1
|
Zeng J, Zhou H, Wan H, Yang J. Single-cell omics: moving towards a new era in ischemic stroke research. Eur J Pharmacol 2025; 1000:177725. [PMID: 40350018 DOI: 10.1016/j.ejphar.2025.177725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Ischemic stroke (IS) is a highly complex and heterogeneous disease involving multiple pathophysiological events. A better understanding of the pathophysiology of IS will enhance preventive, diagnostic and therapeutic strategies. Despite significant advances in modern medicine, the molecular mechanisms of IS are still largely unknown. The high-throughput omics approach opens new avenues for identifying IS biomarkers and elucidating disease pathogenesis mechanisms. Single-cell omics enables a more thorough and in-depth analysis of the cellular interactions and properties in IS. This will lead to a better understanding of the onset, treatment and prognosis of IS. In this paper, we first reviewed the disease signatures and mechanisms research of IS. Subsequently, the use of single-cell omics to comprehend the mechanisms of IS was discussed, along with some recent developments in the field. To further delineate the upstream pathogenic alterations and downstream molecular impacts of IS, we also discussed the current use of machine learning approaches to single-cell omics data analysis. Particularly, single-cell omics is being used to inform risk assessment, early patient diagnosis and treatment strategies, and their potential impact on precision medicine. Thus, we summarized the role of single-cell omics in precision medicine. Despite the relative youth of the field, the development of single-cell omics promises to provide a powerful tool for elucidating the pathogenesis of IS.
Collapse
Affiliation(s)
- Jieqiong Zeng
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Ecological and Environmental, Hubei Industrial Polytechnic, Shiyan, 442000, China
| | - Huifen Zhou
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Haitong Wan
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Jiehong Yang
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
2
|
Staszewski J, Dębiec A, Strilciuc S, Gniadek-Olejniczak K, Piusinska-Macoch R, Balo D, Harston G, Stępień A, Brzozowski K, Zięcina P, Narloch J, Wierzbicki M, Piasecki P. Efficacy of Cerebrolysin Treatment as an Add-On Therapy to Mechanical Thrombectomy in Patients with Acute Ischemic Stroke Due to Large Vessel Occlusion in Anterior Circulation: Results of a 3-Month Follow-up of a Prospective, Open Label, Single-Center Study. Transl Stroke Res 2025:10.1007/s12975-025-01355-z. [PMID: 40325343 DOI: 10.1007/s12975-025-01355-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 04/12/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025]
Abstract
This study hypothesized that Cerebrolysin, a multimodal neuroprotective agent, enhances the efficacy and safety of mechanical thrombectomy (MT) in both acute ischemic stroke (AIS) and recovery stroke phases in selected patients with good collateral status (CTA-CS 2-3) and effective recanalization (mTICI 2b-3). A single-center, prospective, open-label, single-arm study with blinded outcome assessment of 50 consecutive patients with moderate-to-severe AIS treated with MT ≤ 6 h of stroke onset followed by Cerebrolysin (30 ml iv within 8 h of onset and continued to day 21, first cycle) and in a recovery phase (between 69-90 days, second cycle) compared to 50 historical controls matched by propensity scores. Key outcomes included functional independence (mRS 0-2 at 90 days), safety endpoints, and neurological recovery (NIHSS at 24 h and 7 day post MT). Patients receiving Cerebrolysin achieved higher rates of mRS 0-2 at 90 days (68% vs. 44%, p = 0.016, OR 2.7, 95% CI 1.2-6.1; NNT: 4.2), had reduced risk of secondary ICH (14% vs. 40%, p = 0.02; RR 0.37, 95% CI 0.14-0.95), and had lower NIHSS on day 7 (median [IQR]: 3 [4] vs. 6 [9], p = 0.01). There was a significant difference in Barthel Index scores between the Cerebrolysin group and the control group at 30 days (median [IQR]: 77 [32] vs. 63 [50], p = 0.03) and at 3 months (86 [22] vs. 75 [29], p = 0.01) primarily driven by the increase in the mobility and transfer components. Multivariate analysis identified Cerebrolysin as an independent predictor of favorable outcomes at 3 months (OR 7.5, 95% CI 1.8-30.9), particularly in patients with diabetes (interaction OR 9.6, 95% CI 1.01-92). The overall mortality rates at 30 and 90 days were similar in both groups (2% vs 6% and 8% vs 12%, p > 0.1). Cerebrolysin improved functional outcomes at 90 days, accelerated neurological recovery, and reduced complications post-MT in patients with small ischemic core, good collateral circulation, and effective recanalization at baseline. These findings warrant further randomized trials to validate its efficacy and explore its long-term benefits.Registration: URL: https://www.clinicaltrials.gov ; unique identifier: NCT04904341.
Collapse
Affiliation(s)
- Jacek Staszewski
- Clinic of Neurology, Military Institute of Medicine - National Research Institute, Szaserow 128, 04-141, Warsaw, Poland.
- Faculty of Medicine, University of Warsaw, Warsaw, Poland.
| | - Aleksander Dębiec
- Clinic of Neurology, Military Institute of Medicine - National Research Institute, Szaserow 128, 04-141, Warsaw, Poland
| | - Stefan Strilciuc
- Department of Genomics, MEDFUTURE Institute for Biomedical Research, Iuliu Hațieganu University of Medicine and Pharmacy, 400337, Cluj-Napoca, Romania
| | | | - Renata Piusinska-Macoch
- Clinic of Neurology, Military Institute of Medicine - National Research Institute, Szaserow 128, 04-141, Warsaw, Poland
| | - David Balo
- Brainomix Ltd., and, Oxford University Hospitals NHSFT, Oxford, UK
| | - George Harston
- Brainomix Ltd., and, Oxford University Hospitals NHSFT, Oxford, UK
| | - Adam Stępień
- Clinic of Neurology, Military Institute of Medicine - National Research Institute, Szaserow 128, 04-141, Warsaw, Poland
| | - Krzysztof Brzozowski
- Department of Radiology, Military Institute of Medicine - National Research Institute, Warsaw, Poland
| | - Piotr Zięcina
- Department of Radiology, Military Institute of Medicine - National Research Institute, Warsaw, Poland
| | - Jerzy Narloch
- Department of Radiology, Military Institute of Medicine - National Research Institute, Warsaw, Poland
| | - Marek Wierzbicki
- Department of Radiology, Military Institute of Medicine - National Research Institute, Warsaw, Poland
| | - Piotr Piasecki
- Department of Radiology, Military Institute of Medicine - National Research Institute, Warsaw, Poland
| |
Collapse
|
3
|
Adil M, Jiba U, Khan A, Shahrukh M, Hasan N, Ahmad FJ. Advancements in ischemic stroke management: Transition from traditional to nanotechnological approaches. J Drug Deliv Sci Technol 2024; 102:106318. [DOI: 10.1016/j.jddst.2024.106318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
4
|
Ulger O, Eş I, Proctor CM, Algin O. Stroke studies in large animals: Prospects of mitochondrial transplantation and enhancing efficiency using hydrogels and nanoparticle-assisted delivery. Ageing Res Rev 2024; 100:102469. [PMID: 39191353 DOI: 10.1016/j.arr.2024.102469] [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: 06/12/2024] [Revised: 08/08/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024]
Abstract
One of the most frequent reasons for mortality and disability today is acute ischemic stroke, which occurs by an abrupt disruption of cerebral circulation. The intricate damage mechanism involves several factors, such as inflammatory response, disturbance of ion balance, loss of energy production, excessive reactive oxygen species and glutamate release, and finally, neuronal death. Stroke research is now carried out using several experimental models and potential therapeutics. Furthermore, studies are being conducted to address the shortcomings of clinical care. A great deal of research is being done on novel pharmacological drugs, mitochondria targeting compounds, and different approaches including brain cooling and new technologies. Still, there are many unanswered questions about disease modeling and treatment strategies. Before these new approaches may be used in therapeutic settings, they must first be tested on large animals, as most of them have been done on rodents. However, there are several limitations to large animal stroke models used for research. In this review, the damage mechanisms in acute ischemic stroke and experimental acute ischemic stroke models are addressed. The current treatment approaches and promising experimental methods such as mitochondrial transplantation, hydrogel-based interventions, and strategies like mitochondria encapsulation and chemical modification, are also examined in this work.
Collapse
Affiliation(s)
- Oner Ulger
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, Ankara 06010, Turkiye; Gulhane Training and Research Hospital, University of Health Sciences, Ankara 06010, Turkiye.
| | - Ismail Eş
- Department of Engineering Science, Institute of Biomedical Engineering (IBME), University of Oxford, Oxford OX3 7DQ, UK
| | - Christopher M Proctor
- Department of Engineering Science, Institute of Biomedical Engineering (IBME), University of Oxford, Oxford OX3 7DQ, UK
| | - Oktay Algin
- Interventional MR Clinical R&D Institute, Ankara University, Ankara 06100, Turkiye; Department of Radiology, Medical Faculty, Ankara University, Ankara 06100, Turkiye; National MR Research Center (UMRAM), Bilkent University, Ankara 06800, Turkiye
| |
Collapse
|
5
|
Liu W, Liu L, Li H, Xie Y, Bai J, Guan J, Qi H, Sun J. Targeted pathophysiological treatment of ischemic stroke using nanoparticle-based drug delivery system. J Nanobiotechnology 2024; 22:499. [PMID: 39164747 PMCID: PMC11337765 DOI: 10.1186/s12951-024-02772-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024] Open
Abstract
Ischemic stroke poses significant challenges in terms of mortality and disability rates globally. A key obstacle to the successful treatment of ischemic stroke lies in the limited efficacy of administering therapeutic agents. Leveraging the unique properties of nanoparticles for brain targeting and crossing the blood-brain barrier, researchers have engineered diverse nanoparticle-based drug delivery systems to improve the therapeutic outcomes of ischemic stroke. This review provides a concise overview of the pathophysiological mechanisms implicated in ischemic stroke, encompassing oxidative stress, glutamate excitotoxicity, neuroinflammation, and cell death, to elucidate potential targets for nanoparticle-based drug delivery systems. Furthermore, the review outlines the classification of nanoparticle-based drug delivery systems according to these distinct physiological processes. This categorization aids in identifying the attributes and commonalities of nanoparticles that target specific pathophysiological pathways in ischemic stroke, thereby facilitating the advancement of nanomedicine development. The review discusses the potential benefits and existing challenges associated with employing nanoparticles in the treatment of ischemic stroke, offering new perspectives on designing efficacious nanoparticles to enhance ischemic stroke treatment outcomes.
Collapse
Affiliation(s)
- Wei Liu
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Lubin Liu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Hong Li
- Clinical Laboratory, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, 266033, China
| | - Yutong Xie
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Ju Bai
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Jialiang Guan
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Hongzhao Qi
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| | - Jinping Sun
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
6
|
Bao L, Liu Y, Jia Q, Chu S, Jiang H, He S. Argon neuroprotection in ischemic stroke and its underlying mechanism. Brain Res Bull 2024; 212:110964. [PMID: 38670471 DOI: 10.1016/j.brainresbull.2024.110964] [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: 02/28/2024] [Revised: 04/04/2024] [Accepted: 04/21/2024] [Indexed: 04/28/2024]
Abstract
Ischemic stroke (IS), primarily caused by cerebrovascular obstruction, results in severe neurological deficits and has emerged as a leading cause of death and disability worldwide. Recently, there has been increasing exploration of the neuroprotective properties of the inert gas argon. Argon has exhibited impressive neuroprotection in many in vivo and ex vivo experiments without signs of adverse effects, coupled with the advantages of being inexpensive and easily available. However, the efficient administration strategy and underlying mechanisms of neuroprotection by argon in IS are still unclear. This review summarizes current research on the neuroprotective effects of argon in IS with the goal to provide effective guidance for argon application and to elucidate the potential mechanisms of argon neuroprotection. Early and appropriate argon administration at as high a concentration as possible offers favorable neuroprotection in IS. Argon inhalation has been shown to provide some long-term protection benefits. Argon provides the anti-oxidative stress, anti-inflammatory and anti-apoptotic cytoprotective effects mainly around Toll-like receptor 2/4 (TLR2/4), mediated by extracellular signal-regulated kinase 1/2 (ERK1/2), nuclear factor (erythroid-derived 2)-like 2 (Nrf2), nuclear factor kappa-B (NF-ĸB) and B-cell leukemia/lymphoma 2 (Bcl-2). Therefore, argon holds significant promise as a novel clinical neuroprotective gas agent for ischemic stroke after further researches to identify the optimal application strategy and elucidate the underlying mechanism.
Collapse
Affiliation(s)
- Li Bao
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China; Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Yongxin Liu
- Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Qi Jia
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China; Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Sihao Chu
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China; Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Han Jiang
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China; Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Shuang He
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China.
| |
Collapse
|
7
|
Medicherla C, Pashun R, Al-Mufti F. Review of Cerebral Collateral Circulation and Insight into Cardiovascular Strategies to Limit Collateral Damage in Ischemic Stroke. Cardiol Rev 2024; 32:188-193. [PMID: 37729598 DOI: 10.1097/crd.0000000000000614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Cerebral collateral circulation is a dynamic and adaptive process by which alternative vascular pathways supply perfusion to ischemic brain tissue in the event of an arterial occlusion. This complicated network of blood vessels that acts as a natural bypass plays a pivotal role in stroke pathophysiology and has become a key area of study given its significance in stroke treatment and patient outcomes. In this review, we will study the factors influencing the formation, recruitment, and endurance of collateral vessels; discuss imaging modalities for quantitative and qualitative assessment of this network; explore the role of collaterals in stroke management; and highlight several cardiovascular strategies to minimize damage to collaterals and optimize stroke outcomes.
