1
|
Usman M, He G, Lu H. Experimental study on the mechanism of cerebral edema development and MM-MRI manifestation in burned rats. J Neuroradiol 2025; 52:101323. [PMID: 39961526 DOI: 10.1016/j.neurad.2025.101323] [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: 08/16/2024] [Revised: 02/02/2025] [Accepted: 02/02/2025] [Indexed: 02/24/2025]
Abstract
OBJECTIVE The goal of this study was to investigate post-burn cerebral edema, establish its MM-MRI manifestation, and explore the underlying molecular mechanisms. METHODS Rats were randomly assigned to four groups (8h, 24h, 48h, and 72h post-burn) and subjected to thermal burns to induce skin injury, following the rat burn model. Treatment was administered based on Parkland's formula. At specific time points, rats were evaluated using MM-MRI sequences (T1 WI, T2 WI, T2 FLAIR, DWI, and ADC mapping) alongside histological analysis (H&E, TEM) and molecular techniques (IHC, IF, and WB). RESULTS All experimental groups exhibited significantly increased post-burn cerebral edema compared to the sham control group. While no significant changes were observed on T1WI, T2 WI, and T2 FLAIR images, post-burn cerebral edema was clearly visible on DWI and ADC maps in the region of interest (ROI) the basal ganglia. Histological analysis (H&E, TEM) corroborated these findings. Notably, all experimental groups (8h, 24h, 48h, and 72h) showed upregulated expression of AQP4 compared to controls, as evidenced by IHC, IF, and WB. Further, astrocyte end-feet and endothelial cells exhibited significant swelling may be due to AQP4 overexpression, leading to increased intracellular water content. CONCLUSION This study confirms the presence of post-burn cerebral edema in the early stages following burn trauma, might be mediated by AQP4, as supported by histological findings. Radiological results indicate that DWI and ADC mapping are sensitive methods for diagnosing and monitoring post-burn cerebral edema.
Collapse
Affiliation(s)
- Muhammad Usman
- Central Hospital Affiliated with Chongqing University of Technology, Gonglian yicun No.1 street lijiatuo, Banan district, Chongqing, 400054, PR China; Department of Anatomy, Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, 400016, PR China.
| | - Guiqiong He
- Department of Anatomy, Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, 400016, PR China.
| | - Hong Lu
- Department of Radiology, Central Hospital Affiliated to Chongqing University of Technology, Gonglian Yicun No. 1 Street Lijiatuo, Banan District, Chongqing 400054, PR China.
| |
Collapse
|
2
|
Kharkongor R, Stephen J, Khan U, Radhakrishnan R. Exposure to an enriched environment and fucoidan supplementation ameliorate learning and memory function in rats subjected to global cerebral ischemia. Neurosci Lett 2025; 847:138094. [PMID: 39736397 DOI: 10.1016/j.neulet.2024.138094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/01/2025]
Abstract
An enriched environment (EE) constitutes a proficient strategy that instigates social, cognitive, and motor faculties, fostering healing and heightening learning and memory function after ischemia, while fucoidan derived from brown seaweed encompasses a diverse array of bioactivities and is known to possess neuroprotective properties. This study aims to investigate the effectiveness of combining fucoidan and EE in a rat model of vascular dementia to overcome cognitive challenges. The rats were randomly assigned as Sham, Lesion - 4-vessel occlusion (4VO) i.e., transient global cerebral ischemia (tGCI), 4VO + F50mg/kg, 4VO + EE, and 4VO + F50mg/kg + EE. At the end of the study periods, the rats were exposed to the Novel object task, T-maze, and the Morris water maze. The profile of hippocampal pyramidal neurons and their dendrites was assessed through the CFV, and Golgi cox stained brain sections. Neuroinflammatory markers (IL-1β, IL-6, NF-κB, TNF-α) and synaptogenic markers (BDNF, SYP, PSD-95) were evaluated through western blot analysis. The levels of oxidative stress marker (LPO) and antioxidants (SOD, CAT, GSH, GST, GPX) in the hippocampus were quantified through biochemical assay. The findings revealed that the cognitive deficits were significantly reduced in both the 4VO + F50mg/kg and 4VO + F50mg/kg + EE treatment groups and inflammatory markers were reduced with increased antioxidant levels and synaptogenic markers when compared with the lesion group. However, through this study, the combination therapy involving fucoidan and exposure to an EE was proven effective in preserving neural integrity and restoring cognitive function against the damage caused by oxidative stress and inflammation following tGCI.
Collapse
Affiliation(s)
- Ronyson Kharkongor
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - JenishaChris Stephen
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - UlfathTasneem Khan
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - Rameshkumar Radhakrishnan
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai, India.
| |
Collapse
|
3
|
Boyle BR, Berghella AP, Blanco-Suarez E. Astrocyte Regulation of Neuronal Function and Survival in Stroke Pathophysiology. ADVANCES IN NEUROBIOLOGY 2024; 39:233-267. [PMID: 39190078 DOI: 10.1007/978-3-031-64839-7_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
The interactions between astrocytes and neurons in the context of stroke play crucial roles in the disease's progression and eventual outcomes. After a stroke, astrocytes undergo significant changes in their morphology, molecular profile, and function, together termed reactive astrogliosis. Many of these changes modulate how astrocytes relate to neurons, inducing mechanisms both beneficial and detrimental to stroke recovery. For example, excessive glutamate release and astrocytic malfunction contribute to excitotoxicity in stroke, eventually causing neuronal death. Astrocytes also provide essential metabolic support and neurotrophic signals to neurons after stroke, ensuring homeostatic stability and promoting neuronal survival. Furthermore, several astrocyte-secreted molecules regulate synaptic plasticity in response to stroke, allowing for the rewiring of neural circuits to compensate for damaged areas. In this chapter, we highlight the current understanding of the interactions between astrocytes and neurons in response to stroke, explaining the varied mechanisms contributing to injury progression and the potential implications for future therapeutic interventions.
Collapse
Affiliation(s)
- Bridget R Boyle
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrea P Berghella
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Elena Blanco-Suarez
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Neurological Surgery, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Stommel AM, Högler S, Mueller M, Magnet IAM, Kodajova P, Ullram B, Szinovatz A, Panzer FP, Engenhart-Seyrl A, Kaschmekat J, Schütz T, Holzer M, Weihs W. A ventricular fibrillation cardiac arrest model with extracorporeal cardiopulmonary resuscitation in rats: 8 minutes arrest time leads to increased myocardial damage but does not increase neuronal damage compared to 6 minutes. Front Vet Sci 2023; 10:1276588. [PMID: 38026669 PMCID: PMC10655001 DOI: 10.3389/fvets.2023.1276588] [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: 08/12/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Extracorporeal cardiopulmonary resuscitation (ECPR) is an emerging strategy in highly selected patients with refractory cardiac arrest (CA). Animal models can help to identify new therapeutic strategies to improve neurological outcome and cardiac function after global ischemia in CA. Aim of the study was to establish a reproducible ECPR rat model of ventricular fibrillation CA (VFCA) that leads to consistent neuronal damage with acceptable long-term survival rates, which can be used for future research. Materials and methods Male Sprague Dawley rats were resuscitated with ECPR from 6 min (n = 15) and 8 min (n = 16) VFCA. Animals surviving for 14 days after return of spontaneous resuscitation (ROSC) were compared with sham operated animals (n = 10); neurological outcome was assessed daily until day 14. In the hippocampal cornu ammonis 1 region viable neurons were counted. Microglia and astrocyte reaction was assessed by Iba1 and GFAP immunohistochemistry, and collagen fibers in the myocardium were detected in Azan staining. QuPath was applied for quantification. Results Of the 15 rats included in the 6 min CA group, all achieved ROSC (100%) and 10 (67%) survived to 14 days; in the 8 min CA group, 15 (94%) achieved ROSC and 5 (31%) reached the endpoint. All sham animals (n = 10) survived 2 weeks. The quantity of viable neurons was significantly decreased, while the area displaying Iba1 and GFAP positive pixels was significantly increased in the hippocampus across both groups that experienced CA. Interestingly, there was no difference between the two CA groups regarding these changes. The myocardium in the 8 min CA group exhibited significantly more collagen fibers compared to the sham animals, without differences between 6- and 8-min CA groups. However, this significant increase was not observed in the 6 min CA group. Conclusion Our findings indicate a uniform occurrence of neuronal damage in the hippocampus across both CA groups. However, there was a decrease in survival following an 8-min CA. Consequently, a 6-min duration of CA resulted in predictable neurological damage without significant cardiac damage and ensured adequate survival rates up to 14 days. This appears to offer a reliable model for investigating neuroprotective therapies.
Collapse
Affiliation(s)
| | - Sandra Högler
- Department of Pathobiology, Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Matthias Mueller
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Petra Kodajova
- Department of Pathobiology, Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Benjamin Ullram
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Alexander Szinovatz
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Felix Paul Panzer
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Julia Kaschmekat
- Department of Pathobiology, Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Tamara Schütz
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Michael Holzer
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Weihs
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
5
|
Ghazvineh S, Salimi M, Dehghan S, Asemi-Rad A, Dehdar K, Salimi A, Jamaati H, Raoufy MR. Stimulating olfactory epithelium mitigates mechanical ventilation-induced hippocampal inflammation and apoptosis. Hippocampus 2023; 33:880-885. [PMID: 36864704 DOI: 10.1002/hipo.23523] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/13/2023] [Accepted: 02/19/2023] [Indexed: 03/04/2023]
Abstract
Mechanical ventilation (MV), as a life-saving procedure in critical patients, is a risk factor to develop of neurocognitive dysfunction and triggers of inflammation and apoptosis in the brain. Since diversion of breathing route to the tracheal tube diminishes brain activity entrained by physiological nasal breathing, we hypothesized that simulating nasal breathing using rhythmic air-puff (AP) into the nasal cavity of mechanically ventilated rats can reduce hippocampal inflammation and apoptosis in association with restoring respiration-coupled oscillations. We found that stimulating olfactory epithelium through applying rhythmic nasal AP, in association with reviving respiration-coupled brain rhythm, mitigates MV-induced hippocampal apoptosis and inflammation involving microglia and astrocytes. The current translational study opens a window for a novel therapeutic approach to reduce neurological complications induced by MV.
Collapse
Affiliation(s)
- Sepideh Ghazvineh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Morteza Salimi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Samaneh Dehghan
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Eye Research Center, The Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Azam Asemi-Rad
- Department of Anatomy, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Kolsoum Dehdar
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Salimi
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamidreza Jamaati
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Raoufy
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Faculty of Medical Sciences, Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
6
|
Yi X, Tang X, Li T, Chen L, He H, Wu X, Xiang C, Cao M, Wang Z, Wang Y, Wang Y, Huang X. Therapeutic potential of the sphingosine kinase 1 inhibitor, PF-543. Biomed Pharmacother 2023; 163:114401. [PMID: 37167721 DOI: 10.1016/j.biopha.2023.114401] [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: 11/23/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 05/13/2023] Open
Abstract
PF-543 is a sphingosine kinase 1(SPHK1)inhibitor developed by Pfizer and is currently considered the most potent selective SPHK1 inhibitor. SPHK1 catalyses the production of sphingosine 1-phosphate (S1P) from sphingosine. It is the rate-limiting enzyme of S1P production, and there is substantial evidence to support a very important role for sphingosine kinase in health and disease. This review is the first to summarize the role and mechanisms of PF-543 as an SPHK1 inhibitor in anticancer, antifibrotic, and anti-inflammatory processes, providing new therapeutic leads and ideas for future research and clinical trials.
