1
|
Shu H, Liao Q, Chen Z, Liang M, Zhang S, Liu J, Wu Y, Hu P, Luo M, Zhu W, Zhu X, Yang L, Yan T. Flavonoids serve as a promising therapeutic agent for ischemic stroke. Brain Res 2025; 1853:149528. [PMID: 39999903 DOI: 10.1016/j.brainres.2025.149528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 02/27/2025]
Abstract
Ischemic stroke (IS) continues to be a major public health concern and is characterized by significantly high mortality and disabling rates. Inhibiting nerve cells death and enhancing the repair of ischemic tissue are important treatment concepts for IS. Currently, the mainstream treatment strategies mainly focus on short-term care, which underscores the urgent need for novel therapeutic strategies for long-term care. Emerging data reveal that flavonoids have surfaced as promising candidates for IS patients' long-term care. Flavonoids can alleviate neuroinflammation and anti-apoptosis due to their characteristic pharmacological mechanisms. Clinical evidence suggests that long-term flavonoids intake improves IS patients' long-term outcomes. Though the effect of flavonoids in IS treatment has been explored for decades, the neuroprotective pharmacodynamics have not been well established. Thereby, the aim of current review is to summarize the pathways involved in neuroprotective effect of flavonoids. This review will also advance the potential of flavonoids as a viable clinical candidate for the treatment of IS.
Collapse
Affiliation(s)
- Hongxin Shu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qiuye Liao
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Zhihao Chen
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Mingyu Liang
- School of life sciences, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Si Zhang
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Junzhe Liu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Yanze Wu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ping Hu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ming Luo
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Wenping Zhu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xingen Zhu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Li Yang
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Tengfeng Yan
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
2
|
Kim HS, Jee SA, Einisadr A, Seo Y, Seo HG, Jang BS, Park HH, Chung WS, Kim BG. Detrimental influence of Arginase-1 in infiltrating macrophages on poststroke functional recovery and inflammatory milieu. Proc Natl Acad Sci U S A 2025; 122:e2413484122. [PMID: 39951507 PMCID: PMC11848331 DOI: 10.1073/pnas.2413484122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 12/31/2024] [Indexed: 02/16/2025] Open
Abstract
Poststroke inflammation critically influences functional outcomes following ischemic stroke. Arginase-1 (Arg1) is considered a marker for anti-inflammatory macrophages, associated with the resolution of inflammation and promotion of tissue repair in various pathological conditions. However, its specific role in poststroke recovery remains to be elucidated. This study investigates the functional impact of Arg1 expressed in macrophages on poststroke recovery and inflammatory milieu. We observed a time-dependent increase in Arg1 expression, peaking at 7 d after photothrombotic stroke in mice. Cellular mapping analysis revealed that Arg1 was predominantly expressed in LysM-positive infiltrating macrophages. Using a conditional knockout (cKO) mouse model, we examined the role of Arg1 expressed in infiltrating macrophages. Contrary to its presumed beneficial effects, Arg1 cKO in LysM-positive macrophages significantly improved skilled forelimb motor function recovery after stroke. Mechanistically, Arg1 cKO attenuated fibrotic scar formation, enhanced peri-infarct remyelination, and increased synaptic density while reducing microglial synaptic elimination in the peri-infarct cortex. Gene expression analysis of fluorescence-activated single cell sorting (FACS)-sorted CD45low microglia revealed decreased transforming growth factor-β (TGF-β) signaling and proinflammatory cytokine activity in peri-infarct microglia from Arg1 cKO animals. In vitro coculture experiments demonstrated that Arg1 activity in macrophages modulates microglial synaptic phagocytosis, providing evidence for macrophage-microglia interaction. These findings present unique insights into the function of Arg1 in central nervous system injury and highlight an interaction between infiltrating macrophages and resident microglia in shaping the poststroke inflammatory milieu. Our study identifies Arg1 in macrophages as a potential therapeutic target for modulating poststroke inflammation and improving functional recovery.
Collapse
Affiliation(s)
- Hyung Soon Kim
- Department of Brain Science, Ajou University School of Medicine, Suwon16499, Republic of Korea
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon16499, Republic of Korea
| | - Seung Ah Jee
- Department of Brain Science, Ajou University School of Medicine, Suwon16499, Republic of Korea
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon16499, Republic of Korea
| | - Ariandokht Einisadr
- Department of Brain Science, Ajou University School of Medicine, Suwon16499, Republic of Korea
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon16499, Republic of Korea
| | - Yeojin Seo
- Department of Brain Science, Ajou University School of Medicine, Suwon16499, Republic of Korea
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon16499, Republic of Korea
| | - Hyo Gyeong Seo
- Department of Brain Science, Ajou University School of Medicine, Suwon16499, Republic of Korea
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon16499, Republic of Korea
| | - Byeong Seong Jang
- Department of Brain Science, Ajou University School of Medicine, Suwon16499, Republic of Korea
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon16499, Republic of Korea
| | - Hee Hwan Park
- Department of Brain Science, Ajou University School of Medicine, Suwon16499, Republic of Korea
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon16499, Republic of Korea
| | - Won-Suk Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon34141, Republic of Korea
- Center for Vascular Biology, Institute for Basic Science, Daejeon34126, Republic of Korea
| | - Byung Gon Kim
- Department of Brain Science, Ajou University School of Medicine, Suwon16499, Republic of Korea
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon16499, Republic of Korea
- Department of Neurology, Ajou University School of Medicine, Suwon16499, Republic of Korea
| |
Collapse
|
3
|
Fernández-Garza LE, González-Aquines A, Botello-Hernández E, Pérez-Vázquez G, Cristobal-Niño M, Góngora-Rivera F. Segmented neutrophil-to-monocyte ratio and systemic immune-inflammation index associated with the severity and functional prognosis of acute ischemic stroke. Int J Neurosci 2025; 135:228-236. [PMID: 38088139 DOI: 10.1080/00207454.2023.2294705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/09/2023] [Accepted: 12/09/2023] [Indexed: 12/26/2023]
Abstract
PURPOSE/AIM OF THE STUDY To identify the inflammation indexes associated with the severity and functional prognosis in ischemic stroke. MATERIAL AND METHODS A prospective study was conducted with ischemic stroke cases included in the i-ReNe clinical registry. Patients were divided into groups according to the severity on admission measured by the National Institutes of Health Stroke Scale (NIHSS) and the functional prognosis at 30 and 90 days of discharge measured by the modified Rankin Scale (mRS). RESULTS We included 145 patients with a mean age of 61.5 ± 12.75, 97 (66.9%) were men. The leukocyte and neutrophil counts, Neutrophil-to-Lymphocyte ratio (NLR), Derived Neutrophil-to-Lymphocyte ratio (dNLR), Platelet-to-Lymphocyte ratio (PLR), Segmented Neutrophil-to-Monocyte ratio (SeMo ratio), and Systemic Immune-inflammation index (SII) were higher in moderate-to-severe stroke (NIHSS ≥6). NLR, PLR, SeMo ratio, and SII were higher in the group with severe disability and death at 30 days (mRS ≥4). In the multiple logistic regression analyses, SeMo ratio >14.966 and SII >623.723 were associated with moderate-to-severe stroke (NIHSS ≥6). In addition, SeMo ratio >7.845 was associated with severe disability and death at 30 days (mRS ≥4). CONCLUSIONS Systemic inflammation indexes could be rapid and low-cost markers used in the initial evaluation of ischemic stroke, whose values could help to stratify patients according to their severity and functional prognosis. This is the first study to establish a relationship between ischemic stroke and the SeMo ratio.
Collapse
Affiliation(s)
- Luis E Fernández-Garza
- Neurology Department, University Hospital "Dr. José Eleuterio González", Autonomous University of Nuevo Leon, Monterrey, NL, Mexico
| | - Alejandro González-Aquines
- Neurology Department, University Hospital "Dr. José Eleuterio González", Autonomous University of Nuevo Leon, Monterrey, NL, Mexico
| | - Edgar Botello-Hernández
- Neurology Department, University Hospital "Dr. José Eleuterio González", Autonomous University of Nuevo Leon, Monterrey, NL, Mexico
| | - Gil Pérez-Vázquez
- Neurology Department, University Hospital "Dr. José Eleuterio González", Autonomous University of Nuevo Leon, Monterrey, NL, Mexico
| | - Mario Cristobal-Niño
- Neurology Department, University Hospital "Dr. José Eleuterio González", Autonomous University of Nuevo Leon, Monterrey, NL, Mexico
| | - Fernando Góngora-Rivera
- Neurology Department, University Hospital "Dr. José Eleuterio González", Autonomous University of Nuevo Leon, Monterrey, NL, Mexico
- Stroke Unit, University Hospital "Dr. José Eleuterio González", Autonomous University of Nuevo Leon, Monterrey, NL, Mexico
- Neuromodulation and Brain Plasticity Unit (UNYPC), Center for Research and Development in Health Sciences (CIDICS), Autonomous University of Nuevo Leon, Monterrey, NL, Mexico
| |
Collapse
|
4
|
Vatte S, Ugale R. HIF-1, an important regulator in potential new therapeutic approaches to ischemic stroke. Neurochem Int 2023; 170:105605. [PMID: 37657765 DOI: 10.1016/j.neuint.2023.105605] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/03/2023]
Abstract
Ischemic stroke is a leading cause of disability and mortality worldwide due to the narrow therapeutic window of the only approved therapies like intravenous thrombolysis and thrombectomy. Hypoxia inducible factor-1α (HIF-1α) is a sensitive regulator of oxygen homeostasis, and its expression is rapidly induced after hypoxia/ischemia. It plays an extensive role in the pathophysiology of stroke by regulating multiple pathways including glucose metabolism, angiogenesis, neuronal survival, neuroinflammation and blood brain barrier regulation. Here, we give a brief overview of the HIF-1α-targeting strategies currently under investigation and summarise recent research on how HIF-1α is regulated in various brain cells, including neurons and microglia, at various stages in ischemic stroke. The roles of HIF-1 in stroke varies with ischemic time and degree of ischemia, are still up for debate. More focus has been placed on prospective HIF-1α targeting drugs, such as HIF-1α activator, HIF-1α stabilizers, and natural compounds. In this review, we have highlighted the regulation of HIF-1α in the novel therapeutic approaches for treatment of stroke.
Collapse
Affiliation(s)
- Sneha Vatte
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, 440033, India.
| | - Rajesh Ugale
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, 440033, India.
| |
Collapse
|
5
|
Kim JY, Hwang M, Choi NY, Koh SH. Inhibition of the NLRP3 Inflammasome Activation/Assembly through the Activation of the PI3K Pathway by Naloxone Protects Neural Stem Cells from Ischemic Condition. Mol Neurobiol 2023; 60:5330-5342. [PMID: 37300646 DOI: 10.1007/s12035-023-03418-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 05/31/2023] [Indexed: 06/12/2023]
Abstract
Naloxone is a well-known opioid antagonist and has been suggested to have neuroprotective effects in cerebral ischemia. We investigated whether naloxone exhibits anti-inflammatory and neuroprotective effects in neural stem cells (NSCs) injured by oxygen-glucose deprivation (OGD), whether it affects the NOD-like receptor protein 3 (NLRP3) inflammasome activation/assembly, and whether the role of the phosphatidylinositol 3-kinase (PI3K) pathway is important in the control of NLRP3 inflammasome activation/assembly by naloxone. Primary cultured NSCs were subjected to OGD and treated with different concentrations of naloxone. Cell viability, proliferation, and the intracellular signaling proteins associated with the PI3K pathway and NLRP3 inflammasome activation/assembly were evaluated in OGD-injured NSCs. OGD significantly reduced survival, proliferation, and migration and increased apoptosis of NSCs. However, treatment with naloxone significantly restored survival, proliferation, and migration and decreased apoptosis of NSCs. Moreover, OGD markedly increased NLRP3 inflammasome activation/assembly and cleaved caspase-1 and interleukin-1β levels in NSCs, but naloxone significantly attenuated these effects. These neuroprotective and anti-inflammatory effects of naloxone were eliminated when cells were treated with PI3K inhibitors. Our results suggest that NLRP3 inflammasome is a potential therapeutic target and that naloxone reduces ischemic injury in NSCs by inhibiting NLRP3 inflammasome activation/assembly mediated by the activation of the PI3K signaling pathway.
Collapse
Affiliation(s)
- Ji Young Kim
- Department of Nuclear Medicine, Hanyang University College of Medicine, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do, 11923, Republic of Korea
| | - Mina Hwang
- Department of Neurology, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do, 11923, Republic of Korea
| | - Na-Young Choi
- Department of Neurology, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do, 11923, Republic of Korea
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do, 11923, Republic of Korea.
| |
Collapse
|
6
|
Gao Y, Fang C, Wang J, Ye Y, Li Y, Xu Q, Kang X, Gu L. Neuroinflammatory Biomarkers in the Brain, Cerebrospinal Fluid, and Blood After Ischemic Stroke. Mol Neurobiol 2023; 60:5117-5136. [PMID: 37258724 DOI: 10.1007/s12035-023-03399-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
The most frequent type of stroke, known as ischemic stroke (IS), is a significant global public health issue. The pathological process of IS and post-IS episodes has not yet been fully explored, but neuroinflammation has been identified as one of the key processes. Biomarkers are objective indicators used to assess normal or pathological processes, evaluate responses to treatment, and predict outcomes, and some biomarkers can also be used as therapeutic targets. After IS, various molecules are produced by different cell types, such as microglia, astrocytes, infiltrating leukocytes, endothelial cells, and damaged neurons, that participate in the neuroinflammatory response within the ischemic brain region. These molecules may either promote or inhibit neuroinflammation and may be released into extracellular spaces, including cerebrospinal fluid (CSF) and blood, due to reasons such as BBB damage. These neuroinflammatory molecules should be valued as biomarkers to monitor whether their expression levels in the blood, CSF, and brain correlate with the diagnosis and prognosis of IS patients or whether they have potential as therapeutic targets. In addition, although some molecules do not directly participate in the process of neuroinflammation, they have been reported to have potential diagnostic or therapeutic value against post-IS neuroinflammation, and these molecules will also be listed. In this review, we summarize the neuroinflammatory biomarkers in the brain, CSF, and blood after an IS episode and the potential value of these biomarkers for the diagnosis, treatment, and prognosis of IS patients.
