can be considered as a practical strategy for stroke therapy. Zhao et al establish the structure, stability, toxicity and distribution of vespakinin-M (VK) as well as its penetration into the blood–brain barrier in mice. They go on to show that VK promotes functional recovery in mice after ischemia stroke and shed light on the potential underlying mechanisms, which together indicates the potential therapeutic value of targeting VK in stroke therapy.
Collapse
|
271
|
Endurance Training and Exogenous Adenosine Infusion Mitigate Hippocampal Inflammation and Cell Death in a Rat Model of Cerebral Ischemia/Reperfusion Injury. ARCHIVES OF NEUROSCIENCE 2022. [DOI: 10.5812/ans.119236] [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
Background: Cerebral ischemia can cause irreversible structural and functional damages to the brain, especially to the hippocampus. Preconditioning with endurance training and endogenous adenosine infusion may reduce ischemia-associated damages. Objectives: This study aimed to evaluate the effect of preconditioning with endurance training and endogenous adenosine infusion on cell death in the hippocampal CA1 region following ischemia/reperfusion injuries in a rat model. Methods: Male Wistar rats were divided into five groups: (1) control (n = 8); (2) ischemia (n = 12); (3) endurance training + ischemia (n = 12); (4) adenosine infusion + ischemia (n = 12); and (5) endurance training + adenosine infusion + ischemia (n = 12). The rats in the training groups ran on a treadmill five days per week for eight weeks. In the adenosine infusion groups, the rats were injected 0.1 mg/mL/kg of adenosine intraperitoneally. Also, in the ischemic groups, both common carotid arteries were clamped for 45 minutes. Cresyl violet staining and real-time polymerase chain reaction (PCR) assay were used to evaluate cell death and cytokine gene expression, respectively. Results: Based on the present results, treatments, including endurance training + ischemia, adenosine infusion + ischemia, and endurance training + adenosine infusion + ischemia reduced the level of interleukin-6 (IL-6) and glutamate gene expression, respectively, compared to the group of ischemia only. In contrast, the expression of nerve growth factor (NGF) and adenosine receptor (A2A) genes increased by seven, four, and two folds in the endurance training + ischemia, adenosine infusion + ischemia, and endurance training + adenosine infusion + ischemia groups, respectively, compared to the group of ischemia only. Conclusions: Endurance training on a treadmill and exogenous adenosine infusion synergistically diminished cell death and reduced the expression of pro-inflammatory cytokines, while promoting the neurotrophic factor expression. When endurance training and adenosine infusion were used as stimulants before the induction of cerebral ischemia, they significantly reduced cell death.
Collapse
|
272
|
Takahashi S, Mashima K. Neuroprotection and Disease Modification by Astrocytes and Microglia in Parkinson Disease. Antioxidants (Basel) 2022; 11:antiox11010170. [PMID: 35052674 PMCID: PMC8773262 DOI: 10.3390/antiox11010170] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/03/2022] [Accepted: 01/13/2022] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress and neuroinflammation are common bases for disease onset and progression in many neurodegenerative diseases. In Parkinson disease, which is characterized by the degeneration of dopaminergic neurons resulting in dopamine depletion, the pathogenesis differs between hereditary and solitary disease forms and is often unclear. In addition to the pathogenicity of alpha-synuclein as a pathological disease marker, the involvement of dopamine itself and its interactions with glial cells (astrocyte or microglia) have attracted attention. Pacemaking activity, which is a hallmark of dopaminergic neurons, is essential for the homeostatic maintenance of adequate dopamine concentrations in the synaptic cleft, but it imposes a burden on mitochondrial oxidative glucose metabolism, leading to reactive oxygen species production. Astrocytes provide endogenous neuroprotection to the brain by producing and releasing antioxidants in response to oxidative stress. Additionally, the protective function of astrocytes can be modified by microglia. Some types of microglia themselves are thought to exacerbate Parkinson disease by releasing pro-inflammatory factors (M1 microglia). Although these inflammatory microglia may further trigger the inflammatory conversion of astrocytes, microglia may induce astrocytic neuroprotective effects (A2 astrocytes) simultaneously. Interestingly, both astrocytes and microglia express dopamine receptors, which are upregulated in the presence of neuroinflammation. The anti-inflammatory effects of dopamine receptor stimulation are also attracting attention because the functions of astrocytes and microglia are greatly affected by both dopamine depletion and therapeutic dopamine replacement in Parkinson disease. In this review article, we will focus on the antioxidative and anti-inflammatory effects of astrocytes and their synergism with microglia and dopamine.
Collapse
Affiliation(s)
- Shinichi Takahashi
- Department of Neurology and Stroke, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka-shi 350-1298, Japan
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan;
- Correspondence: ; Tel.: +81-42-984-4111 (ext. 7412); Fax: +81-42-984-0664
| | - Kyoko Mashima
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan;
- Department of Neurology, Tokyo Saiseikai Central Hospital, 1-4-17 Mita, Minato-ku, Tokyo 108-0073, Japan
| |
Collapse
|
273
|
Post-stroke treatment with argon preserved neurons and attenuated microglia/macrophage activation long-termly in a rat model of transient middle cerebral artery occlusion (tMCAO). Sci Rep 2022; 12:691. [PMID: 35027642 PMCID: PMC8758662 DOI: 10.1038/s41598-021-04666-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 12/24/2021] [Indexed: 11/18/2022] Open
Abstract
In a previous study from our group, argon has shown to significantly attenuate brain injury, reduce brain inflammation and enhance M2 microglia/macrophage polarization until 7 days after ischemic stroke. However, the long-term effects of argon have not been reported thus far. In the present study, we analyzed the underlying neuroprotective effects and potential mechanisms of argon, up to 30 days after ischemic stroke. Argon administration with a 3 h delay after stroke onset and 1 h after reperfusion demonstrated long-term neuroprotective effect by preserving the neurons at the ischemic boundary zone 30 days after stroke. Furthermore, the excessive microglia/macrophage activation in rat brain was reduced by argon treatment 30 days after ischemic insult. However, long-lasting neurological improvement was not detectable. More sensorimotor functional measures, age- and disease-related models, as well as further histological and molecular biological analyses will be needed to extend the understanding of argon’s neuroprotective effects and mechanism of action after ischemic stroke.