Collapse
Affiliation(s)
- Xueliang Yi
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; University of Electronic Science and Technology of China, China
| | - Xuemei Tang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Tianlong Li
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lin Chen
- University of Electronic Science and Technology of China, China
| | - Hongli He
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; University of Electronic Science and Technology of China, China
| | - Xiaoxiao Wu
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Chunlin Xiang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Min Cao
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zixiang Wang
- University of Electronic Science and Technology of China, China
| | - Yi Wang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; University of Electronic Science and Technology of China, China.
| | - Yiping Wang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; University of Electronic Science and Technology of China, China.
| | - Xiaobo Huang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; University of Electronic Science and Technology of China, China.
| |
Collapse
|
7
|
Yu S, Wu C, Zhu Y, Diao M, Hu W. Rat model of asphyxia-induced cardiac arrest and resuscitation. Front Neurosci 2023; 16:1087725. [PMID: 36685224 PMCID: PMC9846144 DOI: 10.3389/fnins.2022.1087725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/07/2022] [Indexed: 01/05/2023] Open
Abstract
Neurologic injury after cardiopulmonary resuscitation is the main cause of the low survival rate and poor quality of life among patients who have experienced cardiac arrest. In the United States, as the American Heart Association reported, emergency medical services respond to more than 347,000 adults and more than 7,000 children with out-of-hospital cardiac arrest each year. In-hospital cardiac arrest is estimated to occur in 9.7 per 1,000 adult cardiac arrests and 2.7 pediatric events per 1,000 hospitalizations. Yet the pathophysiological mechanisms of this injury remain unclear. Experimental animal models are valuable for exploring the etiologies and mechanisms of diseases and their interventions. In this review, we summarize how to establish a standardized rat model of asphyxia-induced cardiac arrest. There are four key focal areas: (1) selection of animal species; (2) factors to consider during modeling; (3) intervention management after return of spontaneous circulation; and (4) evaluation of neurologic function. The aim was to simplify a complex animal model, toward clarifying cardiac arrest pathophysiological processes. It also aimed to help standardize model establishment, toward facilitating experiment homogenization, convenient interexperimental comparisons, and translation of experimental results to clinical application.
Collapse
|
8
|
Meyer E, Rieder P, Gobbo D, Candido G, Scheller A, de Oliveira RMW, Kirchhoff F. Cannabidiol Exerts a Neuroprotective and Glia-Balancing Effect in the Subacute Phase of Stroke. Int J Mol Sci 2022; 23:12886. [PMID: 36361675 PMCID: PMC9659180 DOI: 10.3390/ijms232112886] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/16/2022] [Accepted: 10/20/2022] [Indexed: 11/22/2022] Open
Abstract
Pharmacological agents limiting secondary tissue loss and improving functional outcomes after stroke are still limited. Cannabidiol (CBD), the major non-psychoactive component of Cannabis sativa, has been proposed as a neuroprotective agent against experimental cerebral ischemia. The effects of CBD mostly relate to the modulation of neuroinflammation, including glial activation. To investigate the effects of CBD on glial cells after focal ischemia in vivo, we performed time-lapse imaging of microglia and astroglial Ca2+ signaling in the somatosensory cortex in the subacute phase of stroke by in vivo two-photon laser-scanning microscopy using transgenic mice with microglial EGFP expression and astrocyte-specific expression of the genetically encoded Ca2+ sensor GCaMP3. CBD (10 mg/kg, intraperitoneally) prevented ischemia-induced neurological impairment, reducing the neurological deficit score from 2.0 ± 1.2 to 0.8 ± 0.8, and protected against neurodegeneration, as shown by the reduction (more than 70%) in Fluoro-Jade C staining (18.8 ± 7.5 to 5.3 ± 0.3). CBD reduced ischemia-induced microglial activation assessed by changes in soma area and total branch length, and exerted a balancing effect on astroglial Ca2+ signals. Our findings indicate that the neuroprotective effects of CBD may occur in the subacute phase of ischemia, and reinforce its strong anti-inflammatory property. Nevertheless, its mechanism of action on glial cells still requires further studies.
Collapse
Affiliation(s)
- Erika Meyer
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
- Laboratory of Brain Ischemia and Neuroprotection, Department of Pharmacology and Therapeutics, State University of Maringá, Maringá 87020900, Brazil
| | - Phillip Rieder
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
| | - Davide Gobbo
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
| | - Gabriella Candido
- Laboratory of Brain Ischemia and Neuroprotection, Department of Pharmacology and Therapeutics, State University of Maringá, Maringá 87020900, Brazil
| | - Anja Scheller
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
| | - Rúbia Maria Weffort de Oliveira
- Laboratory of Brain Ischemia and Neuroprotection, Department of Pharmacology and Therapeutics, State University of Maringá, Maringá 87020900, Brazil
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
| |
Collapse
|
9
|
Moyse E, Krantic S, Djellouli N, Roger S, Angoulvant D, Debacq C, Leroy V, Fougere B, Aidoud A. Neuroinflammation: A Possible Link Between Chronic Vascular Disorders and Neurodegenerative Diseases. Front Aging Neurosci 2022; 14:827263. [PMID: 35663580 PMCID: PMC9161208 DOI: 10.3389/fnagi.2022.827263] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/22/2022] [Indexed: 12/12/2022] Open
Abstract
Various age-related diseases involve systemic inflammation, i.e. a stereotyped series of acute immune system responses, and aging itself is commonly associated with low-grade inflammation or inflamm’aging. Neuroinflammation is defined as inflammation-like processes inside the central nervous system, which this review discusses as a possible link between cardiovascular disease-related chronic inflammation and neurodegenerative diseases. To this aim, neuroinflammation mechanisms are first summarized, encompassing the cellular effectors and the molecular mediators. A comparative survey of the best-known physiological contexts of neuroinflammation (neurodegenerative diseases and transient ischemia) reveals some common features such as microglia activation. The recently published transcriptomic characterizations of microglia have pointed a marker core signature among neurodegenerative diseases, but also unraveled the discrepancies with neuroinflammations related with acute diseases of vascular origin. We next review the links between systemic inflammation and neuroinflammation, beginning with molecular features of respective pro-inflammatory cells, i.e. macrophages and microglia. Finally, we point out a gap of knowledge concerning the atherosclerosis-related neuroinflammation, which is for the most surprising given that atherosclerosis is established as a major risk factor for neurodegenerative diseases.
Collapse
Affiliation(s)
- Emmanuel Moyse
- University of Tours, EA4245, Transplantation, Immunologie, Inflammation, Tours, France
| | - Slavica Krantic
- Centre de Recherche Saint-Antoine (CRSA), Immune System and Neuroinflammation Laboratory, Hôpital Saint-Antoine, Inserm U938, Sorbonne Université, Paris, France
| | - Nesrine Djellouli
- University of Tours, EA4245, Transplantation, Immunologie, Inflammation, Tours, France
| | - Sébastien Roger
- University of Tours, EA4245, Transplantation, Immunologie, Inflammation, Tours, France
| | - Denis Angoulvant
- University of Tours, EA4245, Transplantation, Immunologie, Inflammation, Tours, France
- Department of Cardiology, Tours University Hospital, Tours, France
| | - Camille Debacq
- Division of Geriatric Medicine, Tours University Hospital, Tours, France
| | - Victoire Leroy
- Division of Geriatric Medicine, Tours University Hospital, Tours, France
- University of Tours, EA7505, Education, Ethics, Health, Tours, France
| | - Bertrand Fougere
- Division of Geriatric Medicine, Tours University Hospital, Tours, France
- University of Tours, EA7505, Education, Ethics, Health, Tours, France
- *Correspondence: Bertrand Fougere,
| | - Amal Aidoud
- University of Tours, EA4245, Transplantation, Immunologie, Inflammation, Tours, France
- Division of Geriatric Medicine, Tours University Hospital, Tours, France
| |
Collapse
|
10
|
Treatment of Modified Dahuang Fuzi Decoction on Cognitive Impairment Induced by Chronic Kidney Disease through Regulating AhR/NF- κB/JNK Signal Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:8489699. [PMID: 35463092 PMCID: PMC9023153 DOI: 10.1155/2022/8489699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 02/27/2022] [Accepted: 03/09/2022] [Indexed: 11/18/2022]
Abstract
Aim An increasing widespread of chronic kidney disease (CKD) has been established lately around the globe. In addition to renal function loss, CKD can also cause cognitive impairment (CI). Modified Dahuang Fuzi Decoction (MDFD) is used as a traditional Chinese therapy for CKD. The effect of MDFD on cognitive impairment induced by chronic kidney disease (CKD-CI), and therapeutic mechanisms were investigated. Methods The CKD animals' model was developed in the 5/6 nephrectomized mice. Sham operation and model groups received normal saline, while positive control and MDFD high/medium/low dose received Aricept (10 mg/kg/day) and different doses of MDFD (24, 16, and 8 g/kg/day), respectively. Cognitive function was detected with the Morris water maze test, while related factors were determined by ELISA. Histopathology and mechanism were studied using HE, western blot, and qRT-PCR. Results In the CKD-CI mice model, escape latency decreased significantly, whereas time of crossing platform and time spent within the platform quadrant increased substantially (P < 0.05) after MDFD treatment. Moreover, renal function and brain injury in CKD-CI improved dose-dependently, while the effect of MDFD-L was worse. Proteins such as aryl hydrocarbon receptor, nuclear factor-kappa B and c-Jun-N-terminal kinase, and mRNA in the kidney and brain of all the treatment groups decreased substantially (P < 0.05). Expression of tropomyosin receptor kinase B and brain-derived neurotrophic factor at protein and mRNA levels in the brain were significantly enhanced (P < 0.05). Conclusion MDFD presumably activated the BDNF/TrkB pathway by inhibiting the AhR/NF-κB/JNK signaling pathway to treat CKD-CI.
Collapse
|
11
|
Drabek T. Hemoadsorption after cardiac arrest - not quite the "shelter from the cytokine storm" we were hoping for. Resuscitation 2022; 173:179-181. [PMID: 35227817 DOI: 10.1016/j.resuscitation.2022.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 10/19/2022]
Affiliation(s)
- Tomas Drabek
- Safar Center for Resuscitation Research, Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, UPMC Presbyterian Hospital, 200 Lothrop St. Suite C-200, Pittsburgh, PA 15213, USA.
| |
Collapse
|
12
|
Li JL, Cao Y, Nie H. The Effect of Mild Hypothermia on Nogo-A and Neurological Function in the Brain after Cardiopulmonary Resuscitation in Rats. Fetal Pediatr Pathol 2022; 41:198-207. [PMID: 32589081 DOI: 10.1080/15513815.2020.1783407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
ObjectiveWe investigated the dynamic changes of Nogo-A protein in brain and the effects of mild therapeutic hypothermia (MTH) on its expression after cardiopulmonary resuscitation (CPR). Methods: Western-blotting and neurological scoring of 45 rats subjected to cardiac arrest and CPR with and without MTR were performed to investigate the changes in the expression of Nogo-A protein in the hippocampus and cortex over a period of time ranging from 6 h to 72 h after restoration of spontaneous circulation (ROSC). Results: Nogo-A expression levels were increased at 6 h after CPR in the hippocampus and cortex, peaked at 24 h in the cortex, and at 48 h in the hippocampus. The expression of Nogo-A in the MTR group was significantly lower at 12 h (p < 0.05) compared to those with no MTR after ROSC. Conclusions: MTR blunts the expression of Nogo-A protein in the hippocampus and cortex after cardiac arrest and resuscitation, and MTR may provide cerebral protection after ischemia.