Collapse
Affiliation(s)
- Yikun Gao
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Congcong Fang
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jin Wang
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yingze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yina Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qingxue Xu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xianhui Kang
- Department of Anesthesia, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310006, China.
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
7
|
Dabi YT, Ajagbe AO, Degechisa ST. Toll-like receptors in pathogenesis of neurodegenerative diseases and their therapeutic potential. Immun Inflamm Dis 2023; 11:e839. [PMID: 37102648 PMCID: PMC10116887 DOI: 10.1002/iid3.839] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/17/2023] [Accepted: 03/29/2023] [Indexed: 04/28/2023] Open
Abstract
Toll-like receptors (TLRs) are a family of pattern-recognition receptors triggered by pathogen-derived and tissue-damage-related ligands. TLRs were previously believed to only be expressed in immune cells. However, it is now confirmed that they are ubiquitously expressed in cells within the body including neurons, astrocytes, and microglia of the central nervous system (CNS). Activation of TLRs is capable of inducing immunologic and inflammatory responses to injury or infection of CNS. This response is self-limiting that usually resolves once the infection has been eradicated or the tissue damage has been repaired. However, the persistence of inflammation-inducing insults or a failure in normal resolution mechanisms may result in overwhelming inflammation which may induce neurodegeneration. This implies that TLRs may play a role in mediating the link between inflammation and neurodegenerative diseases namely Alzheimer's disease, Parkinson's disease, Huntington's disease, stroke, and amyotrophic lateral sclerosis. So, new therapeutic approaches that specifically target TLRs may be developed by better understanding TLR expression mechanisms in the CNS and their connections to particular neurodegenerative disorders. Therefore, this review paper discussed the role of TLRs in neurodegenerative diseases.
Collapse
Affiliation(s)
- Yosef Tsegaye Dabi
- Department of Medical Laboratory Science, Institute of Health SciencesWollega UniversityNekemteEthiopia
| | - Abayomi O. Ajagbe
- Department of Anatomy, College of Health Sciences, Faculty of Basic Medical SciencesNile University of NigeriaAbujaNigeria
| | - Sisay T. Degechisa
- Department of Medical Biochemistry, School of Medicine, College of Health SciencesAddis Ababa UniversityAddis AbabaEthiopia
- Department of Medical Laboratory Sciences, College of Medicine and Health SciencesArba Minch UniversityArba MinchEthiopia
| |
Collapse
|
8
|
Thapa K, Shivam K, Khan H, Kaur A, Dua K, Singh S, Singh TG. Emerging Targets for Modulation of Immune Response and Inflammation in Stroke. Neurochem Res 2023; 48:1663-1690. [PMID: 36763312 DOI: 10.1007/s11064-023-03875-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 02/11/2023]
Abstract
The inflammatory and immunological responses play a significant role after stroke. The innate immune activation stimulated by microglia during stroke results in the migration of macrophages and lymphocytes into the brain and are responsible for tissue damage. The immune response and inflammation following stroke have no defined targets, and the intricacies of the immunological and inflammatory processes are only partially understood. Innate immune cells enter the brain and meninges during the acute phase, which can cause ischemia damage. Activation of systemic immunity is caused by danger signals sent into the bloodstream by injured brain cells, which is followed by a significant immunodepression that encourages life-threatening infections. Neuropsychiatric sequelae, a major source of post-stroke morbidity, may be induced by an adaptive immune response that is initiated by antigen presentation during the chronic period and is directed against the brain. Thus, the current review discusses the role of immune response and inflammation in stroke pathogenesis, their role in the progression of injury during the stroke, and the emerging targets for the modulation of the mechanism of immune response and inflammation that may have possible therapeutic benefits against stroke.
Collapse
Affiliation(s)
- Komal Thapa
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.,School of Pharmacy, Chitkara University, Rajpura, Himachal Pradesh, 174103, India
| | - Kumar Shivam
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia.,Faculty of Health, Australian Research Centre in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, 2007, Australia
| | - Sachin Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar - Delhi G.T. Road, Phagwara, Punjab, 144411, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
9
|
Truong SHT, Bonnici B, Rupasinghe S, Kemp-Harper BK, Samuel CS, Broughton BRS. Post-stroke administration of H2 relaxin reduces functional deficits, neuronal apoptosis and immune cell infiltration into the mouse brain. Pharmacol Res 2023; 187:106611. [PMID: 36526079 DOI: 10.1016/j.phrs.2022.106611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/11/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022]
Abstract
Brain inflammation and apoptosis contribute to neuronal damage and loss following ischaemic stroke, leading to cognitive and functional disability. It is well-documented that the human gene-2 (H2)-relaxin hormone exhibits pleiotropic properties via its cognate receptor, Relaxin Family Peptide Receptor 1 (RXFP1), including anti-inflammatory and anti-apoptotic effects, thus making it a potential therapeutic for stroke. Hence, the current study investigated whether post-stroke H2-relaxin administration could improve functional and histological outcomes. 8-12-week-old male C57BL/6 mice were subjected to sham operation or photothrombotic stroke and intravenously-administered with either saline (vehicle) or 0.02, 0.2 or 2 mg/kg doses of recombinant H2-relaxin at 6, 24 and 48 h post-stroke. Motor function was assessed using the hanging wire and cylinder test pre-surgery, and at 24 and 72 h post-stroke. Brains were removed after 72 h and infarct volume was assessed via thionin staining, and RXFP1 expression, leukocyte infiltration and apoptosis were determined by immunofluorescence. RXFP1 was identified on neurons, astrocytes and macrophages, and increased post-stroke. Whilst H2-relaxin did not alter infarct volume, it did cause a dose-dependent improvement in motor function at 24 and 72 h post-stroke. Moreover, 2 mg/kg H2-relaxin significantly decreased the number of apoptotic cells as well as macrophages and neutrophils within the ischaemic hemisphere, but did not alter T or B cells numbers. The anti-inflammatory and anti-apoptotic effects of H2-relaxin when administered at 6 h post-cerebral ischaemia may provide a novel therapeutic option for patients following ischaemic stroke.
Collapse
Affiliation(s)
- Shirley H T Truong
- Cardiovascular & Pulmonary Pharmacology Group, Department of Pharmacology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Benjamin Bonnici
- Cardiovascular & Pulmonary Pharmacology Group, Department of Pharmacology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Samoda Rupasinghe
- Cardiovascular & Pulmonary Pharmacology Group, Department of Pharmacology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Barbara K Kemp-Harper
- Cardiovascular & Pulmonary Pharmacology Group, Department of Pharmacology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Chrishan S Samuel
- Fibrosis Group, Department of Pharmacology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Brad R S Broughton
- Cardiovascular & Pulmonary Pharmacology Group, Department of Pharmacology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
10
|
Chen S, Cheng J, Ye Q, Ye Z, Zhang Y, Liu Y, Huang G, Chen F, Yang M, Wang C, Duan T, Liu X, Zhang Z. Day 1 neutrophil-to-lymphocyte ratio (NLR) predicts stroke outcome after intravenous thrombolysis and mechanical thrombectomy. Front Neurol 2022; 13:941251. [PMID: 36016545 PMCID: PMC9396211 DOI: 10.3389/fneur.2022.941251] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/05/2022] [Indexed: 11/25/2022] Open
Abstract
Background The neutrophil-to-lymphocyte ratio (NLR) is a biomarker reflecting the balance between inflammation (as indicated by the neutrophil count) and adaptive immunity (as indicated by the lymphocyte count). We aimed to estimate ability of NLR at admission and at day 1 for predicting stroke outcome after two reperfusion therapies: intravenous thrombolysis (IVT) and mechanical thrombectomy (MT). Methods A retrospective analysis was performed on patients who received recombinant human tissue plasminogen activator (IVT) and/or underwent MT for acute ischemic stroke (AIS) at the First Affiliated Hospital of Wenzhou Medical University (Wenzhou, China) from January 2018 to December 2020. Blood samples were taken on admission to hospital and on day 1 after stroke onset. Binary logistic regression models were applied to investigate potential associations between NLR at admission or day 1 and the following outcomes: symptomatic intracerebral hemorrhage (sICH), dependence, and mortality at 90 days. The ability of NLR to predict AIS outcome was analyzed using receiver operating characteristic (ROC) curves. Results Data for 927 patients (576 IVT and 351 MT) were reviewed. High admission NLR was associated with dependence in IVT treatment [adjusted odds ratio (OR) 1.21, 95% confidence interval (CI) 1.14–1.23] and 90-day mortality in MT patients (OR 1.09, 95% CI 1.04–1.13). In IVT patients, high NLR at day 1 predicted dependence (OR 1.09, 95% CI 1.02–1.11), sICH (OR = 1.07, 95% CI 1.01–1.12), and 90-day mortality (OR 1.06, 95% CI 1.01–1.15). In MT patients, high NLR at day 1 also predicted dependence (OR 1.08, 95% CI 1.02–1.11) and sICH (OR 1.03, 95% CI 1.01–1.09). ROC analysis confirmed that NLR at day 1 could predict dependence (cut-off 4.2; sensitivity 68.7%; specificity 79.6%), sICH (cut-off 5.1; sensitivity 57.9%, specificity 73.5%), and death (cut-off 5.4; sensitivity 78.8%; specificity 76.4%) in IVT patients. Z values of area under the curves were compared between admissioin and day 1 NLR in IVT patients and showed day 1 NLR can better predict dependence (Z = 2.8, p = 0.004) and 90-day death (Z = 2.8, p = 0.005). Conclusions NLR is a readily available biomarker that can predict AIS outcome after reperfusion treatment and day 1 NLR is even better than admission NLR.
Collapse
Affiliation(s)
- Siyan Chen
- Department of Neurology, Wenzhou Medical University Affiliated the First Hospital, Wenzhou, China
| | - Jianhua Cheng
- Department of Neurology, Wenzhou Medical University Affiliated the First Hospital, Wenzhou, China
| | - Qiang Ye
- Department of Neurology, Wenzhou Medical University Affiliated the First Hospital, Wenzhou, China
| | - Zusen Ye
- Department of Neurology, Wenzhou Medical University Affiliated the First Hospital, Wenzhou, China
| | - Yanlei Zhang
- Department of Neurology, Wenzhou Medical University Affiliated the First Hospital, Wenzhou, China
| | - Yuntao Liu
- Department of Neurology, Wenzhou Medical University Affiliated the First Hospital, Wenzhou, China
| | - Guiqian Huang
- Department of Neurology, Wenzhou Medical University Affiliated the First Hospital, Wenzhou, China
| | - Feichi Chen
- Department of Neurology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Ming Yang
- Department of Neurology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Chuanliu Wang
- Department of Neurology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Tingting Duan
- Department of Neurology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Xiang Liu
- Department of Neurology, Wencheng County People Hospital, Wenzhou, China
| | - Zheng Zhang
- Department of Neurology, Wenzhou Medical University Affiliated the First Hospital, Wenzhou, China
- Department of Neurology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
- *Correspondence: Zheng Zhang
| |
Collapse
|
11
|
Li R, Zhou Y, Zhang S, Li J, Zheng Y, Fan X. The natural (poly)phenols as modulators of microglia polarization via TLR4/NF-κB pathway exert anti-inflammatory activity in ischemic stroke. Eur J Pharmacol 2022; 914:174660. [PMID: 34863710 DOI: 10.1016/j.ejphar.2021.174660] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/04/2021] [Accepted: 11/29/2021] [Indexed: 12/24/2022]
Abstract
Increasing evidences suggest that inflammation plays a key role in the pathogenesis of stroke, a devastating disease second only to cardiac ischemia as a cause of death worldwide. Microglia are the first non-neuronal cells on the scene during the innate immune response to acute ischemic stroke. Microglia respond to acute brain injury by activating and developing classic M1-like (pro-inflammatory) or alternative M2-like (anti-inflammatory) phenotypes. M1 microglia produce pro-inflammatory cytokines to exacerbate neural death, astrocyte apoptosis, and blood brain barrier (BBB) disruption, while M2 microglia play the opposite role. NF-κB, a central regulator of the inflammatory response, was responsible for microglia M1 and M2 polarization. NF-κB p65 and p50 form a heterodimer to initiate a pro-inflammatory cytokine response, which enhances M1 activation and impair M2 response of microglia. TLR4, expressed on the surface of microglia, plays an important role in activating NF-κB, ultimately causing the M1 response of microglia. Therefore, modulation of microglial phenotypes via TLR4/NF-κB signaling pathway may be a promising therapeutic approach for ischemic stroke. Dietary (poly)phenols are present in various foods, which have shown promising protective effects on ischemic stroke. In vivo studies strongly suggest that many (poly)phenols have a pronounced impact on ischemic stroke, as demonstrated by lower neuroinflammation. Thus, this review focuses on the anti-inflammatory properties of dietary (poly)phenols and discusses their effects on the polarization of microglia through modulating TLR4/NF-κB signaling pathway in the ischemic stroke.