Collapse
Affiliation(s)
| | - Raymond Pashun
- Department of Cardiology, New York University Langone Health, New York, NY
| | - Fawaz Al-Mufti
- Department of Neurology, Neurosurgery, and Radiology, New York Medical College, Valhalla, NY
| |
Collapse
|
8
|
Hernández-Bernal F, Estenoz-García D, Gutiérrez-Ronquillo JH, Martín-Bauta Y, Catasús-Álvarez K, Gutiérrez-Castillo M, Guevara-Rodríguez M, Castro-Jeréz A, Fuentes-González Y, Pinto-Cruz Y, Valenzuela-Silva C, Muzio-González VL, Pérez-Saad H, Subirós-Martínez N, Guillén-Nieto GE, Garcia-del-Barco-Herrera D. Combination therapy of Epidermal Growth Factor and Growth Hormone-Releasing Hexapeptide in acute ischemic stroke: a phase I/II non-blinded, randomized clinical trial. Front Neurol 2024; 15:1303402. [PMID: 38638315 PMCID: PMC11024445 DOI: 10.3389/fneur.2024.1303402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 03/14/2024] [Indexed: 04/20/2024] Open
Abstract
Objective This study tested the hypothesis that a neuroprotective combined therapy based on epidermal growth factor (EGF) and growth hormone-releasing hexapeptide (GHRP6) could be safe for acute ischemic stroke patients, admitting up to 30% of serious adverse events (SAE) with proven causality. Methods A multi-centric, randomized, open-label, controlled, phase I-II clinical trial with parallel groups was conducted (July 2017 to January 2018). Patients aged 18-80 years with a computed tomography-confirmed ischemic stroke and less than 12 h from the onset of symptoms were randomly assigned to the study groups I (75 μg rEGF + 3.5 mg GHRP6 i.v., n=10), II (75 μg rEGF + 5 mg GHRP6 i.v., n=10), or III (standard care control, n=16). Combined therapy was given BID for 7 days. The primary endpoint was safety over 6 months. Secondary endpoints included neurological (NIHSS) and functional [Barthel index and modified Rankin scale (mRS)] outcomes. Results The study population had a mean age of 66 ± 11 years, with 21 men (58.3%), a baseline median NIHSS score of 9 (95% CI: 8-11), and a mean time to treatment of 7.3 ± 2.8 h. Analyses were conducted on an intention-to-treat basis. SAEs were reported in 9 of 16 (56.2%) patients in the control group, 3 of 10 (30%) patients in Group I (odds ratio (OR): 0.33; 95% CI: 0.06-1.78), and 2 of 10 (20%) patients in Group II (OR: 0.19; 95% CI: 0.03-1.22); only two events in one patient in Group I were attributed to the intervention treatment. Compliance with the study hypothesis was greater than 0.90 in each group. Patients treated with EGF + GHRP6 had a favorable neurological and functional evolution at both 90 and 180 days, as evidenced by the inferential analysis of NIHSS, Barthel, and mRS and by their moderate to strong effect size. At 6 months, proportion analysis evidenced a higher survival rate for patients treated with the combined therapy. Ancillary analysis including merged treated groups and utility-weighted mRS also showed a benefit of this combined therapy. Conclusion EGF + GHRP6 therapy was safe. The functional benefits of treatment in this study supported a Phase III study. Clinical Trial Registration RPCEC00000214 of the Cuban Public Registry of Clinical Trials, Unique identifier: IG/CIGB-845I/IC/1601.
Collapse
Affiliation(s)
- Francisco Hernández-Bernal
- Clinical Trial Direction, Center for Genetic Engineering and Biotechnology, Havana, Cuba
- Department of Comprehensive General Medicine, Latin American School of Medicine (ELAM), Havana, Cuba
| | | | | | - Yenima Martín-Bauta
- Clinical Trial Direction, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Karen Catasús-Álvarez
- Clinical Trial Direction, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | | | | | | | | | | | | | | | - Héctor Pérez-Saad
- Neuroprotection Project, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Nelvys Subirós-Martínez
- Neuroprotection Project, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Gerardo E. Guillén-Nieto
- Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Havana, Cuba
- Department of Physiology, Latin American School of Medicine (ELAM), Havana, Cuba
| | - Diana Garcia-del-Barco-Herrera
- Neuroprotection Project, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Havana, Cuba
- Department of Physiology, Latin American School of Medicine (ELAM), Havana, Cuba
| |
Collapse
|
9
|
Stavchansky VV, Yuzhakov VV, Sevan'kaeva LE, Fomina NK, Koretskaya AE, Denisova AE, Mozgovoy IV, Gubsky LV, Filippenkov IB, Myasoedov NF, Limborska SA, Dergunova LV. Melanocortin Derivatives Induced Vascularization and Neuroglial Proliferation in the Rat Brain under Conditions of Cerebral Ischemia. Curr Issues Mol Biol 2024; 46:2071-2092. [PMID: 38534749 DOI: 10.3390/cimb46030133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/28/2024] Open
Abstract
Stroke remains the second leading cause of death worldwide. The development of new therapeutic agents focused on restoring vascular function and neuroprotection of viable tissues is required. In this study the neuroprotective activity of melanocortin-like ACTH(4-7)PGP and ACTH(6-9)PGP peptides was investigated in rat brain at 24 h after transient middle cerebral artery occlusion (tMCAO). The severity of ischemic damage, changes in the proliferative activity of neuroglial cells and vascularization of rat brain tissue were analyzed. The administration of peptides resulted in a significant increase in the volume density of neurons in the perifocal zone of infarction compared to rats subjected to ischemia and receiving saline. Immunohistochemical analysis of the proliferative activity of neuroglia cells using PCNA antibodies showed a significant increase in the number of proliferating cells in the penumbra and in the intact cerebral cortex of rats receiving peptide treatment. The effect of peptides on vascularization was examined using CD31 antibodies under tMCAO conditions, revealing a significant increase in the volume density of vessels and their sizes in the penumbra after administration of ACTH(4-7)PGP and ACTH(6-9)PGP. These findings confirm the neuroprotective effect of peptides due to the activation of neuroglia proliferation and the enhancement of collateral blood flow.
Collapse
Affiliation(s)
- Vasily V Stavchansky
- National Research Center "Kurchatov Institute", Kurchatov Sq. 2, Moscow 123182, Russia
| | - Vadim V Yuzhakov
- A. Tsyb Medical Radiological Research Center-Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Koroleva Str. 4B, Obninsk 249036, Russia
| | - Larisa E Sevan'kaeva
- A. Tsyb Medical Radiological Research Center-Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Koroleva Str. 4B, Obninsk 249036, Russia
| | - Natalia K Fomina
- A. Tsyb Medical Radiological Research Center-Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Koroleva Str. 4B, Obninsk 249036, Russia
| | - Anastasia E Koretskaya
- A. Tsyb Medical Radiological Research Center-Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Koroleva Str. 4B, Obninsk 249036, Russia
| | - Alina E Denisova
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, Ostrovitianov Str. 1, Moscow 117997, Russia
| | - Ivan V Mozgovoy
- National Research Center "Kurchatov Institute", Kurchatov Sq. 2, Moscow 123182, Russia
| | - Leonid V Gubsky
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, Ostrovitianov Str. 1, Moscow 117997, Russia
| | - Ivan B Filippenkov
- National Research Center "Kurchatov Institute", Kurchatov Sq. 2, Moscow 123182, Russia
| | - Nikolay F Myasoedov
- National Research Center "Kurchatov Institute", Kurchatov Sq. 2, Moscow 123182, Russia
| | - Svetlana A Limborska
- National Research Center "Kurchatov Institute", Kurchatov Sq. 2, Moscow 123182, Russia
| | - Lyudmila V Dergunova
- National Research Center "Kurchatov Institute", Kurchatov Sq. 2, Moscow 123182, Russia
| |
Collapse
|
10
|
Xie X, Wang L, Dong S, Ge S, Zhu T. Immune regulation of the gut-brain axis and lung-brain axis involved in ischemic stroke. Neural Regen Res 2024; 19:519-528. [PMID: 37721279 PMCID: PMC10581566 DOI: 10.4103/1673-5374.380869] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/11/2023] [Accepted: 06/12/2023] [Indexed: 09/19/2023] Open
Abstract
Local ischemia often causes a series of inflammatory reactions when both brain immune cells and the peripheral immune response are activated. In the human body, the gut and lung are regarded as the key reactional targets that are initiated by brain ischemic attacks. Mucosal microorganisms play an important role in immune regulation and metabolism and affect blood-brain barrier permeability. In addition to the relationship between peripheral organs and central areas and the intestine and lung also interact among each other. Here, we review the molecular and cellular immune mechanisms involved in the pathways of inflammation across the gut-brain axis and lung-brain axis. We found that abnormal intestinal flora, the intestinal microenvironment, lung infection, chronic diseases, and mechanical ventilation can worsen the outcome of ischemic stroke. This review also introduces the influence of the brain on the gut and lungs after stroke, highlighting the bidirectional feedback effect among the gut, lungs, and brain.
Collapse
Affiliation(s)
- Xiaodi Xie
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Lei Wang
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Shanshan Dong
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - ShanChun Ge
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Ting Zhu
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
11
|
Dong X, Li C, Yao Y, Liu F, Jiang P, Gao Y. Xingnaojing injection alleviates cerebral ischemia/reperfusion injury through regulating endoplasmic reticulum stress in Vivo and in Vitro. Heliyon 2024; 10:e25267. [PMID: 38327400 PMCID: PMC10847655 DOI: 10.1016/j.heliyon.2024.e25267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/09/2024] Open
Abstract
Background Xingnaojing (XNJ) injection, an extract derived from traditional Chinese medicine, is commonly used to treat ischemic stroke (IS). Previous studies have shown that XNJ has the ability to alleviate apoptosis in cerebral ischemia-reperfusion injury. However, the potential mechanisms have not been clarified. Objective To identify the neuroprotective effect of XNJ and explore whether XNJ inhibits cell apoptosis associated with endoplasmic reticulum stress (ERS) after IS. Methods In this study, cultured hippocampal neurons from mouse embryos and Sprague-Dawley rats were assigned randomly to four groups: sham, model, XNJ, and edaravone. The treatment groups were administered 2 h after modelling. Neurological deficit scores and motor performance tests were performed after 24 h of modelling. Additionally, pathomorphology, cell apoptosis and calcium content were evaluated. To ascertain the expression of ERS proteins, western blotting and polymerase chain reaction were employed. Results The results indicated that XNJ treatment resulted in a notable decrease in infarct volume, apoptosis and missteps compared with the model group. XNJ also exhibited improvements in neurological function, grip strength and motor time. The calcium content significantly reduced in XNJ group. The XNJ administration resulted in a reduction in the levels of proteins associated with ERS including CHOP, GRP78, Bax, caspase-12, caspase-9, and cleaved-caspase-3, but an increase of the Bcl-2/Bax ratio. Furthermore, the downregulation of mRNA expression of CHOP, GRP78, caspase-12, caspase-9, and caspase-3 was confirmed in both cultured neurons and rat model. Conclusion These findings suggest that XNJ may alleviate apoptosis by modulating the ERS-induced apoptosis pathway, making it a potential novel therapeutic approach for ischemic stroke.
Collapse
Affiliation(s)
- Xinglu Dong
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Chuanpeng Li
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
- Chinese Medicine Key Research Room of Brain Disorders Syndrome and Treatment of the National Administration of Traditional Chinese Medicine, Beijing, China
| | - Yaoyao Yao
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
- Chinese Medicine Key Research Room of Brain Disorders Syndrome and Treatment of the National Administration of Traditional Chinese Medicine, Beijing, China
| | - Fengzhi Liu
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
- Chinese Medicine Key Research Room of Brain Disorders Syndrome and Treatment of the National Administration of Traditional Chinese Medicine, Beijing, China
| | - Ping Jiang
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Ying Gao
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
- Chinese Medicine Key Research Room of Brain Disorders Syndrome and Treatment of the National Administration of Traditional Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
12
|
Zhang W, Zhang P, Xu LH, Gao K, Zhang JL, Yao MN, Li RL, Guo C, Wang JW, Wu QX. Ethanol extract of Verbena officinalis alleviates MCAO-induced ischaemic stroke by inhibiting IL17A pathway-regulated neuroinflammation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155237. [PMID: 38056148 DOI: 10.1016/j.phymed.2023.155237] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/28/2023] [Accepted: 11/22/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND The prevention and treatment of ischaemic stroke is a worldwide challenge, and effective clinical treatment strategies are lacking. Studies have demonstrated the efficacy of Verbena officinalis in managing cerebrovascular disorders. However, the neuroprotective bioactive components and mechanisms remain unclear. PURPOSE To investigate the pharmacological combinatorial components and mechanism underlying the anti-ischemic stroke effect of the ethanol extract of Verbena officinalis (VO Ex). STUDY DESIGN AND METHODS The components of VO Ex were identified by HPLC. A middle cerebral artery occlusion (MCAO) induced brain injury model was used to assess the therapeutic effect of VO Ex. The activity of the chemical components of VO Ex was evaluated using a primary astrocyte injury model induced by oxygen-glucose deprivation/reperfusion (OGD/R). RNA sequencing was used to reveal the potential targets of VO Ex against cerebral ischemia-reperfusion injury (CIRI), and the results were verified by qRT-PCR and western blotting. The key components and target binding ability were predicted by molecular docking. Finally, the mechanism of combinatorial components was verified by experiments. RESULTS The HPLC results indicated that the main ingredients of VO Ex were hastatoside, verbenalin, acteoside, luteolin, apigenin and hispidulin. In vivo experiments showed that VO Ex improved MCAO-induced acute cerebral ischemic injury. Transcriptomic data and biological experiments suggested that VO Ex exerted therapeutic effects through IL17A signalling pathways. The in vitro experiments indicated that verbenalin, acteoside, luteolin, apigenin and hispidulin exhibited neuroprotective activities. The novel formula of VALAH, derived from the aforementioned active ingredients, exhibited superior efficacy compared to each individual component. Molecular docking and mechanistic studies have confirmed that VALAH functions in the treatment of ischaemic stroke by suppressing the activation of the IL17A signalling pathway. CONCLUSION This work is the first to reveal that VO Ex effectively inhibits the IL17A signaling pathway and mitigates neuroinflammation following ischemic stroke. Moreover, we identified the novel formula VALAH as the bioactive combinatorial components for VO Ex. Further research suggests that the activity of VALAH is associated with IL17A-mediated regulation of neuroinflammation. This finding provides new insights into the efficacious components and mechanisms of traditional Chinese medicine.
Collapse
Affiliation(s)
- Wei Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China; Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Peng Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Le-Hua Xu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Kai Gao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Juan-Li Zhang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Min-Na Yao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Rui-Li Li
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Chao Guo
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jing-Wen Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Quan-Xiang Wu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
13
|
Wei X, Wen Y, Hu Y, Guo X. Total Saponins of Panax Notoginseng Modulate the Astrocyte Inflammatory Signaling Pathway and Attenuate Inflammatory Injury Induced by Oxygen- Glucose Deprivation/Reperfusion Injury in Rat Brain Microvascular Endothelial Cells. Curr Stem Cell Res Ther 2024; 19:267-276. [PMID: 37218204 DOI: 10.2174/1574888x18666230509113912] [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: 12/07/2022] [Revised: 03/08/2023] [Accepted: 03/20/2023] [Indexed: 05/24/2023]
Abstract
OBJECTIVE Reperfusion after cerebral ischemia causes brain injury. Total saponins of Panax notoginseng (PNS) have potential roles in protecting against cerebral ischemia-reperfusion injury. However, whether PNS regulates astrocytes on oxygen-glucose deprivation/reperfusion (OGD/R) injury in rat brain microvascular endothelial cells (BMECs) and its mechanism still need further clarification. METHODS Rat C6 glial cells were treated with PNS at different doses. Cell models were established by exposing C6 glial cells and BMECs to OGD/R. Cell viability was assessed, and levels of nitrite concentration, inflammatory factors (iNOS, IL-1β, IL-6, IL-8, TNF-α), and oxidative stress-related factors (MDA, SOD, GSH-Px, T-AOC) were subsequently measured through CCK8, Grice analysis, Western blot, and ELISA, respectively. The co-cultured C6 and endothelial cells were treated with PNS for 24 hours before model establishment. Then transendothelial electrical resistance (TEER), lactate dehydrogenase (LDH) activity, brain-derived neurotrophic factor (BDNF) content, and mRNA and protein levels and positive rates of tight junction proteins [Claudin-5, Occludin, ZO-1] were measured by a cell resistance meter, corresponding kits, ELISA, RT-qPCR, Western blot, and immunohistochemistry, respectively. RESULTS PNS had no cytotoxicity. PNS reduced iNOS, IL-1β, IL-6, IL-8, and TNF-α levels in astrocytes, promoted T-AOC level and SOD and GSH-Px activities, and inhibited MDA levels, thus inhibiting oxidative stress in astrocytes. In addition, PNS alleviated OGD/R injury, reduced Na-Flu permeability, and enhanced TEER, LDH activity, BDNF content, and levels of tight junction proteins Claudin-5, Occludin, ZO-1 in the culture system of astrocytes and rat BMECs after OGD/R. CONCLUSION PNS repressed astrocyte inflammation and attenuated OGD/R injury in rat BMECs.