Collapse
Affiliation(s)
- Jia-Li Li
- Department of Emergency, The Second Affifiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, Sichuan, China
| | - Yu Cao
- Department of Emergency, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Hu Nie
- Department of Emergency, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
13
|
Yang Y, Song J, Liu N, Wei G, Liu S, Zhang S, Jiang N, Yang H, Du G. Salvianolic acid A relieves cognitive disorder after chronic cerebral ischemia: Involvement of Drd2/Cryab/NF-κB pathway. Pharmacol Res 2022; 175:105989. [PMID: 34800628 DOI: 10.1016/j.phrs.2021.105989] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/03/2021] [Accepted: 11/12/2021] [Indexed: 12/14/2022]
Abstract
Chronic cerebral ischemia (CCI) refers to long-term hypoperfusion of cerebral blood flow with the main clinical manifestations of progressive cognitive impairment. The pathological mechanism of CCI is complex, and there is a lack of effective treatments. Salvianolic acid A (SalA) is a neuroprotective extract of Salvia miltiorrhiza with the effects of anti-inflammation and anti-apoptosis. In this study, the effect of SalA on cognitive function and Drd2/Cryab/NF-κB signaling pathway in rats with CCI was investigated. Morris water maze and open field test were used to observe the effects of SalA on the cognitive function of CCI rats. The pathological changes in the brain were observed by HE, Nissl, and LFB staining. TUNEL staining, enzyme-linked immunosorbent assay, and western blot analysis were used to detect the inflammatory and apoptosis in the cortex and hippocampus. The expression of Drd2/Cryab/NF-κB pathway-related molecules and Drd2 localization were detected by western blotting and dual immunofluorescence, respectively. SH-SY5Y cells were exposed to chronic hypoglycemic and hypoxic injury in vitro, and Drd2 inhibitor haloperidol was used to verify the involved pathway. The results showed that SalA could improve the cognitive function of CCI rats, reduce pathological damage of cortex and hippocampus, inhibit neuroinflammation and apoptosis, and suppress the activation of NF-κB by regulating Drd2/Cryab pathway. And SalA inhibited NF-κB activation and nuclear translocation in SH-SY5Y cells by upregulating Drd2/Cryab pathway, which was reversed by haloperidol interference. In conclusion, SalA could relieve CCI-induced cognitive impairment in rats, at least partly through the Drd2/Cryab/NF-κB pathway.
Collapse
Affiliation(s)
- Yujiao Yang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang 110016, PR China; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Junke Song
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| | - NanNan Liu
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 280 Waihuan East Road, Panyu District, Guangdong 510006, PR China
| | - Guangyi Wei
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 280 Waihuan East Road, Panyu District, Guangdong 510006, PR China
| | - Shan Liu
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 280 Waihuan East Road, Panyu District, Guangdong 510006, PR China
| | - Sen Zhang
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Nan Jiang
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; School of Pharmacy, Henan University, North Section of Jinming Avenue, Kaifeng 475004, Henan, PR China
| | - Haiguang Yang
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Guanhua Du
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang 110016, PR China; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| |
Collapse
|
14
|
Bassi TG, Rohrs EC, Fernandez KC, Ornowska M, Nicholas M, Gani M, Evans D, Reynolds SC. Transvenous Diaphragm Neurostimulation Mitigates Ventilation-associated Brain Injury. Am J Respir Crit Care Med 2021; 204:1391-1402. [PMID: 34491883 PMCID: PMC8865722 DOI: 10.1164/rccm.202101-0076oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 09/01/2021] [Indexed: 11/16/2022] Open
Abstract
Rationale: Mechanical ventilation (MV) is associated with hippocampal apoptosis and inflammation, and it is important to study strategies to mitigate them. Objectives: To explore whether temporary transvenous diaphragm neurostimulation (TTDN) in association with MV mitigates hippocampal apoptosis and inflammation after 50 hours of MV. Methods: Normal-lung porcine study comparing apoptotic index, inflammatory markers, and neurological-damage serum markers between never-ventilated subjects, subjects undergoing 50 hours of MV plus either TTDN every other breath or every breath, and subjects undergoing 50 hours of MV (MV group). MV settings in volume control were Vt of 8 ml/kg, and positive end-expiratory pressure of 5 cm H2O. Measurements and Main Results: Apoptotic indices, microglia percentages, and reactive astrocyte percentages were greater in the MV group in comparison with the other groups (P < 0.05). Transpulmonary pressure at baseline and at study end were both lower in the group receiving TTDN every breath, but lung injury scores and systemic inflammatory markers were not different between the groups. Serum concentrations of four neurological-damage markers were lower in the group receiving TTDN every breath than in the MV group (P < 0.05). Heart rate variability declined significantly in the MV group and increased significantly in both TTDN groups over the course of the experiments. Conclusions: Our study found that mechanical ventilation is associated with hippocampal apoptosis and inflammation, independent of lung injury and systemic inflammation. Also, in a porcine model, TTDN results in neuroprotection after 50 hours, and the degree of neuroprotection increases with greater exposure to TTDN.
Collapse
Affiliation(s)
- Thiago G. Bassi
- Simon Fraser University, Burnaby, British Columbia, Canada
- Lungpacer Medical Inc., Vancouver, British Columbia, Canada; and
| | - Elizabeth C. Rohrs
- Simon Fraser University, Burnaby, British Columbia, Canada
- Fraser Health Authority, Royal Columbian Hospital, New Westminster, British Columbia, Canada
| | - Karl C. Fernandez
- Simon Fraser University, Burnaby, British Columbia, Canada
- Fraser Health Authority, Royal Columbian Hospital, New Westminster, British Columbia, Canada
| | | | - Michelle Nicholas
- Simon Fraser University, Burnaby, British Columbia, Canada
- Fraser Health Authority, Royal Columbian Hospital, New Westminster, British Columbia, Canada
| | - Matt Gani
- Lungpacer Medical Inc., Vancouver, British Columbia, Canada; and
| | - Doug Evans
- Lungpacer Medical Inc., Vancouver, British Columbia, Canada; and
| | - Steven C. Reynolds
- Simon Fraser University, Burnaby, British Columbia, Canada
- Fraser Health Authority, Royal Columbian Hospital, New Westminster, British Columbia, Canada
| |
Collapse
|
15
|
Zhang G, Lai Z, Gu L, Xu K, Wang Z, Duan Y, Chen H, Zhang M, Zhang J, Zhao Z, Wang S. Delta Opioid Receptor Activation with Delta Opioid Peptide [d-Ala2, d-Leu5] Enkephalin Contributes to Synaptic Improvement in Rat Hippocampus against Global Ischemia. Cell Transplant 2021; 30:9636897211041585. [PMID: 34470528 PMCID: PMC8419564 DOI: 10.1177/09636897211041585] [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] [Indexed: 11/28/2022] Open
Abstract
Global cerebral ischemia induced by cardiac arrest usually leads to poor neurological outcomes. Numerous studies have focused on ways to prevent ischemic damage in the brain, however clinical therapies are still limited. Our previous studies revealed that delta opioid receptor (DOR) activation with [d-Ala2, d-Leu5] enkephalin (DADLE), a DOR agonist, not only significantly promotes neuronal survival on day 3, but also improves spatial memory deficits on days 5-9 after ischemia. However, the neurological mechanism underlying DADLE-induced cognitive recovery remains unclear. This study first examined the changes in neuronal survival in the CA1 region at the advanced time point (day 7) after ischemia/reperfusion (I/R) injury and found a significant amelioration of damaged CA1 neurons in the rats treated with DADLE (2.5 nmol) when administered at the onset of reperfusion. The structure and function of CA1 neurons on days 3 and 7 post-ischemia showed significant improvements in both the density of the injured dendritic spines and the basic transmission of the impaired CA3-CA1 synapses following DADLE treatment. The molecular changes involved in DADLE-mediated synaptic modulation on days 3 and 7 post-ischemia implied the time-related differential regulation of PKCα-MARCKS on the dendritic spine structure and of BDNF- ERK1/2-synapsin I on synaptic function, in response to ischemic/reperfusion injury as well as to DADLE treatment. Importantly, all the beneficial effects of DADLE on ischemia-induced cellular, synaptic, and molecular deficits were eliminated by the DOR inhibitor naltrindole (2.5 nmol). Taken together, this study suggested that DOR activation-induced protective signaling pathways of PKCα-MARCKS involved in the synaptic morphology and BDNF-ERK-synapsin I in synaptic transmission may be engaged in the cognitive recovery in rats suffering from advanced cerebral ischemia.
Collapse
Affiliation(s)
- Guangming Zhang
- Department of Anesthesiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Zelin Lai
- Shanghai Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Lingling Gu
- Shanghai Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Kejia Xu
- Department of Anesthesiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Zhenlu Wang
- Shanghai Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Yale Duan
- Shanghai Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Huifen Chen
- Department of Clinical Laboratory, Shanghai First Maternity and Infant Hospital
| | - Min Zhang
- Tongji University School of Medicine, Shanghai 201204, China
| | - Jun Zhang
- Department of Clinical Laboratory, Shanghai First Maternity and Infant Hospital.,Tongji University School of Medicine, Shanghai 201204, China
| | - Zheng Zhao
- Shanghai Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Shuyan Wang
- Department of Anesthesiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| |
Collapse
|
16
|
Vilhena ER, Bonato JM, Schepers M, Kunieda JKC, Milani H, Vanmierlo T, Prickaerts J, de Oliveira RMW. Positive effects of roflumilast on behavior, neuroinflammation, and white matter injury in mice with global cerebral ischemia. Behav Pharmacol 2021; 32:459-471. [PMID: 34320520 DOI: 10.1097/fbp.0000000000000640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Inhibition of phosphodiesterase 4 (PDE4) is a promising pharmacological strategy for the treatment of cerebral ischemic conditions. To increase the relevance and increase the translational value of preclinical studies, it is important to conduct experiments using different animal species and strains, different animal models, and to evaluate long-term functional outcomes after cerebral ischemia. In the present study, the effects of the selective PDE4 inhibitor roflumilast were evaluated in vivo and in vitro. Balb/c mice were subjected to bilateral common carotid artery occlusion (BCCAO) and tested during 21 days in multiple behavioral tasks to investigate the long-term effects of roflumilast on functional recovery. The effects of roflumilast were also investigated on hippocampal cell loss, white matter injury, and expression of neuroinflammatory markers. Roflumilast prevented cognitive and emotional deficits induced by BCCAO in mice. Roflumilast also prevented neurodegeneration and reduced the white matter damage in the brain of ischemic animals. Besides, roflumilast decreased Iba-1 (microglia marker) levels and increased Arginase-1 (Arg-1; microglia M2 phenotype marker) levels in the hippocampus of these mice. Likewise, roflumilast suppressed inducible nitric oxide synthase (microglia M1 phenotype marker) expression and increased Arg-1 levels in a primary mouse microglia culture. These findings support evidence that PDE4 inhibition by roflumilast might be beneficial in cerebral ischemic conditions. The neuroprotective effects of roflumilast appear to be mediated by a decrease in neuroinflammation.