Collapse
Affiliation(s)
- Ruoqi Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yuan Zhou
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Shanshan Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jieying Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yingyi Zheng
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
12
|
He Q, Ma Y, Liu J, Zhang D, Ren J, Zhao R, Chang J, Guo ZN, Yang Y. Biological Functions and Regulatory Mechanisms of Hypoxia-Inducible Factor-1α in Ischemic Stroke. Front Immunol 2021; 12:801985. [PMID: 34966392 PMCID: PMC8710457 DOI: 10.3389/fimmu.2021.801985] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/26/2021] [Indexed: 12/15/2022] Open
Abstract
Ischemic stroke is caused by insufficient cerebrovascular blood and oxygen supply. It is a major contributor to death or disability worldwide and has become a heavy societal and clinical burden. To date, effective treatments for ischemic stroke are limited, and innovative therapeutic methods are urgently needed. Hypoxia inducible factor-1α (HIF-1α) is a sensitive regulator of oxygen homeostasis, and its expression is rapidly induced after hypoxia/ischemia. It plays an extensive role in the pathophysiology of stroke, including neuronal survival, neuroinflammation, angiogenesis, glucose metabolism, and blood brain barrier regulation. In addition, the spatiotemporal expression profile of HIF-1α in the brain shifts with the progression of ischemic stroke; this has led to contradictory findings regarding its function in previous studies. Therefore, unveiling the Janus face of HIF-1α and its target genes in different type of cells and exploring the role of HIF-1α in inflammatory responses after ischemia is of great importance for revealing the pathogenesis and identifying new therapeutic targets for ischemic stroke. Herein, we provide a succinct overview of the current approaches targeting HIF-1α and summarize novel findings concerning HIF-1α regulation in different types of cells within neurovascular units, including neurons, endothelial cells, astrocytes, and microglia, during the different stages of ischemic stroke. The current representative translational approaches focused on neuroprotection by targeting HIF-1α are also discussed.
Collapse
Affiliation(s)
- Qianyan He
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yinzhong Ma
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jie Liu
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Dianhui Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Jiaxin Ren
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Ruoyu Zhao
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - JunLei Chang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhen-Ni Guo
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yi Yang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Li Y, Ren M, Wang J, Ma R, Chen H, Xie Q, Li H, Li J, Wang J. Progress in Borneol Intervention for Ischemic Stroke: A Systematic Review. Front Pharmacol 2021; 12:606682. [PMID: 34017247 PMCID: PMC8129537 DOI: 10.3389/fphar.2021.606682] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 04/14/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Borneol is a terpene and bicyclic organic compound that can be extracted from plants or chemically synthesized. As an important component of proprietary Chinese medicine for the treatment of stroke, its neuroprotective effects have been confirmed in many experiments. Unfortunately, there is no systematic review of these studies. This study aimed to systematically examine the neuroprotective effects of borneol in the cascade reaction of experimental ischemic stroke at different periods. Methods: Articles on animal experiments and cell-based research on the actions of borneol against ischemic stroke in the past 20°years were collected from Google Scholar, Web of Science, PubMed, ScienceDirect, China National Knowledge Infrastructure (CNKI), and other biomedical databases. Meta-analysis was performed on key indicators in vivo experiments. After sorting the articles, we focused on the neuroprotective effects and mechanism of action of borneol at different stages of cerebral ischemia. Results: Borneol is effective in the prevention and treatment of nerve injury in ischemic stroke. Its mechanisms of action include improvement of cerebral blood flow, inhibition of neuronal excitotoxicity, blocking of Ca2+ overload, and resistance to reactive oxygen species injury in the acute ischemic stage. In the subacute ischemic stage, borneol may antagonize blood-brain barrier injury, intervene in inflammatory reactions, and prevent neuron excessive death. In the late stage, borneol promotes neurogenesis and angiogenesis in the treatment of ischemic stroke. Conclusion: Borneol prevents neuronal injury after cerebral ischemia via multiple action mechanisms, and it can mobilize endogenous nutritional factors to hasten repair and regeneration of brain tissue. Because the neuroprotective effects of borneol are mediated by various therapeutic factors, deficiency caused by a single-target drug is avoided. Besides, borneol promotes other drugs to pass through the blood-brain barrier to exert synergistic therapeutic effects.
Collapse
Affiliation(s)
- Yong Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mihong Ren
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiajun Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rong Ma
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hai Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Xie
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hongyan Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinxiu Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jian Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
14
|
Hou D, Wang C, Ye X, Zhong P, Wu D. Persistent inflammation worsens short-term outcomes in massive stroke patients. BMC Neurol 2021; 21:62. [PMID: 33568099 PMCID: PMC7874622 DOI: 10.1186/s12883-021-02097-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/31/2021] [Indexed: 11/10/2022] Open
Abstract
Background Persistent inflammation is an important driver of disease progression and affects prognosis. Some indicators of inflammation predict short-term outcomes. The relationship between prognosis, especially mortality, and persistent inflammation in massive stroke has not been studied, and this has been the subject of our research. Methods From April 1, 2017 to February 1, 2020, consecutive patients were prospectively enrolled. Clinical data, laboratory data, imaging data and follow-up infections morbidity were compared between 2 groups according to modified Rankin scale (mRS) scores (mRS < 3 and ≥ 3) at 1 month. The binomial logistic analysis was used to determine independent factors of 1-month prognosis. Short-term functional outcome, mortality and infection rates in massive stroke with and without persistent inflammation were compared. Results One hundred thirty-nine patients with massive stroke were included from 800 patients. We found that admission blood glucose levels (p = 0.005), proportions of cerebral hemispheric (p = 0.001), posterior circulatory (p = 0.035), and lacunar (p = 0.022) ischemia were higher in poor outcome patients; neutrophil-to-lymphocyte ratio (odd ratio = 1.87, 95%CI 1.14–3.07, p = 0.013) and blood glucose concentrations (odd ratio = 1.34, 95%CI 1.01–1.79, p = 0.043) can independently predict the short-term prognosis in massive stroke patients. We also found that the incidence of pulmonary infection (p = 0.009), one-month mortality (p = 0.003) and adverse outcomes (p = 0.0005) were higher in patients with persistent inflammation. Conclusions This study suggested that persistent inflammation is associated with poor prognosis, 1-month mortality and the occurrence of in-hospital pulmonary infection and that higher baseline inflammation level predicts short-term poor outcomes in massive stroke. Supplementary Information The online version contains supplementary material available at 10.1186/s12883-021-02097-9.
Collapse
Affiliation(s)
- Duanlu Hou
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Shanghai, 200240, China
| | - Chunjie Wang
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Shanghai, 200240, China.,Jiangchuan Community Health Service Center of Minhang District, Shanghai, China
| | - Xiaofei Ye
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Ping Zhong
- Department of Neurology, Shidong Hospital of Yangpu District, Shanghai, No.999, Shiguang Road, Shanghai, 200438, China.
| | - Danhong Wu
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Shanghai, 200240, China.
| |
Collapse
|
15
|
Mesencephalic Electrical Stimulation Reduces Neuroinflammation after Photothrombotic Stroke in Rats by Targeting the Cholinergic Anti-Inflammatory Pathway. Int J Mol Sci 2021; 22:ijms22031254. [PMID: 33514001 PMCID: PMC7865599 DOI: 10.3390/ijms22031254] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 01/22/2021] [Indexed: 11/25/2022] Open
Abstract
Inflammation is crucial in the pathophysiology of stroke and thus a promising therapeutic target. High-frequency stimulation (HFS) of the mesencephalic locomotor region (MLR) reduces perilesional inflammation after photothrombotic stroke (PTS). However, the underlying mechanism is not completely understood. Since distinct neural and immune cells respond to electrical stimulation by releasing acetylcholine, we hypothesize that HFS might trigger the cholinergic anti-inflammatory pathway via activation of the α7 nicotinic acetylcholine receptor (α7nAchR). To test this hypothesis, rats underwent PTS and implantation of a microelectrode into the MLR. Three hours after intervention, either HFS or sham-stimulation of the MLR was applied for 24 h. IFN-γ, TNF-α, and IL-1α were quantified by cytometric bead array. Choline acetyltransferase (ChAT)+ CD4+-cells and α7nAchR+-cells were quantified visually using immunohistochemistry. Phosphorylation of NFĸB, ERK1/2, Akt, and Stat3 was determined by Western blot analyses. IFN-γ, TNF-α, and IL-1α were decreased in the perilesional area of stimulated rats compared to controls. The number of ChAT+ CD4+-cells increased after MLR-HFS, whereas the amount of α7nAchR+-cells was similar in both groups. Phospho-ERK1/2 was reduced significantly in stimulated rats. The present study suggests that MLR-HFS may trigger anti-inflammatory processes within the perilesional area by modulating the cholinergic system, probably via activation of the α7nAchR.
Collapse
|
16
|
Nalamolu KR, Challa SR, Fornal CA, Grudzien NA, Jorgenson LC, Choudry MM, Smith NJ, Palmer CJ, Pinson DM, Klopfenstein JD, Veeravalli KK. Attenuation of the Induction of TLRs 2 and 4 Mitigates Inflammation and Promotes Neurological Recovery After Focal Cerebral Ischemia. Transl Stroke Res 2021; 12:923-936. [PMID: 33426628 DOI: 10.1007/s12975-020-00884-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/16/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022]
Abstract
The intense inflammatory response triggered in the brain after focal cerebral ischemia is detrimental. Recently, we showed that the suppression of toll-like receptors (TLRs) 2 and 4 attenuates infarct size and reduces the expression of pro-inflammatory cytokines in the ischemic brain. In this study, we further examined the effect of unsuppressed induction of TLRs 2 and 4 on the expression of its downstream signaling molecules and pro-inflammatory cytokines 1 week after reperfusion. The primary purpose of this study was to investigate the effect of simultaneous knockdown of TLRs 2 and 4 on M1/M2 microglial polarization dynamics and post-stroke neurological deficits and the recovery. Transient focal cerebral ischemia was induced in young adult male Sprague-Dawley rats by the middle cerebral artery occlusion (MCAO) procedure using a monofilament suture. Appropriate cohorts of rats were treated with a nanoparticle formulation of TLR2shRNA and TLR4shRNA (T2sh+T4sh) expressing plasmids (1 mg/kg each of T2sh and T4sh) or scrambled sequence inserted vector (vehicle control) expressing plasmids (2 mg/kg) intravenously via tail vein immediately after reperfusion. Animals from various cohorts were euthanized during reperfusion, and the ischemic brain tissue was isolated and utilized for PCR followed by agarose gel electrophoresis, real-time PCR, immunoblot, and immunofluorescence analysis. Appropriate groups were subjected to a battery of standard neurological tests at regular intervals until 14 days after reperfusion. The increased expression of both TLRs 2 and 4 and their downstream signaling molecules including the pro-inflammatory cytokines was observed even at 1-week after reperfusion. T2sh+T4sh treatment immediately after reperfusion attenuated the post-ischemic inflammation, preserved the motor function, and promoted recovery of the sensory and motor functions. We conclude that the post-ischemic induction of TLRs 2 and 4 persists for at least 7 days after reperfusion, contributes to the severity of acute inflammation, and impedes neurological recovery. Unlike previous studies in TLRs 2 or 4 knockout models, results of this study in a pharmacologically relevant preclinical rodent stroke model have translational significance.
Collapse
Affiliation(s)
- Koteswara Rao Nalamolu
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, 1 Illini Dr, Peoria, IL, 61605, USA
- Department of Pharmaceutical and Biomedical Sciences, California Health Sciences University, Clovis, CA, USA
| | - Siva Reddy Challa
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, 1 Illini Dr, Peoria, IL, 61605, USA
| | - Casimir A Fornal
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, 1 Illini Dr, Peoria, IL, 61605, USA
| | - Natalia A Grudzien
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, 1 Illini Dr, Peoria, IL, 61605, USA
| | - Laura C Jorgenson
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, 1 Illini Dr, Peoria, IL, 61605, USA
| | - Mouneeb M Choudry
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, 1 Illini Dr, Peoria, IL, 61605, USA
| | - Nathan J Smith
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Cassandra J Palmer
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, 1 Illini Dr, Peoria, IL, 61605, USA
| | - David M Pinson
- Department of Health Sciences Education, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Jeffrey D Klopfenstein
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, 1 Illini Dr, Peoria, IL, 61605, USA
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Krishna Kumar Veeravalli
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, 1 Illini Dr, Peoria, IL, 61605, USA.
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL, USA.