Collapse
|
274
|
Poh L, Sim WL, Jo DG, Dinh QN, Drummond GR, Sobey CG, Chen CLH, Lai MKP, Fann DY, Arumugam TV. The role of inflammasomes in vascular cognitive impairment. Mol Neurodegener 2022; 17:4. [PMID: 35000611 PMCID: PMC8744307 DOI: 10.1186/s13024-021-00506-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 12/02/2021] [Indexed: 12/11/2022] Open
Abstract
There is an increasing prevalence of Vascular Cognitive Impairment (VCI) worldwide, and several studies have suggested that Chronic Cerebral Hypoperfusion (CCH) plays a critical role in disease onset and progression. However, there is a limited understanding of the underlying pathophysiology of VCI, especially in relation to CCH. Neuroinflammation is a significant contributor in the progression of VCI as increased systemic levels of the proinflammatory cytokine interleukin-1β (IL-1β) has been extensively reported in VCI patients. Recently it has been established that CCH can activate the inflammasome signaling pathways, involving NLRP3 and AIM2 inflammasomes that critically regulate IL-1β production. Given that neuroinflammation is an early event in VCI, it is important that we understand its molecular and cellular mechanisms to enable development of disease-modifying treatments to reduce the structural brain damage and cognitive deficits that are observed clinically in the elderly. Hence, this review aims to provide a comprehensive insight into the molecular and cellular mechanisms involved in the pathogenesis of CCH-induced inflammasome signaling in VCI.
Collapse
Affiliation(s)
- Luting Poh
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei Liang Sim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Quynh Nhu Dinh
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| | - Grant R. Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| | - Christopher G. Sobey
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| | - Christopher Li-Hsian Chen
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mitchell K. P. Lai
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Y. Fann
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Healthy Longevity, National University Health System (NUHS), Singapore, Singapore
| | - Thiruma V. Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| |
Collapse
|
275
|
Parvez S, Kaushik M, Ali M, Alam MM, Ali J, Tabassum H, Kaushik P. Dodging blood brain barrier with "nano" warriors: Novel strategy against ischemic stroke. Theranostics 2022; 12:689-719. [PMID: 34976208 PMCID: PMC8692911 DOI: 10.7150/thno.64806] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 10/14/2021] [Indexed: 12/13/2022] Open
Abstract
Ischemic stroke (IS) is one of the leading causes of death and disability resulting in inevitable burden globally. Ischemic injury initiates cascade of pathological events comprising energy dwindling, failure of ionic gradients, failure of blood brain barrier (BBB), vasogenic edema, calcium over accumulation, excitotoxicity, increased oxidative stress, mitochondrial dysfunction, inflammation and eventually cell death. In spite of such complexity of the disease, the only treatment approved by US Food and Drug Administration (FDA) is tissue plasminogen activator (t-PA). This therapy overcome blood deficiency in the brain along with side effects of reperfusion which are responsible for considerable tissue injury. Therefore, there is urgent need of novel therapeutic perspectives that can protect the integrity of BBB and salvageable brain tissue. Advancement in nanomedicine is empowering new approaches that are potent to improve the understanding and treatment of the IS. Herein, we focus nanomaterial mediated drug delivery systems (DDSs) and their role to bypass and cross BBB especially via intranasal drug delivery. The various nanocarriers used in DDSs are also discussed. In a nut shell, the objective is to provide an overview of use of nanomedicine in the diagnosis and treatment of IS to facilitate the research from benchtop to bedside.
Collapse
|
276
|
Ovcjak A, Xiao A, Kim JS, Xu B, Szeto V, Turlova E, Abussaud A, Chen NH, Miller SP, Sun HS, Feng ZP. Ryanodine receptor inhibitor dantrolene reduces hypoxic-ischemic brain injury in neonatal mice. Exp Neurol 2022; 351:113985. [DOI: 10.1016/j.expneurol.2022.113985] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 01/07/2022] [Accepted: 01/13/2022] [Indexed: 11/04/2022]
|
277
|
Hamblin MH, Murad R, Yin J, Vallim G, Lee JP. Modulation of gene expression on a transcriptome-wide level following human neural stem cell transplantation in aged mouse stroke brains. Exp Neurol 2022; 347:113913. [PMID: 34752785 PMCID: PMC8647207 DOI: 10.1016/j.expneurol.2021.113913] [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: 10/12/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Neural stem cell (NSC) transplantation offers great potential for treating ischemic stroke. Clinically, ischemia followed by reperfusion results in robust cerebrovascular injury that upregulates proinflammatory factors, disrupts neurovascular units, and causes brain cell death. NSCs possess multiple actions that can be exploited for reducing the severity of neurovascular injury. Our previous studies in young adult mice showed that human NSC transplantation during the subacute stage diminishes stroke pathophysiology and improves behavioral outcome. METHODS We employed a well-established and commonly used stroke model, middle cerebral artery occlusion with subsequent reperfusion (MCAO/R). Here, we assessed the outcomes of hNSC transplantation 48 h post-MCAO (24 h post-transplant) in aged mouse brains in response to stroke because aging is a crucial risk factor for cerebral ischemia. Next, we tested whether administration of the integrin α5β1 inhibitor, ATN-161, prior to hNSC transplantation further affects stoke outcome as compared with NSCs alone. RNA sequencing (RNA-seq) was used to assess the impact of hNSC transplantation on differentially expressed genes (DEGs) on a transcriptome-wide level. RESULTS Here, we report that hNSC-engrafted brains with or without ATN-161 showed significantly reduced infarct size, and attenuated the induction of proinflammatory factors and matrix metalloproteases. RNA-seq analysis revealed DEGs and molecular pathways by which hNSCs induce a beneficial post-stroke outcome in aged stroke brains. 811 genes were differentially expressed (651 downregulated and 160 upregulated) in hNSC-engrafted stroke brains. Functional pathway analysis identified enriched and depleted pathways in hNSC-engrafted aged mouse stroke brains. Depletion of pathways following hNSC-engraftment included signaling involving neuroinflammation, acute phase response, leukocyte extravasation, and phagosome formation. On the other hand, enrichment of pathways in hNSC-engrafted brains was associated with PPAR signaling, LXR/RXR activation, and inhibition of matrix metalloproteases. Hierarchical cluster analysis of DEGs in hNSC-engrafted brains indicate decreased expression of genes encoding TNF receptors, proinflammatory factors, apoptosis factors, adhesion and leukocyte extravasation, and Toll-like receptors. CONCLUSIONS Our study is the first to show global transcripts differentially expressed following hNSC transplantation in the subacute phase of stroke in aged mice. The outcome of our transcriptome study would be useful to develop new therapies ameliorating early-stage stroke injury.