Collapse
Affiliation(s)
- Emanuella R Vilhena
- Department of Pharmacology and Therapeutics, State University of Maringá, Paraná, Brazil
| | - Jéssica M Bonato
- Department of Pharmacology and Therapeutics, State University of Maringá, Paraná, Brazil
| | - Melissa Schepers
- Neuroimmune Connect and Repair Lab., Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Juliana K C Kunieda
- Department of Pharmacology and Therapeutics, State University of Maringá, Paraná, Brazil
| | - Humberto Milani
- Department of Pharmacology and Therapeutics, State University of Maringá, Paraná, Brazil
| | - Tim Vanmierlo
- Neuroimmune Connect and Repair Lab., Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Rúbia M W de Oliveira
- Department of Pharmacology and Therapeutics, State University of Maringá, Paraná, Brazil
| |
Collapse
|
17
|
CA1 Hippocampal Pyramidal Cells in Rats, Resuscitated From 8 Minutes of Ventricular Fibrillation Cardiac Arrest, Recover After 20 Weeks of Survival: A Retrospective Pilot Study. Shock 2021; 54:531-538. [PMID: 32931694 DOI: 10.1097/shk.0000000000001469] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
PURPOSE The cornu ammonis 1 (CA1) region of the hippocampus is specifically vulnerable to global ischemia. We hypothesized that histopathological outcome in a ventricular fibrillation cardiac arrest (VFCA) rat model depends on the time point of the examination. METHODS Male Sprague-Dawley rats were put into VFCA for 8 min, received chest compressions for 2 min, and were defibrillated to achieve return of spontaneous circulation. Animals surviving for 80 min, 14 days and 140 days were compared with controls. Viable neurons were counted in a 500 μm sector of the CA1 region and layer thickness measured. Microglia cells and astrocytes were counted in a 250×300 μm aspect. RESULTS Control and 80 min surviving animals had similar numbers of pyramidal neurons in the CA1 region. In 14 days and 140 days survivors neuron numbers and layer thickness were severely diminished compared with controls (P < 0.001). Two-thirds of the 140 days survivors showed significantly more viable neurons than the last third. Microglia was increased in 14 days survivors compared with controls and 140 days survivors, while astrocytes increased in 14 days and 140 days survivors compared with controls (P < 0.001). 140 days survivors had significantly higher astrocyte counts compared with 14 days survivors. CONCLUSIONS The amount and type of brain lesions present after global ischemia depend on the survival time. A consistent reduction in pyramidal cells in the CA1 region was present in all animals 14 days after VFCA, but in two-thirds of animals a repopulation of pyramidal cells seems to have taken place after 140 days.
Collapse
|
18
|
Fenton-Navarro B, Garduño Ríos D, Torner L, Letechipía-Vallejo G, Cervantes M. Melatonin Decreases Circulating Levels of Galectin-3 and Cytokines, Motor Activity, and Anxiety Following Acute Global Cerebral Ischemia in Male Rats. Arch Med Res 2021; 52:505-513. [DOI: 10.1016/j.arcmed.2021.01.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/21/2020] [Accepted: 01/21/2021] [Indexed: 12/23/2022]
|
19
|
Wang N, Yu H, Song Q, Mao P, Li K, Bao G. Sesamol-loaded stearic acid-chitosan nanomicelles mitigate the oxidative stress-stimulated apoptosis and induction of pro-inflammatory cytokines in motor neuronal of the spinal cord through NF-ĸB signaling pathway. Int J Biol Macromol 2021; 186:23-32. [PMID: 34214577 DOI: 10.1016/j.ijbiomac.2021.06.171] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/15/2021] [Accepted: 06/26/2021] [Indexed: 10/21/2022]
Abstract
As natural potential antioxidants suffer from low cellular uptake, the development of drug-loaded nanoplatforms may provide useful information about the treatment of spinal cord injury (SCI). In the present study, sesamol (SM)-loaded stearic acid (SA) -chitosan (CS) nanomicelles were fabricated and well-characterized. Afterwards, the neuroprotective effects of SM@SA-CS nanomicelles against lipopolysaccharide (LPS)-induced oxidative stress in NSC-34 cells was assessed by different cellular and molecular pathways. It was deduced that the size of synthesized SM@SA-CS was in the range of 10-20 nm and the hydrodynamic radii of SA-CA and SM@SA-CA nanomicelles were 53.12 ± 6.21 nm and 59.12 ± 7.31 nm, respectively. Furthermore, SM@SA-CS nanomicelles displayed a sustained drug release at physiological pH, potential dissolution rate and stability even up to 15 days. Cellular assay showed that SM@SA-CS nanomicelles co-incubation with LPS for 24 h in comparison with free drug remarkably regulated cell survival, membrane leakage, generation of ROS, activity of non-enzymatic and enzymatic antioxidant systems, and apoptotic and inflammatory signaling pathway through NF-ĸB signaling pathway. These data indicated that SM@SA-CS nanomicelles can be developed as a promising platform for the mitigation of oxidative stress-mediated apoptosis in neural cells.
Collapse
Affiliation(s)
- Ning Wang
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Hai Yu
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Qian Song
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Ping Mao
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Kuo Li
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Gang Bao
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
20
|
Li F, Xu Y, Li X, Wang X, Yang Z, Li W, Cheng W, Yan G. Triblock Copolymer Nanomicelles Loaded with Curcumin Attenuates Inflammation via Inhibiting the NF-κB Pathway in the Rat Model of Cerebral Ischemia. Int J Nanomedicine 2021; 16:3173-3183. [PMID: 34007172 PMCID: PMC8121676 DOI: 10.2147/ijn.s300379] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/22/2021] [Indexed: 12/29/2022] Open
Abstract
AIM Cerebral ischemic injury is one of the debilitating diseases showing that inflammation plays an important role in worsening ischemic damage. Therefore, studying the effects of some potential anti-inflammatory compounds can be very important in the treatment of cerebral ischemic injury. METHODS This study investigated anti-inflammatory effects of triblock copolymer nanomicelles loaded with curcumin (abbreviated as NC) in the brain of rats following transient cerebral ischemia/reperfusion (I/R) injury in stroke. After preparation of NC, their protective effects against bilateral common carotid artery occlusion (BCCAO) were explored by different techniques. Concentrations of free curcumin (C) and NC in liver, kidney, brain, and heart organs, as well as in plasma, were measured using a spectrofluorometer. Western blot analysis was then used to measure NF-κB-p65 protein expression levels. Also, ELISA assay was used to examine the level of cytokines IL-1β, IL-6, and TNF-α. Lipid peroxidation levels were assessed using MDA assay and H&E staining was used for histopathological examination of the hippocampus tissue sections. RESULTS The results showed a higher level of NC compared to C in plasma and organs including the brain, heart, and kidneys. Significant upregulation of NF-κB, IL-1β, IL-6, and TNF-α expressions compared to control was observed in rats after induction of I/R, which leads to an increase in inflammation. However, NC was able to downregulate significantly the level of these inflammatory cytokines compared to C. Also, the level of lipid peroxidation in pre-treated rats with 80mg/kg NC was significantly reduced. CONCLUSION Our findings in the current study demonstrate a therapeutic effect of NC in an animal model of cerebral ischemia/reperfusion (I/R) injury in stroke through the downregulation of NF-κB-p65 protein and inflammatory cytokines.
Collapse
Affiliation(s)
- Fengguang Li
- Department of Neurology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, 430081, People’s Republic of China
| | - Yan Xu
- Department of Pharmacy, General Hospital of Central Theater Command, Wuhan, 430010, People’s Republic of China
| | - Xing Li
- Department of Neurology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, 430081, People’s Republic of China
| | - Xinghua Wang
- Department of Neurology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, 430081, People’s Republic of China
| | - Zhigang Yang
- Department of Neurology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, 430081, People’s Republic of China
| | - Wanli Li
- Department of Neurology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, 430081, People’s Republic of China
| | - Wei Cheng
- Department of Neurology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, 430081, People’s Republic of China
| | - Gangli Yan
- Department of Neurology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, 430081, People’s Republic of China
| |
Collapse
|
21
|
Shan Y, Hu J, Lv H, Cui X, Di W. miR-221 Exerts Neuroprotective Effects in Ischemic Stroke by Inhibiting the Proinflammatory Response. J Stroke Cerebrovasc Dis 2021; 30:105489. [PMID: 33276305 DOI: 10.1016/j.jstrokecerebrovasdis.2020.105489] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/03/2020] [Accepted: 11/16/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Ischemic stroke is clearly affected by microRNAs (miRNAs) due to dysfunction of their regulatory networks. Our clinical data confirmed decreased miR-221 levels in plasma collected from patients with acute ischemia compared with plasma from healthy controls. Therefore, we further aimed to demonstrate the regulatory mechanisms by which miR-221 exerts its neuroprotective effects in acute ischemic brain injury. METHODS Middle cerebral artery occlusion (MCAO) was used to establish focal cerebral ischemia in adult male C57BL/6 mice. A miR-221 mimic or a negative mimic control was injected by intracerebroventricular administration 24 h prior to MCAO. After 48 h, cerebral infarction volume and neurological scores were calculated, and to determine the extent of neuroprotection by miR-221, neurobehavioral tests were performed. Quantitative real-time PCR, ELISA, and flow cytometry were also performed to identify the expression of inflammation-related cytokines and chemokines as well as infiltration/activation of various immune cells in the brain. RESULTS The results showed that MCAO mice treated with a miR-221 mimic exhibited significantly decreased cerebral infarction volume and increased amelioration of behavioral deficits. Moreover, the expression of proinflammatory cytokines (TNF-α, MCP-1, VCAM-1, and IL-6) and chemokines (CCL2 and CCL3) was significantly decreased in the miR-221 mimic-treated group. In addition, the flow cytometry data showed that macrophage infiltration and microglial activation were blocked by miR-221 treatment. CONCLUSION our results indicate that miR-221 could decrease brain damage in the setting of acute ischemic stroke by inhibiting the proinflammatory response, which furthered our understanding of the molecular basis of miR-221 and provided a new potential therapeutic target for the treatment of ischemic stroke .
Collapse
Affiliation(s)
- Yuan Shan
- Department of Neurology, Shaanxi Provincial People's Hospital, No. 256 West Friendship Rd, Xi'an, China, 710068
| | - Jun Hu
- Department of Neurology, Shaanxi Provincial People's Hospital, No. 256 West Friendship Rd, Xi'an, China, 710068
| | - Hua Lv
- Department of Neurology, Shaanxi Provincial People's Hospital, No. 256 West Friendship Rd, Xi'an, China, 710068
| | - Xiaoli Cui
- Department of Neurology, Shaanxi Provincial People's Hospital, No. 256 West Friendship Rd, Xi'an, China, 710068
| | - Wei Di
- Department of Neurology, Shaanxi Provincial People's Hospital, No. 256 West Friendship Rd, Xi'an, China, 710068.
| |
Collapse
|
22
|
Uray T, Dezfulian C, Palmer AA, Miner KM, Leak RK, Stezoski JP, Janesko‐Feldman K, Kochanek PM, Drabek T. Cardiac Arrest Induced by Asphyxia Versus Ventricular Fibrillation Elicits Comparable Early Changes in Cytokine Levels in the Rat Brain, Heart, and Serum. J Am Heart Assoc 2021; 10:e018657. [PMID: 33599149 PMCID: PMC8174297 DOI: 10.1161/jaha.120.018657] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 01/04/2021] [Indexed: 12/14/2022]
Abstract
Background Current postresuscitative care after cardiac arrest (CA) does not address the cause of CA. We previously reported that asphyxial CA (ACA) and ventricular fibrillation CA (VFCA) elicit unique injury signatures. We hypothesized that the early cytokine profiles of the serum, heart, and brain differ in response to ACA versus VFCA. Methods and Results Adult male rats were subjected to 10 minutes of either ACA or VFCA. Naives and shams (anesthesia and surgery without CA) served as controls (n=12/group). Asphyxiation produced an ≈4-minute period of progressive hypoxemia followed by a no-flow duration of ≈6±1 minute. Ventricular fibrillation immediately induced no flow. Return of spontaneous circulation was achieved earlier after ACA compared with VFCA (42±18 versus 105±22 seconds; P<0.001). Brain cytokines in naives were, in general, low or undetectable. Shams exhibited a modest effect on select cytokines. Both ACA and VFCA resulted in robust cytokine responses in serum, heart, and brain at 3 hours. Significant regional differences pinpointed the striatum as a key location of neuroinflammation. No significant differences in cytokines, neuron-specific enolase, S100b, and troponin T were observed across CA models. Conclusions Both models of CA resulted in marked systemic, heart, and brain cytokine responses, with similar degrees of change across the 2 CA insults. Changes in cytokine levels after CA were most pronounced in the striatum compared with other brain regions. These collective observations suggest that the amplitude of the changes in cytokine levels after ACA versus VFCA may not mediate the differences in secondary injuries between these 2 CA phenotypes.