- Department of Neurology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA.
| |
Collapse
|
17
|
Xue W, Duan X, Hao Y, Liang X, Qiu G. Eriocitrin alleviates the arterial occlusion-mediated cerebral ischemic-reperfusion injury through the modulation of apoptotic proteins and immune markers in mice. Pharmacogn Mag 2021. [DOI: 10.4103/pm.pm_577_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
18
|
Gou X, Ying J, Yue Y, Qiu X, Hu P, Qu Y, Li J, Mu D. The Roles of High Mobility Group Box 1 in Cerebral Ischemic Injury. Front Cell Neurosci 2020; 14:600280. [PMID: 33384585 PMCID: PMC7770223 DOI: 10.3389/fncel.2020.600280] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 11/25/2020] [Indexed: 12/11/2022] Open
Abstract
High mobility group box 1 (HMGB1) is a ubiquitous nuclear protein that plays an important role in stabilizing nucleosomes and DNA repair. HMGB1 can be passively released from necrotic neurons or actively secreted by microglia, macrophages/monocytes, and neutrophils. Cerebral ischemia is a major cause of mortality and disability worldwide, and its outcome depends on the number of neurons dying due to hypoxia in the ischemic area. HMGB1 contributes to the pathogenesis of cerebral ischemia via mediating neuroinflammatory responses to cerebral ischemic injury. Extracellular HMGB1 regulates many neuroinflammatory events by interacting with its different cell surface receptors, such as receptors for advanced glycation end products, toll-like receptor (TLR)-2, and TLR-4. Additionally, HMGB1 can be redox-modified, thus exerting specific cellular functions in the ischemic brain and has different roles in the acute and late stages of cerebral ischemic injury. However, the role of HMGB1 in cerebral ischemia is complex and remains unclear. Herein, we summarize and review the research on HMGB1 in cerebral ischemia, focusing especially on the role of HMGB1 in hypoxic ischemia in the immature brain and in white matter ischemic injury. We also outline the possible mechanisms of HMGB1 in cerebral ischemia and the main strategies to inhibit HMGB1 pertaining to its potential as a novel critical molecular target in cerebral ischemic injury.
Collapse
Affiliation(s)
- Xiaoyun Gou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Junjie Ying
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Yan Yue
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Xia Qiu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Peng Hu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Jinhui Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Zhang YM, Qu XY, Tao LN, Zhai JH, Gao H, Song YQ, Zhang SX. XingNaoJing injection ameliorates cerebral ischaemia/reperfusion injury via SIRT1-mediated inflammatory response inhibition. PHARMACEUTICAL BIOLOGY 2020; 58:16-24. [PMID: 31854225 PMCID: PMC6968491 DOI: 10.1080/13880209.2019.1698619] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Context: XingNaoJing injection (XNJ), extracted from a traditional compound Chinese medicine Angong niuhuang pill, is well known for treating stroke in the clinic, but the specific effects and mechanisms remain unclear.Objective: We investigated the mechanistic basis for the protective effect of XNJ on cerebral ischaemia/reperfusion (I/R) injury.Materials and methods: Five groups of 10 SD rats underwent 2 h of middle cerebral artery occlusion (MCAO) followed by 24 h reperfusion. XNJ at 10 and 15 mL/kg was intraperitoneally administered 24 h before ischaemia and at the onset of reperfusion respectively. The silent information regulator 1 (SIRT1) inhibitor EX527 was intracerebroventricularly injected 0.5 h before reperfusion. Cerebral infarction size, neurological scores, morphological changes, and expression levels of inflammatory mediators and SIRT1 were measured. Furthermore, human brain microvascular endothelial cells (HBMECs) were subjected to 3 h oxygen and glucose deprivation (OGD) followed by 24 h reoxygenation to mimic cerebral I/R in vitro. EX527 pre-treatment occurred 1 h before OGD. SIRT1 and inflammatory mediator levels were analyzed.Results: Both XNJ doses significantly decreased cerebral infarct area (40.11% vs. 19.66% and 9.87%) and improved neurological scores and morphological changes. Inflammatory mediator levels were remarkably decreased in both model systems after XNJ treatment. XNJ also enhanced SIRT1 expression. Notably, the SIRT1 inhibitor EX527 attenuated the XNJ-mediated decrease in inflammation in vivo and in vitro.Conclusions: XNJ improved cerebral I/R injury through inhibiting the inflammatory response via the SIRT1 pathway, which may be a useful target in treating cerebral I/R injury.
Collapse
Affiliation(s)
- Yue-Ming Zhang
- Department of Pharmacy, the First Hospital of Jilin University, Changchun, China
| | - Xiao-Yu Qu
- Department of Pharmacy, the First Hospital of Jilin University, Changchun, China
| | - Li-Na Tao
- Department of Pharmacy, the First Hospital of Jilin University, Changchun, China
| | - Jing-Hui Zhai
- Department of Pharmacy, the First Hospital of Jilin University, Changchun, China
| | - Huan Gao
- Department of Pharmacy, the First Hospital of Jilin University, Changchun, China
| | - Yan-Qing Song
- Department of Pharmacy, the First Hospital of Jilin University, Changchun, China
- CONTACT Yan-Qing Song
| | - Si-Xi Zhang
- Department of Pharmacy, the First Hospital of Jilin University, Changchun, China
- Si-Xi Zhang Department of Pharmacy, the First Hospital of Jilin University, 71# Xinmin Street, Changchun130021, Jilin Province, PR China
| |
Collapse
|
20
|
Famakin BM, Vemuganti R. Toll-Like Receptor 4 Signaling in Focal Cerebral Ischemia: a Focus on the Neurovascular Unit. Mol Neurobiol 2020; 57:2690-2701. [PMID: 32306272 DOI: 10.1007/s12035-020-01906-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/19/2020] [Indexed: 12/14/2022]
Abstract
A robust innate immune activation leads to downstream expression of inflammatory mediators that amplify tissue damage and consequently increase the morbidity after stroke. The Toll-like receptor 4 (TLR4) pathway is a major innate immune pathway activated acutely and chronically after stroke. Hence, understanding the intricacies of the temporal profile, specific control points, and cellular specificity of TLR4 activation is crucial for the development of any novel therapeutics targeting the endogenous innate immune response after focal cerebral ischemia. The goal of this review is to summarize the current findings related to TLR4 signaling after stroke with a specific focus on the components of the neurovascular unit such as astrocytes, neurons, endothelial cells, and pericytes. In addition, this review will examine the effects of focal cerebral ischemia on interaction of these neurovascular unit components.
Collapse
Affiliation(s)
| | - R Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- William S. Middleton VA Hospital, Madison, WI, USA
| |
Collapse
|
21
|
Jayaraj RL, Azimullah S, Beiram R, Jalal FY, Rosenberg GA. Neuroinflammation: friend and foe for ischemic stroke. J Neuroinflammation 2019; 16:142. [PMID: 31291966 PMCID: PMC6617684 DOI: 10.1186/s12974-019-1516-2] [Citation(s) in RCA: 909] [Impact Index Per Article: 151.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/10/2019] [Indexed: 12/13/2022] Open
Abstract
Stroke, the third leading cause of death and disability worldwide, is undergoing a change in perspective with the emergence of new ideas on neurodegeneration. The concept that stroke is a disorder solely of blood vessels has been expanded to include the effects of a detrimental interaction between glia, neurons, vascular cells, and matrix components, which is collectively referred to as the neurovascular unit. Following the acute stroke, the majority of which are ischemic, there is secondary neuroinflammation that both promotes further injury, resulting in cell death, but conversely plays a beneficial role, by promoting recovery. The proinflammatory signals from immune mediators rapidly activate resident cells and influence infiltration of a wide range of inflammatory cells (neutrophils, monocytes/macrophages, different subtypes of T cells, and other inflammatory cells) into the ischemic region exacerbating brain damage. In this review, we discuss how neuroinflammation has both beneficial as well as detrimental roles and recent therapeutic strategies to combat pathological responses. Here, we also focus on time-dependent entry of immune cells to the ischemic area and the impact of other pathological mediators, including oxidative stress, excitotoxicity, matrix metalloproteinases (MMPs), high-mobility group box 1 (HMGB1), arachidonic acid metabolites, mitogen-activated protein kinase (MAPK), and post-translational modifications that could potentially perpetuate ischemic brain damage after the acute injury. Understanding the time-dependent role of inflammatory factors could help in developing new diagnostic, prognostic, and therapeutic neuroprotective strategies for post-stroke inflammation.
Collapse
Affiliation(s)
- Richard L. Jayaraj
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Sheikh Azimullah
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Rami Beiram
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Fakhreya Y. Jalal
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Gary A. Rosenberg
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131 USA
| |
Collapse
|
22
|
Parada E, Casas AI, Palomino-Antolin A, Gómez-Rangel V, Rubio-Navarro A, Farré-Alins V, Narros-Fernandez P, Guerrero-Hue M, Moreno JA, Rosa JM, Roda JM, Hernández-García BJ, Egea J. Early toll-like receptor 4 blockade reduces ROS and inflammation triggered by microglial pro-inflammatory phenotype in rodent and human brain ischaemia models. Br J Pharmacol 2019; 176:2764-2779. [PMID: 31074003 DOI: 10.1111/bph.14703] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 04/09/2019] [Accepted: 04/12/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Ischaemic stroke is a leading cause of death, disability, and a high unmet medical need. Post-reperfusion inflammation and an up-regulation of toll-like receptor 4 (TLR4), an upstream sensor of innate immunity, are associated with poor outcome in stroke patients. Here, we identified the therapeutic effect of targeting the LPS/TLR4 signal transduction pathway. EXPERIMENTAL APPROACH We tested the effect of the TLR4 inhibitor, eritoran (E5564) in different in vitro ischaemia-related models: human organotypic cortex culture, rat organotypic hippocampal cultures, and primary mixed glia cultures. We explored the therapeutic window of E5564 in the transient middle cerebral artery occlusion model of cerebral ischaemia in mice. KEY RESULTS In vivo, administration of E5564 1 and 4 hr post-ischaemia reduced the expression of different pro-inflammatory chemokines and cytokines, infarct volume, blood-brain barrier breakdown, and improved neuromotor function, an important clinically relevant outcome. In the human organotypic cortex culture, E5564 reduced the activation of microglia and ROS production evoked by LPS. CONCLUSION AND IMPLICATIONS TLR4 signalling has a causal role in the inflammation associated with a poor post-stroke outcome. Importantly, its inhibition by eritoran (E5564) provides neuroprotection both in vitro and in vivo, including in human tissue, suggesting a promising new therapeutic approach for ischaemic stroke.
Collapse
Affiliation(s)
- Esther Parada
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, Madrid, Spain.,Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, Madrid, Spain
| | - Ana I Casas
- Department of Pharmacology and Personalised Medicine, CARIM, Maastricht University, Maastricht, The Netherlands
| | - Alejandra Palomino-Antolin
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, Madrid, Spain.,Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, Madrid, Spain
| | - Vanessa Gómez-Rangel
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, Madrid, Spain.,Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, Madrid, Spain
| | - Alfonso Rubio-Navarro
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Victor Farré-Alins
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, Madrid, Spain.,Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, Madrid, Spain
| | - Paloma Narros-Fernandez
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, Madrid, Spain.,Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, Madrid, Spain
| | - Melania Guerrero-Hue
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan Antonio Moreno
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Juliana M Rosa
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, Madrid, Spain.,Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, Madrid, Spain
| | - José M Roda
- Servicio de Neurocirugía, Hospital Universitario La Paz, Madrid, Spain
| | | | - Javier Egea
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, Madrid, Spain.,Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, Madrid, Spain
| |
Collapse
|
23
|
Lambertsen KL, Finsen B, Clausen BH. Post-stroke inflammation-target or tool for therapy? Acta Neuropathol 2019; 137:693-714. [PMID: 30483945 PMCID: PMC6482288 DOI: 10.1007/s00401-018-1930-z] [Citation(s) in RCA: 304] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 11/03/2018] [Accepted: 11/04/2018] [Indexed: 12/22/2022]
Abstract
Inflammation is currently considered a prime target for the development of new stroke therapies. In the acute phase of ischemic stroke, microglia are activated and then circulating immune cells invade the peri-infarct and infarct core. Resident and infiltrating cells together orchestrate the post-stroke inflammatory response, communicating with each other and the ischemic neurons, through soluble and membrane-bound signaling molecules, including cytokines. Inflammation can be both detrimental and beneficial at particular stages after a stroke. While it can contribute to expansion of the infarct, it is also responsible for infarct resolution, and influences remodeling and repair. Several pre-clinical and clinical proof-of-concept studies have suggested the effectiveness of pharmacological interventions that target inflammation post-stroke. Experimental evidence shows that targeting certain inflammatory cytokines, such as tumor necrosis factor, interleukin (IL)-1, IL-6, and IL-10, holds promise. However, as these cytokines possess non-redundant protective and immunoregulatory functions, their neutralization or augmentation carries a risk of unwanted side effects, and clinical translation is, therefore, challenging. This review summarizes the cell biology of the post-stroke inflammatory response and discusses pharmacological interventions targeting inflammation in the acute phase after a stroke that may be used alone or in combination with recanalization therapies. Development of next-generation immune therapies should ideally aim at selectively neutralizing pathogenic immune signaling, enhancing tissue preservation, promoting neurological recovery and leaving normal function intact.
Collapse
Affiliation(s)
- Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense, Denmark.
- Department of Clinical Research, BRIDGE-Brain Research-Inter-Disciplinary Guided Excellence, University of Southern Denmark, 5000, Odense C, Denmark.