Collapse
Affiliation(s)
- Xiaobing Wei
- Department of Neurosurgery, Huizhou Third People's Hospital, Guangzhou Medical University, No. 1 Xuebei Street, Huicheng District, Huizhou, Guangdong, China
| | - Yiqi Wen
- Department of Neurosurgery, Huizhou Third People's Hospital, Guangzhou Medical University, No. 1 Xuebei Street, Huicheng District, Huizhou, Guangdong, China
| | - Yongzhen Hu
- Department of Neurosurgery, Huizhou Third People's Hospital, Guangzhou Medical University, No. 1 Xuebei Street, Huicheng District, Huizhou, Guangdong, China
| | - Xuli Guo
- Department of Internal Medicine-Oncology, Huizhou Municipal Central Hospital, No. 41 Eleng North Road, Huicheng District, Huizhou, Guangdong, China
| |
Collapse
|
14
|
López-Morales MA, Castelló-Ruiz M, Burguete MC, Hervás D, Pérez-Pinzón MA, Salom JB. Effect and mechanisms of resveratrol in animal models of ischemic stroke: A systematic review and Bayesian meta-analysis. J Cereb Blood Flow Metab 2023; 43:2013-2028. [PMID: 37802493 PMCID: PMC10925864 DOI: 10.1177/0271678x231206236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 08/03/2023] [Accepted: 09/20/2023] [Indexed: 10/10/2023]
Abstract
Resveratrol (RSV) holds promise as cerebroprotective treatment in cerebral ischemia. This systematic review aims to assess the effects and mechanisms of RSV in animal models of ischemic stroke. We searched Medline, Embase and Web of Science to identify 75 and 57 eligible rodent studies for qualitative and quantitative syntheses, respectively. Range of evidence met 10 of 13 STAIR criteria. Median (Q1, Q3) quality score was 7 (5, 8) on the CAMARADES 15-item checklist. Bayesian meta-analysis showed SMD estimates (95% CI) favoring RSV: infarct size (-1.72 [-2.03; -1.41]), edema size (-1.61 [-2.24; -0.98]), BBB impairment (-1.85 [-2.54; -1.19]), neurofunctional impairment (-1.60 [-1.92; -1.29]), and motor performance (1.39 [0.64; 2.08]); and less probably neuronal survival (0.63 [-1.40; 2.48]) and apoptosis (-0.96 [-2.87; 1.02]). Species (rat vs mouse) was associated to a larger benefit. Sensitivity analyses confirmed robustness of the estimates. Reduction of oxidative stress, inflammation, and apoptosis underlie these effects. Our results quantitatively state the beneficial effects of RSV on structural and functional outcomes in rodent stroke models, update the evidence on the mechanisms of action, and provide an exhaustive list of targeted signaling pathways. Current evidence highlights the need for conducting further high-quality preclinical research to better inform clinical research.
Collapse
Affiliation(s)
- Mikahela A López-Morales
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - María Castelló-Ruiz
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, Valencia, Spain
| | - María C Burguete
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - David Hervás
- Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain
| | - Miguel A Pérez-Pinzón
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Department of Neurology, Miller School of Medicine, University of Miami, Miami, USA
| | - Juan B Salom
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| |
Collapse
|
15
|
Montes P, Ortíz-Islas E, Rodríguez-Pérez CE, Ruiz-Sánchez E, Silva-Adaya D, Pichardo-Rojas P, Campos-Peña V. Neuroprotective-Neurorestorative Effects Induced by Progesterone on Global Cerebral Ischemia: A Narrative Review. Pharmaceutics 2023; 15:2697. [PMID: 38140038 PMCID: PMC10747486 DOI: 10.3390/pharmaceutics15122697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/14/2023] [Accepted: 11/18/2023] [Indexed: 12/24/2023] Open
Abstract
Progesterone (P4) is a neuroactive hormone having pleiotropic effects, supporting its pharmacological potential to treat global (cardiac-arrest-related) cerebral ischemia, a condition associated with an elevated risk of dementia. This review examines the current biochemical, morphological, and functional evidence showing the neuroprotective/neurorestorative effects of P4 against global cerebral ischemia (GCI). Experimental findings show that P4 may counteract pathophysiological mechanisms and/or regulate endogenous mechanisms of plasticity induced by GCI. According to this, P4 treatment consistently improves the performance of cognitive functions, such as learning and memory, impaired by GCI. This functional recovery is related to the significant morphological preservation of brain structures vulnerable to ischemia when the hormone is administered before and/or after a moderate ischemic episode; and with long-term adaptive plastic restoration processes of altered brain morphology when treatment is given after an episode of severe ischemia. The insights presented here may be a guide for future basic research, including the study of P4 administration schemes that focus on promoting its post-ischemia neurorestorative effect. Furthermore, considering that functional recovery is a desired endpoint of pharmacological strategies in the clinic, they could support the study of P4 treatment for decreasing dementia in patients who have suffered an episode of GCI.
Collapse
Affiliation(s)
- Pedro Montes
- Laboratorio de Neuroinmunoendocrinología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico
| | - Emma Ortíz-Islas
- Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (E.O.-I.); (C.E.R.-P.)
| | - Citlali Ekaterina Rodríguez-Pérez
- Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (E.O.-I.); (C.E.R.-P.)
| | - Elizabeth Ruiz-Sánchez
- Laboratorio de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico;
| | - Daniela Silva-Adaya
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico;
| | - Pavel Pichardo-Rojas
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030, USA;
| | - Victoria Campos-Peña
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico;
| |
Collapse
|
16
|
Lino MM, Rondão T, Banerjee A, Aires I, Rodrigues M, Reis T, Santinha A, Fernandes D, Serrenho D, Sobrino T, Sargento-Freitas J, Pereira FC, Carvalho AL, Ferreira L. Small extracellular vesicles administered directly in the brain promote neuroprotection and decreased microglia reactivity in a stroke mouse model. NANOSCALE 2023; 15:18212-18217. [PMID: 37933179 DOI: 10.1039/d3nr03861k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Herein, we investigate the bioactivity of small extracellular vesicles (sEVs), focusing on their local effect in the brain. sEVs from mononuclear cells (MNCs) showed superior effects in vitro to sEVs from mesenchymal stem cells (MSCs) and were able to promote neuroprotection and decrease microglia reactivity in a stroke mouse model.
Collapse
Affiliation(s)
- Miguel M Lino
- CNC - Centre for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.
| | - Tiago Rondão
- CNC - Centre for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.
| | - Arnab Banerjee
- CNC - Centre for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.
| | - Inês Aires
- CNC - Centre for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.
| | - Magda Rodrigues
- CNC - Centre for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.
| | - Tiago Reis
- CNC - Centre for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.
| | - António Santinha
- CNC - Centre for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.
| | - Dominique Fernandes
- CNC - Centre for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.
| | - Débora Serrenho
- CNC - Centre for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Portugal
| | - Tomás Sobrino
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | | | - Frederico C Pereira
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- iCBR, Coimbra Institute for Clinical and Biomedical Research, CIBB - Centre for Innovative Biomedicine and Biotechnology, Coimbra, Portugal
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Portugal
| | - Ana Luísa Carvalho
- CNC - Centre for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Lino Ferreira
- CNC - Centre for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
17
|
Li Z, Gong P, Zhang M, Li C, Xiao P, Yu M, Wang X, An L, Bi F, Song X, Wang X. Multi-parametric MRI assessment of melatonin regulating the polarization of microglia in rats after cerebral ischemia/reperfusion injury. Brain Res Bull 2023; 204:110807. [PMID: 37923146 DOI: 10.1016/j.brainresbull.2023.110807] [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: 08/19/2023] [Revised: 10/15/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
OBJECTIVES Multi-parametric magnetic resonance imaging (MRI) can provide comprehensive and valuable information for precise diagnosis and treatment evaluation of a number of diseases. In this study, the neuroprotective effects of melatonin (Mel) on a rat model of cerebral ischemia/reperfusion injury (CIRI) were assessed by multi-parametric MRI combined with histopathological techniques for longitudinal monitoring of the lesion microenvironment. METHODS Sixty Sprague Dawley (SD) rats were randomly divided into three groups: the Sham, CIRI and CIRI + Mel groups. At multiple time points after ischemia, MRI scanning was performed on a 7.0 Tesla MRI scanner. Multi-parametric MRI includes T2-weighted imaging (T2WI), diffusion weighted imaging (DWI), and chemical exchange saturation transfer (CEST)-MRI. CEST effects were calculated by the Lorentzian difference method, 3.5 ppm indicates amide protons of mobile proteins/peptide (Amide-CEST) and 2.0 ppm indicates amine protons (Guan-CEST). Multiple histopathological techniques were used to examine the histopathological changes and explore the therapeutic effects of Mel. RESULTS T2WI and DWI-MRI could localize the infarct foci and areas in CIRI rats, which was further validated by staining, 2, 3, 5-triphenyl tetrazolium chloride (TTC) staining, hematoxylin and eosin (H&E) staining, and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick end labelling (TUNEL) staining. After Mel treatment, T2WI and DWI-MRI showed smaller infarct volume, and neurons displayed improved morphology with less apoptosis rates. Notably, Amide-CEST and Guan-CEST signal decreased as early as 2 h after CIRI (all P <0.001), reflecting the change of pH after ischemia. After Mel treatment, both Amide-CEST and Guan-CEST signal increased in ischemic cortex and striatum compared with control group (all P < 0.001). The immunofluorescence staining and western blotting analysis suggested the expression of M2 microglia increased after Mel treatment; While,after Mel treatment the inflammatory factor interleukin-1β (IL-1β) decreased compared with control CIRI rats. CONCLUSIONS Multi-parametric MRI was shown to be an effective method to monitor the brain damage in a rat model of CIRI and assess the therapeutic effects of Mel treatment. Amide-CEST and Guan-CEST were especially sensitive to the changes in brain microenvironment during the early stage after CIRI. Furthermore, the neuroprotective effect of Mel treatment is associated with its promotion of the microglia polarized to M2 type in CIRI rats.
Collapse
Affiliation(s)
- Zhen Li
- School of Medical Imaging, Weifang Medical University, Weifang 261053, Shandong Province, China; Medical Imaging Center, Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| | - Ping Gong
- Medical Imaging Center, Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| | - Mengbei Zhang
- School of Medical Imaging, Weifang Medical University, Weifang 261053, Shandong Province, China; Department of Radiology, Zibo Central Hospital, Zibo 255020, Shandong Province, China
| | - Chen Li
- School of Medical Imaging, Weifang Medical University, Weifang 261053, Shandong Province, China
| | - Peilun Xiao
- Department of Anatomy, School of Basic Medicine, Weifang Medical University, Weifang 261053, Shandong Province, China
| | - Miao Yu
- School of Medical Imaging, Weifang Medical University, Weifang 261053, Shandong Province, China
| | - Xizhen Wang
- Medical Imaging Center, Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| | - Lin An
- School of Medical Imaging, Weifang Medical University, Weifang 261053, Shandong Province, China
| | - Fangfang Bi
- Department of Neurology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, Guangdong Province, China.
| | - Xiaolei Song
- Center for Biomedical Imaging Research, School of Medicine, Tsinghua University, Beijing 100084, China.
| | - Xiaoli Wang
- School of Medical Imaging, Weifang Medical University, Weifang 261053, Shandong Province, China; Medical Imaging Center, Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China.
| |
Collapse
|
18
|
Li M, Gao Y, Jiang M, Zhang H, Zhang Y, Wu Y, Zhou W, Wu D, Wu C, Wu L, Bao L, Ge X, Qi Z, Wei M, Li A, Ding Y, Zhang J, Pan G, Wu Y, Cheng Y, Zheng Y, Ji X. Dual-sized hollow particle incorporated fibroin thermal insulating coatings on catheter for cerebral therapeutic hypothermia. Bioact Mater 2023; 26:116-127. [PMID: 36879558 PMCID: PMC9984786 DOI: 10.1016/j.bioactmat.2023.02.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 02/19/2023] [Accepted: 02/19/2023] [Indexed: 02/27/2023] Open
Abstract
Selective endovascular hypothermia has been used to provide cooling-induced cerebral neuroprotection, but current catheters do not support thermally-insulated transfer of cold infusate, which results in an increased exit temperature, causes hemodilution, and limits its cooling efficiency. Herein, air-sprayed fibroin/silica-based coatings combined with chemical vapor deposited parylene-C capping film was prepared on catheter. This coating features in dual-sized-hollow-microparticle incorporated structures with low thermal conductivity. The infusate exit temperature is tunable by adjusting the coating thickness and infusion rate. No peeling or cracking was observed on the coatings under bending and rotational scenarios in the vascular models. Its efficiency was verified in a swine model, and the outlet temperature of coated catheter (75 μm thickness) was 1.8-2.0 °C lower than that of the uncoated one. This pioneering work on catheter thermal insulation coatings may facilitate the clinical translation of selective endovascular hypothermia for neuroprotection in patients with acute ischemic stroke.