Collapse
Affiliation(s)
- Thomas Uray
- Safar Center for Resuscitation ResearchUniversity of Pittsburgh School of MedicinePittsburghPA
- Department of Critical Care MedicineUniversity of Pittsburgh School of MedicinePittsburghPA
- Department of Emergency MedicineVienna General HospitalMedical University of ViennaViennaAustria
| | - Cameron Dezfulian
- Safar Center for Resuscitation ResearchUniversity of Pittsburgh School of MedicinePittsburghPA
- Department of Critical Care MedicineUniversity of Pittsburgh School of MedicinePittsburghPA
| | - Abigail A. Palmer
- Safar Center for Resuscitation ResearchUniversity of Pittsburgh School of MedicinePittsburghPA
- Lake Erie College of Osteopathic MedicineEriePA
| | - Kristin M. Miner
- Graduate School of Pharmaceutical SciencesDuquesne UniversityPittsburghPA
| | - Rehana K. Leak
- Graduate School of Pharmaceutical SciencesDuquesne UniversityPittsburghPA
| | - Jason P. Stezoski
- Safar Center for Resuscitation ResearchUniversity of Pittsburgh School of MedicinePittsburghPA
- Department of Critical Care MedicineUniversity of Pittsburgh School of MedicinePittsburghPA
- Department of Anesthesiology and Perioperative MedicineUniversity of Pittsburgh School of MedicinePittsburghPA
| | - Keri Janesko‐Feldman
- Safar Center for Resuscitation ResearchUniversity of Pittsburgh School of MedicinePittsburghPA
- Department of Critical Care MedicineUniversity of Pittsburgh School of MedicinePittsburghPA
| | - Patrick M. Kochanek
- Safar Center for Resuscitation ResearchUniversity of Pittsburgh School of MedicinePittsburghPA
- Department of Critical Care MedicineUniversity of Pittsburgh School of MedicinePittsburghPA
| | - Tomas Drabek
- Safar Center for Resuscitation ResearchUniversity of Pittsburgh School of MedicinePittsburghPA
- Department of Anesthesiology and Perioperative MedicineUniversity of Pittsburgh School of MedicinePittsburghPA
| |
Collapse
|
23
|
Bonato JM, Meyer E, de Mendonça PSB, Milani H, Prickaerts J, Weffort de Oliveira RM. Roflumilast protects against spatial memory impairments and exerts anti-inflammatory effects after transient global cerebral ischemia. Eur J Neurosci 2021; 53:1171-1188. [PMID: 33340424 DOI: 10.1111/ejn.15089] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022]
Abstract
Phosphodiesterase 4 (PDE4) inhibitors have been shown to present beneficial effects in cerebral ischemic injury because of their ability to improve cognition and target different phases and mechanisms of cerebral ischemia, including apoptosis, neurogenesis, angiogenesis, and inflammation. The present study investigated whether repeated treatment with the PDE4 inhibitor roflumilast rescued memory loss and attenuated neuroinflammation in rats following transient global cerebral ischemia (TGCI). TGCI caused memory impairments, neuronal loss (reflected by Neuronal nuclei (NeuN) immunoreactivity), and compensatory neurogenesis (reflected by doublecortin (DCX) immunoreactivity) in the hippocampus. Also, increases in the protein expression of the phosphorylated response element-binding protein (pCREB) and inflammatory markers such as the glial fibrillary acidic protein (GFAP) and ionized calcium-binding adaptor molecule 1 (Iba-1), were detected in the hippocampus in TGCI rats. Repeated treatment with roflumilast (0.003 and 0.01 mg/kg) prevented spatial memory deficits without promoting hippocampal protection in ischemic animals. Roflumilast increased the levels of pCREB, arginase-1, interleukin (IL) 4, and IL-10 in the hippocampus 21 days after TGCI. These data suggest a protective effect of roflumilast against functional sequelae of cerebral ischemia, which might be related to its anti-inflammatory properties.
Collapse
Affiliation(s)
- Jéssica M Bonato
- Department of Pharmacology and Therapeutics, State University of Maringá, Maringá, Brazil
| | - Erika Meyer
- Department of Pharmacology and Therapeutics, State University of Maringá, Maringá, Brazil
| | | | - Humberto Milani
- Department of Pharmacology and Therapeutics, State University of Maringá, Maringá, Brazil
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | | |
Collapse
|
24
|
Huang J, Liu Y, Cheng L, Li J, Zhang T, Zhao G, Zhang H. Glucagon-like peptide-1 cleavage product GLP-1(9-36) reduces neuroinflammation from stroke via the activation of insulin-like growth factor 1 receptor in astrocytes. Eur J Pharmacol 2020; 887:173581. [PMID: 32949596 DOI: 10.1016/j.ejphar.2020.173581] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 12/20/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) is an endogenous gut hormone and a key regulator in maintaining glucose homeostasis by stimulating insulin secretion. Its natural cleavage product GLP-1 (9-36), which was formerly considered a "bio-inactive" metabolite mainly due to its low affinity for GLP-1 receptor, possesses unique properties such as cardiovascular protection. Little is known about the effects and mechanisms of GLP-1 (9-36) in cerebral ischemia and reperfusion injury. Here, we report that systemic application of GLP-1 (9-36) in adult mice facilitated functional recovery and reduced infarct volume, astrogliosis, and neuronal apoptosis following middle cerebral artery occlusion and reperfusion. Interestingly, these effects were still observed in GLP-1 receptor knockout (Glp-1rKO) mice but were partially reversed in insulin-like growth factor 1 (IGF-1) receptor knockdown (Igf-1rKD) mice. Primary astrocytes were cultured and subjected to oxygen-glucose deprivation/reoxygenation (OGD/R), and enzyme-linked immunosorbent assay indicated that GLP-1 (9-36) pretreatment reduces tumor necrosis factor-α, interleukin (IL)-1β, and IL-6 levels. This effect was not diminished in Glp-1rKO astrocytes but was reversed in Igf-1rKO astrocytes, emphasizing that the anti-inflammatory effect of GLP-1 (9-36) in astrocytes is independent of GLP-1 receptor signaling and is instead mediated by IGF-1 receptor. Immunoprecipitation experiments showed that GLP-1 (9-36) directly interacts with IGF-1 receptor in astrocytes. Western blot data indicated that GLP-1 (9-36) activates IGF-1 receptor and downstream PI3K-AKT pathway in astrocytes upon OGD/R injury, which was abrogated by preincubation with IGF-1 receptor autophosphorylation inhibitor picropodophyllin. Thus, our findings suggest that GLP-1 (9-36) improved stroke outcome by reducing inflammation in astrocytes via interaction with IGF-1 receptor.
Collapse
Affiliation(s)
- Jing Huang
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, China; Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yunhan Liu
- Department of Neurology Impatient, Second Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Liusiyuan Cheng
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, China
| | - Jihong Li
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, China
| | - Tangrui Zhang
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, China
| | - Gang Zhao
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Huinan Zhang
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, China.
| |
Collapse
|
25
|
Warenits AM, Hatami J, Müllebner A, Ettl F, Teubenbacher U, Magnet IAM, Bauder B, Janata A, Miller I, Moldzio R, Kramer AM, Sterz F, Holzer M, Högler S, Weihs W, Duvigneau JC. Motor Cortex and Hippocampus Display Decreased Heme Oxygenase Activity 2 Weeks After Ventricular Fibrillation Cardiac Arrest in Rats. Front Med (Lausanne) 2020; 7:513. [PMID: 33015090 PMCID: PMC7511667 DOI: 10.3389/fmed.2020.00513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/24/2020] [Indexed: 01/07/2023] Open
Abstract
Heme oxygenase (HO) and biliverdin reductase (BVR) activities are important for neuronal function and redox homeostasis. Resuscitation from cardiac arrest (CA) frequently results in neuronal injury and delayed neurodegeneration that typically affect vulnerable brain regions, primarily hippocampus (Hc) and motor cortex (mC), but occasionally also striatum and cerebellum. We questioned whether these delayed effects are associated with changes of the HO/BVR system. We therefore analyzed the activities of HO and BVR in the brain regions Hc, mC, striatum and cerebellum of rats subjected to ventricular fibrillation CA (6 min or 8 min) after 2 weeks following resuscitation, or sham operation. From all investigated regions, only Hc and mC showed significantly decreased HO activities, while BVR activity was not affected. In order to find an explanation for the changed HO activity, we analyzed protein abundance and mRNA expression levels of HO-1, the inducible, and HO-2, the constitutively expressed isoform, in the affected regions. In both regions we found a tendency for a decreased immunoreactivity of HO-2 using immunoblots and immunohistochemistry. Additionally, we investigated the histological appearance and the expression of markers indicative for activation of microglia [tumor necrosis factor receptor type I (TNFR1) mRNA and immunoreactivity for ionized calcium-binding adapter molecule 1 (Iba1])], and activation of astrocytes [immunoreactivity for glial fibrillary acidic protein (GFAP)] in Hc and mC. Morphological changes were detected only in Hc displaying loss of neurons in the cornu ammonis 1 (CA1) region, which was most pronounced in the 8 min CA group. In this region also markers indicating inflammation and activation of pro-death pathways (expression of HO-1 and TNFR1 mRNA, as well as Iba1 and GFAP immunoreactivity) were upregulated. Since HO products are relevant for maintaining neuronal function, our data suggest that neurodegenerative processes following CA may be associated with a decreased capacity to convert heme into HO products in particularly vulnerable brain regions.