Collapse
Affiliation(s)
- Milton H Hamblin
- Tulane University Health Sciences Center, Tulane University, New Orleans, LA 70112, USA.
| | - Rabi Murad
- Bioinformatics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Jun Yin
- Bioinformatics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Gustavo Vallim
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jean-Pyo Lee
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| |
Collapse
|
278
|
del Zoppo GJ, Moskowitz MA, Nedergaard M. The Neurovascular Unit and Responses to Ischemia. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00007-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
279
|
Near-Lifespan Tracking of Cerebral Microvascular Degeneration in Aging to Alzheimer’s Continuum. ADVANCES IN GERIATRIC MEDICINE AND RESEARCH 2022; 4. [PMID: 35466329 PMCID: PMC9022674 DOI: 10.20900/agmr20220003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder affecting millions of people worldwide and is currently incurable. As the population ages, AD and related dementia are becoming the biggest epidemic in medical history: the number of people aged 65 and older with AD is projected to increase between two- and three-fold by 2050. Imaging and biomarker studies suggest that the pathophysiological processes of AD begin more than a decade before the diagnosis of dementia, opening the possibility of early, preemptive prediction. For accurate prediction, it is important although challenging to fully understand how multiple etiologies and age-related prodromal processes contribute to the onset of Alzheimer’s continuum, across a long period comparable to the lifespan. Addressing this challenge was one of the overarching transformative concepts at the 2015 AD Research Summit, “to develop new programs on systems biology and integrative physiology to gain a deeper understanding of the complex biology of the disease.” Among other factors, cerebral microvascular degeneration (CMD) may play a key role in the onset and development of Alzheimer’s continuum, potentially prior to, along with, or independently of the beta-amyloid (Aβ) accumulation. Despite its importance for early detection and as a therapeutic target for early intervention, it is unknown whether CMD is a causal factor for AD pathogenesis or an early consequence of multifactorial conditions that lead to AD at a later stage. Here, this Viewpoint suggests that we should fill two critical knowledge gaps: (1) Temporal relationships between various CMDs and other key factors before/during/after the onset of Alzheimer’s continuum have not been established; (2) Little integrative study down to the capillary vessel level has been conducted on how individual defects in various microvascular structural and flow properties distinctly correlate with and/or contribute to neuronal degeneration. As the first step toward filling these gaps, I propose utilizing recent advances in microscopic imaging and image analysis techniques to longitudinally track a comprehensive set of CMDs over the lifespan in model animals, along with Aβ, tau, neuronal degeneration, and cognitive impairment when possible.
Collapse
|
280
|
Chen C, Duan F, Xie Y, Wan Q, Liu H, Gong J, Huang L, Song Z. Nuciferine attenuates acute ischemic stroke in a rat model: a metabolomic approach for the mechanistic study. Mol Omics 2022; 18:765-778. [DOI: 10.1039/d2mo00158f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nuciferine is a promise therapeutic candidate for ischemic stroke. 1H NMR metabolomics was conducted in this study to further elucidate its pharmacological mechanism, which is helpful to be used as a potential treatment for stroke clinically.
Collapse
Affiliation(s)
- Chang Chen
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, P. R. China
| | - Feipeng Duan
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, P. R. China
| | - Yongyan Xie
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, P. R. China
| | - Quan Wan
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, P. R. China
| | - Haiyun Liu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, P. R. China
| | - Jinpeng Gong
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, P. R. China
| | - Liping Huang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, P. R. China
| | - Zonghua Song
- Chinese Pharmacopoeia Commission, Beijing 100061, P. R. China
| |
Collapse
|
281
|
Garcia-Bonilla L, Iadecola C, Anrather J. Inflammation and Immune Response. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00010-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
282
|
Yang T, Guo R, Ofengeim D, Hwang JY, Zukin RS, Chen J, Zhang F. Molecular and Cellular Mechanisms of Ischemia-Induced Neuronal Death. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00005-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
283
|
Chen H, Zhou L. Treatment of ischemic stroke with modified mesenchymal stem cells. Int J Med Sci 2022; 19:1155-1162. [PMID: 35919816 PMCID: PMC9339408 DOI: 10.7150/ijms.74161] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/15/2022] [Indexed: 11/27/2022] Open
Abstract
Ischemic stroke is one of the leading causes of death and disability. Ischemia triggers a cascade of events leading to cell death and cerebral infarction. Mesenchymal stem cell (MSC) therapy is a promising treatment modality to promote the development of nerve and blood vessels and improve nerve function. However, MSCs have a limited therapeutic effect in the harsh microenvironment of ischemic brain tissue. Modified MSC therapy shows better therapeutic effect under different pathological conditions, and is expected to be translated into clinical practice. In this article, we review the latest advances in the development of modified MSCs for the treatment of cerebral ischemia. In particular, we summarize the targets involved in migration, homing, antioxidant stress, anti-inflammatory, nerve and vascular regeneration, providing new ideas for clinical transformation.