Collapse
Affiliation(s)
- Ming Li
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yuan Gao
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, 100191, China
| | - Miaowen Jiang
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Hongkang Zhang
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Yang Zhang
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yan Wu
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Wenhao Zhou
- Shaanxi Key Laboratory of Biomedical Metallic Materials, Northwest Institute for Nonferrous Metal Research, Xi'an, 710016, China
| | - Di Wu
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Chuanjie Wu
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Longfei Wu
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Luzi Bao
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Xiaoxiao Ge
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Zhengfei Qi
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Ming Wei
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Ang Li
- Department of Biomedical Engineering, Columbia University, New York City, NY, 10027, USA
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Jicheng Zhang
- Gong Yi Van-research Innovation Composite Material Co. Ltd, Zheng Zhou, 451299, China
| | - Guangzhen Pan
- Gong Yi Van-research Innovation Composite Material Co. Ltd, Zheng Zhou, 451299, China
| | - Yu Wu
- Gong Yi Van-research Innovation Composite Material Co. Ltd, Zheng Zhou, 451299, China
| | - Yan Cheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Xunming Ji
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.,School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, 100191, China.,Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China.,Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| |
Collapse
|
19
|
Ho YJ, Cheng HL, Liao LD, Lin YC, Tsai HC, Yeh CK. Oxygen-loaded microbubble-mediated sonoperfusion and oxygenation for neuroprotection after ischemic stroke reperfusion. Biomater Res 2023; 27:65. [PMID: 37415210 DOI: 10.1186/s40824-023-00400-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/21/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND Ischemic stroke-reperfusion (S/R) injury is a crucial issue in the protection of brain function after thrombolysis. The vasodilation induced by ultrasound (US)-stimulated microbubble cavitation has been applied to reduce S/R injury through sonoperfusion. The present study uses oxygen-loaded microbubbles (OMBs) with US stimulation to provide sonoperfusion and local oxygen therapy for the reduction of brain infarct size and neuroprotection after S/R. METHODS The murine S/R model was established by photodynamic thrombosis and thrombolysis at the remote branch of the anterior cerebral artery. In vivo blood flow, partial oxygen pressure (pO2), and brain infarct staining were examined to analyze the validity of the animal model and OMB treatment results. The animal behaviors and measurement of the brain infarct area were used to evaluate long-term recovery of brain function. RESULTS The percentage of blood flow was 45 ± 3%, 70 ± 3%, and 86 ± 2% after 60 min stroke, 20 min reperfusion, and 10 min OMB treatment, respectively, demonstrating sonoperfusion, and the corresponding pO2 level was 60 ± 1%, 76 ± 2%, and 79 ± 4%, showing reoxygenation. After 14 days of treatment, a 87 ± 3% reduction in brain infarction and recovery of limb coordination were observed in S/R mice. The expression of NF-κB, HIF-1α, IL-1β, and MMP-9 was inhibited and that of eNOS, BDNF, Bcl2, and IL-10 was enhanced, indicating activation of anti-inflammatory and anti-apoptosis responses and neuroprotection. Our study demonstrated that OMB treatment combines the beneficial effects of sonoperfusion and local oxygen therapy to reduce brain infarction and activate neuroprotection to prevent S/R injury.
Collapse
Affiliation(s)
- Yi-Ju Ho
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Hsiang-Lung Cheng
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu, 30013, Taiwan
| | - Lun-De Liao
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Taiwan
| | - Yu-Chun Lin
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Hong-Chieh Tsai
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, No.5Fuxing St.Guishan Dist., Taoyuan City, 333, Taiwan.
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu, 30013, Taiwan.
| |
Collapse
|
20
|
Zhang P, You S, Ding X, Luan P, Xu J, Cui Q, Wang F, Li R, Zhu Y, Zhang J. Protective effect and underlying mechanism of muscone on acute cerebral ischemia-reperfusion injury in rats. JOURNAL OF ETHNOPHARMACOLOGY 2023; 308:116287. [PMID: 36841376 DOI: 10.1016/j.jep.2023.116287] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 02/12/2023] [Accepted: 02/14/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Musk is a widely used traditional Chinese medicine, which has resuscitation, activating blood, and disperse swelling effects. Musk is commonly used in the prevention of myocardial infarction and ischemic stroke, and muscone is its main active component. AIM OF THE STUDY The effect and mechanism of muscone to improve the condition of ischemic stroke is not clear, accordingly, we verified its efficacy in ischemia-reperfused rats, and investigated its mechanism by PC12 and THP-1 cells. METHODS A transient middle cerebral artery occlusion (tMCAO) rat model was established for in vivo experiments. 2,3,5-Triphenyl Tetrazolium Chloride (TTC) staining was used to calculate infarct rate. Neuroprotection and angiogenesis were assessed by Hematoxylin-eosin (HE) staining, nissl staining, immunofluorescence staining, and quantitative real-time PCR (qRT-PCR). Oxygen glucose deprivation-reperfusion (OGD/R) model of PC12 cells was established for neuroprotection analysis, where CCK-8 assay was used to measure cell viability, flow cytometry and Hoechst 33258 staining were used to demonstrate apoptosis, and protein levels were detected by Western blot. For angiogenesis analysis, enzyme-linked immunosorbent assay (ELISA) and qRT-PCR were used to detect angiogenic factors expressed by THP-1. Cell viability assay, scratch wound assay, and tube formation assay were used to evaluate angiogenic effect of HUVECs treated with medium of THP-1. And the angiogenic pathway in HUVECs was detected by Western blot. RESULTS According to the results, in cerebral ischemia-reperfusion rats, the infarct rate and tissue damage were significantly reduced by muscone, and the expression of neurotrophic factors and angiogenesis-related factors were all elevated. In OGD/R-PC12 cell models, muscone could increase cell viability and inhibit apoptosis via Bax/Bcl-2/Caspase-3 pathway. In THP-1-mediated angiogenesis of HUVECs, muscone promoted the secretion of angiogenesis-related factors in THP-1 and thus indirectly promoted the proliferation, migration and tube formation of HUVECs, and then regulated phosphorylation of VEGFR2 and Akt in HUVECs. CONCLUSIONS Our study indicated that muscone may be a potential neuroprotective and proangiogenic agent in cerebral ischemia.
Collapse
Affiliation(s)
- Pei Zhang
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional, Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Suxin You
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional, Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xinyue Ding
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional, Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Putuo District Central Hospital of Shanghai, Shanghai, 200062, China
| | - Pengwei Luan
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional, Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jiazhen Xu
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional, Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qianfei Cui
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional, Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Feiyun Wang
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional, Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ruixiang Li
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional, Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuying Zhu
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional, Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Jiange Zhang
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional, Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
21
|
Zhou Q, Dong Y, Wang K, Wang Z, Ma B, Yang B. A comprehensive analysis of the hub genes for oxidative stress in ischemic stroke. Front Neurosci 2023; 17:1166010. [PMID: 37229425 PMCID: PMC10203175 DOI: 10.3389/fnins.2023.1166010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/06/2023] [Indexed: 05/27/2023] Open
Abstract
Ischemic stroke (IS), resulting from the occlusion of the cerebral artery and subsequent interruption of blood flow, represents a major and critical threat to public health. Oxidative stress (OS) has been confirmed to play a role in the IS pathological process and neural death. Understanding the essential role of OS-related genes in ischemic stroke is critical to understanding the current perception of the pathophysiological process in IS. Herein, by integrating three IS datasets (GSE16561, GSE22255, and GSE58294), we divided IS samples into the low- and high-OS groups by calculating the OS score identified by the oxidative stress gene set. The functional enrichment analysis of differentially expressed genes (DEGs) between the low- and high-OS groups indicated that DEGs were associated with hypoxia, the inflammatory response, and oxidative phosphorylation pathways. Furthermore, nine hub genes (namely TLR1, CXCL1, MMP9, TLR4, IL1R2, EGR1, FOS, CXCL10, and DUSP1) were identified through the Girvan-Newman algorithm and cytoHubba algorithms. Nine hub genes were highly expressed in IS samples and positively related to neutrophils and macrophages. Drug-sensitive analysis targeting hub genes defined allopurinol and nickel sulfate as potential candidates for impairing the neural death caused by oxidative stress in IS. Finally, we employed five machine learning methods to check the efficacy of the predictive model identified by nine hub genes. The results showed that our model had superior power for predicting the OS activity of IS patients. TLR4 was found to have excellent diagnostic value and a wide-spectrum interaction with other hub genes. Our research emphasized the impact of oxidative stress on ischemic stroke, which supports the idea that antioxidants hold great promise in ischemic stroke therapy.
Collapse
Affiliation(s)
- Qing Zhou
- Rehabilitation Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Dong
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Wang
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ziyan Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bingquan Ma
- Rehabilitation Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bo Yang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
22
|
Kittur FS, Hung CY, Li PA, Sane DC, Xie J. Asialo-rhuEPO as a Potential Neuroprotectant for Ischemic Stroke Treatment. Pharmaceuticals (Basel) 2023; 16:610. [PMID: 37111367 PMCID: PMC10143832 DOI: 10.3390/ph16040610] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Neuroprotective drugs to protect the brain against cerebral ischemia and reperfusion (I/R) injury are urgently needed. Mammalian cell-produced recombinant human erythropoietin (rhuEPOM) has been demonstrated to have excellent neuroprotective functions in preclinical studies, but its neuroprotective properties could not be consistently translated in clinical trials. The clinical failure of rhuEPOM was thought to be mainly due to its erythropoietic activity-associated side effects. To exploit its tissue-protective property, various EPO derivatives with tissue-protective function only have been developed. Among them, asialo-rhuEPO, lacking terminal sialic acid residues, was shown to be neuroprotective but non-erythropoietic. Asialo-rhuEPO can be prepared by enzymatic removal of sialic acid residues from rhuEPOM (asialo-rhuEPOE) or by expressing human EPO gene in glycoengineered transgenic plants (asialo-rhuEPOP). Both types of asialo-rhuEPO, like rhuEPOM, displayed excellent neuroprotective effects by regulating multiple cellular pathways in cerebral I/R animal models. In this review, we describe the structure and properties of EPO and asialo-rhuEPO, summarize the progress on neuroprotective studies of asialo-rhuEPO and rhuEPOM, discuss potential reasons for the clinical failure of rhuEPOM with acute ischemic stroke patients, and advocate future studies needed to develop asialo-rhuEPO as a multimodal neuroprotectant for ischemic stroke treatment.
Collapse
Affiliation(s)
- Farooqahmed S. Kittur
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| | - Chiu-Yueh Hung
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| | - P. Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| | - David C. Sane
- Carilion Clinic and Virginia Tech Carilion School of Medicine, Roanoke, VA 24014, USA;
| | - Jiahua Xie
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| |
Collapse
|
23
|
Rajendram P, Ikram A, Fisher M. Combined Therapeutics: Future Opportunities for Co-therapy with Thrombectomy. Neurotherapeutics 2023; 20:693-704. [PMID: 36943636 PMCID: PMC10275848 DOI: 10.1007/s13311-023-01369-1] [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] [Accepted: 03/14/2023] [Indexed: 03/23/2023] Open
Abstract
Stroke is an urgent public health issue with millions of patients worldwide living with its devastating effects. The advent of thrombolysis and endovascular thrombectomy has transformed the hyperacute care of these patients. However, a significant proportion of patients receiving these therapies still goes on to have unfavorable outcomes and many more remain ineligible for these therapies based on our current guidelines. The future of stroke care will depend on an expansion of the scope of thrombolysis and endovascular thrombectomy to patients outside traditional time windows, more distal occlusions, and large vessel occlusions with mild clinical deficits, for whom clinical trial results have not proven therapeutic efficacy. Novel cytoprotective therapies targeting the ischemic cascade and reperfusion injury therapy, in combination with our existing treatment modalities, should be explored to further improve outcomes for these patients with acute ischemic stroke. In this review, we will review the current status of thrombolysis and thrombectomy, suggest additional data that is needed to enhance these therapies, and discuss how cytoprotection might be combined with thrombectomy.
Collapse
Affiliation(s)
- Phavalan Rajendram
- Division of Stroke and Cerebrovascular Diseases, Department of Neurology, Harvard Medical School, Beth Israel Deaconess Medical Center, Palmer Building Room 127, 330 Brookline Avenue, Boston, MA, 02215-5400, USA.
| | - Asad Ikram
- Division of Stroke and Cerebrovascular Diseases, Department of Neurology, Harvard Medical School, Beth Israel Deaconess Medical Center, Palmer Building Room 127, 330 Brookline Avenue, Boston, MA, 02215-5400, USA
| | - Marc Fisher
- Division of Stroke and Cerebrovascular Diseases, Department of Neurology, Harvard Medical School, Beth Israel Deaconess Medical Center, Palmer Building Room 127, 330 Brookline Avenue, Boston, MA, 02215-5400, USA
| |
Collapse
|
24
|
Qiao C, Liu Z, Qie S. The Implications of Microglial Regulation in Neuroplasticity-Dependent Stroke Recovery. Biomolecules 2023; 13:biom13030571. [PMID: 36979506 PMCID: PMC10046452 DOI: 10.3390/biom13030571] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/23/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Stroke causes varying degrees of neurological deficits, leading to corresponding dysfunctions. There are different therapeutic principles for each stage of pathological development. Neuroprotection is the main treatment in the acute phase, and functional recovery becomes primary in the subacute and chronic phases. Neuroplasticity is considered the basis of functional restoration and neurological rehabilitation after stroke, including the remodeling of dendrites and dendritic spines, axonal sprouting, myelin regeneration, synapse shaping, and neurogenesis. Spatiotemporal development affects the spontaneous rewiring of neural circuits and brain networks. Microglia are resident immune cells in the brain that contribute to homeostasis under physiological conditions. Microglia are activated immediately after stroke, and phenotypic polarization changes and phagocytic function are crucial for regulating focal and global brain inflammation and neurological recovery. We have previously shown that the development of neuroplasticity is spatiotemporally consistent with microglial activation, suggesting that microglia may have a profound impact on neuroplasticity after stroke and may be a key therapeutic target for post-stroke rehabilitation. In this review, we explore the impact of neuroplasticity on post-stroke restoration as well as the functions and mechanisms of microglial activation, polarization, and phagocytosis. This is followed by a summary of microglia-targeted rehabilitative interventions that influence neuroplasticity and promote stroke recovery.
Collapse
Affiliation(s)
- Chenye Qiao
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Zongjian Liu
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Shuyan Qie
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| |
Collapse
|
25
|
Liu A, Hu J, Yeh TS, Wang C, Tang J, Huang X, Chen B, Huangfu L, Yu W, Zhang L. Neuroprotective Strategies for Stroke by Natural Products: Advances and Perspectives. Curr Neuropharmacol 2023; 21:2283-2309. [PMID: 37458258 PMCID: PMC10556387 DOI: 10.2174/1570159x21666230717144752] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 09/09/2023] Open
Abstract
Cerebral ischemic stroke is a disease with high prevalence and incidence. Its management focuses on rapid reperfusion with intravenous thrombolysis and endovascular thrombectomy. Both therapeutic strategies reduce disability, but the therapy time window is short, and the risk of bleeding is high. Natural products (NPs) have played a key role in drug discovery, especially for cancer and infectious diseases. However, they have made little progress in clinical translation and pose challenges to the treatment of stroke. Recently, with the investigation of precise mechanisms in cerebral ischemic stroke and the technological development of NP-based drug discovery, NPs are addressing these challenges and opening up new opportunities in cerebral stroke. Thus, in this review, we first summarize the structure and function of diverse NPs, including flavonoids, phenols, terpenes, lactones, quinones, alkaloids, and glycosides. Then we propose the comprehensive neuroprotective mechanism of NPs in cerebral ischemic stroke, which involves complex cascade processes of oxidative stress, mitochondrial damage, apoptosis or ferroptosis-related cell death, inflammatory response, and disruption of the blood-brain barrier (BBB). Overall, we stress the neuroprotective effect of NPs and their mechanism on cerebral ischemic stroke for a better understanding of the advances and perspective in NPs application that may provide a rationale for the development of innovative therapeutic regimens in ischemic stroke.