Collapse
Affiliation(s)
| | - Jasmin Hatami
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Andrea Müllebner
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria.,Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Florian Ettl
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Ursula Teubenbacher
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | | | - Barbara Bauder
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - Andreas Janata
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Ingrid Miller
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Rudolf Moldzio
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | | | - Fritz Sterz
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Michael Holzer
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Sandra Högler
- Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Weihs
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
26
|
Modulation of oxidative stress, inflammatory and apoptotic response by curcumin against cerebral ischemia reperfusion injury in a mouse model. INTERDISCIPLINARY NEUROSURGERY 2020. [DOI: 10.1016/j.inat.2020.100741] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
27
|
Gao XZ, Zhang ZX, Han GL. MiR-29a-3p Enhances the Viability of Rat Neuronal Cells that Injured by Oxygen-Glucose Deprivation/Reoxygenation Treatment Through Targeting TNFRSF1A and Regulating NF-κB Signaling Pathway. J Stroke Cerebrovasc Dis 2020; 29:105210. [PMID: 33066952 DOI: 10.1016/j.jstrokecerebrovasdis.2020.105210] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/21/2020] [Accepted: 07/25/2020] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE We attempt to investigate the role of TNFRSF1A and its underlying mechanism in oxygen-glucose deprivation/reoxygenation (OGD/R)-induced injury in rat pheochromocytoma PC12 cells. METHODS Public datasets GSE61616 and GSE106680 were downloaded from GEO database. PC12 cells were used to construct OGD/R models. QRT-PCR and western blot were implemented to test the relative mRNA and protein levels, respectively. The miRNA online prediction website TargetScan was used to predict TNFRSF1A upstream regulated miRNAs, which were then confirmed by luciferase reporter assay. The changes in cell viability and apoptosis were evaluated using cell counting kit 8 (CCK-8), lactose dehydrogenase (LDH), and flow cytometry assays. RESULTS Bioinformatics analysis demonstrated that the expression of TNFRSF1A was upregulated in CI/RI and middle cerebral artery occlusion models compared with control, respectively. And a significant upregulation was also observed in OGD/R-damaged PC12 cells. Depletion of TNFRSF1A can notably enhance the cells proliferation after OGD/R treatment, while enlargement of TNFRSF1A presented the opposite outcomes. Moreover, miR-29a-3p was shown to be the upstream regulatory miRNA of TNFRSF1A. The levels of TNFRSF1A were inversely mediated by miR-29a-3p. Overexpression of miR-29a-3p can raise the cell viability, decrease the LDH activity, and reduce the apoptotic ratio in OGD/R-treated cells. Besides, TNFRSF1A can attenuate the protective effect of miR-29a-3p on OGD/R-treated cells. Furthermore, miR-29a-3p mimic inhibited, while overexpression of TNFRSF1A promoted the activation of NF-κB signaling pathway, and TNFRSF1A can attenuate the suppressive effect of miR-29a-3p on the NF-κB pathway. CONCLUSION Our research illustrated that the potential regulatory role of miR-29a-3p/TNFRSF1A axis in neurons cells suffered from OGD/R, and their effects on NF-κB signaling pathway, providing a possible bio-target for protecting cells from OGD/R damage .
Collapse
Affiliation(s)
- Xiao-Zeng Gao
- Department of Anesthesiology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Zhao-Xia Zhang
- Department of Geriatrics, Shanxian centrol Hospital, Heze, Shandong 274300, P.R. China
| | - Guang-Liang Han
- Department of Neurosurgery, Shengli Oilfield Central Hospital of Binzhou Medical College, Dongying, Shandong 257034, P.R. China.
| |
Collapse
|
28
|
Lai Z, Gu L, Yu L, Chen H, Yu Z, Zhang C, Xu X, Zhang M, Zhang M, Ma M, Zhao Z, Zhang J. Delta opioid peptide [d-Ala2, d-Leu5] enkephalin confers neuroprotection by activating delta opioid receptor-AMPK-autophagy axis against global ischemia. Cell Biosci 2020; 10:79. [PMID: 32549974 PMCID: PMC7294676 DOI: 10.1186/s13578-020-00441-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 06/05/2020] [Indexed: 01/09/2023] Open
Abstract
Background Ischemic stroke poses a severe risk to human health worldwide, and currently, clinical therapies for the disease are limited. Delta opioid receptor (DOR)-mediated neuroprotective effects against ischemia have attracted increasing attention in recent years. Our previous studies revealed that DOR activation by [d-Ala2, d-Leu5] enkephalin (DADLE), a selective DOR agonist, can promote hippocampal neuronal survival on day 3 after ischemia. However, the specific molecular and cellular mechanisms underlying the DOR-induced improvements in ischemic neuronal survival remain unclear. Results We first detected the cytoprotective effects of DADLE in an oxygen–glucose deprivation/reperfusion (OGD/R) model and observed increased viability of OGD/R SH-SY5Y neuronal cells. We also evaluated changes in the DOR level following ischemia/reperfusion (I/R) injury and DADLE treatment and found that DADLE increased DOR levels after ischemia in vivo and vitro. The effects of DOR activation on postischemic autophagy were then investigated, and the results of the animal experiment showed that DOR activation by DADLE enhanced autophagy after ischemia, as indicated by elevated LC3 II/I levels and reduced P62 levels. Furthermore, the DOR-mediated protective effects on ischemic CA1 neurons were abolished by the autophagy inhibitor 3-methyladenine (3-MA). Moreover, the results of the cell experiments revealed that DOR activation not only augmented autophagy after OGD/R injury but also alleviated autophagic flux dysfunction. The molecular pathway underlying DOR-mediated autophagy under ischemic conditions was subsequently studied, and the in vivo and vitro data showed that DOR activation elevated autophagy postischemia by triggering the AMPK/mTOR/ULK1 signaling pathway, while the addition of the AMPK inhibitor compound C eliminated the protective effects of DOR against I/R injury. Conclusion DADLE-evoked DOR activation enhanced neuronal autophagy through activating the AMPK/mTOR/ULK1 signaling pathway to improve neuronal survival and exert neuroprotective effects against ischemia.
Collapse
Affiliation(s)
- Zelin Lai
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, 200062 China
| | - Lingling Gu
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, 200062 China
| | - Lu Yu
- Comprehensive Department of Traditional Chinese Medicine, Putuo Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
| | - Huifen Chen
- Department of Clinical Laboratory, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 201204 China
| | - Zhenhua Yu
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, 200062 China
| | - Cheng Zhang
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, 200062 China
| | - Xiaoqing Xu
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, 200062 China
| | - Mutian Zhang
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, 200062 China
| | - Min Zhang
- Department of Clinical Laboratory, Shanghai Public Health Clinical Center, Affiliated to Fudan University, Shanghai, 201508 China
| | - Mingliang Ma
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, 200062 China.,Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062 China
| | - Zheng Zhao
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, 200062 China
| | - Jun Zhang
- Department of Clinical Laboratory, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 201204 China.,Department of Clinical Laboratory, Shanghai Public Health Clinical Center, Affiliated to Fudan University, Shanghai, 201508 China
| |
Collapse
|
29
|
Cho SB, Eum WS, Shin MJ, Kwon HJ, Park JH, Choi YJ, Park J, Han KH, Kang JH, Kim DS, Cho SW, Kim DW, Choi SY. Transduced Tat-aldose Reductase Protects Hippocampal Neuronal Cells against Oxidative Stress-induced Damage. Exp Neurobiol 2019; 28:612-627. [PMID: 31698553 PMCID: PMC6844837 DOI: 10.5607/en.2019.28.5.612] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 08/06/2019] [Accepted: 09/02/2019] [Indexed: 12/15/2022] Open
Abstract
Aldose reductase (AR) protein, a member of the NADPH-dependent aldo-keto reductase family, reduces a wide range of aldehydes and enhances cell survival by inhibition of oxidative stress. Oxidative stress is known as one of the major pathological factor in ischemia. Since the precise function of AR protein in ischemic injury is fully unclear, we examined the function of AR protein in hippocampal neuronal (HT-22) cells and in an animal model of ischemia in this study. Cell permeable Tat-AR protein was produced by fusion of protein transduction domain in Tat for delivery into the cells. Tat-AR protein transduced into HT-22 cells and significantly inhibited cell death and regulated the mitogen-activate protein kinases (MAPKs), Bcl-2, Bax, and Caspase-3 under oxidative stress condition. In an ischemic animal model, Tat-AR protein transduced into the brain tissues through the blood-brain barrier (BBB) and drastically decreased neuronal cell death in hippocampal CA1 region. These results indicate that transduced Tat-AR protein has protective effects against oxidative stress-induced neuronal cell death in vitro and in vivo, suggesting that Tat-AR protein could be used as potential therapeutic agent in ischemic injury.
Collapse
Affiliation(s)
- Su Bin Cho
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Won Sik Eum
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Min Jea Shin
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Korea
| | - Jung Hwan Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Yeon Joo Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Jinseu Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Kyu Hyung Han
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Ju Hyeon Kang
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31538, Korea
| | - Duk-Soo Kim
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31538, Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
30
|
Zhang M, Ma Y, Chai L, Mao H, Zhang J, Fan X. Storax Protected Oxygen-Glucose Deprivation/Reoxygenation Induced Primary Astrocyte Injury by Inhibiting NF-κB Activation in vitro. Front Pharmacol 2019; 9:1527. [PMID: 30687092 PMCID: PMC6337066 DOI: 10.3389/fphar.2018.01527] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 12/12/2018] [Indexed: 01/22/2023] Open
Abstract
Stroke is the second leading cause of death and the leading cause of long-term disability in the world. There is an urgent unmet need to develop a range of neuroprotective strategies to restrain the damage that occurs in the hours and days following a stroke. Storax, a natural resin extracted from injuring Liquidambar orientalis Mill, has been used to treat acute stroke in traditional Chinese medicine for many centuries. Storax has demonstrated the neuroprotective effects in cerebrovascular diseases. However, the neuroprotective mechanisms activated by storax in ischemia/reperfusion-injured astrocytes have not been elucidated. In this study, we established an oxygen-glucose deprivation/reoxygenation (OGD/R)-induced astrocytes injury model to investigate the effects of storax on OGD/R-induced astrocytes injury and potential mechanisms. Experimental results showed that storax alleviated expression of inflammatory cytokines and protected primary cortical astrocytes injured by OGD/R. Furthermore, storax could inhibit NF-κB activation in injured astrocytes by OGD/R and inhibition of NF-κB with Bay-11-7082 obscured the neuroprotective effects of storax. In conclusion, storax alleviated expression of inflammatory cytokines and protected primary cortical astrocytes injured by OGD/R, which was partially mediated by NF-κB signaling pathway activation.
Collapse
Affiliation(s)
- Meng Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yan Ma
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lijuan Chai
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Haoping Mao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junhua Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiang Fan
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
31
|
Memory deficits and hippocampal inflammation in cerebral hypoperfusion and reperfusion in male rats: Neuroprotective role of vanillic acid. Life Sci 2018; 211:126-132. [PMID: 30195619 DOI: 10.1016/j.lfs.2018.08.065] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 08/20/2018] [Accepted: 08/27/2018] [Indexed: 11/20/2022]
Abstract
Ischemic stroke is one of the leading causes of neurological deterioration and mortality worldwide. Neuroprotective strategies are being investigated to minimize cognitive deficits after ischemic events. Here we investigated the neuroprotective potential of vanillic acid (VA) in an animal model of transient bilateral common carotid artery occlusion and reperfusion (BCCAO/R). Adult male Wistar rats (250-300 g) were randomly divided in 4 groups and submitted to either cerebral hypoperfusion-reperfusion or a sham surgery after two-weeks of pretreatment with VA and/or normal saline. To induce the animal model of hypoperfusion, bilateral common carotid arteries were occluded (2VO model) for 30 min, followed by 72 h of reperfusion. Subsequently, their cognitive performance was evaluated in a Morris water maze (MWM) test, and also hippocampi were removed for ELISA assays and TUNEL staining test. The results showed that 2VO significantly reduced the spatial memory performance in MWM. As well as, BCCAO/R increased the level of IL-6, TNF-α and TUNEL positive cells, and also decreased the contents of IL-10 in the hippocampus of vehicle- pretreated groups as compared to the sham-operated groups. Furthermore, 14 consecutive days pretreatment with VA significantly restored the spatial memory, decreased the levels of IL-6, TNF-α and TUNEL positive cells and also increased the IL-10 levels in the hippocampi of the BCCAO/R rats. VA alone did not show any change neither in the status of various cytokines nor behavioral and TUNEL staining tests over sham values. Our data confirm that VA could potentially serve as a novel, promising, and accessible neuroprotective agent against cerebrovascular insufficiency states and vascular dementia.