Collapse
Affiliation(s)
- Hao Chen
- Department of Neurovascular Surgery, First Hospital of Jilin University, 1xinmin Avenue Changchun130021, Jilin Province, China
| | - Liangfu Zhou
- Department of Neurovascular Surgery, First Hospital of Jilin University, 1xinmin Avenue Changchun130021, Jilin Province, China
| |
Collapse
|
284
|
Song J. Comparison of Cerebral Cortex Transcriptome Profiles in Ischemic Stroke and Alzheimer’s Disease Models. Clin Nutr Res 2022; 11:159-170. [PMID: 35949563 PMCID: PMC9348914 DOI: 10.7762/cnr.2022.11.3.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/20/2022] [Accepted: 06/24/2022] [Indexed: 11/19/2022] Open
Abstract
Ischemic stroke and Alzheimer’s disease (AD) are representative geriatric diseases with a rapidly increasing prevalence worldwide. Recent studies have reported an association between ischemic stroke neuropathology and AD neuropathology. Ischemic stroke shares some similar characteristics with AD, such as glia activation-induced neuroinflammation, amyloid beta accumulation, and neuronal cell loss, as well as some common risk factors with AD progression. Although there are considerable similarities in neuropathology between ischemic stroke and AD, no studies have ever compared specific genetic changes of brain cortex between ischemic stroke and AD. Therefore, in this study, I compared the cerebral cortex transcriptome profile of 5xFAD mice, an AD mouse model, with those of middle cerebral artery occlusion (MCAO) mice, an ischemic stroke mouse model. The data showed that the expression of many genes with important functional implications in MCAO mouse brain cortex were related to synaptic dysfunction and neuronal cell death in 5xFAD mouse model. In addition, changes in various protein-coding RNAs involved in synaptic plasticity, amyloid beta accumulation, neurogenesis, neuronal differentiation, glial activation, inflammation and neurite outgrowth were observed. The findings could serve as an important basis for further studies to elucidate the pathophysiology of AD in patients with ischemic stroke.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea
| |
Collapse
|
285
|
Qi Y, Xing Y, Wang L, Zhang J, Cao Y, Liu L, Chen Y. Multimodal Monitoring in Large Hemispheric Infarction: Quantitative Electroencephalography Combined With Transcranial Doppler for Prognosis Prediction. Front Neurol 2021; 12:724571. [PMID: 34956039 PMCID: PMC8693413 DOI: 10.3389/fneur.2021.724571] [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: 06/13/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Background: We aimed to explore whether transcranial Doppler (TCD) combined with quantitative electroencephalography (QEEG) can improve prognosis evaluation in patients with a large hemispheric infarction (LHI) and to establish an accurate prognosis prediction model. Methods: We prospectively assessed 90-day mortality in patients with LHI. Brain function was monitored using TCD-QEEG at the bedside of the patient. Results: Of the 59 (55.3 ± 10.6 years; 17 men) enrolled patients, 37 (67.3%) patients died within 90 days. The Cox regression analyses revealed that the Glasgow Coma Scale (GCS) score ≤ 8 [hazard ratio (HR), 3.228; 95% CI, 1.335–7.801; p = 0.009], TCD-terminal internal carotid artery as the offending vessel (HR, 3.830; 95% CI, 1.301–11.271; p = 0.015), and QEEG-a (delta + theta)/(alpha + beta) ratio ≥ 3 (HR, 3.647; 95% CI, 1.170–11.373; p = 0.026) independently predicted survival duration. Combining these three factors yielded an area under the receiver operating characteristic curve of 0.905 and had better predictive accuracy than those of individual variables (p < 0.05). Conclusion: TCD and QEEG complement the GCS score to create a reliable multimodal method for monitoring prognosis in patients with LHI.
Collapse
Affiliation(s)
- Yajie Qi
- Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Department of Neurosurgery, Northern Jiangsu People's Hospital, Clinical Medical College of Yangzhou University, Yangzhou, China
| | - Yingqi Xing
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Diagnostic Center of Vascular Ultrasound, Beijing, China
| | - Lijuan Wang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Jie Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yanting Cao
- Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Department of Neurology, Linyi People's Hospital, Linyi, China
| | - Li Liu
- Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Department of Neurology, Changchun People's Hospital, Changchun, China
| | - Ying Chen
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
286
|
ADAM8 Activates NLRP3 Inflammasome to Promote Cerebral Ischemia-Reperfusion Injury. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:3097432. [PMID: 34956566 PMCID: PMC8702307 DOI: 10.1155/2021/3097432] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/03/2021] [Accepted: 11/15/2021] [Indexed: 11/22/2022]
Abstract
Stroke is the leading cause of death and disability in humans. Strokes are classified as either ischemic or hemorrhagic. Ischemic stroke accounts for 70–80% of the cases. Inflammation is a key factor in ischemic brain injury. Studies have shown that inflammatory response induced by NLRP3 inflammasome is one of the root causes of brain damage in mice with cerebral ischemia. However, its specific mechanism in cerebral ischemia is still unclear. ADAM8 (a disintegrin and metalloproteases 8) is a transmembrane protein with different functions. It plays an important role in tumors and neuroinflammation-related diseases. However, the role and molecular mechanism of ADAM8 in cerebral ischemia injury are still unclear. This study aims to evaluate the role of ADAM8 in cerebral ischemic injury and explore its signal transduction mechanism. This experiment shows that ADAM8 can significantly cause neurological deficits in MCAO mice and can substantially cause ipsilateral cerebral edema and cerebral infarction in MCAO mice. In addition, ADAM8 can significantly induce cortical cell apoptosis in MCAO mice, leading to the loss of neurons and the expression of proinflammatory factors COX2, iNOS, TNFα, and IL-6. Importantly, we confirmed that ADAM8 mediates the inflammatory response by promoting the activation of NLRP3 inflammasome, microglia, and astrocytes. These results indicate that ADAM8 may be a candidate drug target for the prevention and treatment of the cerebral ischemic injury.