Collapse
Affiliation(s)
- Aifen Liu
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Jingyan Hu
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Tzu-Shao Yeh
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong 226019, China
| | - Chengniu Wang
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Jilong Tang
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Xiaohong Huang
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Bin Chen
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Liexiang Huangfu
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Weili Yu
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Lei Zhang
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
- Department of Pharmaceutical Botany, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
26
|
Hu X, Qian Z, Chen J, Chen M, Zhong W, Shen C, Hu Z, Li R. Effects of edaravone dexborneol on neurological function and serum inflammatory factor levels in patients with acute anterior circulation large vessel occlusion stroke. Transl Neurosci 2023; 14:20220312. [PMID: 37854582 PMCID: PMC10579784 DOI: 10.1515/tnsci-2022-0312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/23/2023] [Accepted: 09/04/2023] [Indexed: 10/20/2023] Open
Abstract
The goal of this study is to evaluate and analyze the effects of edaravone (EDV) dexborneol on neurological function and serum inflammatory factor levels among patients with acute anterior circulation big artery blockage stroke. A total of 142 patients with acute anterior circulation large vessel occlusion (LVO) were randomly allocated to the study group (69 patients) or the control group (73 patients). In the study group, patients were treated with 37.5 mg EDV dexborneol twice a day for 10-14 days, based on the control group. The primary efficacy outcome was the National Institutes of Health Stroke Scale score change from baseline to 90 days and the proportion of modified Rankin Scale (mRS)score ≤1 at 90 days after randomization. The secondary outcome included the decrease in inflammatory factors at 14 days. The primary safety outcome was the incidence of hemorrhagic transformation assessed according to Heidelberg bleeding classification within 7 days. A higher percentage of patients with HIHSS score ≤5 at 90 days in the EDV dexcamphorol group was observed than in the control group (75.36% vs 64.38%; P = 0.015). A higher percentage of patients with mRS score ≤1 at 90 days in the EDV dexcamphorol group was observed than in the control group (63.77% vs 50.68%; P = 0.012). After treatment, the levels of IL-6 and hs-CRP were significantly lower following treatment and compared to the control group (P < 0.05). In patients receiving the EDV dexborneol group, a significantly decreased risk of radiographic intracranial hemorrhage was found compared with the control group (20.29% vs 39.73%; P = 0.0006). In conclusion, EDV dexborneol can improve the clinical outcomes of patients with acute anterior circulation LVO stroke, which can be used as an effective supplement to thrombectomy therapy.
Collapse
Affiliation(s)
- Xiaohong Hu
- Neurology Department, Longyan First Affiliated Hospital of Fujian Medical University, No. 105, Jiuyi North Road, Longyan, 364000, China
| | - Zhenhong Qian
- Neurology Department, Liancheng County Hospital, No. 1, Miaoqian Road, Liancheng, 366200, China
| | - Jianhui Chen
- Emergency Department, Longyan First Affiliated Hospital of Fujian Medical University, No. 105, Jiuyi North Road, Longyan, 364000, China
| | - Mingsheng Chen
- Neurology Department, Longyan First Affiliated Hospital of Fujian Medical University, No. 105, Jiuyi North Road, Longyan, 364000, China
| | - Wenying Zhong
- Neurology Department, Longyan First Affiliated Hospital of Fujian Medical University, No. 105, Jiuyi North Road, Longyan, 364000, China
| | - Chaoxiong Shen
- Neurology Department, Longyan First Affiliated Hospital of Fujian Medical University, No. 105, Jiuyi North Road, Longyan, 364000, China
| | - Zhizhou Hu
- Neurology Department, Longyan First Affiliated Hospital of Fujian Medical University, No. 105, Jiuyi North Road, Longyan, 364000, China
| | - Rongtong Li
- Neurology Department, Longyan First Affiliated Hospital of Fujian Medical University, No. 105, Jiuyi North Road, Longyan, 364000, China
| |
Collapse
|
27
|
Choudhary N, Tewari D, Nabavi SF, Kashani HRK, Lorigooini Z, Filosa R, Khan FB, Masoudian N, Nabavi SM. Plant based food bioactives: A boon or bane for neurological disorders. Crit Rev Food Sci Nutr 2022; 64:3279-3325. [PMID: 36369694 DOI: 10.1080/10408398.2022.2131729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Neurological disorders are the foremost occurring diseases across the globe resulting in progressive dysfunction, loss of neuronal structure ultimately cell death. Therefore, attention has been drawn toward the natural resources for the search of neuroprotective agents. Plant-based food bioactives have emerged as potential neuroprotective agents for the treatment of neurodegenerative disorders. This comprehensive review primarily focuses on various plant food bioactive, mechanisms, therapeutic targets, in vitro and in vivo studies in the treatment of neurological disorders to explore whether they are boon or bane for neurological disorders. In addition, the clinical perspective of plant food bioactives in neurological disorders are also highlighted. Scientific evidences point toward the enormous therapeutic efficacy of plant food bioactives in the prevention or treatment of neurological disorders. Nevertheless, identification of food bioactive components accountable for the neuroprotective effects, mechanism, clinical trials, and consolidation of information flow are warranted. Plant food bioactives primarily act by mediating through various pathways including oxidative stress, neuroinflammation, apoptosis, excitotoxicity, specific proteins, mitochondrial dysfunction, and reversing neurodegeneration and can be used for the prevention and therapy of neurodegenerative disorders. In conclusion, the plant based food bioactives are boon for neurological disorders.
Collapse
Affiliation(s)
- Neeraj Choudhary
- Department of Pharmacognosy, Adesh Institute of Pharmacy and Biomedical Sciences, Adesh University, Bathinda, Punjab, India
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Seyed Fazel Nabavi
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
- Nutringredientes Research Center, Federal Institute of Education, Science and Technology (IFCE), Baturite, Ceara, Brazil
| | - Hamid Reza Khayat Kashani
- Department of Neurosurgery, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Lorigooini
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Rosanna Filosa
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
- Department of Science and Technology, University of Sannio, 82100, Benevento, Italy
| | - Farheen Badrealam Khan
- Department of Biology, College of Science, The United Arab Emirates University, Al Ain, 15551 United Arab Emirates
| | - Nooshin Masoudian
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
| | - Seyed Mohammad Nabavi
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
- Nutringredientes Research Center, Federal Institute of Education, Science and Technology (IFCE), Baturite, Ceara, Brazil
| |
Collapse
|
28
|
Zhang D, Li T, Wang A, Feng L, Lai X, Cao K, Zhou L, Yang B, Cui F, Li Q, Dou J, Qi B, Zhang C, Gao Y. Efficacy and safety of LongShengZhi capsule on functional recovery after acute ischemic stroke (LONGAN): Protocol and statistical analysis plan for a randomized, double-blind, placebo-controlled trial. Front Pharmacol 2022; 13:916421. [PMID: 36091794 PMCID: PMC9448855 DOI: 10.3389/fphar.2022.916421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Due to limited time windows and technical requirements, only a small percentage of patients can receive reperfusion therapy for acute ischemic stroke (AIS). Previous studies have shown that LongShengZhi (LSZ) capsule can improve neurological outcomes in patients after AIS, yet those results have not been finally verified through rigorous randomized controlled trials. Thus, this trial was designed to further clarify the efficacy and safety of LSZ capsule for patients with AIS. Methods: LSZ capsule on Functional Recovery after Acute Ischemic Stroke (LONGAN) trial is a prospective, multicenter, randomized, placebo-controlled, double-blind, parallel-group, superiority trial that enrolls patients from stroke and rehabilitation units in China. We will enroll 1,376 patients aged 18 years or older with AIS within 7 days of symptom onset and a National Institute of Health Stroke Scale (NIHSS) score of 4-15. Eligible patients will be randomized to receive either 2 g LSZ capsules three times a day or placebo LSZ capsules for 90 days. The primary outcome is the proportion of patients with favorable outcomes, as measured by the modified Rankin Scale (mRS) 90 days after randomization. The main safety outcome is the proportion of severe adverse events. Conclusion: This study will be the first randomized, double-blind trial to evaluate the efficacy and safety of LSZ capsule in patients with AIS. In order to improve the transparency and reproducibility of the trial, the data will be analyzed in accordance with this pre-specified plan for statistical analysis to reduce bias due to selective analysis and reporting. This trial aims to provide high-quality evidence for the efficacy and safety of LSZ capsule for AIS.
Collapse
Affiliation(s)
- Dandan Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| | - Tingting Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Anxin Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Luda Feng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xinxing Lai
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| | - Kegang Cao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| | - Li Zhou
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Baolin Yang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Fangyuan Cui
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qingbin Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jinjuan Dou
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Baoyun Qi
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Chi Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Gao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
29
|
Noori T, Shirooie S, Sureda A, Sobarzo-Sanchez E, Dehpour AR, Saldías M, Akkol EK. Regulation of DAPK1 by Natural Products: An Important Target in Treatment of Stroke. Neurochem Res 2022; 47:2142-2157. [PMID: 35674928 DOI: 10.1007/s11064-022-03628-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 05/01/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022]
Abstract
Stroke is a sudden neurological disorder that occurs due to impaired blood flow to an area of the brain. Stroke can be caused by the blockage or rupture of a blood vessel in the brain, called ischemic stroke and hemorrhagic stroke, respectively. Stroke is more common in men than women. Atrial fibrillation, hypertension, kidney disease, high cholesterol and lipids, genetic predisposition, inactivity, poor nutrition, diabetes mellitus, family history and smoking are factors that increase the risk of stroke. Restoring blood flow by repositioning blocked arteries using thrombolytic agents or endovascular therapy are the most effective treatments for stroke. However, restoring circulation after thrombolysis can cause fatal edema or intracranial hemorrhage, and worsen brain damage in a process known as ischemia-reperfusion injury. Therefore, there is a pressing need to find and develop more effective treatments for stroke. In the past, the first choice of treatment was based on natural compounds. Natural compounds are able to reduce the symptoms and reduce various diseases including stroke that attract the attention of the pharmaceutical industry. Nowadays, as a result of the numerous studies carried out in the field of herbal medicine, many useful and valuable effects of plants have been identified. The death-associated protein kinase (DAPK) family is one of the vital families of serine/threonine kinases involved in the regulation of some biological functions in human cells. DAPK1 is the most studied kinase within the DAPKs family as it is involved in neuronal and recovery processes. Dysregulation of DAPK1 in the brain is involved in the developing neurological diseases such as stroke. Natural products can function in a variety of ways, including reducing cerebral edema, reducing brain endothelial cell death, and inhibiting TNFα and interleukin-1β (IL-1β) through regulating the DAPK1 signal against stroke. Due to the role of DAPK1 in neurological disorders, the aim of this article was to investigate the role of DAPK1 in stroke and its modulation by natural compounds.
Collapse
Affiliation(s)
- Tayebeh Noori
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Samira Shirooie
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress (NUCOX) and Health Research Institute of Balearic Islands (IdISBa), University of Balearic Islands, 07122, Palma de Mallorca, Balearic Islands, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Eduardo Sobarzo-Sanchez
- Instituto de Investigación y Postgrado, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago, Chile
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Marianela Saldías
- Instituto de Investigación y Postgrado, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago, Chile
| | - Esra Küpeli Akkol
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, 06330, Ankara, Turkey
| |
Collapse
|
30
|
Qin C, Yang S, Chu YH, Zhang H, Pang XW, Chen L, Zhou LQ, Chen M, Tian DS, Wang W. Signaling pathways involved in ischemic stroke: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2022; 7:215. [PMID: 35794095 PMCID: PMC9259607 DOI: 10.1038/s41392-022-01064-1] [Citation(s) in RCA: 388] [Impact Index Per Article: 129.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/01/2022] [Accepted: 06/15/2022] [Indexed: 02/07/2023] Open
Abstract
Ischemic stroke is caused primarily by an interruption in cerebral blood flow, which induces severe neural injuries, and is one of the leading causes of death and disability worldwide. Thus, it is of great necessity to further detailly elucidate the mechanisms of ischemic stroke and find out new therapies against the disease. In recent years, efforts have been made to understand the pathophysiology of ischemic stroke, including cellular excitotoxicity, oxidative stress, cell death processes, and neuroinflammation. In the meantime, a plethora of signaling pathways, either detrimental or neuroprotective, are also highly involved in the forementioned pathophysiology. These pathways are closely intertwined and form a complex signaling network. Also, these signaling pathways reveal therapeutic potential, as targeting these signaling pathways could possibly serve as therapeutic approaches against ischemic stroke. In this review, we describe the signaling pathways involved in ischemic stroke and categorize them based on the pathophysiological processes they participate in. Therapeutic approaches targeting these signaling pathways, which are associated with the pathophysiology mentioned above, are also discussed. Meanwhile, clinical trials regarding ischemic stroke, which potentially target the pathophysiology and the signaling pathways involved, are summarized in details. Conclusively, this review elucidated potential molecular mechanisms and related signaling pathways underlying ischemic stroke, and summarize the therapeutic approaches targeted various pathophysiology, with particular reference to clinical trials and future prospects for treating ischemic stroke.
Collapse
Affiliation(s)
- Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yun-Hui Chu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Wei Pang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lian Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Man Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
31
|
Staszewski J, Stȩpień A, Piusińska-Macoch R, Dȩbiec A, Gniadek-Olejniczak K, Frankowska E, Maliborski A, Chadaide Z, Balo D, Król B, Namias R, Harston G, Mróz J, Piasecki P. Efficacy of Cerebrolysin Treatment as an Add-On Therapy to Mechanical Thrombectomy in Patients With Acute Ischemic Stroke Due to Large Vessel Occlusion: Study Protocol for a Prospective, Open Label, Single-Center Study With 12 Months of Follow-Up. Front Neurol 2022; 13:910697. [PMID: 35860483 PMCID: PMC9289167 DOI: 10.3389/fneur.2022.910697] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/30/2022] [Indexed: 12/18/2022] Open
Abstract
This study is designed to determine the efficacy of Cerebrolysin treatment as an add-on therapy to mechanical thrombectomy (MT) in reducing global disability in subjects with acute ischemic stroke (AIS). We have planned a single center, prospective, open-label, single-arm study with a 12-month follow-up of 50 patients with moderate to severe AIS, with a small established infarct core and with good collateral circulation who achieve significant reperfusion following MT and who receive additional Cerebrolysin within 8 h of stroke onset compared to 50 historical controls treated with MT alone, matched for age, clinical severity, occlusion location, baseline perfusion lesion volume, onset to reperfusion time, and use of iv thrombolytic therapy. The primary outcome measure will be the overall proportion of subjects receiving Cerebrolysin compared to the control group experiencing a favorable functional outcome (by modified Rankin Scale 0-2) at 90 days, following stroke onset. The secondary objectives are to determine the efficacy of Cerebrolysin as compared to the control group in reducing the risk of symptomatic secondary hemorrhagic transformation, improving neurological outcomes (NIHSS 0-2 at day 7, day 30, and 90), reducing mortality rates (over the 90-day and 12 months study period), and improving: activities of daily living (by Barthel Index), health-related quality of life (EQ-5D-5L) assessed at day 30, 90, and at 12 months. The other measures of efficacy in the Cerebrolysin group will include: assessment of final stroke volume and penumbral salvage (measured by CT/CTP at 30 days) and its change compared to baseline volume, changes over time in language function (by the 15-item Boston Naming Test), hemispatial neglect (by line bisection test), global cognitive function (by The Montreal Cognitive Assessment), and depression (by Hamilton Depression Rating Scale) between day 30 and day 90 assessments). The patients will receive 30 ml of Cerebrolysin within 8 h of AIS stroke onset and continue treatment once daily until day 21 (first cycle) and they will receive a second cycle of treatment (30 ml/d for 21 days given in the Outpatient Department or Neurorehabilitation Clinic) from day 69 to 90.