Collapse
|
32
|
Godinho J, de Sa-Nakanishi AB, Moreira LS, de Oliveira RMW, Huzita CH, Mello JCP, da Silva AOF, Nakamura CV, Previdelli IS, Ribeiro MHDM, Milani H. Ethyl-acetate fraction of Trichilia catigua protects against oxidative stress and neuroinflammation after cerebral ischemia/reperfusion. JOURNAL OF ETHNOPHARMACOLOGY 2018; 221:109-118. [PMID: 29660468 DOI: 10.1016/j.jep.2018.04.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/19/2018] [Accepted: 04/12/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Trichilia catigua A. Juss (Meliaceae) preparations have been used in folk medicine to alleviate fatigue, stress, and improve memory. Antinociceptive, antiinflammatory, and in vitro neuroprotective effects have been observed in animals. Cerebral ischemia/reperfusion (I/R) leads to severe neuropsychological deficits that are largely associated with oxidative stress, inflammation and neurodegeneration. We reported previously that an ethyl-acetate fraction (EAF) of T. catigua reduced brain ischemia-induced learning and memory impairments in the absence of histological protection. AIM OF THE STUDY Continuing those studies, here we aimed to investigate the antioxidant and antiinflammatory properties of T. catigua in an in vivo model of I/R. MATERIAL AND METHODS Rats were subjected to 15 min of brain ischemia (4-VO model) followed by up to 15 days of reperfusion. Vehicle was given by gavage 30 min before ischemia and at 1 h of reperfusion. In a first experiment, brain ischemia-induced changes in oxidative stress markers, i.e., reduced glutathione (GSH), oxidized glutathione (GSSG), superoxide dismutase (SOD), catalase (CAT), malondialdehyde (MDA), and protein carbonyl groups (PCGs) were measured on days 1, 3, and 5 post-ischemia. Similar time course analysis was done for neuroinflammation markers, i.e., microglia (OX42 immunorreactivity) and astrocytes (GFAP immunorreactivity), in the hippocampus. In a second experiment, the time points at which these markers of oxidative stress and neuroinflammation peaked were used to test the effects of T. catigua (400 mg/kg, p.o.). RESULTS Oxidative stress markers peaked on day 1 post-ischemia. GSH decreased (-23.2%) while GSSG increased (+ 71.1%), which yielded a significant reduction in the GSH/GSSG ratio (-39.1%). The activity of CAT was largely reduced by ischemia (-54.6% to -65.1%), while the concentration of PCG almost doubled in the brain of ischemic rats (+99.10%) in comparison to sham. Treatment with the EAF of T. catigua normalized these changes in oxidative markers to the control levels (GSH: +27.5%; GSSG: -23.8%; GSH/GSSG: +44.6%; PCG: -80.3%). In the hippocampus, neuroinflammation markers peaked on day 5 post-ischemia, with microglial and astrocytic responses increasing to 54.8% and 37.1%, respectively. The elevation in glial cells response was completely prevented by EAF. CONCLUSION These results demonstrate that T. catigua has both antioxidant and antiinflammatory activities after transient global cerebral ischemia in rats, which may contribute to the previously reported memory protective effect of T. catigua.
Collapse
Affiliation(s)
- Jacqueline Godinho
- Department of Pharmacology and Therapeutics, State University of Maringá, Maringá, Paraná, Brazil
| | | | | | | | - Claudia Hitomi Huzita
- Department of Pharmacology and Therapeutics, State University of Maringá, Maringá, Paraná, Brazil
| | - João Carlos P Mello
- Department of Pharmacy, State University of Maringá, Maringá, Paraná, Brazil
| | | | - Celso Vataru Nakamura
- Department of Basic Health Sciences, State University of Maringá, Maringá, Paraná, Brazil
| | | | | | - Humberto Milani
- Department of Pharmacology and Therapeutics, State University of Maringá, Maringá, Paraná, Brazil.
| |
Collapse
|
33
|
Dheer A, Jain V, Kushwah N, Kumar R, Prasad D, Singh S. Temporal and Spatial Changes in Glial Cells During Chronic Hypobaric Hypoxia: Role in Neurodegeneration. Neuroscience 2018; 383:235-246. [DOI: 10.1016/j.neuroscience.2018.04.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/16/2018] [Accepted: 04/18/2018] [Indexed: 01/05/2023]
|
34
|
Espinosa-Garcia C, Sayeed I, Yousuf S, Atif F, Sergeeva EG, Neigh GN, Stein DG. Stress primes microglial polarization after global ischemia: Therapeutic potential of progesterone. Brain Behav Immun 2017. [PMID: 28648389 DOI: 10.1016/j.bbi.2017.06.012] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Despite the fact that stress is associated with increased risk of stroke and worsened outcome, most preclinical studies have ignored this comorbid factor, especially in the context of testing neuroprotective treatments. Preclinical research suggests that stress primes microglia, resulting in an enhanced reactivity to a subsequent insult and potentially increasing vulnerability to stroke. Ischemia-induced activated microglia can be polarized into a harmful phenotype, M1, which produces pro-inflammatory cytokines, or a protective phenotype, M2, which releases anti-inflammatory cytokines and neurotrophic factors. Selective modulation of microglial polarization by inhibiting M1 or stimulating M2 may be a potential therapeutic strategy for treating cerebral ischemia. Our laboratory and others have shown progesterone to be neuroprotective against ischemic stroke in rodents, but it is not known whether it will be as effective under a comorbid condition of chronic stress. Here we evaluated the neuroprotective effect of progesterone on the inflammatory response in the hippocampus after exposure to stress followed by global ischemia. We focused on the effects of microglial M1/M2 polarization and pro- and anti-inflammatory mediators in stressed ischemic animals. Male Sprague-Dawley rats were exposed to 8 consecutive days of social defeat stress and then subjected to global ischemia or sham surgery. The rats received intraperitoneal injections of progesterone (8mg/kg) or vehicle at 2h post-ischemia followed by subcutaneous injections at 6h and once every 24h post-injury for 7days. The animals were killed at 7 and 14days post-ischemia, and brains were removed and processed to assess outcome measures using histological, immunohistochemical and molecular biology techniques. Pre-ischemic stress (1) exacerbated neuronal loss and neurodegeneration as well as microglial activation in the selectively vulnerable CA1 hippocampal region, (2) dysregulated microglial polarization, leading to upregulation of both M1 and M2 phenotype markers, (3) increased pro-inflammatory cytokine expression, and (4) reduced anti-inflammatory cytokine and neurotrophic factor expression in the ischemic hippocampus. Treatment with progesterone significantly attenuated stress-induced microglia priming by modulating polarized microglia and the inflammatory environment in the hippocampus, the area most vulnerable to ischemic injury. Our findings can be taken to suggest that progesterone holds potential as a candidate for clinical testing in ischemic stroke where high stress may be a contributing factor.
Collapse
Affiliation(s)
| | - Iqbal Sayeed
- Department of Emergency Medicine, Emory University, Atlanta, GA 30322, USA.
| | - Seema Yousuf
- Department of Emergency Medicine, Emory University, Atlanta, GA 30322, USA.
| | - Fahim Atif
- Department of Emergency Medicine, Emory University, Atlanta, GA 30322, USA.
| | - Elena G Sergeeva
- Department of Emergency Medicine, Emory University, Atlanta, GA 30322, USA.
| | - Gretchen N Neigh
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA 30322, USA.
| | - Donald G Stein
- Department of Emergency Medicine, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
35
|
Huang L, Ma Q, Li Y, Li B, Zhang L. Inhibition of microRNA-210 suppresses pro-inflammatory response and reduces acute brain injury of ischemic stroke in mice. Exp Neurol 2017; 300:41-50. [PMID: 29111308 DOI: 10.1016/j.expneurol.2017.10.024] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 10/20/2017] [Accepted: 10/25/2017] [Indexed: 12/11/2022]
Abstract
Stroke is a leading cause of mortality and chronic neurologic disability. Yet, the successful treatment remains limited. In this study, we investigated the efficacy and the mechanism of a novel treatment, microRNA-210 (miR-210) inhibition, in protecting acute ischemic brain injury in adult mice. Focal cerebral ischemia was induced by middle cerebral artery occlusion (MCAO) in adult male C57BL/6 mice. MiR-210-LNA (miR-210 inhibitor) or the negative control was administered via intracerebroventricular injection 24h prior or 4h after MCAO. Cerebral infarction volume and behavioral deficits were determined 48h after MCAO. The expression of inflammation-related genes and infiltration/activation of various immune cells in the brain were assessed by RT-qPCR, flow cytometry, and immunohistochemistry. Acute ischemic stroke significantly increased miR-210 levels in the brain, which was abolished by miR-210-LNA administered prior to MCAO. Pre- and post-MCAO treatments with miR-210-LNA significantly decreased cerebral infarction and ameliorated behavioral deficits induced by MCAO. Long-term behavioral recovery was also improved by miR-210-LNA post-treatment. At the same time, inhibition of miR-210 significantly reduced the expression of pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6) and chemokines (CCL2 and CCL3), but had no significant effect on anti-inflammatory factors (TGF-β and IL-10). In addition, MCAO-induced macrophage infiltration and microglial activation in the brain were inhibited by the miR-210-LNA treatment. In summary, inhibition of miR-210 suppresses pro-inflammatory response and reduces brain damage in the acute phase of ischemic stroke, providing new insight in molecular basis of a novel therapeutic strategy of miR-210 inhibition in the treatment of acute ischemic stroke.
Collapse
Affiliation(s)
- Lei Huang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA 92350, United States
| | - Qingyi Ma
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA 92350, United States
| | - Yong Li
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA 92350, United States
| | - Bo Li
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA 92350, United States
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA 92350, United States.
| |
Collapse
|
36
|
Intranasal post-cardiac arrest treatment with orexin-A facilitates arousal from coma and ameliorates neuroinflammation. PLoS One 2017; 12:e0182707. [PMID: 28957432 PMCID: PMC5619710 DOI: 10.1371/journal.pone.0182707] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 07/24/2017] [Indexed: 12/31/2022] Open
Abstract
Cardiac arrest (CA) entails significant risks of coma resulting in poor neurological and behavioral outcomes after resuscitation. Significant subsequent morbidity and mortality in post-CA patients are largely due to the cerebral and cardiac dysfunction that accompanies prolonged whole-body ischemia post-CA syndrome (PCAS). PCAS results in strong inflammatory responses including neuroinflammation response leading to poor outcome. Currently, there are no proven neuroprotective therapies to improve post-CA outcomes apart from therapeutic hypothermia. Furthermore, there are no acceptable approaches to promote cortical or cognitive arousal following successful return of spontaneous circulation (ROSC). Hypothalamic orexinergic pathway is responsible for arousal and it is negatively affected by neuroinflammation. However, whether activation of the orexinergic pathway can curtail neuroinflammation is unknown. We hypothesize that targeting the orexinergic pathway via intranasal orexin-A (ORXA) treatment will enhance arousal from coma and decrease the production of proinflammatory cytokines resulting in improved functional outcome after resuscitation. We used a highly validated CA rat model to determine the effects of intranasal ORXA treatment 30-minute post resuscitation. At 4hrs post-CA, the mRNA levels of proinflammatory markers (IL1β, iNOS, TNF-α, GFAP, CD11b) and orexin receptors (ORX1R and ORX2R) were examined in different brain regions. CA dramatically increased proinflammatory markers in all brain regions particularly in the prefrontal cortex, hippocampus and hypothalamus. Post-CA intranasal ORXA treatment significantly ameliorated the CA-induced neuroinflammatory markers in the hypothalamus. ORXA administration increased production of orexin receptors (ORX1R and ORX2R) particularly in hypothalamus. In addition, ORXA also resulted in early arousal as measured by quantitative electroencephalogram (EEG) markers, and recovery of the associated behavioral neurologic deficit scale score (NDS). Our results indicate that intranasal delivery of ORXA post-CA has an anti-inflammatory effect and accelerates cortical EEG and behavioral recovery. Beneficial outcomes from intranasal ORXA treatment lay the groundwork for therapeutic clinical approach to treating post-CA coma.