- Department of Neurology, Odense University Hospital, 5000, Odense, Denmark.
| | - Bente Finsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense, Denmark
- Department of Clinical Research, BRIDGE-Brain Research-Inter-Disciplinary Guided Excellence, University of Southern Denmark, 5000, Odense C, Denmark
| | - Bettina Hjelm Clausen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense, Denmark
- Department of Clinical Research, BRIDGE-Brain Research-Inter-Disciplinary Guided Excellence, University of Southern Denmark, 5000, Odense C, Denmark
| |
Collapse
|
24
|
Lei JR, Tu XK, Wang Y, Tu DW, Shi SS. Resveratrol downregulates the TLR4 signaling pathway to reduce brain damage in a rat model of focal cerebral ischemia. Exp Ther Med 2019; 17:3215-3221. [PMID: 30936996 DOI: 10.3892/etm.2019.7324] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 11/23/2018] [Indexed: 12/21/2022] Open
Abstract
Previous studies have demonstrated that inflammation and disruption of the blood-brain barrier (BBB) are important pathological processes during focal cerebral ischemia. Therefore, the present study evaluated the neuroprotective effects of resveratrol against brain damage, inflammation and BBB disruption in rats with focal cerebral ischemia and assessed the potential underlying molecular mechanisms. Sprague-Dawley rats underwent cerebral ischemia/reperfusion (IR) and then received intraperitoneal resveratrol (10 and 100 mg/kg) 2 h following the onset of ischemia. Following 24 h of ischemia, neurological deficit scores, cerebral infarctions, morphological characteristics, cerebral water content, myeloperoxidase (MPO) activity and Evans blue extravasation were assessed. Additionally, the protein expression levels of Toll-like receptor 4 (TLR4) and nuclear factor (NF)-κB p65 were detected using western blot analyses, the mRNA expression levels of cyclooxygenase-2 (COX-2) and matrix metalloproteinase-9 (MMP-9) were examined by reverse-transcription polymerase chain reaction, and tumor necrosis factor (TNF)-α and interleukin (IL)-1β blood levels were determined by ELISA. Resveratrol significantly reduced neurological deficit scores, cerebral infarct sizes, neuronal injury, MPO activity and EB content. Cerebral ischemia increased the expression levels of TLR4, NF-κB p65, COX-2, MMP-9, TNF-α and IL-1β, but all of these factors were reduced by resveratrol. In conclusion, the present data suggest that resveratrol reduces inflammation, BBB disruption and brain damage in rats following focal cerebral ischemia. Additionally, the neuroprotective effects of resveratrol against cerebral ischemia may be associated with downregulation of the TLR4 pathway.
Collapse
Affiliation(s)
- Jun-Rong Lei
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China.,Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Xian-Kun Tu
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Yang Wang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - De-Wen Tu
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Song-Sheng Shi
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
25
|
Yang J, Gu L, Guo X, Huang J, Chen Z, Huang G, Kang Y, Zhang X, Long J, Su L. LncRNA ANRIL Expression and ANRIL Gene Polymorphisms Contribute to the Risk of Ischemic Stroke in the Chinese Han Population. Cell Mol Neurobiol 2018; 38:1253-1269. [PMID: 29881905 PMCID: PMC11481959 DOI: 10.1007/s10571-018-0593-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 05/23/2018] [Indexed: 12/13/2022]
Abstract
The aim of the present study was to explore the role of lncRNA ANRIL in the pathogenesis of ischemic stroke (IS) and coronary artery disease (CAD) and to determine the association between ANRIL variants and the genetic susceptibility of IS and CAD in the Chinese Han population. A genetic association study including 550 IS patients, 550 CAD patients, and 550 healthy controls was conducted. The expression levels of lncRNA ANRIL, CDKN2A, and CDKN2B were detected using qRT-PCR. Genotyping was performed by Sequenom MassARRAY on an Agena platform. Our study showed that IS patients had an increased lncRNA ANRIL expression (P = 0.002) and a decreased CDKN2A expression (P < 0.001) compared with normal controls. A significant difference with regard to the genotype distribution of rs2383207 was found between male IS patients and controls (P = 0.011). The minor allele of rs2383207 significantly increased the IS risk under a recessive model (OR = 1.52, 95% CI = 1.05-2.21, P = 0.027). The minor allele of rs1333049 was significantly associated with the risk of IS among the male patients under a recessive model (OR = 1.56, 95% CI = 1.04-2.35, P = 0.031). However, no significant association was found between the ANRIL variants and the risk of CAD (all P > 0.050). In addition, we found a decreased lncRNA ANRIL expression in IS patients who carried the GG genotype of rs1333049 compared with IS patients who carried the CC or CG genotype (P = 0.041). In summary, we found that IS patients had an increased lncRNA ANRIL expression and a decreased CDKN2A expression compared with the controls, which might play an impellent role in pathological processes of IS. The ANRIL variants rs2383207 and rs1333049 were significantly associated with the risk of IS among males but not females in the Chinese Han population.
Collapse
Affiliation(s)
- Jialei Yang
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Lian Gu
- First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, People's Republic of China
| | - Xiaojing Guo
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Jiao Huang
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Zhaoxia Chen
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Guifeng Huang
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Yiwen Kang
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Xiaoting Zhang
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Jianxiong Long
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
| | - Li Su
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
| |
Collapse
|
26
|
Xu H, Qin W, Hu X, Mu S, Zhu J, Lu W, Luo Y. Lentivirus-mediated overexpression of OTULIN ameliorates microglia activation and neuroinflammation by depressing the activation of the NF-κB signaling pathway in cerebral ischemia/reperfusion rats. J Neuroinflammation 2018; 15:83. [PMID: 29544517 PMCID: PMC5856386 DOI: 10.1186/s12974-018-1117-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 03/06/2018] [Indexed: 01/05/2023] Open
Abstract
Background Ischemic stroke-induced neuroinflammation is mainly mediated by microglial cells. The nuclear factor kappa B (NF-κB) pathway is the key transcriptional pathway that initiates inflammatory responses following cerebral ischemia. OTULIN, a critical negative regulator of the NF-κΒ signaling pathway, exerts robust effects on peripheral immune cell-mediated inflammation and is regarded as an essential mediator for repressing inflammation in vivo. The effect of OTULIN on inflammatory responses in the central nervous system (CNS) was previously unstudied. This current study investigated the anti-inflammatory effect of OTULIN both in vitro and in vivo in ischemic stroke models. Methods Sprague-Dawley (SD) rats were subjected to transient middle cerebral artery occlusion (tMCAO) or an intraperitoneal injection of lipopolysaccharide (LPS). Overexpression of the OTULIN gene was utilized to observe the effect of OTULIN on ischemic stroke outcomes. The effect of OTULIN overexpression on microglia-mediated neuroinflammation was examined in rat primary microglia (PM) and in the microglial cell line N9 after induction by oxygen-glucose deprivation (OGD)-treated neuronal medium. The activation and inflammatory responses of microglia were detected using immunofluorescence, ELISA, and qRT-PCR. The details of molecular mechanism were assessed using Western blotting. Results In the tMCAO rats, the focal cerebral ischemia/reperfusion injury induced a continuous increase in OTULIN expression within 72 h, and OTULIN expression was increased in activated microglial cells. OTULIN overexpression obviously decreased the cerebral infarct volume, improved the neurological function deficits, and reduced neuronal loss at 72 h after reperfusion, and it also inhibited the activation of microglia and attenuated the release of TNF-α, IL-1β, and IL-6 by suppressing the NF-κB pathway at 24 h after tMCAO. In vitro, OTULIN overexpression inhibited the microglia-mediated neuroinflammation by reducing the production of TNF-α, IL-1β, and IL-6 via depressing the NF-κB pathway in both PM and N9 cells. Conclusions OTULIN provides a potential therapeutic target for ischemic brain injury by ameliorating the excessive activation of microglial cells and neuroinflammation through repressing the NF-κB signaling pathway. Electronic supplementary material The online version of this article (10.1186/s12974-018-1117-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hongbei Xu
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wenyi Qin
- Department of Integrated Chinese and Western Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiao Hu
- Department of Neurology, Guizhou Provincial People's hospital, Guizhou, 50002, China
| | - Song Mu
- Department of Anus & Intestine surgery, the Affiliated Hospital of Guizhou Medical University, Guizhou, 550004, China
| | - Jun Zhu
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wenhao Lu
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yong Luo
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China. .,Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
27
|
Nalamolu KR, Smith NJ, Chelluboina B, Klopfenstein JD, Pinson DM, Wang DZ, Vemuganti R, Veeravalli KK. Prevention of the Severity of Post-ischemic Inflammation and Brain Damage by Simultaneous Knockdown of Toll-like Receptors 2 and 4. Neuroscience 2018; 373:82-91. [PMID: 29337240 DOI: 10.1016/j.neuroscience.2018.01.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/06/2017] [Accepted: 01/04/2018] [Indexed: 10/18/2022]
Abstract
Toll-like receptor 2 (TLR2) and TLR4 belong to a family of highly conserved pattern recognition receptors and are well-known upstream sensors of signaling pathways of innate immunity. TLR2 and TLR4 upregulation is thought to be associated with poor outcome in stroke patients. We currently show that transient focal ischemia in adult rats induces TLR2 and TLR4 expression within hours and shRNA-mediated knockdown of TLR2 and TLR4 alone and in combination decreases the infarct size and swelling. We further show that TLR2 and TLR4 knockdown also prevented the induction of their downstream signaling molecules MyD88, IRAK1, and NFκB p65 as well as the pro-inflammatory cytokines IL-1β, IL-6, and TNFα. This study thus shows that attenuation of the severity of TLR2- and TLR4-mediated post-stroke inflammation ameliorates ischemic brain damage.
Collapse
Affiliation(s)
- Koteswara Rao Nalamolu
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA
| | - Nathan J Smith
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA
| | - Bharath Chelluboina
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA
| | - Jeffrey D Klopfenstein
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA; Department of Neurosurgery, University of Illinois College of Medicine, Peoria, IL, USA; Comprehensive Stroke Center, Illinois Neurological Institute, OSF HealthCare System, Saint Francis Medical Center, Peoria, IL, USA
| | - David M Pinson
- Department of Pathology, University of Illinois College of Medicine, Peoria, IL, USA
| | - David Z Wang
- Department of Neurology, University of Illinois College of Medicine, Peoria, IL, USA; Comprehensive Stroke Center, Illinois Neurological Institute, OSF HealthCare System, Saint Francis Medical Center, Peoria, IL, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, School of Medicine and Public Health University of Wisconsin, Madison, WI, USA; William S. Middleton VA Hospital, Madison, WI, USA
| | - Krishna Kumar Veeravalli
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA; Department of Neurosurgery, University of Illinois College of Medicine, Peoria, IL, USA; Department of Neurology, University of Illinois College of Medicine, Peoria, IL, USA.
| |
Collapse
|
28
|
Allen RS, Sayeed I, Oumarbaeva Y, Morrison KC, Choi PH, Pardue MT, Stein DG. Progesterone treatment shows greater protection in brain vs. retina in a rat model of middle cerebral artery occlusion: Progesterone receptor levels may play an important role. Restor Neurol Neurosci 2018; 34:947-963. [PMID: 27802245 DOI: 10.3233/rnn-160672] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND/OBJECTIVE To determine whether inflammation increases in retina as it does in brain following middle cerebral artery occlusion (MCAO), and whether the neurosteroid progesterone, shown to have protective effects in both retina and brain after MCAO, reduces inflammation in retina as well as brain. METHODS MCAO rats treated systemically with progesterone or vehicle were compared with shams. Protein levels of cytosolic NF-κB, nuclear NF-κB, phosphorylated NF-κB, IL-6, TNF-α, CD11b, progesterone receptor A and B, and pregnane X receptor were assessed in retinas and brains at 24 and 48 h using western blots. RESULTS Following MCAO, significant increases were observed in the following inflammatory markers: pNF-κB and CD11b at 24 h in both brain and retina, nuclear NF-κB at 24 h in brain and 48 h in retina, and TNF-α at 24 h in brain.Progesterone treatment in MCAO animals significantly attenuated levels of the following markers in brain: pNF-κB, nuclear NF-κB, IL-6, TNF-α, and CD11b, with significantly increased levels of cytosolic NF-κB. Retinas from progesterone-treated animals showed significantly reduced levels of nuclear NF-κB and IL-6 and increased levels of cytosolic NF-κB, with a trend for reduction in other markers. Post-MCAO, progesterone receptors A and B were upregulated in brain and downregulated in retina. CONCLUSION Inflammatory markers increased in both brain and retina after MCAO, with greater increases observed in brain. Progesterone treatment reduced inflammation, with more dramatic reductions observed in brain than retina. This differential effect may be due to differences in the response of progesterone receptors in brain and retina after injury.