Collapse
|
287
|
Gong S, Ma H, Zheng F, Huang J, Zhang Y, Yu B, Li F, Kou J. Inhibiting YAP in Endothelial Cells From Entering the Nucleus Attenuates Blood-Brain Barrier Damage During Ischemia-Reperfusion Injury. Front Pharmacol 2021; 12:777680. [PMID: 34899341 PMCID: PMC8662521 DOI: 10.3389/fphar.2021.777680] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Blood-brain barrier (BBB) damage is a critical event in ischemic stroke, contributing to aggravated brain damage. Endothelial cell form a major component of the BBB, but its regulation in stroke has yet to be clarified. We investigated the function of Yes-associated protein 1 (YAP) in the endothelium on BBB breakdown during cerebral ischemia/reperfusion (I/R) injury. The effects of YAP on BBB dysfunction were explored in middle cerebral artery occlusion/reperfusion (MCAO/R)-injury model mice and using brain microvascular endothelial cells (BMEC) exposed to oxygen-glucose deprivation/reoxygenation (OGD/R) injury. The degree of brain injury was estimated using staining (2,3,5-Triphenyltetrazolium chloride, hematoxylin and eosin) and the detection of cerebral blood flow. BBB breakdown was investigated by examining the leakage of Evans Blue dye and evaluating the expression of tight junction (TJ)-associated proteins and matrix metallopeptidase (MMP) 2 and 9. YAP expression was up-regulated in the nucleus of BMEC after cerebral I/R injury. Verteporfin (YAP inhibitor) down-regulated YAP expression in the nucleus and improved BBB hyperpermeability and TJ integrity disruption stimulated by cerebral I/R. YAP-targeted small interfering RNA (siRNA) exerted the same effects in BMEC cells exposed to OGD/R injury. Our findings provide new insights into the contributions made by YAP to the maintenance of BBB integrity and highlight the potential for YAP to serve as a therapeutic target to modulate BBB integrity following ischemic stroke and related cerebrovascular diseases.
Collapse
Affiliation(s)
- Shuaishuai Gong
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Huifen Ma
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Fan Zheng
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Juan Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yuanyuan Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Boyang Yu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Fang Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Junping Kou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
288
|
Hu Y, Tao W. Microenvironmental Variations After Blood-Brain Barrier Breakdown in Traumatic Brain Injury. Front Mol Neurosci 2021; 14:750810. [PMID: 34899180 PMCID: PMC8662751 DOI: 10.3389/fnmol.2021.750810] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is linked to several pathologies. The blood-brain barrier (BBB) breakdown is considered to be one of the initial changes. Further, the microenvironmental alteration following TBI-induced BBB breakdown can be multi-scaled, constant, and dramatic. The microenvironmental variations after disruption of BBB includes several pathological changes, such as cerebral blood flow (CBF) alteration, brain edema, cerebral metabolism imbalances, and accumulation of inflammatory molecules. The modulation of the microenvironment presents attractive targets for TBI recovery, such as reducing toxic substances, inhibiting inflammation, and promoting neurogenesis. Herein, we briefly review the pathological alterations of the microenvironmental changes following BBB breakdown and outline potential interventions for TBI recovery based on microenvironmental modulation.
Collapse
Affiliation(s)
- Yue Hu
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weiwei Tao
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
289
|
Singer T, Ding S, Ding S. Astroglia Abnormalities in Post-stroke Mood Disorders. ADVANCES IN NEUROBIOLOGY 2021; 26:115-138. [PMID: 34888833 DOI: 10.1007/978-3-030-77375-5_6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Stroke is the leading cause of human death and disability. After a stroke, many patients may have some physical disability, including difficulties in moving, speaking, and seeing, but patients may also exhibit changes in mood manifested by depression, anxiety, and cognitive changes which we call post-stroke mood disorders (PSMDs). Astrocytes are the most diverse and numerous glial cell type in the central nervous system (CNS). They provide structural, nutritional, and metabolic support to neurons and regulate synaptic activity under normal conditions. Astrocytes are also critically involved in focal ischemic stroke (FIS). They undergo many changes after FIS. These changes may affect acute neuronal death and brain damage as well as brain recovery and PSMD in the chronic phase after FIS. Studies using postmortem brain specimens and animal models of FIS suggest that astrocytes/reactive astrocytes are involved in PSMD. This chapter provides an overview of recent advances in the molecular base of astrocyte in PSMD. As astrocytes exhibit high plasticity after FIS, we suggest that targeting local astrocytes may be a promising strategy for PSMD therapy.
Collapse
Affiliation(s)
- Tracey Singer
- Dalton Cardiovascular Research Center, Columbia, MO, USA
| | - Sarah Ding
- Dalton Cardiovascular Research Center, Columbia, MO, USA
| | - Shinghua Ding
- Dalton Cardiovascular Research Center, Columbia, MO, USA.
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
290
|
Hayes CA, Ashmore BG, Vijayasankar A, Marshall JP, Ashpole NM. Insulin-Like Growth Factor-1 Differentially Modulates Glutamate-Induced Toxicity and Stress in Cells of the Neurogliovascular Unit. Front Aging Neurosci 2021; 13:751304. [PMID: 34887742 PMCID: PMC8650493 DOI: 10.3389/fnagi.2021.751304] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 10/08/2021] [Indexed: 11/29/2022] Open
Abstract
The age-related reduction in circulating levels of insulin-like growth factor-1 (IGF-1) is associated with increased risk of stroke and neurodegenerative diseases in advanced age. Numerous reports highlight behavioral and physiological deficits in blood-brain barrier function and neurovascular communication when IGF-1 levels are low. Administration of exogenous IGF-1 reduces the extent of tissue damage and sensorimotor deficits in animal models of ischemic stroke, highlighting the critical role of IGF-1 as a regulator of neurovascular health. The beneficial effects of IGF-1 in the nervous system are often attributed to direct actions on neurons; however, glial cells and the cerebrovasculature are also modulated by IGF-1, and systemic reductions in circulating IGF-1 likely influence the viability and function of the entire neuro-glio-vascular unit. We recently observed that reduced IGF-1 led to impaired glutamate handling in astrocytes. Considering glutamate excitotoxicity is one of the main drivers of neurodegeneration following ischemic stroke, the age-related loss of IGF-1 may also compromise neural function indirectly by altering the function of supporting glia and vasculature. In this study, we assess and compare the effects of IGF-1 signaling on glutamate-induced toxicity and reactive oxygen species (ROS)-produced oxidative stress in primary neuron, astrocyte, and brain microvascular endothelial cell cultures. Our findings verify that neurons are highly susceptible to excitotoxicity, in comparison to astrocytes or endothelial cells, and that a prolonged reduction in IGFR activation increases the extent of toxicity. Moreover, prolonged IGFR inhibition increased the susceptibility of astrocytes to glutamate-induced toxicity and lessened their ability to protect neurons from excitotoxicity. Thus, IGF-1 promotes neuronal survival by acting directly on neurons and indirectly on astrocytes. Despite increased resistance to excitotoxic death, both astrocytes and cerebrovascular endothelial cells exhibit acute increases in glutamate-induced ROS production and mitochondrial dysfunction when IGFR is inhibited at the time of glutamate stimulation. Together these data highlight that each cell type within the neuro-glio-vascular unit differentially responds to stress when IGF-1 signaling was impaired. Therefore, the reductions in circulating IGF-1 observed in advanced age are likely detrimental to the health and function of the entire neuro-glio-vascular unit.