Collapse
Affiliation(s)
- Jacek Staszewski
- Clinic of Neurology, Military Institute of Medicine, Warsaw, Poland
| | - Adam Stȩpień
- Clinic of Neurology, Military Institute of Medicine, Warsaw, Poland
| | | | | | | | - Emilia Frankowska
- Department of Radiology, Military Institute of Medicine, Warsaw, Poland
| | - Artur Maliborski
- Department of Radiology, Military Institute of Medicine, Warsaw, Poland
| | - Zoltan Chadaide
- Brainomix Ltd., and Oxford University Hospitals NHSFT, Oxford, United Kingdom
| | - David Balo
- Brainomix Ltd., and Oxford University Hospitals NHSFT, Oxford, United Kingdom
| | - Beata Król
- Brainomix Ltd., and Oxford University Hospitals NHSFT, Oxford, United Kingdom
| | - Rafael Namias
- Brainomix Ltd., and Oxford University Hospitals NHSFT, Oxford, United Kingdom
| | - George Harston
- Brainomix Ltd., and Oxford University Hospitals NHSFT, Oxford, United Kingdom
| | - Józef Mróz
- Neurorehabilitation Clinic, Military Institute of Medicine, Warsaw, Poland
| | - Piotr Piasecki
- Department of Radiology, Military Institute of Medicine, Warsaw, Poland
| |
Collapse
|
32
|
Purroy F, Beretta S, England TJ, Hess DC, Pico F, Shuaib A. Editorial: Remote Ischemic Conditioning (Pre, Per, and Post) as an Emerging Strategy of Neuroprotection in Ischemic Stroke. Front Neurol 2022; 13:932891. [PMID: 35812090 PMCID: PMC9260686 DOI: 10.3389/fneur.2022.932891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Francisco Purroy
- Clinical Neurosciences Group IRBLleida, Stroke Unit, Hospital Universitari Arnau de Vilanova de Lleida, Universitat de Lleida, Lleida, Spain
- *Correspondence: Francisco Purroy
| | - Simone Beretta
- Department of Neurology, San Gerardo Hospital Monza, Monza, Italy
| | | | - David Charles Hess
- Medical College of Georgia, Augusta University Augusta, Augusta, GA, United States
| | - Fernando Pico
- Centre Hospitalier de Versailles Le Chesnay, Le Chesnay, France
| | - Ashfaq Shuaib
- Department of Neurology, Medical College of Georgia, University of Alberta Edmonton, Edmonton, AB, Canada
| |
Collapse
|
33
|
Guo Y, Mao M, Li Q, Yu X, Zhou L. Extracts of Ginkgo flavonoids and ginkgolides improve cerebral ischaemia-reperfusion injury through the PI3K/Akt/Nrf2 signalling pathway and multicomponent in vivo processes. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 99:154028. [PMID: 35279613 DOI: 10.1016/j.phymed.2022.154028] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 02/22/2022] [Accepted: 02/28/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Cerebral ischaemia-reperfusion injury (CIRI) is a common disease characterized by severe attacks and a high disabling rate worldwide. Oxidative stress injury has been proposed as a major risk factor for CIRI. Ginkgo biloba extract (GBE) has been shown to elicit vascular protective effects, the main components of which are Ginkgo flavonoids (GF) and ginkgolides (GL). Our previous study showed that GF and GL played a central role in protecting CIRI, but the mechanism remains unclear. This study aimed to further reveal the protective effect mechanism of GF and GL in rats with CIRI. METHODS The antioxidant activity in vitro was assessed by the DPPH method. The model used in this study was established by middle cerebral artery occlusion (MCAO) and reperfusion; the level of CIRI was assessed by nerve function score and TTC staining; we measured the oxidative stress indices in the brain cortex, including LDH, GSH-Px, and the protein contents of Akt, p-Akt, Nrf2, and HO-1; HPLC-MS was used to detect drug concentrations in rat plasma at different times after administration of GF and GL; and the pharmacokinetic parameters of each component were calculated by Drug and Statistic Version 3.2.6 (DAS 3.2.6) software and SPSS 17.0. RESULTS Regarding the DPPH free radical scavenging ability, GF performed better free radical scavenging ability than GL. In terms of the nerve function score and TTC staining, there were no statistically significant differences among the GF, GL and combined groups; however, there were significant differences in reducing the activity of LDH and increasing the activity of GSH-Px in the three administration groups. For the expression of Akt, p-Akt, Nrf2, and HO-1, the combined group had a significant effect compared with that in the GF or GL group. In addition, there was a significant multicomponent interaction in vivo in the combined group compared with the GF or GL group. CONCLUSION After GF and GL were used in combination, the effect of anti-CIRI was more pronounced. This result indicated that GF and GL might improve CIRI by activating the PI3K/Akt/Nrf2 signalling pathway and promoting multicomponent interactions in vivo.
Collapse
Affiliation(s)
- Ying Guo
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Mingjiang Mao
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qiuying Li
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiahui Yu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Liping Zhou
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| |
Collapse
|
34
|
Yoneda S, Fukuta T, Ozono M, Kogure K. Enhancement of cerebroprotective effects of lipid nanoparticles encapsulating FK506 on cerebral ischemia/reperfusion injury by particle size regulation. Biochem Biophys Res Commun 2022; 611:53-59. [PMID: 35477093 DOI: 10.1016/j.bbrc.2022.04.080] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/18/2022] [Indexed: 11/02/2022]
Abstract
Delivery of cerebroprotective agents using liposomes has been demonstrated to be useful for treating cerebral ischemia/reperfusion (I/R) injury. We previously reported that intravenous administration of liposomes with diameters of 100 nm showed higher accumulation in the I/R region compared with larger liposomes (>200 nm) by passage through the disintegrated blood-brain barrier, suggesting a size-dependence for liposome-mediated drug delivery. Based on these findings, we hypothesized that regulation of liposomal particle size (<100 nm) may enhance the therapeutic efficacy of encapsulated drugs on cerebral I/R injury. Herein, we prepared lipid nanoparticles (LNP) with particle sizes <100 nm by the microfluidics method and compared their therapeutic potential with LNP exhibiting sizes >100 nm in cerebral I/R model rats. Intravenously administered smaller LNP (ca. 60 nm) exhibited wider accumulation and diffusivity in the brain parenchyma of the I/R region compared with larger LNP (>100 nm). Importantly, treatment with LNP encapsulating the cerebroprotective agent FK506 (FK-LNP) with particle sizes <100 nm showed greater cerebroprotective effects than FK-LNP with sizes >100 nm, and also significantly ameliorated brain injury. These results suggest that particle size regulation of LNP to sizes <100 nm can enhance the therapeutic effect of encapsulated drugs for treatment of cerebral I/R injury, and that FK-LNP could be a promising cerebroprotective agent.
Collapse
Affiliation(s)
- Shintaro Yoneda
- Department of Pharmaceutical Health Chemistry, Graduate School of Pharmaceutical Sciences, Tokushima University, Shomachi 1, Tokushima, 770-8505, Japan
| | - Tatsuya Fukuta
- Department of Physical Pharmaceutics, School of Pharmaceutical Sciences, Wakayama Medical University, 25-1 Shichiban-cho, Wakayama, 640-8156, Japan.
| | - Mizune Ozono
- Department of Pharmaceutical Health Chemistry, Graduate School of Biomedical Sciences, Tokushima University, Shomachi 1, Tokushima, 770-8505, Japan
| | - Kentaro Kogure
- Department of Pharmaceutical Health Chemistry, Graduate School of Biomedical Sciences, Tokushima University, Shomachi 1, Tokushima, 770-8505, Japan
| |
Collapse
|
35
|
Vos EM, Geraedts VJ, van der Lugt A, Dippel DWJ, Wermer MJH, Hofmeijer J, van Es ACGM, Roos YBWEM, Peeters-Scholte CMPCD, van den Wijngaard IR. Systematic Review - Combining Neuroprotection With Reperfusion in Acute Ischemic Stroke. Front Neurol 2022; 13:840892. [PMID: 35370911 PMCID: PMC8969766 DOI: 10.3389/fneur.2022.840892] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/17/2022] [Indexed: 12/22/2022] Open
Abstract
Background Clinical trials of neuroprotection in acute ischemic stroke (AIS) have provided disappointing results. Reperfusion may be a necessary condition for positive effects of neuroprotective treatments. This systematic review provides an overview of efficacy of neuroprotective agents in combination with reperfusion therapy in AIS. Methods A literature search was performed on the following databases, namely PubMed, Embase, Web of Science, Cochrane Library, Emcare. All databases were searched up to September 23rd 2021. All randomized controlled trials in which patients were treated with neuroprotective strategies within 12 h of stroke onset in combination with intravenous thrombolysis (IVT), endovascular therapy (EVT), or both were included. Results We screened 1,764 titles/abstracts and included 30 full reports of unique studies with a total of 16,160 patients. In 15 studies neuroprotectants were tested for clinical efficacy, where all patients had to receive reperfusion therapies, either IVT and/or EVT. Heterogeneity in reported outcome measures was observed. Treatment was associated with improved clinical outcome for: 1) uric acid in patients treated with EVT and IVT, 2) nerinetide in patients who underwent EVT without IVT, 3) imatinib in stroke patients treated with IVT with or without EVT, 4) remote ischemic perconditioning and IVT, and 5) high-flow normobaric oxygen treatment after EVT, with or without IVT. Conclusion Studies specifically testing effects of neuroprotective agents in addition to IVT and/or EVT are scarce. Future neuroprotection studies should report standardized functional outcome measures and combine neuroprotective agents with reperfusion therapies in AIS or aim to include prespecified subgroup analyses for treatment with IVT and/or EVT.
Collapse
Affiliation(s)
- E. M. Vos
- Department of Neurology, The Hague Medical Center, The Hague, Netherlands
- *Correspondence: E. M. Vos
| | - V. J. Geraedts
- Department of Neurology, The Hague Medical Center, The Hague, Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
| | - A. van der Lugt
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - D. W. J. Dippel
- Department of Neurology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - M. J. H. Wermer
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
| | - J. Hofmeijer
- Department of Neurology, Rijnstate Hospital, Arnhem, Netherlands
- Department of Clinical Neurophysiology, Technical Medical Centre, University of Twente, Enschede, Netherlands
| | - A. C. G. M. van Es
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
- Department of Radiology, The Hague Medical Center, The Hague, Netherlands
| | - Y. B. W. E. M. Roos
- Department of Neurology, Amsterdam University Medical Center, Amsterdam, Netherlands
| | | | - I. R. van den Wijngaard
- Department of Neurology, The Hague Medical Center, The Hague, Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
36
|
Zeng J, Bao T, Yang K, Zhu X, Wang S, Xiang W, Ge A, Zeng L, Ge J. The mechanism of microglia-mediated immune inflammation in ischemic stroke and the role of natural botanical components in regulating microglia: A review. Front Immunol 2022; 13:1047550. [PMID: 36818470 PMCID: PMC9933144 DOI: 10.3389/fimmu.2022.1047550] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 12/05/2022] [Indexed: 02/05/2023] Open
Abstract
Ischemic stroke (IS) is one of the most fatal diseases. Neuroimmunity, inflammation, and oxidative stress play important roles in various complex mechanisms of IS. In particular, the early proinflammatory response resulting from the overactivation of resident microglia and the infiltration of circulating monocytes and macrophages in the brain after cerebral ischemia leads to secondary brain injury. Microglia are innate immune cells in the brain that constantly monitor the brain microenvironment under normal conditions. Once ischemia occurs, microglia are activated to produce dual effects of neurotoxicity and neuroprotection, and the balance of the two effects determines the fate of damaged neurons. The activation of microglia is defined as the classical activation (M1 type) or alternative activation (M2 type). M1 type microglia secrete pro-inflammatory cytokines and neurotoxic mediators to exacerbate neuronal damage, while M2 type microglia promote a repairing anti-inflammatory response. Fine regulation of M1/M2 microglial activation to minimize damage and maximize protection has important therapeutic value. This review focuses on the interaction between M1/M2 microglia and other immune cells involved in the regulation of IS phenotypic characteristics, and the mechanism of natural plant components regulating microglia after IS, providing novel candidate drugs for regulating microglial balance and IS drug development.
Collapse
Affiliation(s)
- Jinsong Zeng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Tingting Bao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | | | - Shanshan Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Wang Xiang
- Department of Rheumatology, The First People's Hospital Changde City, Changde, Hunan, China
| | - Anqi Ge
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Liuting Zeng
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China.,Hunan Academy of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
37
|
Jurcau A, Simion A. Neuroinflammation in Cerebral Ischemia and Ischemia/Reperfusion Injuries: From Pathophysiology to Therapeutic Strategies. Int J Mol Sci 2021; 23:14. [PMID: 35008440 PMCID: PMC8744548 DOI: 10.3390/ijms23010014] [Citation(s) in RCA: 242] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 12/18/2021] [Accepted: 12/18/2021] [Indexed: 02/07/2023] Open
Abstract
Its increasing incidence has led stroke to be the second leading cause of death worldwide. Despite significant advances in recanalization strategies, patients are still at risk for ischemia/reperfusion injuries in this pathophysiology, in which neuroinflammation is significantly involved. Research has shown that in the acute phase, neuroinflammatory cascades lead to apoptosis, disruption of the blood-brain barrier, cerebral edema, and hemorrhagic transformation, while in later stages, these pathways support tissue repair and functional recovery. The present review discusses the various cell types and the mechanisms through which neuroinflammation contributes to parenchymal injury and tissue repair, as well as therapeutic attempts made in vitro, in animal experiments, and in clinical trials which target neuroinflammation, highlighting future therapeutic perspectives.