Collapse
Affiliation(s)
- Rachael S Allen
- Department of Emergency Medicine, Emory University, Atlanta, GA, USA.,Department of Ophthalmology, Emory University, Atlanta, GA, USA.,Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, USA
| | - Iqbal Sayeed
- Department of Emergency Medicine, Emory University, Atlanta, GA, USA
| | - Yuliya Oumarbaeva
- Department of Emergency Medicine, Emory University, Atlanta, GA, USA
| | | | - Paul H Choi
- Department of Emergency Medicine, Emory University, Atlanta, GA, USA
| | - Machelle T Pardue
- Department of Ophthalmology, Emory University, Atlanta, GA, USA.,Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, USA
| | - Donald G Stein
- Department of Emergency Medicine, Emory University, Atlanta, GA, USA
| |
Collapse
|
29
|
Protective Role of Apocynin via Suppression of Neuronal Autophagy and TLR4/NF-κB Signaling Pathway in a Rat Model of Traumatic Brain Injury. Neurochem Res 2017; 42:3296-3309. [DOI: 10.1007/s11064-017-2372-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 06/14/2017] [Accepted: 08/01/2017] [Indexed: 10/19/2022]
|
30
|
Mechanism of Lycium barbarum polysaccharides on primary cultured rat hippocampal neurons. Cell Tissue Res 2017; 369:455-465. [DOI: 10.1007/s00441-017-2648-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 04/25/2017] [Indexed: 01/27/2023]
|
31
|
Nobiletin improves propofol-induced neuroprotection via regulating Akt/mTOR and TLR 4/NF-κB signaling in ischemic brain injury in rats. Biomed Pharmacother 2017; 91:494-503. [PMID: 28478273 DOI: 10.1016/j.biopha.2017.04.048] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 04/09/2017] [Accepted: 04/10/2017] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Stroke is regarded as one of the main health concerns globally, presenting with high mortality and morbidity rates. Cerebral ischemic damage and infarction are critically associated with stroke. Various mechanisms related to inflammation, oxidative stress and excitotoxicity are found to be involved in ischemic damage. Very short time period for treatment has necessitated in development of more effective neuroprotective agents. Study aimed in investigated the effects of nobiletin on experimentally induced ischemic brain injury and also to assess whether nobiletin potentiated the neuroprotective effects of propofol. METHODS Male Sprague-Dawley rats were subjected to ischemia/reperfusion (I/R) injury. Induction of cerebral infarction and I/R was done by middle cerebral artery occlusion (MCAO). Nobiletin (100 or 200mg/kg b.wt.) was intragastrically administered to rats for 9 days before ischemia induction and on the day of induction nobiletin was administered an hour prior. Separate group of rats were post-conditioned with propofol (50mg/kg/h; i.v.) for 30min following 24h of reperfusion. RESULTS Propofol post-conditioning either with or without administration of nobiletin prior I/R injury attenuated pulmonary edema, neuronal apoptosis and reduced cerebral infarct volume. Overproduction of pro-inflammatory cytokines (TNF-α, IL-1β and IL-6) and nitric oxide following I/R were reduced. Propofol either alone or with prior nobiletin treatment had down-regulated TLR4 and TLR4-mediated NF-κB signaling and caused activation of Akt/mTOR cascade. CONCLUSION Propofol post-conditioning either with nobiletin prior I/R injury was found to be more effective than propofol alone, suggesting the positive effects of nobiletin on propofol-mediated anti-inflammatory and neuroprotective effects.
Collapse
|
32
|
Shanshan Y, Beibei J, Li T, Minna G, Shipeng L, Li P, Yong Z. Phospholipase A2 of Peroxiredoxin 6 Plays a Critical Role in Cerebral Ischemia/Reperfusion Inflammatory Injury. Front Cell Neurosci 2017; 11:99. [PMID: 28424593 PMCID: PMC5380807 DOI: 10.3389/fncel.2017.00099] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 03/23/2017] [Indexed: 12/14/2022] Open
Abstract
Microglia-mediated inflammation is an important step in the progression of cerebral ischemia/reperfusion injury and the associated production of receptors of immunomoudulation, including Toll-like receptors (TLRs). Peroxiredoxin 6 (Prdx6) has been demonstrated as the endogenous antioxidant protein for its peroxidase properties. However, the role of the independent phospholipase A2 (iPLA2) activity of Prdx6 in stroke has not been well studied. In this study, we evaluated whether blocking the calcium-iPLA2 activity of Prdx6 using siRNA and inhibitors (1-hexadecyl-3-(trifluoroethgl)-sn-glycerol-2 phosphomethanol, MJ33) would have a critical effect on inflammatory brain damage. We conducted oxygen-glucose deprivation (OGD)/recovery (R) in vitro and middle cerebral artery occlusion (MCAO) in vivo in a microglia/neuron co-culture system and in rats. In vitro, we found that Prdx6-iPLA2 activity was associated with the secretion of neurotoxic inflammatory mediators interleukin1β (IL-1β), interleukin-17 (IL-17) and interleukin-23 (IL-23) and elevated expression of Toll-like receptor 2/4 (TLR2/4), leading to the formation of nuclear factor-kappa B (NF-κB), inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in microglial cells. In vivo, combined treatment with Prdx6-iPLA2 activity inhibitor MJ33 showed a greater diminution in neurologic deficits, cerebral infarction, brain water content and inflammatory molecules than Prdx6-siRNA treatment alone. Our findings provide new insight into Prdx6-iPLA2 function in the brain. Inhibition of Prdx6-iPLA2 activity by gene therapy and/or pharmacology may constitute a promising new therapeutic approach to the treatment of stroke.
Collapse
Affiliation(s)
- Yu Shanshan
- Department of Pathology, Chongqing Medical UniversityChongqing, China.,Molecular Medical Laboratory, Chongqing Medical UniversityChongqing, China.,Institute of Neuroscience, Chongqing Medical UniversityChongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Jiang Beibei
- Department of Pathology, Chongqing Medical UniversityChongqing, China.,Molecular Medical Laboratory, Chongqing Medical UniversityChongqing, China.,Institute of Neuroscience, Chongqing Medical UniversityChongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Tan Li
- Department of Pathology, Chongqing Medical UniversityChongqing, China.,Molecular Medical Laboratory, Chongqing Medical UniversityChongqing, China.,Institute of Neuroscience, Chongqing Medical UniversityChongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Gao Minna
- Department of Pathology, Chongqing Medical UniversityChongqing, China.,Molecular Medical Laboratory, Chongqing Medical UniversityChongqing, China.,Institute of Neuroscience, Chongqing Medical UniversityChongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Lei Shipeng
- Department of Respiratory Medicine, Jiangjin Center HospitalChongqing, China
| | - Peng Li
- Department of Pathology, Chongqing Medical UniversityChongqing, China.,Molecular Medical Laboratory, Chongqing Medical UniversityChongqing, China.,Institute of Neuroscience, Chongqing Medical UniversityChongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Zhao Yong
- Department of Pathology, Chongqing Medical UniversityChongqing, China.,Molecular Medical Laboratory, Chongqing Medical UniversityChongqing, China.,Institute of Neuroscience, Chongqing Medical UniversityChongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| |
Collapse
|
33
|
Thomsen BB, Gredal H, Wirenfeldt M, Kristensen BW, Clausen BH, Larsen AE, Finsen B, Berendt M, Lambertsen KL. Spontaneous ischaemic stroke lesions in a dog brain: neuropathological characterisation and comparison to human ischaemic stroke. Acta Vet Scand 2017; 59:7. [PMID: 28086932 PMCID: PMC5237225 DOI: 10.1186/s13028-016-0275-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 12/31/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Dogs develop spontaneous ischaemic stroke with a clinical picture closely resembling human ischaemic stroke patients. Animal stroke models have been developed, but it has proved difficult to translate results obtained from such models into successful therapeutic strategies in human stroke patients. In order to face this apparent translational gap within stroke research, dogs with ischaemic stroke constitute an opportunity to study the neuropathology of ischaemic stroke in an animal species. CASE PRESENTATION A 7 years and 8 months old female neutered Rottweiler dog suffered a middle cerebral artery infarct and was euthanized 3 days after onset of neurological signs. The brain was subjected to histopathology and immunohistochemistry. Neuropathological changes were characterised by a pan-necrotic infarct surrounded by peri-infarct injured neurons and reactive microglia/macrophages and astrocytes. CONCLUSIONS The neuropathological changes reported in the present study were similar to findings in human patients with ischaemic stroke. The dog with spontaneous ischaemic stroke is of interest as a complementary spontaneous animal model for further neuropathological studies.
Collapse
|
34
|
Lee HI, Park JH, Park MY, Kim NG, Park KJ, Choi BT, Shin YI, Shin HK. Pre-conditioning with transcranial low-level light therapy reduces neuroinflammation and protects blood-brain barrier after focal cerebral ischemia in mice. Restor Neurol Neurosci 2016; 34:201-14. [PMID: 26889965 DOI: 10.3233/rnn-150559] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
PURPOSE Transcranial low-level light therapy (LLLT) has gained interest as a non-invasive, inexpensive and safe method of modulating neurological and psychological functions in recent years. This study was designed to examine the preventive effects of LLLT via visible light source against cerebral ischemia at the behavioral, structural and neurochemical levels. METHODS The mice received LLLT twice a day for 2 days prior to photothrombotic cortical ischemia. RESULTS LLLT significantly reduced infarct size and edema and improved neurological and motor function 24 h after ischemic injury. In addition, LLLT markedly inhibited Iba-1- and GFAP-positive cells, which was accompanied by a reduction in the expression of inflammatory mediators and inhibition of MAPK activation and NF-κB translocation in the ischemic cortex. Concomitantly, LLLT significantly attenuated leukocyte accumulation and infiltration into the infarct perifocal region. LLLT also prevented BBB disruption after ischemic events, as indicated by a reduction of Evans blue leakage and water content. These findings were corroborated by immunofluorescence staining of the tight junction-related proteins in the ischemic cortex in response to LLLT. CONCLUSIONS Non-invasive intervention of LLLT in ischemic brain injury may provide a significant functional benefit with an underlying mechanism possibly being suppression of neuroinflammation and reduction of BBB disruption.
Collapse
Affiliation(s)
- Hae In Lee
- Department of Rehabilitation Medicine, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Jung Hwa Park
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Min Young Park
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Nam Gyun Kim
- Medical Research Center of Color Seven, Seoul, Republic of Korea
| | - Kyoung-Jun Park
- Medical Research Center of Color Seven, Seoul, Republic of Korea
| | - Byung Tae Choi
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea.,Division of Meridian and Structural Medicine, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea.,Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Yong-Ii Shin
- Department of Rehabilitation Medicine, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea.,Research Institute for Convergence of Biomedical Science and Technology, Pusan National UniversityYangsan Hospital, Yangsan, Gyeongnam, Republic of Korea.,Department of Rehabilitation Medicine, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Hwa Kyoung Shin
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea.,Division of Meridian and Structural Medicine, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea.,Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam, Republic of Korea.,Department of Rehabilitation Medicine, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| |
Collapse
|
35
|
Anti-Inflammatory Effects of Traditional Chinese Medicines against Ischemic Injury in In Vivo Models of Cerebral Ischemia. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:5739434. [PMID: 27703487 PMCID: PMC5040804 DOI: 10.1155/2016/5739434] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 07/25/2016] [Indexed: 12/11/2022]
Abstract
Inflammation plays a crucial role in the pathophysiology of acute ischemic stroke. In the ischemic cascade, resident microglia are rapidly activated in the brain parenchyma and subsequently trigger inflammatory mediator release, which facilitates leukocyte-endothelial cell interactions in inflammation. Activated leukocytes invade the endothelial cell junctions and destroy the blood-brain barrier integrity, leading to brain edema. Toll-like receptors (TLRs) stimulation in microglia/macrophages through the activation of intercellular signaling pathways secretes various proinflammatory cytokines and enzymes and then aggravates cerebral ischemic injury. The secreted cytokines activate the proinflammatory transcription factors, which subsequently regulate cytokine expression, leading to the amplification of the inflammatory response and exacerbation of the secondary brain injury. Traditional Chinese medicines (TCMs), including TCM-derived active compounds, Chinese herbs, and TCM formulations, exert neuroprotective effects against inflammatory responses by downregulating the following: ischemia-induced microglial activation, microglia/macrophage-mediated cytokine production, proinflammatory enzyme production, intercellular adhesion molecule-1, matrix metalloproteinases, TLR expression, and deleterious transcription factor activation. TCMs also aid in upregulating anti-inflammatory cytokine expression and neuroprotective transcription factor activation in the ischemic lesion in the inflammatory cascade during the acute phase of cerebral ischemia. Thus, TCMs exert potent anti-inflammatory properties in ischemic stroke and warrant further investigation.