Collapse
Affiliation(s)
- Cellas A Hayes
- Department of BioMolecular Sciences, University of Mississippi School of Pharmacy, University of Mississippi, Oxford, MS, United States
| | - Brandon G Ashmore
- Department of BioMolecular Sciences, University of Mississippi School of Pharmacy, University of Mississippi, Oxford, MS, United States
| | - Akshaya Vijayasankar
- Department of BioMolecular Sciences, University of Mississippi School of Pharmacy, University of Mississippi, Oxford, MS, United States
| | - Jessica P Marshall
- Department of BioMolecular Sciences, University of Mississippi School of Pharmacy, University of Mississippi, Oxford, MS, United States
| | - Nicole M Ashpole
- Department of BioMolecular Sciences, University of Mississippi School of Pharmacy, University of Mississippi, Oxford, MS, United States.,Research Institute of Pharmaceutical Sciences, University of Mississippi School of Pharmacy, University of Mississippi, Oxford, MS, United States
| |
Collapse
|
291
|
Abbasi-Habashi S, Jickling GC, Winship IR. Immune Modulation as a Key Mechanism for the Protective Effects of Remote Ischemic Conditioning After Stroke. Front Neurol 2021; 12:746486. [PMID: 34956045 PMCID: PMC8695500 DOI: 10.3389/fneur.2021.746486] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Remote ischemic conditioning (RIC), which involves a series of short cycles of ischemia in an organ remote to the brain (typically the limbs), has been shown to protect the ischemic penumbra after stroke and reduce ischemia/reperfusion (IR) injury. Although the exact mechanism by which this protective signal is transferred from the remote site to the brain remains unclear, preclinical studies suggest that the mechanisms of RIC involve a combination of circulating humoral factors and neuronal signals. An improved understanding of these mechanisms will facilitate translation to more effective treatment strategies in clinical settings. In this review, we will discuss potential protective mechanisms in the brain and cerebral vasculature associated with RIC. We will discuss a putative role of the immune system and circulating mediators of inflammation in these protective processes, including the expression of pro-and anti-inflammatory genes in peripheral immune cells that may influence the outcome. We will also review the potential role of extracellular vesicles (EVs), biological vectors capable of delivering cell-specific cargo such as proteins and miRNAs to cells, in modulating the protective effects of RIC in the brain and vasculature.
Collapse
Affiliation(s)
- Sima Abbasi-Habashi
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Glen C Jickling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Division of Neurology, Faculty of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Ian R Winship
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
292
|
He J, Ling Q, Chen Y. Construction and Application of a Model for Predicting the Risk of Delirium in Postoperative Patients With Type a Aortic Dissection. Front Surg 2021; 8:772675. [PMID: 34869569 PMCID: PMC8636852 DOI: 10.3389/fsurg.2021.772675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/11/2021] [Indexed: 12/19/2022] Open
Abstract
Background: Postoperative delirium (POD), an alteration in a patient's consciousness pattern, can affect the treatment and prognosis of a disease. Objective: To construct a prediction model for delirium in patients with type A aortic dissection after surgery and to validate its effectiveness. Methods: A retrospective cohort design was used to study 438 patients undergoing surgical treatment for type A aortic dissection from April 2019 to June 2020 in tertiary care hospitals. POD (n = 78) and non-delirium groups (n = 360) were compared and analyzed for each index in the perioperative period. A prediction model was established using multifactorial logistic regression, and 30 patients' perioperative data were collected for model validation. Results: Eight predictors were included in this study: smoking, diabetes, previous cardiovascular surgery, ejection fraction (EF), time to aortic block, acute kidney injury, low cardiac output syndrome, and pulmonary complications. The area under the receiver operating characteristic (ROC) curve of the constructed prediction model was 0.98 ± 0.005, and the Youden index was 0.91. The validation results showed 97% sensitivity, 100% specificity, and 93% accuracy. The expression of the model was Z = Smoking assignment* – 2.807 – 6.009*Diabetes assignment – 2.994*Previous cardiovascular surgery assignment – 0.129*Ejection fraction assignment + 0.071*Brain perfusion time assignment – 2.583*Acute kidney injury assignment – 2.916*Low cardiac output syndrome assignment – 3.461*Pulmonary related complications assignment + 20.576. Conclusion: The construction of an effective prediction model for the risk of delirium in patients after type A aortic stratification can help identify patients at high risk of POD early. It also provides a reference for healthcare professionals in the prevention and care of these patients.
Collapse
Affiliation(s)
- Junfeng He
- Department of Nursing, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qing Ling
- Department of Nursing, Nanjing First Hospital, Nanjing, China
| | - Yuhong Chen
- Department of Nursing, Nanjing First Hospital, Nanjing, China
| |
Collapse
|
293
|
Stuckey SM, Ong LK, Collins-Praino LE, Turner RJ. Neuroinflammation as a Key Driver of Secondary Neurodegeneration Following Stroke? Int J Mol Sci 2021; 22:ijms222313101. [PMID: 34884906 PMCID: PMC8658328 DOI: 10.3390/ijms222313101] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/25/2021] [Accepted: 12/01/2021] [Indexed: 01/13/2023] Open
Abstract
Ischaemic stroke involves the rapid onset of focal neurological dysfunction, most commonly due to an arterial blockage in a specific region of the brain. Stroke is a leading cause of death and common cause of disability, with over 17 million people worldwide suffering from a stroke each year. It is now well-documented that neuroinflammation and immune mediators play a key role in acute and long-term neuronal tissue damage and healing, not only in the infarct core but also in distal regions. Importantly, in these distal regions, termed sites of secondary neurodegeneration (SND), spikes in neuroinflammation may be seen sometime after the initial stroke onset, but prior to the presence of the neuronal tissue damage within these regions. However, it is key to acknowledge that, despite the mounting information describing neuroinflammation following ischaemic stroke, the exact mechanisms whereby inflammatory cells and their mediators drive stroke-induced neuroinflammation are still not fully understood. As a result, current anti-inflammatory treatments have failed to show efficacy in clinical trials. In this review we discuss the complexities of post-stroke neuroinflammation, specifically how it affects neuronal tissue and post-stroke outcome acutely, chronically, and in sites of SND. We then discuss current and previously assessed anti-inflammatory therapies, with a particular focus on how failed anti-inflammatories may be repurposed to target SND-associated neuroinflammation.