Collapse
Affiliation(s)
- Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania;
- Neurology Ward, Clinical Municipal Hospital “dr. G. Curteanu” Oradea, 410154 Oradea, Romania
| | - Aurel Simion
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania;
- Neurorehabilitation Ward, Clinical Municipal Hospital “dr. G. Curteanu” Oradea, 410154 Oradea, Romania
| |
Collapse
|
38
|
Konduri P, van Voorst H, Bucker A, van Kranendonk K, Boers A, Treurniet K, Berkhemer O, Yoo AJ, van Zwam W, van Oostenbrugge R, van der Lugt A, Dippel D, Roos Y, Bot J, Majoie C, Marquering H. Posttreatment Ischemic Lesion Evolution Is Associated With Reduced Favorable Functional Outcome in Patients With Stroke. Stroke 2021; 52:3523-3531. [PMID: 34289708 DOI: 10.1161/strokeaha.120.032331] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND AND PURPOSE Ischemic lesion volume can increase even 24 hours after onset of an acute ischemic stroke. In this study, we investigated the association of lesion evolution with functional outcome and the influence of successful recanalization on this association. METHODS We included patients from the MR CLEAN trial (Multicenter Randomized Clinical Trial of Endovascular Treatment for Acute Ischemic Stroke in the Netherlands) who received good quality noncontrast CT images 24 hours and 1 week after stroke onset. The ischemic lesion delineations included infarct, edema, and hemorrhagic transformation. Lesion evolution was defined as the difference between the volumes measured on the 1-week and 24-hour noncontrast CTs. The association of lesion evolution with functional outcome was evaluated using unadjusted and adjusted logistic regression. Adjustments were made for baseline, clinical, and imaging parameters that were associated P<0.10) in univariate analysis with favorable functional outcome, defined as modified Rankin Scale score of ≤2. Interaction analysis was performed to evaluate the influence of successful recanalization, defined as modified Arterial Occlusion Lesion score of 3 points, on this association. RESULTS Of the 226 patients who were included, 69 (31%) patients achieved the favorable functional outcome. Median lesion evolution was 22 (interquartile range, 10-45) mL. Lesion evolution was significantly inversely correlated with favourable functional outcome: unadjusted odds ratio, 0.76 (95% CI, 0.66-0.86; per 10 mL of lesion evolution; P<0.01) and adjusted odds ratio: 0.85 (95% CI, 0.72-0.97; per 10 mL of lesion evolution; P=0.03). There was no significant interaction of successful recanalization on the association of lesion evolution and favorable functional outcome (odds ratio, 1.01 [95% CI, 0.77-1.36]; P=0.94). CONCLUSIONS In our population, subacute ischemic lesion evolution is associated with unfavorable functional outcome. This study suggests that even 24 hours after onset of stroke, deterioration of the brain continues, which has a negative effect on functional outcome. This finding may warrant additional treatment in the subacute phase.
Collapse
Affiliation(s)
- Praneeta Konduri
- Department of Biomedical Engineering and Physics (P.K., H.v.V., A.B., H.M.), Amsterdam UMC, location AMC, the Netherlands
- Department of Radiology and Nuclear Medicine (P.K., H.v.V., H.v.V., K.v.K., K.T., O.B., C.M., H.M.), Amsterdam UMC, location AMC, the Netherlands
| | - Henk van Voorst
- Department of Biomedical Engineering and Physics (P.K., H.v.V., A.B., H.M.), Amsterdam UMC, location AMC, the Netherlands
- Department of Radiology and Nuclear Medicine (P.K., H.v.V., H.v.V., K.v.K., K.T., O.B., C.M., H.M.), Amsterdam UMC, location AMC, the Netherlands
| | - Amber Bucker
- Department of Biomedical Engineering and Physics (P.K., H.v.V., A.B., H.M.), Amsterdam UMC, location AMC, the Netherlands
| | - Katinka van Kranendonk
- Department of Radiology and Nuclear Medicine (P.K., H.v.V., H.v.V., K.v.K., K.T., O.B., C.M., H.M.), Amsterdam UMC, location AMC, the Netherlands
| | - Anna Boers
- Department of Radiology, University Medical Center Groningen, the Netherlands (A.B.)
- Nico-lab, Amsterdam, Netherlands (A.B.)
| | - Kilian Treurniet
- Department of Radiology and Nuclear Medicine (P.K., H.v.V., H.v.V., K.v.K., K.T., O.B., C.M., H.M.), Amsterdam UMC, location AMC, the Netherlands
- Department of Radiology, Haaglanden Medisch Centrum, The Hague, the Netherlands (K.T.)
| | - Olvert Berkhemer
- Department of Radiology and Nuclear Medicine (P.K., H.v.V., H.v.V., K.v.K., K.T., O.B., C.M., H.M.), Amsterdam UMC, location AMC, the Netherlands
- Department of Neurology (O.B., D.D.), Erasmus MC University Medical Center, Rotterdam, Netherlands
- Department of Radiology & Nuclear Medicine (O.B., A.v.d.L.), Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Albert J Yoo
- Department of Radiology, Texas Stroke Institute, Dallas-Fort Worth (A.J.Y.)
| | - Wim van Zwam
- Department of Radiology (W.v.Z.), Maastricht University Medical Center and Cardiovascular Research Institute Maastricht (CARIM)
| | - Robert van Oostenbrugge
- Department of Neurology (R.v.O.), Maastricht University Medical Center and Cardiovascular Research Institute Maastricht (CARIM)
| | - Aad van der Lugt
- Department of Radiology & Nuclear Medicine (O.B., A.v.d.L.), Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Diederik Dippel
- Department of Neurology (O.B., D.D.), Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Yvo Roos
- Department of Neurology (Y.R.), Amsterdam UMC, location AMC, the Netherlands
| | - Joost Bot
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit van Amsterdam (J.B.)
| | - Charles Majoie
- Department of Radiology and Nuclear Medicine (P.K., H.v.V., H.v.V., K.v.K., K.T., O.B., C.M., H.M.), Amsterdam UMC, location AMC, the Netherlands
| | - Henk Marquering
- Department of Biomedical Engineering and Physics (P.K., H.v.V., A.B., H.M.), Amsterdam UMC, location AMC, the Netherlands
- Department of Radiology and Nuclear Medicine (P.K., H.v.V., H.v.V., K.v.K., K.T., O.B., C.M., H.M.), Amsterdam UMC, location AMC, the Netherlands
| |
Collapse
|
39
|
Konduri P, van Kranendonk K, Boers A, Treurniet K, Berkhemer O, Yoo AJ, van Zwam W, van Oostenbrugge R, van der Lugt A, Dippel D, Roos Y, Bot J, Majoie C, Marquering H. The Role of Edema in Subacute Lesion Progression After Treatment of Acute Ischemic Stroke. Front Neurol 2021; 12:705221. [PMID: 34354669 PMCID: PMC8329530 DOI: 10.3389/fneur.2021.705221] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/18/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Ischemic lesions commonly continue to progress even days after treatment, and this lesion growth is associated with unfavorable functional outcome in acute ischemic stroke patients. The aim of this study is to elucidate the role of edema in subacute lesion progression and its influence on unfavorable functional outcome by quantifying net water uptake. Methods: We included all 187 patients from the MR CLEAN trial who had high quality follow-up non-contrast CT at 24 h and 1 week. Using a CT densitometry-based method to calculate the net water uptake, we differentiated total ischemic lesion volume (TILV) into edema volume (EV) and edema-corrected infarct volume (ecIV). We calculated these volumes at 24 h and 1 week after stroke and determined their progression in the subacute period. We assessed the effect of 24-h lesion characteristics on EV and ecIV progression. We evaluated the influence of edema and edema-corrected infarct progression on favorable functional outcome after 90 days (modified Rankin Scale: 0-2) after correcting for potential confounders. Lastly, we compared these volumes between subgroups of patients with and without successful recanalization using the Mann-Whitney U-test. Results: Median TILV increased from 37 (IQR: 18-81) ml to 68 (IQR: 30-130) ml between 24 h and 1 week after stroke, while the net water uptake increased from 22 (IQR: 16-26)% to 27 (IQR: 22-32)%. The TILV progression of 20 (8.8-40) ml was mostly caused by ecIV with a median increase of 12 (2.4-21) ml vs. 6.5 (2.7-15) ml of EV progression. Larger TILV, EV, and ecIV volumes at 24 h were all associated with more edema and lesion progression. Edema progression was associated with unfavorable functional outcome [aOR: 0.53 (0.28-0.94) per 10 ml; p-value: 0.05], while edema-corrected infarct progression showed a similar, non-significant association [aOR: 0.80 (0.62-0.99); p-value: 0.06]. Lastly, edema progression was larger in patients without successful recanalization, whereas ecIV progression was comparable between the subgroups. Conclusion: EV increases in evolving ischemic lesions in the period between 1 day and 1 week after acute ischemic stroke. This progression is larger in patients without successful recanalization and is associated with unfavorable functional outcome. However, the extent of edema cannot explain the total expansion of ischemic lesions since edema-corrected infarct progression is larger than the edema progression.
Collapse
Affiliation(s)
- Praneeta Konduri
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, Amsterdam, Netherlands.,Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Katinka van Kranendonk
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Anna Boers
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, Amsterdam, Netherlands.,Nico.lab, Amsterdam, Netherlands
| | - Kilian Treurniet
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, Netherlands.,Department of Radiology and Nuclear Medicine, Haaglanden Medisch Centrum, The Hague, Netherlands
| | - Olvert Berkhemer
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, Netherlands.,Department of Neurology, Erasmus MC-University Medical Center, Rotterdam, Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Albert J Yoo
- Department of Radiology, Texas Stroke Institute, Dallas-Fort Worth, TX, United States
| | - Wim van Zwam
- Department of Radiology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, Netherlands
| | - Robert van Oostenbrugge
- Department of Neurology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, Netherlands
| | - Aad van der Lugt
- Department of Radiology and Nuclear Medicine, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Diederik Dippel
- Department of Neurology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Yvo Roos
- Department of Neurology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Joost Bot
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Vrije Universiteit van Amsterdam, Amsterdam, Netherlands
| | - Charles Majoie
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Henk Marquering
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, Amsterdam, Netherlands.,Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | | |
Collapse
|
40
|
Moser U. Tetrahydrocannabinol and cannabidiol as an oromucosal spray in a 1:1 ratio: a therapeutic option for patients with central post-stroke pain syndrome? BMJ Case Rep 2021; 14:14/7/e243072. [PMID: 34230048 DOI: 10.1136/bcr-2021-243072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Central pain after stroke due to brainstem infarction is very rare. Treatment is difficult and specific guidelines are lacking. This is the report of a 61-year-old female patient who, after a posterolateral left medulla oblongata insult with incomplete Wallenberg syndrome, subsequently developed a burning and tingling pain in the contralateral leg and a burning and shooting pain in the ipsilateral face in trigeminal branches 1 and 2. More than 3 years of therapy with amitriptyline, gabapentin, pregabalin and various grade II and III opioids was ineffective or showed intolerable side effects. The administration of tetrahydrocannabinol and cannabidiol as an oromucosal spray in a 1:1 ratio improved the pain situation and quality of life quickly and permanently. The encouraging results in the present case may suggest that treatment with medical cannabis should be considered in similar cases when standard therapies are insufficient.
Collapse
|
41
|
Zakharov VV, Vakhnina NV, Gogoleva AG. [The criteria for effectiveness of reperfusion therapy and neuroprotective therapy in ischemic stroke]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:86-92. [PMID: 34037360 DOI: 10.17116/jnevro202112104186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The narrative review discusses the data on efficacy and safety of reperfusion therapy (RT) and neuroprotective therapy in ischemic stroke. The influence of therapy on mortality, residual neurologic deficit and disability is analyzed. It was shown that RT (thrombolysis or mechanical thromboextraction) leads to significant decrease of residual neurologic deficit or disability. The influence of RT on mortality is controversial. There is some evidence that RT can increase early mortality due to hemorrhagic complications. Neuroprotective therapy is much less studied in stroke but is recognized as safe. Neuroprotective therapy (i.e. cerebrolysin) can diminish residual neurologic deficit and disability, while it has no influence on mortality.
Collapse
Affiliation(s)
- V V Zakharov
- Sechenov First Moscow State Medical University, Moscow, Russia.,Sklifosovsky Institute of Clinical Medicine, Moscow, Russia
| | - N V Vakhnina
- Sechenov First Moscow State Medical University, Moscow, Russia.,Sklifosovsky Institute of Clinical Medicine, Moscow, Russia
| | - A G Gogoleva
- Sechenov First Moscow State Medical University, Moscow, Russia.,Sklifosovsky Institute of Clinical Medicine, Moscow, Russia
| |
Collapse
|
42
|
Choudhary RC, Shoaib M, Sohnen S, Rolston DM, Jafari D, Miyara SJ, Hayashida K, Molmenti EP, Kim J, Becker LB. Pharmacological Approach for Neuroprotection After Cardiac Arrest-A Narrative Review of Current Therapies and Future Neuroprotective Cocktail. Front Med (Lausanne) 2021; 8:636651. [PMID: 34084772 PMCID: PMC8167895 DOI: 10.3389/fmed.2021.636651] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/12/2021] [Indexed: 11/13/2022] Open
Abstract
Cardiac arrest (CA) results in global ischemia-reperfusion injury damaging tissues in the whole body. The landscape of therapeutic interventions in resuscitation medicine has evolved from focusing solely on achieving return of circulation to now exploring options to mitigate brain injury and preserve brain function after CA. CA pathology includes mitochondrial damage and endoplasmic reticulum stress response, increased generation of reactive oxygen species, neuroinflammation, and neuronal excitotoxic death. Current non-pharmacologic therapies, such as therapeutic hypothermia and extracorporeal cardiopulmonary resuscitation, have shown benefits in protecting against ischemic brain injury and improving neurological outcomes post-CA, yet their application is difficult to institute ubiquitously. The current preclinical pharmacopeia to address CA and the resulting brain injury utilizes drugs that often target singular pathways and have been difficult to translate from the bench to the clinic. Furthermore, the limited combination therapies that have been attempted have shown mixed effects in conferring neuroprotection and improving survival post-CA. The global scale of CA damage and its resultant brain injury necessitates the future of CA interventions to simultaneously target multiple pathways and alleviate the hemodynamic, mitochondrial, metabolic, oxidative, and inflammatory processes in the brain. This narrative review seeks to highlight the current field of post-CA neuroprotective pharmaceutical therapies, both singular and combination, and discuss the use of an extensive multi-drug cocktail therapy as a novel approach to treat CA-mediated dysregulation of multiple pathways, enhancing survival, and neuroprotection.
Collapse
Affiliation(s)
- Rishabh C Choudhary
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States.,Department of Emergency Medicine, Northshore University Hospital, Northwell Health, Manhasset, NY, United States
| | - Muhammad Shoaib
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Samantha Sohnen
- Department of Anesthesiology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Daniel M Rolston
- Department of Emergency Medicine, Northshore University Hospital, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States.,Department of Surgery, North Shore University Hospital, Northwell Health, Manhasset, NY, United States
| | - Daniel Jafari
- Department of Emergency Medicine, Northshore University Hospital, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States.,Department of Surgery, North Shore University Hospital, Northwell Health, Manhasset, NY, United States
| | - Santiago J Miyara
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States
| | - Kei Hayashida
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States.,Department of Emergency Medicine, Northshore University Hospital, Northwell Health, Manhasset, NY, United States
| | | | - Junhwan Kim
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States.,Department of Emergency Medicine, Northshore University Hospital, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Lance B Becker
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States.,Department of Emergency Medicine, Northshore University Hospital, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| |
Collapse
|
43
|
Zha H, Fan Y, Yang L, Yin M, Miao W, He J, Wang Y. Autophagy protects against cerebral ischemic reperfusion injury by inhibiting neuroinflammation. Am J Transl Res 2021; 13:4726-4737. [PMID: 34150053 PMCID: PMC8205746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/04/2020] [Indexed: 06/12/2023]
Abstract
OBJECTIVE To examine the effect of autophagy on cerebral damage caused by different models and test the hypothesis that its protection mechanism acts via inhibiting expression of neuroinflammatory mediators. METHODS Autophagy was induced by rapamycin treatment. Cerebral damage was induced using models of IL-6 treatment, oxygen glucose deprivation/reoxygenation (OGD/R) in vitro, and middle cerebral artery occlusion (MCAO) in vivo. The effect and mechanism of autophagy was examined and assessed in terms of cell viability, infarction size in brain tissue, neurological score, production of inflammatory mediators IL-1β and IL-6, transcription and protein expression of autophagy markers beclin-1 and LC-3II in different experimental groups. RESULTS Autophagy triggered by rapamycin could protect neurons from IL-6-induced injury and astrocytes from OGD/R-induced injury in vitro and in rat brain tissue from MCAO in vivo. Autophagy significantly increased cell viability, attenuated cerebral infarction and improved neurological scores. It also inhibited production of the IL-1β and IL-6 and elevated the expression of beclin-1 and LC-3II. CONCLUSIONS Autophagy can inhibit the inflammatory response and reduce cerebral I/R injury. There was a relationship between the extent of protection and (i) the level of the autophagic response, (ii) the stage of the cerebral I/R injury, and (iii) the time of intervention.