Collapse
|
36
|
Infiltration of invariant natural killer T cells occur and accelerate brain infarction in permanent ischemic stroke in mice. Neurosci Lett 2016; 633:62-68. [PMID: 27637387 DOI: 10.1016/j.neulet.2016.09.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/08/2016] [Accepted: 09/08/2016] [Indexed: 12/13/2022]
Abstract
Invariant natural killer T (iNKT) cells are a unique subset of T cells that have been implicated in inflammation, atopy, autoimmunity, infections, and cancer. Although iNKT cells have been extensively studied over the past decade, its role in the pathogenesis of ischemic brain injury is still largely unknown. In our study, we determined whether iNKT cells infiltration occur in a mouse model of permanent cerebral ischemia. C57BL6/J male mice were treated with either alpha-galactosylceramide (α-GalCer) or vehicle control before undergoing permanent middle cerebral artery occlusion (pMCAO). α-GalCer, a glycolipid antigen, specifically activates iNKT cells by a CD1d-restricted mechanism. Using flow cytometry, 10,000 leukocytes (CD45 high cells) from the ischemic hemisphere and peripheral blood respectively were analyzed to determine the number of NK1.1+CD3+ cells at 3, 12, 24 and 48h post-pMCAO. Cerebral infarct size, brain edema and morphological characteristics were measured at the stipulated time points by 2,3,5-triphenyltetrazolium chloride (TTC) staining, weighing, and H&E staining. The levels of IFN-γ and TNF-α in brain tissue and serum were assessed by immunohistochemistry and ELISA respectively. We found that the number of iNKT cells started increasing from 12h (PB sample) and 24h (ischemic hemisphere sample) respectively in the vehicle treated group. iNKT cells infiltration occurred at an earlier time-point compared in the α-GalCer treated group (T=3H vs T=12H in PB sample; T=12H vs T=24H in ischemic hemisphere sample). Brain water content at 12h and 24h was significantly higher in pMCAO+α-GalCer mice compared to pMCAO+vehicle mice which was in turn higher than mice that underwent sham surgery. Aggravated morphological abnormalities in HE-stained neurons and significantly increased neurons with pyknotic nuclei and cavitation in the ischemic region were observed at 24h in the pMCAO+α-GalCer and pMCAO+vehicle groups. Cerebral infarct volume, neurological deficit Scores and brain edema were significantly increased at 24h in the pMCAO+α-GalCer group compared to pMCAO+vehicle group. In the pMCAO+vehicle group, the serum concentrations of TNF-α and IFN-γ were increased at 12h and 24h post-pMCAO, and remained elevated up to 48h. In mice treated with pMCAO+α-GalCer, TNF-α and IFN-γ were both increased at 12h post-pMCAO, and remained elevated up to 48h. Immunohistochemistry showed that protein expression of TNF-α and IFN-γ in brain tissues was higher in α-GalCer-treated mice. Our results demonstrate that within 48h of focal permanent cerebral ischemia, iNKT cells infiltrate into the brain and contribute to brain infarction.
Collapse
|
37
|
Kawabori M, Yenari MA. Inflammatory responses in brain ischemia. Curr Med Chem 2016; 22:1258-77. [PMID: 25666795 DOI: 10.2174/0929867322666150209154036] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/02/2014] [Accepted: 02/02/2015] [Indexed: 12/20/2022]
Abstract
Brain infarction causes tissue death by ischemia due to occlusion of the cerebral vessels and recent work has shown that post stroke inflammation contributes significantly to the development of ischemic pathology. Because secondary damage by brain inflammation may have a longer therapeutic time window compared to the rescue of primary damage following arterial occlusion, controlling inflammation would be an obvious therapeutic target. A substantial amount of experimentall progress in this area has been made in recent years. However, it is difficult to elucidate the precise mechanisms of the inflammatory responses following ischemic stroke because inflammation is a complex series of interactions between inflammatory cells and molecules, all of which could be either detrimental or beneficial. We review recent advances in neuroinflammation and the modulation of inflammatory signaling pathways in brain ischemia. Potential targets for treatment of ischemic stroke will also be covered. The roles of the immune system and brain damage versus repair will help to clarify how immune modulation may treat stroke.
Collapse
Affiliation(s)
| | - Midori A Yenari
- Dept. of Neurology, University of California, San Francisco and the San Francisco Veterans Affairs Medical Center, 4150 Clement Street, San Francisco, CA 94121, USA.
| |
Collapse
|
38
|
Singh V, Roth S, Veltkamp R, Liesz A. HMGB1 as a Key Mediator of Immune Mechanisms in Ischemic Stroke. Antioxid Redox Signal 2016; 24:635-51. [PMID: 26493086 DOI: 10.1089/ars.2015.6397] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
SIGNIFICANCE Stroke is the leading cause of morbidity and mortality worldwide. Inflammatory cascades have a major impact on outcome and regeneration after ischemic stroke. High-mobility group box 1 (HMGB1) has come into the focus of experimental and clinical stroke research because it is released from necrotic brain tissue and its differential redox forms attract and activate immune cells after ischemic brain injury. HMGB1 is a potent inducer of inflammatory cascades, and thereby, secondary deterioration of neurological outcome. RECENT ADVANCES The role of HMGB1 in sterile inflammation is well established. Emerging evidence suggests that HMGB1 modulates neuroinflammation after experimental brain ischemia and that it may be a useful prognostic biomarker for stroke patients. CRITICAL ISSUES HMGB1 is instantly released from necrotic cells in the ischemic core and activates an early inflammatory response. In addition, brain-released HMGB1 can be redox modified in the circulation and activate peripheral immune cells. HMGB1 concentrations correlate with disease severity and outcome after brain injury. This is the first review depicting the crucial role of HMGB1 in the initiation and perpetuation of secondary immune alterations after experimental and clinical stroke. FUTURE DIRECTIONS HMGB1-dependent signaling pathways are on the verge and have the potential to become a central topic in experimental stroke research. Current and upcoming projects in this field will be paving the way for future translational approaches targeting the center of poststroke inflammation to improve stroke recovery and long-term outcome.
Collapse
Affiliation(s)
- Vikramjeet Singh
- 1 Institute for Stroke and Dementia Research , Klinikum der Universität München, Munich, Germany .,2 Munich Cluster for Systems Neurology (SyNergy) , Munich, Germany
| | - Stefan Roth
- 1 Institute for Stroke and Dementia Research , Klinikum der Universität München, Munich, Germany .,2 Munich Cluster for Systems Neurology (SyNergy) , Munich, Germany
| | - Roland Veltkamp
- 3 Division of Brain Sciences, Imperial College London , London, United Kingdom
| | - Arthur Liesz
- 1 Institute for Stroke and Dementia Research , Klinikum der Universität München, Munich, Germany .,2 Munich Cluster for Systems Neurology (SyNergy) , Munich, Germany
| |
Collapse
|
39
|
Long L, Wang J, Chen N, Zheng S, Shi L, Xu Y, Luo C, Deng Y. Curcumin Ameliorates Reserpine-Induced Gastrointestinal Mucosal Lesions Through Inhibiting IκB-α/NF-κB Pathway and Regulating Expression of Vasoactive Intestinal Peptide and Gastrin in Rats. J Med Food 2016; 19:528-34. [PMID: 26872103 DOI: 10.1089/jmf.2015.3570] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The objective of our study was to investigate whether curcumin protects against reserpine-induced gastrointestinal mucosal lesions (GMLs) in rats and to explore the mechanism of curcumin's action. Sprague-Dawley rats were randomly divided into four groups: control group, reserpine-treated group, reserpine treatment group with curcumin at high dose (200 mg/kg), and reserpine treatment group with curcumin at low dose (100 mg/kg). Rats in reserpine-treated group were induced by intraperitoneally administered reserpine (0.5 mg/kg) for 28 days. TUNEL staining and hematoxylin and eosin staining were used to evaluate the apoptotic cells and morphologic changes. In addition, to explore the mechanism of curcumin in protecting GMLs, we used serum of experimental rats to assess the level of vasoactive intestinal peptide (VIP), gastrin, interleukin-6, interleukin-10, tumor necrosis factor-α and interferon-γ by ELISA and radioimmunoassay. The protein levels of NF-κB, p-IκB-α, IκB-α, Bcl-2, Bax, and cleaved-caspase-3 were examined by western blot analysis. Data were analyzed with SPSS 19.0 software package. Curcumin treatment prevented tissue damage and cell death in the reserpine-treated rats and effectively decreased inflammatory response and balanced the expression of VIP and gastrin in the reserpine-treated rats. NF-κB, p-IκB-α, Bax, and cleaved-caspase-3 were increased in the reserpine group, but the curcumin high-dose group inhibited them. Curcumin can target the IκB-α/NF-κB pathway to inhibit inflammatory response and regulate the level of VIP and gastrin in reserpine-induced GML rats.
Collapse
Affiliation(s)
- Lingli Long
- 1 Research Center of Translational Medicine, The First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, China
| | - Jingnan Wang
- 1 Research Center of Translational Medicine, The First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, China
| | - Ningning Chen
- 2 Department of Spine Surgery, Sun Yat-Sen University , Guangzhou, China
| | - Shuhui Zheng
- 1 Research Center of Translational Medicine, The First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, China
| | - Lanying Shi
- 3 Department of Traditional Chinese Medicine, Sun Yat-Sen University , Guangzhou, China
| | - Yuxia Xu
- 1 Research Center of Translational Medicine, The First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, China
| | - Canqiao Luo
- 4 Pathology, The First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, China
| | - Yubin Deng
- 1 Research Center of Translational Medicine, The First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, China
| |
Collapse
|
40
|
Hakimizadeh E, Kazemi Arababadi M, Shamsizadeh A, Roohbakhsh A, Allahtavakoli M. The Possible Role of Toll-Like Receptor 4 in the Pathology of Stroke. Neuroimmunomodulation 2016; 23:131-136. [PMID: 27287756 DOI: 10.1159/000446481] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 04/18/2016] [Indexed: 11/19/2022] Open
Abstract
Stroke is a prevalent and dangerous health problem, which triggers an intense inflammatory response to Toll-like receptor (TLR) activation. TLRs are the essential components of the response of the innate immunity system, and, therefore, they are one of the key factors involved in recognizing pathogens and internal ligands. Among TLRs, TLR4 significantly participates in the induction of inflammation and brain functions; hence, it has been hypothesized that this molecule is associated with several immune-related brain diseases such as stroke. It has also been proved that animals with TLR4 deficiency have higher protection against ischemia and that the absence of TLR4 reduces neuroinflammation and injuries associated with brain trauma. TLR4 deficiency may play a neuroprotective role in the occurrence of stroke. This article reviews recent information regarding the impact of TLR4 on the pathogenicity of stroke.
Collapse
Affiliation(s)
- Elham Hakimizadeh
- Physiology-Pharmacology Research Center, Rafsanjan University of Medical Sciences,Rafsanjan, Iran
| | | | | | | | | |
Collapse
|
41
|
Jickling GC, Liu D, Ander BP, Stamova B, Zhan X, Sharp FR. Targeting neutrophils in ischemic stroke: translational insights from experimental studies. J Cereb Blood Flow Metab 2015; 35:888-901. [PMID: 25806703 PMCID: PMC4640255 DOI: 10.1038/jcbfm.2015.45] [Citation(s) in RCA: 431] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 01/23/2015] [Accepted: 01/26/2015] [Indexed: 01/08/2023]
Abstract
Neutrophils have key roles in ischemic brain injury, thrombosis, and atherosclerosis. As such, neutrophils are of great interest as targets to treat and prevent ischemic stroke. After stroke, neutrophils respond rapidly promoting blood-brain barrier disruption, cerebral edema, and brain injury. A surge of neutrophil-derived reactive oxygen species, proteases, and cytokines are released as neutrophils interact with cerebral endothelium. Neutrophils also are linked to the major processes that cause ischemic stroke, thrombosis, and atherosclerosis. Thrombosis is promoted through interactions with platelets, clotting factors, and release of prothrombotic molecules. In atherosclerosis, neutrophils promote plaque formation and rupture by generating oxidized-low density lipoprotein, enhancing monocyte infiltration, and degrading the fibrous cap. In experimental studies targeting neutrophils can improve stroke. However, early human studies have been met with challenges, and suggest that selective targeting of neutrophils may be required. Several properties of neutrophil are beneficial and thus may important to preserve in patients with stroke including antimicrobial, antiinflammatory, and neuroprotective functions.