Collapse
Affiliation(s)
- Shannon M. Stuckey
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (S.M.S.); (L.E.C.-P.)
| | - Lin Kooi Ong
- School of Pharmacy, Monash University Malaysia, Subang Jaya 47500, Malaysia;
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, The University of Newcastle, Callaghan 2308, Australia
| | - Lyndsey E. Collins-Praino
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (S.M.S.); (L.E.C.-P.)
| | - Renée J. Turner
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (S.M.S.); (L.E.C.-P.)
- Correspondence: ; Tel.: +61-8-8313-3114
| |
Collapse
|
294
|
Hu ZB, Lu ZX, Zhu F, Jiang CQ, Zhang WS, Pan J, Jin YL, Xu L, Thomas GN, Cheng K, Lam T. Higher total white blood cell and neutrophil counts are associated with an increased risk of fatal stroke occurrence: the Guangzhou biobank cohort study. BMC Neurol 2021; 21:470. [PMID: 34856939 PMCID: PMC8638334 DOI: 10.1186/s12883-021-02495-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 11/16/2021] [Indexed: 12/05/2022] Open
Abstract
Background Chronic inflammatory diseases are linked to an increased risk of stroke events. The white blood cell (WBC) count is a common marker of the inflammatory response. However, it is unclear whether the WBC count, its subpopulations and their dynamic changes are related to the risk of fatal stroke in relatively healthy elderly population. Methods In total, 27,811 participants without a stroke history at baseline were included and followed up for a mean of 11.5 (standard deviation = 2.3) years. After review of available records, 503 stroke deaths (ischaemic 227, haemorrhagic 172 and unclassified 104) were recorded. Cox proportional hazards regression was used to assess the associations between the WBC count, its subpopulations and their dynamic changes (two-phase examination from baseline to the 1st follow-up) and the risk of fatal all stroke, fatal ischaemic stroke and fatal haemorrhagic stroke. Results (i) Regarding the WBC count in relation to the risk of fatal stroke, restricted cubic splines showed an atypically U-curved association between the WBC count and the risk of fatal all stroke occurrence. Compared with those in the lowest WBC count quartile (< 5.3*10^9/L), the participants with the highest WBC count (> 7.2*10^9/L) had a 53 and 67% increased risk for fatal all stroke (adjusted hazard ratio [aHR] = 1.53, 95% confidence interval (CI) 1.16–2.02, P = 0.003) and fatal haemorrhagic stroke (aHR = 1.67, 95% CI 1.10–2.67, P = 0.03), respectively; compared with those in the lowest quartile (< 3.0*10^9/L), the participants with the highest NEUT count (> 4.5*10^9/L) had a 45 and 65% increased risk for fatal all stroke (aHR = 1.45, 95% CI 1.10–1.89, P = 0.008) and fatal ischaemic stroke (aHR = 1.65, 95%CI 1.10–2.47 P = 0.02), respectively. With the additional adjustment for C-reactive protein, the same results as those for all stroke and ischaemic stroke, but not haemorrhagic stroke, were obtained for the WBC count (4 ~ 10*10^9/L) and the NEUT count (the NEUT counts in the top 1% and bottom 1% at baseline were excluded). (ii) Regarding dynamic changes in the WBC count in relation to the risk of fatal stroke, compared with the stable group (− 25% ~ 25%, dynamic changes from two phases of examination (baseline, from September 1st, 2003 to February 28th, 2008; 1st follow-up, from March 31st 2008 to December 31st 2012)), the groups with a 25% increase in the WBC count and NEUT count respectively had a 60% (aHR = 1.60, 95% CI 1.07–2.40, P = 0.02) and 45% (aHR = 1.45, 95% CI1.02–2.05, P = 0.04) increased risk of fatal all stroke occurrence. Conclusions The WBC count, especially the NEUT count, was associated with an increased risk of fatal all stroke occurrence. Longitudinal changes in the WBC count and NEUT count increase in excess of 25% were also associated with an increased risk of fatal all stroke occurrence in the elderly population. Supplementary Information The online version contains supplementary material available at 10.1186/s12883-021-02495-z.
Collapse
Affiliation(s)
- Zhi-Bing Hu
- Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Ze-Xiong Lu
- Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Feng Zhu
- Guangzhou Twelfth People's Hospital, Guangzhou, China.