Collapse
Affiliation(s)
- Hao Zha
- Department of Reproductive and Genetics, Second Affiliated Hospital of Kunming Medical UniversityKunming, China
| | - Yaodong Fan
- Department of Neurosurgery, Third Affiliated Hospital of Kunming Medical UniversityKunming, China
| | - Li Yang
- Department of Histology and Embryology, Kunming Medical UniversityKunming, China
| | - Mei Yin
- Department of Neurology, Second Affiliated Hospital of Kunming Medical UniversityKunming, China
| | - Wei Miao
- Department of Neurology, Second Affiliated Hospital of Kunming Medical UniversityKunming, China
| | - Jin He
- Department of Neurology, Second Affiliated Hospital of Kunming Medical UniversityKunming, China
| | - Ying Wang
- Department of Neurology, Second Affiliated Hospital of Kunming Medical UniversityKunming, China
| |
Collapse
|
44
|
Zhu J, Zhu Z, Ren Y, Dong Y, Li Y, Yang X. LINGO-1 shRNA protects the brain against ischemia/reperfusion injury by inhibiting the activation of NF-κB and JAK2/STAT3. Hum Cell 2021; 34:1114-1122. [PMID: 33830473 PMCID: PMC8197719 DOI: 10.1007/s13577-021-00527-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 04/03/2021] [Indexed: 11/27/2022]
Abstract
LINGO-1 may be involved in the pathogenesis of cerebral ischemia. However, its biological function and underlying molecular mechanism in cerebral ischemia remain to be further defined. In our study, middle cerebral artery occlusion/reperfusion (MACO/R) mice model and HT22 cell oxygen–glucose deprivation/reperfusion (OGD/R) were established to simulate the pathological process of cerebral ischemia in vivo and in vitro and to detect the relevant mechanism. We found that LINGO-1 mRNA and protein were upregulated in mice and cell models. Down-regulation LINGO-1 improved the neurological symptoms and reduced pathological changes and the infarct size of the mice after MACO/R. In addition, LINGO-1 interference alleviated apoptosis and promoted cell proliferation in HT22 of OGD/R. Moreover, down-regulation of LINGO-1 proved to inhibit nuclear translocation of p-NF-κB and reduce the expression level of p-JAK2 and p-STAT3. In conclusion, our data suggest that shLINGO-1 attenuated ischemic injury by negatively regulating NF-KB and JAK2/STAT3 pathways, highlighting a novel therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Jiaying Zhu
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Zhu Zhu
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Yipin Ren
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Yukang Dong
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Yaqi Li
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Xiulin Yang
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China.
| |
Collapse
|
45
|
Subedi L, Gaire BP. Phytochemicals as regulators of microglia/macrophages activation in cerebral ischemia. Pharmacol Res 2021; 165:105419. [DOI: 10.1016/j.phrs.2021.105419] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 12/16/2020] [Accepted: 01/02/2021] [Indexed: 12/12/2022]
|
46
|
Lipoxin A4 regulates microglial M1/M2 polarization after cerebral ischemia-reperfusion injury via the Notch signaling pathway. Exp Neurol 2021; 339:113645. [PMID: 33600815 DOI: 10.1016/j.expneurol.2021.113645] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/30/2021] [Accepted: 02/12/2021] [Indexed: 11/23/2022]
Abstract
Microglia are rapidly activated after acute ischemic stroke, and the polarization of microglial is associated with the prognosis of acute ischemic stroke. Lipoxin A4 (LXA4), an anti-inflammatory agent, has a protective effect against ischemic stroke. However, the role of LXA4 on the polarization of microglial after acute ischemic stroke remains undetermined. We hypothesized that LXA4 may exert the neuroprotective effect though regulating the polarization of microglial. In this study, clinical features of acute ischemic stroke were simulated using a rat model of model of middle cerebral artery occlusion (MCAO) in vivo and the BV2 microglia oxygen-glucose deprivation/reoxygenation model (OGD/R) in vitro. The protective effects of LXA4 on cerebral ischemia-reperfusion injury were determined using TTC staining, HE staining, and TUNEL staining. The expression of targeted genes was assayed using quantitative real-time PCR (qRT-PCR), immunofluorescence, and western blot to investigated the regulation of LXA4 on microglia polarization after acute ischemic stroke. We found that LXA4 exerted protective effects on focal cerebral ischemia-reperfusion injury and reduced the expression of the pro-inflammatory cytokines IL-1β and TNF-α. Furthermore, LXA4 inhibited the expression of Notch-1, Hes1, iNOS and CD32 all of which are associated with the differentiation into M1 microglia. By contrast, LXA4 upregulated the expression of Hes5, Arg-1 and CD206 all of which are associated with M2 phenotype in microglia. In addition, blocking the Notch signaling pathway with the inhibitor DAPT significantly mitigated the effect of LXA4 on microglia differentiation. These data suggest that LXA4 may regulate the polarization of microglia after cerebral ischemia-reperfusion injury through the Notch signaling pathway.
Collapse
|
47
|
Malone K, Diaz Diaz AC, Shearer JA, Moore AC, Waeber C. The effect of fingolimod on regulatory T cells in a mouse model of brain ischaemia. J Neuroinflammation 2021; 18:37. [PMID: 33516262 PMCID: PMC7847573 DOI: 10.1186/s12974-021-02083-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/14/2021] [Indexed: 11/30/2022] Open
Abstract
Background The role of the immune system in stroke is well-recognised. Fingolimod, an immunomodulatory agent licensed for the management of relapsing-remitting multiple sclerosis, has been shown to provide benefit in rodent models of stroke. Its mechanism of action, however, remains unclear. We hypothesised fingolimod increases the number and/or function of regulatory T cells (Treg), a lymphocyte population which promotes stroke recovery. The primary aim of this study was to rigorously investigate the effect of fingolimod on Tregs in a mouse model of brain ischaemia. The effect of fingolimod in mice with common stroke-related comorbidities (ageing and hypercholesteremia) was also investigated. Methods Young (15–17 weeks), aged C57BL/6 mice (72–73 weeks), and ApoE−/− mice fed a high-fat diet (20–21 weeks) underwent permanent electrocoagulation of the left middle cerebral artery. Mice received either saline or fingolimod (0.5 mg/kg or 1 mg/kg) at 2, 24, and 48 h post-ischaemia via intraperitoneal injection. Another cohort of young mice (8–9, 17–19 weeks) received short-term (5 days) or long-term (10 days) fingolimod (0.5 mg/kg) treatment. Flow cytometry was used to quantify Tregs in blood, spleen, and lymph nodes. Immunohistochemistry was used to quantify FoxP3+ cell infiltration into the ischaemic brain. Results Fingolimod significantly increased the frequency of Tregs within the CD4+ T cell population in blood and spleen post-ischaemia in all three mouse cohorts compared to untreated ischemic mice. The highest splenic Treg frequency in fingolimod-treated mice was observed in ApoE−/− mice (9.32 ± 1.73% vs. 7.8 ± 3.01% in young, 6.09 ± 1.64% in aged mice). The highest circulating Treg frequency was also noted in ApoE−/− mice (8.39 ± 3.26% vs. 5.43 ± 2.74% in young, 4.56 ± 1.60% in aged mice). Fingolimod significantly increased the number of FoxP3+ cells in the infarct core of all mice. The most pronounced effects were seen when mice were treated for 10 days post-ischaemia. Conclusions Fingolimod increases Treg frequency in spleen and blood post-ischaemia and enhances the number of FoxP3+ cells in the ischaemic brain. The effect of fingolimod on this regulatory cell population may underlie its neuroprotective activity and could be exploited as part of future stroke therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02083-5.
Collapse
Affiliation(s)
- Kyle Malone
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Andrea C Diaz Diaz
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Jennifer A Shearer
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Anne C Moore
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Christian Waeber
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland. .,School of Pharmacy, University College Cork, Cork, Ireland.
| |
Collapse
|
48
|
Matei N, Camara J, Zhang JH. The Next Step in the Treatment of Stroke. Front Neurol 2021; 11:582605. [PMID: 33551950 PMCID: PMC7862333 DOI: 10.3389/fneur.2020.582605] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/23/2020] [Indexed: 12/12/2022] Open
Abstract
Although many patients do not receive reperfusion therapy because of delayed presentation and/or severity and location of infarct, new reperfusion approaches are expanding the window of intervention. Novel application of neuroprotective agents in combination with the latest methods of reperfusion provide a path to improved stroke intervention outcomes. We examine why neuroprotective agents have failed to translate to the clinic and provide suggestions for new approaches. New developments in recanalization therapy in combination with therapeutics evaluated in parallel animal models of disease will allow for novel, intra-arterial deployment of therapeutic agents over a vastly expanded therapeutic time window and with greater likelihood success. Although the field of neuronal, endothelial, and glial protective therapies has seen numerous large trials, the application of therapies in the context of newly developed reperfusion strategies is still in its infancy. Given modern imaging developments, evaluation of the penumbra will likely play a larger role in the evolving management of stroke. Increasingly more patients will be screened with neuroimaging to identify patients with adequate collateral blood supply allowing for delayed rescue of the penumbra. These patients will be ideal candidates for therapies such as reperfusion dependent therapeutic agents that pair optimally with cutting-edge reperfusion techniques.
Collapse
Affiliation(s)
- Nathanael Matei
- Department of Ophthalmology, University of Southern California, Los Angeles, CA, United States
| | - Justin Camara
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, United States
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, United States.,Department of Anesthesiology, Loma Linda University, Loma Linda, CA, United States.,Department of Neurosurgery, Loma Linda University, Loma Linda, CA, United States
| |
Collapse
|
49
|
Szabó MR, Pipicz M, Csont T, Csonka C. Modulatory Effect of Myokines on Reactive Oxygen Species in Ischemia/Reperfusion. Int J Mol Sci 2020; 21:ijms21249382. [PMID: 33317180 PMCID: PMC7763329 DOI: 10.3390/ijms21249382] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/06/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
There is a growing body of evidence showing the importance of physical activity against acute ischemic events in various organs. Ischemia/reperfusion injury (I/R) is characterized by tissue damage as a result of restriction and subsequent restoration of blood supply to an organ. Oxidative stress due to increased reactive oxygen species formation and/or insufficient antioxidant defense is considered to play an important role in I/R. Physical activity not only decreases the general risk factors for ischemia but also confers direct anti-ischemic protection via myokine production. Myokines are skeletal muscle-derived cytokines, representing multifunctional communication channels between the contracting skeletal muscle and other organs through an endocrine manner. In this review, we discuss the most prominent members of the myokines (i.e., brain-derived neurotrophic factor (BDNF), cathepsin B, decorin, fibroblast growth factors-2 and -21, follistatin, follistatin-like, insulin-like growth factor-1; interleukin-6, interleukin-7, interleukin-15, irisin, leukemia inhibitory factor, meteorin-like, myonectin, musclin, myostatin, and osteoglycin) with a particular interest in their potential influence on reactive oxygen and nitrogen species formation or antioxidant capacity. A better understanding of the mechanism of action of myokines and particularly their participation in the regulation of oxidative stress may widen their possible therapeutic use and, thereby, may support the fight against I/R.
Collapse
Affiliation(s)
- Márton Richárd Szabó
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, 6720 Szeged, Hungary; (M.R.S.); (M.P.); (T.C.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - Márton Pipicz
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, 6720 Szeged, Hungary; (M.R.S.); (M.P.); (T.C.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - Tamás Csont
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, 6720 Szeged, Hungary; (M.R.S.); (M.P.); (T.C.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - Csaba Csonka
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, 6720 Szeged, Hungary; (M.R.S.); (M.P.); (T.C.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
- Department of Sports Medicine, University of Szeged, Tisza Lajos krt 107, 6725 Szeged, Hungary
- Correspondence: ; Tel.: +36-30-5432-693
| |
Collapse
|
50
|
Alam JJ, Krakovsky M, Germann U, Levy A. Continuous administration of a p38α inhibitor during the subacute phase after transient ischemia-induced stroke in the rat promotes dose-dependent functional recovery accompanied by increase in brain BDNF protein level. PLoS One 2020; 15:e0233073. [PMID: 33275615 PMCID: PMC7717516 DOI: 10.1371/journal.pone.0233073] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022] Open
Abstract
There is unmet need for effective stroke therapies. Numerous neuroprotection attempts for acute cerebral ischemia have failed and as a result there is growing interest in developing therapies to promote functional recovery through increasing synaptic plasticity. For this research study, we hypothesized that in addition to its previously reported role in mediating cell death during the acute phase, the alpha isoform of p38 mitogen-activated protein kinase, p38α, may also contribute to interleukin-1β-mediated impairment of functional recovery during the subacute phase after acute ischemic stroke. Accordingly, an oral, brain-penetrant, small molecule p38α inhibitor, neflamapimod, was evaluated as a subacute phase stroke treatment to promote functional recovery. Neflamapimod administration to rats after transient middle cerebral artery occlusion at two dose levels was initiated outside of the previously characterized therapeutic window for neuroprotection of less than 24 hours for p38α inhibitors. Six-week administration of neflamapimod, starting at 48 hours after reperfusion, significantly improved behavioral outcomes assessed by the modified neurological severity score at Week 4 and at Week 6 post stroke in a dose-dependent manner. Neflamapimod demonstrated beneficial effects on additional measures of sensory and motor function. It also resulted in a dose-related increase in brain-derived neurotrophic factor (BDNF) protein levels, a previously reported potential marker of synaptic plasticity that was measured in brain homogenates at sacrifice. Taken together with literature evidence on the role of p38α-dependent suppression by interleukin-1β of BDNF-mediated synaptic plasticity and BDNF production, our findings support a mechanistic model in which inhibition of p38α promotes functional recovery after ischemic stroke by blocking the deleterious effects of interleukin-1β on synaptic plasticity. The dose-related in vivo efficacy of neflamapimod offers the possibility of having a therapy for stroke that could be initiated outside the short time window for neuroprotection and for improving recovery after a completed stroke.
Collapse
Affiliation(s)
- John J. Alam
- EIP Pharma, Inc., Boston, Massachusetts, United States of America
- * E-mail:
| | | | - Ursula Germann
- EIP Pharma, Inc., Boston, Massachusetts, United States of America
| | | |
Collapse
|