Collapse
Affiliation(s)
- Glen C Jickling
- Department of Neurology, University of California at Davis, Sacramento, California, USA
| | - DaZhi Liu
- Department of Neurology, University of California at Davis, Sacramento, California, USA
| | - Bradley P Ander
- Department of Neurology, University of California at Davis, Sacramento, California, USA
| | - Boryana Stamova
- Department of Neurology, University of California at Davis, Sacramento, California, USA
| | - Xinhua Zhan
- Department of Neurology, University of California at Davis, Sacramento, California, USA
| | - Frank R Sharp
- Department of Neurology, University of California at Davis, Sacramento, California, USA
| |
Collapse
|
42
|
Tu XK, Yang WZ, Chen JP, Chen Y, Chen Q, Chen PP, Shi SS. Repetitive ischemic preconditioning attenuates inflammatory reaction and brain damage after focal cerebral ischemia in rats: involvement of PI3K/Akt and ERK1/2 signaling pathway. J Mol Neurosci 2014; 55:912-22. [PMID: 25338292 DOI: 10.1007/s12031-014-0446-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 10/13/2014] [Indexed: 12/29/2022]
Abstract
Ischemic preconditioning (IPC) has been demonstrated to provide a neuroprotection against brain damage produced by focal cerebral ischemia. However, it is elusive whether ischemic preconditioning attenuates ischemic brain damage through modulating phosphatidylinositol 3-kinase/Akt (PI3K/Akt) and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway. In the present study, we first explored the best scheme of repetitive ischemic preconditioning (RIPC) to protect rat brain against ischemic damage and then further investigated the underlying mechanisms in RIPC's neuroprotection. Adult male Sprague-Dawley rats underwent ischemic preconditioning or (and) middle cerebral artery occlusion (MCAO). LY294002 or (and) PD98059 were injected intracerebroventricularly to selectively inhibit the activation of PI3K/Akt or ERK1/2. Neurological deficit scores, cerebral infarct volume, and morphological characteristic were detected at corresponding time after cerebral ischemia. The enzymatic activity of myeloperoxidase (MPO) was measured 24 h after cerebral ischemia. Expressions of p-Akt, t-Akt, p-ERK1/2, t-ERK1/2, nuclear factor-kappa B (NF-κB) p65, and cyclooxygenase-2 (COX-2) in ischemic brain were determined by Western blot. The release of tumor necrosis factor-α (TNF-α) in blood was examined by ELISA. In the various schemes of RIPC, IPC2 × 5 min causes less neuronal damage in the cortex and subcortex of ischemic brain and provides an obvious alleviation of cerebral infarction and neurological deficit after lethal ischemia. IPC2 × 5 min significantly reduces cerebral infarct volume, neurological deficit scores, and MPO activity; all of which were diminished by LY294002 or (and) PD98059. IPC2 × 5 min significantly upregulates the expressions of p-Akt and p-ERK1/2, which were inhibited by LY294002 or (and) PD98059. IPC2 × 5 min significantly downregulates the expressions of NF-κB p65 and COX-2 and attenuates the release of TNF-α; all of which were abolished by LY294002 or (and) PD98059. IPC2 × 5 min is the best scheme of RIPC to protect rat brain against cerebral ischemia. IPC2 × 5 min attenuates brain damage in rats subjected to lethal ischemia, and this neuroprotection is associated with inhibition of neuroinflammation through modulating PI3K/Akt and ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Xian-kun Tu
- Department of Neurosurgery, Fujian Medical University Union Hospital, 29# Xinquan Road, Fuzhou, Fujian, 350001, China,
| | | | | | | | | | | | | |
Collapse
|
43
|
Lambertsen KL, Østergaard K, Clausen BH, Hansen S, Stenvang J, Thorsen SB, Meldgaard M, Kristensen BW, Hansen PB, Sorensen GL, Finsen B. No effect of ablation of surfactant protein-D on acute cerebral infarction in mice. J Neuroinflammation 2014; 11:123. [PMID: 25038795 PMCID: PMC4110550 DOI: 10.1186/1742-2094-11-123] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 07/03/2014] [Indexed: 12/03/2022] Open
Abstract
Background Crosstalk between the immune system in the brain and the periphery may contribute to the long-term outcome both in experimental and clinical stroke. Although, the immune defense collectin surfactant protein-D (SP-D) is best known for its role in pulmonary innate immunity, SP-D is also known to be involved in extrapulmonary modulation of inflammation in mice. We investigated whether SP-D affected cerebral ischemic infarction and ischemia-induced inflammatory responses in mice. Methods The effect of SP-D was studied by comparing the size of ischemic infarction and the inflammatory and astroglial responses in SP-D knock out (KO) and wild type (WT) mice subjected to permanent middle cerebral artery occlusion. SP-D mRNA production was assessed in isolated cerebral arteries and in the whole brain by PCR, and SP-D protein in normal appearing and ischemic human brain by immunohistochemistry. Changes in plasma SP-D and TNF were assessed by ELISA and proximity ligation assay, respectively. Results Infarct volumetric analysis showed that ablation of SP-D had no effect on ischemic infarction one and five days after induction of ischemia. Further, ablation of SP-D had no effect on the ischemia-induced increase in TNF mRNA production one day after induction of ischemia; however the TNF response to the ischemic insult was affected at five days. SP-D mRNA was not detected in parenchymal brain cells in either naïve mice or in mice subjected to focal cerebral ischemia. However, SP-D mRNA was detected in middle cerebral artery cells in WT mice and SP-D protein in vascular cells both in normal appearing and ischemic human brain tissue. Measurements of the levels of SP-D and TNF in plasma in mice suggested that levels were unaffected by the ischemic insult. Microglial-leukocyte and astroglial responses were comparable in SP-D KO and WT mice. Conclusions SP-D synthesis in middle cerebral artery cells is consistent with SP-D conceivably leaking into the infarcted area and affecting local cytokine production. However, there was no SP-D synthesis in parenchymal brain cells and ablation of SP-D had no effect on ischemic cerebral infarction.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Bente Finsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, JB Winsloewsvej 25, 2, DK-5000 Odense C, Denmark.
| |
Collapse
|
44
|
Tu XK, Yang WZ, Chen JP, Chen Y, Ouyang LQ, Xu YC, Shi SS. Curcumin Inhibits TLR2/4-NF-κB Signaling Pathway and Attenuates Brain Damage in Permanent Focal Cerebral Ischemia in Rats. Inflammation 2014; 37:1544-51. [DOI: 10.1007/s10753-014-9881-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
45
|
Shi SS, Yang WZ, Chen Y, Chen JP, Tu XK. Propofol reduces inflammatory reaction and ischemic brain damage in cerebral ischemia in rats. Neurochem Res 2014; 39:793-9. [PMID: 24610527 DOI: 10.1007/s11064-014-1272-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 02/25/2014] [Accepted: 02/26/2014] [Indexed: 10/25/2022]
Abstract
Our previous studies demonstrated that inflammatory reaction and neuronal apoptosis are the most important pathological mechanisms in ischemia-induced brain damage. Propofol has been shown to attenuate ischemic brain damage via inhibiting neuronal apoptosis. The present study was performed to evaluate the effect of propofol on brain damage and inflammatory reaction in rats of focal cerebral ischemia. Sprague-Dawley rats underwent permanent middle cerebral artery occlusion, then received treatment with propofol (10 or 50 mg/kg) or vehicle after 2 h of ischemia. Neurological deficit scores, cerebral infarct size and morphological characteristic were measured 24 h after cerebral ischemia. The enzymatic activity of myeloperoxidase (MPO) was assessed 24 h after cerebral ischemia. Nuclear factor-kappa B (NF-κB) p65 expression in ischemic rat brain was detected by western blot. Cyclooxygenase-2 (COX-2) expression in ischemic rat brain was determined by immunohistochemistry. ELISA was performed to detect the serum concentration of tumor necrosis factor-α (TNF-α). Neurological deficit scores, cerebral infarct size and MPO activity were significantly reduced by propofol administration. Furthermore, expression of NF-κB, COX-2 and TNF-α were attenuated by propofol administration. Our results demonstrated that propofol (10 and 50 mg/kg) reduces inflammatory reaction and brain damage in focal cerebral ischemia in rats. Propofol exerts neuroprotection against ischemic brain damage, which might be associated with the attenuation of inflammatory reaction and the inhibition of inflammatory genes.
Collapse
Affiliation(s)
- Song-sheng Shi
- Department of Neurosurgery, Fujian Medical University Union Hospital, 29# Xinquan Road, Fuzhou, 350001, Fujian, China
| | | | | | | | | |
Collapse
|
46
|
TAT-Hsp70 induces neuroprotection against stroke via anti-inflammatory actions providing appropriate cellular microenvironment for transplantation of neural precursor cells. J Cereb Blood Flow Metab 2013; 33:1778-88. [PMID: 23881248 PMCID: PMC3824176 DOI: 10.1038/jcbfm.2013.126] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 06/25/2013] [Accepted: 06/30/2013] [Indexed: 11/08/2022]
Abstract
Heat-shock protein 70 (Hsp70) protects against cerebral ischemia, which is attributed to its chaperone activity. However, recent reports also describe pro-inflammatory actions of Hsp70 via activation of Toll-like receptors (TLR). Using membrane-permeable transactivator of transcription (TAT)-Hsp70, we analyzed TAT-Hsp70-induced neuroprotection and its underlying mechanism after cerebral ischemia in mice. Infusion of TAT-Hsp70 reduced infarct volume and enhanced blood-brain barrier integrity on day 3 poststroke, when given no later than 12 hours. The latter was associated with reduction of microglial activation, although upregulation of pro-inflammatory TLR-2/4 was observed both in verum and in control animals. Nevertheless, protein abundance and nuclear translocation of downstream nuclear factor kappa B (NF-κB) as well as proteasomal degradation of the NF-κB regulator Ikappa B alpha (IκB-α) were significantly reduced by TAT-Hsp70. TAT-Hsp70-induced neuroprotection and functional recovery were restricted to 4 weeks only. However, TAT-Hsp70 provided an appropriate extracellular milieu for delayed intravenous transplantation of adult neural precursor cells (NPCs). Thus, NPCs that were grafted 28 days poststroke induced long-term neuroprotection for at least 3 months, which was not due to integration of grafted cells but rather due to paracrine effects of transplanted NPCs. Conclusively, TAT-Hsp70 ameliorates postischemic inflammation via proteasome inhibition, thus providing an appropriate extracellular milieu for delayed NPC transplantation and culminating in long-term neuroprotection.
Collapse
|
47
|
TLR2 and TLR4 in the brain injury caused by cerebral ischemia and reperfusion. Mediators Inflamm 2013; 2013:124614. [PMID: 23864765 PMCID: PMC3706022 DOI: 10.1155/2013/124614] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 05/30/2013] [Accepted: 06/11/2013] [Indexed: 02/06/2023] Open
Abstract
Brain injury caused by cerebral ischemia/reperfusion is a complicated pathophysiological course, in which inflammation is thought to play an important role. Toll-like receptors are a type of transmembrane proteins, which can recognize either exogenous pathogen-associated molecular patterns or endogenous stress or damage-associated molecular patterns in the innate immune system and initiate inflammatory responses. Among Toll-like receptors, TLR2 and TLR4 are found to be more important than others in the pathological progression of cerebral injury due to ischemia and reperfusion. This review will focus on the biological characteristics and functions of TLR2 and TLR4 and their downstream signal pathways.
Collapse
|
48
|
Li W, Cheng X, Chen HS, He ZY. Apobec-1 increases cyclooxygenase-2 and aggravates injury in oxygen-deprived neurogenic cells and middle cerebral artery occlusion rats. Neurochem Res 2013; 38:1434-45. [PMID: 23609497 DOI: 10.1007/s11064-013-1043-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 03/19/2013] [Accepted: 04/10/2013] [Indexed: 01/21/2023]
Abstract
Given that cyclooxygenase-2 (COX-2) plays a crucial role during cerebral ischemia and Apobec-1 is a critical regulator of COX-2 mRNA stabilization in gastrointestinal settings, the correlation of COX-2 and Apobec-1 was investigated in neurogenic cells and rat model of cerebral ischemia. After neurogenic SH-SY5Y, NG108-15 and PC12 cells were exposed to oxygen-glucose deprivation, cell viability, LDH leakage and Apobec-1 expression were determined. The effect of Apobec-1 overexpression on injury severity of oxygen-glucose deprivation, COX-2 expression, C-to-U editing of COX-2 mRNA were measured in vitro. Then the correlation of Apobec-1 level and injury severity was analyzed in cells with oxygen-glucose deprivation and in rats with middle cerebral artery occlusion. Apobec-1 expression was elevated along with upregulation of COX-2 and injury severity of oxygen-glucose deprivation in the three cell lines. Apobec-1 overexpression aggravated injury of oxygen-glucose deprivation in vitro and could be correlated to injury severity in vivo. Meanwhile, Apobec-1 increased COX-2 expression and COX-2 mRNA stabilization in neurogenic cells, and failed to catalyze C-to-U editing of COX-2 mRNA. Apobec-1 could upregulate COX-2 expression in neurogenic cells by stabilizing COX-2 mRNA, and might aggravate injury of oxygen-glucose deprivation in neurogenic cells as well as in rats with cerebral ischemia.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurology, First Affiliated Hospital, China Medical University, North Nanjing Street #155, Shenyang, Liaoning Province, China
| | | | | | | |
Collapse
|
49
|
Ström JO, Ingberg E, Theodorsson A, Theodorsson E. Method parameters' impact on mortality and variability in rat stroke experiments: a meta-analysis. BMC Neurosci 2013; 14:41. [PMID: 23548160 PMCID: PMC3637133 DOI: 10.1186/1471-2202-14-41] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 03/22/2013] [Indexed: 12/14/2022] Open
Abstract
Background Even though more than 600 stroke treatments have been shown effective in preclinical studies, clinically proven treatment alternatives for cerebral infarction remain scarce. Amongst the reasons for the discrepancy may be methodological shortcomings, such as high mortality and outcome variability, in the preclinical studies. A common approach in animal stroke experiments is that A) focal cerebral ischemia is inflicted, B) some type of treatment is administered and C) the infarct sizes are assessed. However, within this paradigm, the researcher has to make numerous methodological decisions, including choosing rat strain and type of surgical procedure. Even though a few studies have attempted to address the questions experimentally, a lack of consensus regarding the optimal methodology remains. Methods We therefore meta-analyzed data from 502 control groups described in 346 articles to find out how rat strain, procedure for causing focal cerebral ischemia and the type of filament coating affected mortality and infarct size variability. Results The Wistar strain and intraluminal filament procedure using a silicone coated filament was found optimal in lowering infarct size variability. The direct and endothelin methods rendered lower mortality rate, whereas the embolus method increased it compared to the filament method. Conclusions The current article provides means for researchers to adjust their middle cerebral artery occlusion (MCAo) protocols to minimize infarct size variability and mortality.
Collapse
Affiliation(s)
- Jakob O Ström
- Department of Clinical and Experimental Medicine, Clinical Chemistry, Faculty of Health Sciences, Linköping University, County Council of Östergötland, Linköping, Sweden.
| | | | | | | |
Collapse
|
50
|
Liu LX, Zhou XY, Li CS, Liu LQ, Huang SY, Zhou SN. Selenoprotein S expression in the rat brain following focal cerebral ischemia. Neurol Sci 2013; 34:1671-8. [DOI: 10.1007/s10072-013-1319-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 01/21/2013] [Indexed: 11/30/2022]
|