| | | | - Wei-Sen Zhang
- Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Jin Pan
- Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Ya-Li Jin
- Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Lin Xu
- School of Public Health, the University of Hong Kong, Pokfulam, Hong Kong.,School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - G Neil Thomas
- Institute of Applied Health Research, University of Birmingham, Sun Yat-sen University, Birmingham, UK
| | - Karkeung Cheng
- Institute of Applied Health Research, University of Birmingham, Sun Yat-sen University, Birmingham, UK
| | - Taihing Lam
- Guangzhou Twelfth People's Hospital, Guangzhou, China.,School of Public Health, the University of Hong Kong, Pokfulam, Hong Kong.,School of Public Health, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
295
|
Kaurav H, Sharma A, Upadhyay NK, Kapoor DN. Long term delivery of glibenclamide from in situ forming microparticles for the treatment of ischemic stroke. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
296
|
Li W, Xu D. Application of intelligent rehabilitation equipment in occupational therapy for enhancing upper limb function of patients in the whole phase of stroke. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2021. [DOI: 10.1016/j.medntd.2021.100097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
297
|
He F, Ma C, Feng J, Li X, Xia S, Lin Q, Dai R. Angiogenesis effects of 4-methoxy benzyl alcohol on cerebral ischemia-reperfusion injury via regulation of VEGF-Ang/Tie2 balance. Can J Physiol Pharmacol 2021; 99:1253-1263. [PMID: 34283928 DOI: 10.1139/cjpp-2021-0118] [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: 11/22/2022]
Abstract
Angiogenesis facilitates the formation of microvascular networks and promotes neurological deficit recovery after cerebral ischemia-reperfusion injury (CIRI). This study investigated the angiogenesis effects of 4-methoxy benzyl alcohol (4-MA) on CIRI. The angiogenesis effects of 4-MA and the potential underlying mechanisms were assessed based on a middle cerebral artery occlusion/reperfusion (MCAO/R) rat model and a hind limb ischemic (HLI) mouse model. Immunofluorescence was conducted to detect microvessel density, and Western blotting and polymerase chain reaction were performed to determine the expression of angiogenesis-promoting factors. In addition, we investigated whether the angiogenesis effects of 4-MA caused damage to the blood-brain barrier (BBB). After treatment with 4-MA (20 mg/kg) for 7 days, the neurological deficits recovered and microvessel density in the cerebral cortex increased in the MCAO/R rats. Additionally, 4-MA also regulated the expression of angiogenesis factors, with an increase in vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor 2 (VEGFR-2) expression and a decrease in angiopoietin 1 (Ang-1), Ang-2, and Tie-2 expression in both MCAO/R rats and HLI mice. Moreover, 4-MA increased the expression of angiogenesis-promoting factors without exacerbating BBB cascade damage in MCAO/R rats. Our results indicated that 4-MA may contribute to the formation of microvascular networks, thus promoting neurological deficit recovery after CIRI.
Collapse
Affiliation(s)
- Fangyan He
- Department of Pharmacology, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Chenjing Ma
- Research Institute of Resource Insects, Chinese Academy of Forestry, Kunming, Yunnan 650000, China
| | - Jin Feng
- Department of Pharmacology, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Xiufang Li
- Department of Pharmacology, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Shuangli Xia
- Department of Pharmacology, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Qing Lin
- Department of Pharmacology, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Rong Dai
- Department of Pharmacology, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| |
Collapse
|
298
|
Solovyev R, Kalinin AA, Gabruseva T. 3D convolutional neural networks for stalled brain capillary detection. Comput Biol Med 2021; 141:105089. [PMID: 34920160 DOI: 10.1016/j.compbiomed.2021.105089] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 11/26/2021] [Accepted: 11/26/2021] [Indexed: 01/08/2023]
Abstract
Adequate blood supply is critical for normal brain function. Brain vasculature dysfunctions, including stalled blood flow in cerebral capillaries, are associated with cognitive decline and pathogenesis in Alzheimer's disease. Recent advances in imaging technology enabled generation of high-quality 3D images that can be used to visualize stalled blood vessels. However, localization of stalled vessels in 3D images is often required as the first step for downstream analysis. When performed manually, this process is tedious, time-consuming, and error-prone. Here, we describe a deep learning-based approach for automatic detection of stalled capillaries in brain images based on 3D convolutional neural networks. Our approach includes custom 3D data augmentations and a weights transfer method that re-uses weights from 2D models pre-trained on natural images for initialization of 3D networks. We used an ensemble of several 3D models to produce the winning submission to the "Clog Loss: Advance Alzheimer's Research with Stall Catchers" machine learning competition that challenged the participants with classifying blood vessels in 3D image stacks as stalled or flowing. In this setting, our approach outperformed other methods and demonstrated state-of-the-art results, achieving 85% Matthews correlation coefficient, 85% sensitivity, and 99.3% specificity. The source code for our solution is publicly available.
Collapse
Affiliation(s)
- Roman Solovyev
- Institute for Design Problems in Microelectronics of Russian Academy of Sciences, 3, Sovetskaya Street, Moscow, 124 365, Russian Federation.
| | - Alexandr A Kalinin
- Shenzhen Research Institute of Big Data, Shenzhen, 518 172, Guangdong, China; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48 109, USA
| | | |
Collapse
|
299
|
Ononin inhibits cerebral ischemia/reperfusion injury via suppression of inflammatory responses in experimental rats and SH-SY5Y cells. APPLIED NANOSCIENCE 2021. [DOI: 10.1007/s13204-021-02184-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
300
|
Ramezani M, Simani L, Abedi S, Pakdaman H. Is Selenium Supplementation Beneficial in Acute Ischemic Stroke? Neurologist 2021; 27:51-55. [PMID: 34842573 DOI: 10.1097/nrl.0000000000000365] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Selenium (Se) plays a significant role in brain physiology. The existing human data demonstrate that stroke is associated with significantly reduced Se levels and glutathione peroxidase (GPx) activity. This study proposed to investigate the effect of intravenous Se (Selenase) administration in patients with acute ischemic stroke (AIS) on neurological outcomes, antioxidant enzyme activity, and inflammatory marker levels. METHODS AIS patients (n=50) were recruited from a neurology unit of a university-affiliated hospital. Patients were randomly assigned to receive either Selenase or placebo (saline) for 5 days. The modified ranking scale, the national institute of health stroke scale, and the mini-mental state examination, as primary outcomes, and the serum GPx concentration, total antioxidant activity, and tumor necrosis factor-α levels, as secondary outcomes, were measured at the baseline and on day 30. RESULTS Eventually, 44 patients with AIS completed the intervention study. A notable increase in GPx and total antioxidant activity levels was detected in the treatment group compared with the placebo group (110.63±52.48 m/mL, 1.34±0.30 mmol/L, P<0.05), whereas the serum tumor necrosis factor-α level in the Selenase group was significantly lower than that of the placebo group (58.58±61.33 pg/mL, P<0.05). In addition, Selenase improved the modified ranking scale and national institute of health stroke scale scores significantly (P<0.05 and <0.04, respectively), but no statistical difference was observed between the 2 groups in the mini-mental state examination score. CONCLUSION Selenase, plausibly due to its antioxidant function, results in positive outcomes in terms of neurological deficits, antioxidant enzyme activity, and inflammatory marker levels.
Collapse
Affiliation(s)
- Mahtab Ramezani
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences
- Department of Neurology
| | - Leila Simani
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Hossein Pakdaman
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences
| |
Collapse
|