Collapse
|
37
|
Obadia N, Lessa MA, Daliry A, Silvares RR, Gomes F, Tibiriçá E, Estato V. Cerebral microvascular dysfunction in metabolic syndrome is exacerbated by ischemia-reperfusion injury. BMC Neurosci 2017; 18:67. [PMID: 28886695 PMCID: PMC5591496 DOI: 10.1186/s12868-017-0384-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 08/31/2017] [Indexed: 12/11/2022] Open
Abstract
Background Metabolic syndrome (MetS) is associated with an increased risk of cerebrovascular diseases, including cerebral ischemia. Microvascular dysfunction is an important feature underlying the pathophysiology of cerebrovascular diseases. In this study, we aimed to investigate the impacts of ischemia and reperfusion (IR) injury on the cerebral microvascular function of rats with high-fat diet-induced MetS. Results We examined Wistar rats fed a high-fat diet (HFD) or normal diet (CTL) for 20 weeks underwent 30 min of bilateral carotid artery occlusion followed by 1 h of reperfusion (IR) or sham surgery. Microvascular blood flow was evaluated on the parietal cortex surface through a cranial window by laser speckle contrast imaging, functional capillary density, endothelial function and endothelial–leukocyte interactions by intravital videomicroscopy. Lipid peroxidation was assessed by TBARs analysis, the expression of oxidative enzymes and inflammatory markers in the brain tissue was analyzed by real-time PCR. The cerebral IR in MetS animals induced a functional capillary rarefaction (HFD IR 117 ± 17 vs. CTL IR 224 ± 35 capillary/mm2; p < 0.05), blunted the endothelial response to acetylcholine (HFD IR −16.93% vs. CTL IR 16.19% from baseline inner diameter p < 0.05) and increased the endothelial–leukocyte interactions in the venules in the brain. The impact of ischemia on the cerebral microvascular blood flow was worsened in MetS animals, with a marked reduction of cerebral blood flow, exposing brain tissue to a higher state of hypoxia. Conclusions Our results demonstrate that during ischemia and reperfusion, animals with MetS are more susceptible to alterations in the cerebral microcirculation involving endothelial dysfunction and oxidative stress events.
Collapse
Affiliation(s)
- Nathalie Obadia
- Laboratory of Cardiovascular Investigation, Oswaldo Cruz Foundation, Avenida Brasil, 4365, Rio de Janeiro, RJ, 21045-900, Brazil
| | - Marcos Adriano Lessa
- Laboratory of Cardiovascular Investigation, Oswaldo Cruz Foundation, Avenida Brasil, 4365, Rio de Janeiro, RJ, 21045-900, Brazil
| | - Anissa Daliry
- Laboratory of Cardiovascular Investigation, Oswaldo Cruz Foundation, Avenida Brasil, 4365, Rio de Janeiro, RJ, 21045-900, Brazil
| | - Raquel Rangel Silvares
- Laboratory of Cardiovascular Investigation, Oswaldo Cruz Foundation, Avenida Brasil, 4365, Rio de Janeiro, RJ, 21045-900, Brazil
| | - Fabiana Gomes
- Laboratory of Cardiovascular Investigation, Oswaldo Cruz Foundation, Avenida Brasil, 4365, Rio de Janeiro, RJ, 21045-900, Brazil
| | - Eduardo Tibiriçá
- Laboratory of Cardiovascular Investigation, Oswaldo Cruz Foundation, Avenida Brasil, 4365, Rio de Janeiro, RJ, 21045-900, Brazil.,National Institute of Cardiology, Rio de Janeiro, Brazil
| | - Vanessa Estato
- Laboratory of Cardiovascular Investigation, Oswaldo Cruz Foundation, Avenida Brasil, 4365, Rio de Janeiro, RJ, 21045-900, Brazil. .,Institute of Drug Technology, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.
| |
Collapse
|
38
|
Global gene expression profile of cerebral ischemia-reperfusion injury in rat MCAO model. Oncotarget 2017; 8:74607-74622. [PMID: 29088811 PMCID: PMC5650366 DOI: 10.18632/oncotarget.20253] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 06/30/2017] [Indexed: 12/19/2022] Open
Abstract
It is well-established that reperfusion following cerebral ischemic injury gives rise to secondary injury accompanied by structural and functional damage. However, it remains unclear how global genes changes in cerebral ischemia-reperfusion injury (IRI). This study investigated global gene expression in the hippocampi of Wistar rats following transient cerebral IRI using an RNA-sequencing strategy. The results revealed ≥2-fold up-regulation of 156 genes and ≥2-fold down-regulation of 26 genes at 24 h post-reperfusion. Fifteen differentially expressed genes were selected to confirm the RNA-sequencing results. Gene expression levels were dynamic, with the peak expression level of each gene occurring at different time points post-reperfusion. Gene Ontology (GO) analysis classified the differentially expressed genes as mainly involved in inflammation, stress and immune response, glucose metabolism, proapoptosis, antiapoptosis, and biological processes. KEGG pathway analysis suggested that IRI activated different signaling pathways, including focal adhesion, regulation of actin cytoskeleton, cytokine-cytokine receptor interaction, MAPK signaling, and Jak-STAT signaling. This study describes global gene expression profiles in the hippocampi of Wistar rats using the middle cerebral artery occlusion (MCAO) model. These findings provide new insights into the molecular pathogenesis of IRI and potential drug targets for the prevention and treatment of IRI in the future.
Collapse
|
39
|
Wang L, Liu H, Zhang L, Wang G, Zhang M, Yu Y. Neuroprotection of Dexmedetomidine against Cerebral Ischemia-Reperfusion Injury in Rats: Involved in Inhibition of NF-κB and Inflammation Response. Biomol Ther (Seoul) 2017; 25:383-389. [PMID: 27871154 PMCID: PMC5499616 DOI: 10.4062/biomolther.2015.180] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 05/09/2016] [Accepted: 09/21/2016] [Indexed: 11/05/2022] Open
Abstract
Dexmedetomidine is an α2-adrenergic receptor agonist that exhibits a protective effect on ischemia-reperfusion injury of the heart, kidney, and other organs. In the present study, we examined the neuroprotective action and potential mechanisms of dexmedetomidine against ischemia-reperfusion induced cerebral injury. Transient focal cerebral ischemia-reperfusion injury was induced in Sprague-Dawley rats by middle cerebral artery occlusion. After the ischemic insult, animals then received intravenous dexmedetomidine of 1 µg/kg load dose, followed by 0.05 µg/kg/min infusion for 2 h. After 24 h of reperfusion, neurological function, brain edema, and the morphology of the hippocampal CA1 region were evaluated. The levels and mRNA expressions of interleukin-1β, interleukin-6 and tumor nevrosis factor-α as well as the protein expression of inducible nitric oxide synthase, cyclooxygenase-2, nuclear factor-κBp65, inhibitor of κBα and phosphorylated of κBα in hippocampus were assessed. We found that dexmedetomidine reduced focal cerebral ischemia-reperfusion injury in rats by inhibiting the expression and release of inflammatory cytokines and mediators. Inhibition of the nuclear factor-κB pathway may be a mechanism underlying the neuroprotective action of dexmedetomidine against focal cerebral I/R injury.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Haiyan Liu
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Ligong Zhang
- Department of Anesthesia, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Gongming Wang
- Department of Anesthesia, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Mengyuan Zhang
- Department of Anesthesia, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Yonghui Yu
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| |
Collapse
|
40
|
Wang X, An F, Wang S, An Z, Wang S. Orientin Attenuates Cerebral Ischemia/Reperfusion Injury in Rat Model through the AQP-4 and TLR4/NF-κB/TNF-α Signaling Pathway. J Stroke Cerebrovasc Dis 2017. [PMID: 28645524 DOI: 10.1016/j.jstrokecerebrovasdis.2017.05.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Orientin has been reported to have extensive pharmaceutical effects of antioxidant, anti-inflammatory, antithrombosis, antiapoptosis, and so on. In the present study, we tried to investigate the protective effects of orientin on cerebral ischemia-reperfusion (I/R) injury and explored the possible mechanisms. METHODS Middle cerebral artery occlusion rat model was established and then treated with low, middle, and high concentrations of orientin, respectively, with edaravone as a positive control. The treatment effect of orientin was evaluated by measuring the neurological deficit score, cerebral infarction, brain edema, oxidative stress, excitatory amino acids release, the expression levels of aquaporin-4 (AQP-4), and related inflammatory molecules using different methods including immunohistochemistry, enzyme-linked immunosorbent assay, real-time PCR, and western blot. Moreover, morphological and structural changes were also observed by hematoxylin-eosin staining and transmission electron microscope. RESULTS Orientin provided a significant reduction on neurological deficits, cerebral infarction, cerebral edema, oxidative damage, and neurotoxicity of excitatory amino acids compared to model group (P < .05) in a dose-dependent manner. In addition, orientin substantially downregulated AQP-4 and inflammatory factors expression (P < .05) and improved cell morphology and structure in rats following I/R injury. CONCLUSION Orientin was able to mediate noticeable protection against cerebral I/R injury through the attenuation of oxidative stress and neurotoxicity of amino acids and inhibiting the upregulation of AQP-4 and inflammatory cytokines.
Collapse
Affiliation(s)
- Xiaoru Wang
- Department of Pharmacy, Hebei North University, Zhangjiakou, Hebei Province, China
| | - Fang An
- Graduate Faculty, Hebei North University, Zhangjiakou, Hebei Province, China
| | - Shulin Wang
- Department of Pharmacy, Hebei North University, Zhangjiakou, Hebei Province, China
| | - Zexin An
- Department of Information, First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, China
| | - Shuhua Wang
- Department of Pharmacy, Hebei North University, Zhangjiakou, Hebei Province, China.
| |
Collapse
|
41
|
Mori MA, Meyer E, Soares LM, Milani H, Guimarães FS, de Oliveira RMW. Cannabidiol reduces neuroinflammation and promotes neuroplasticity and functional recovery after brain ischemia. Prog Neuropsychopharmacol Biol Psychiatry 2017; 75:94-105. [PMID: 27889412 DOI: 10.1016/j.pnpbp.2016.11.005] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/22/2016] [Indexed: 11/30/2022]
Abstract
This study investigated the effects of cannabidiol (CBD), a non-psychotomimetic phytochemical present in Cannabis sativa, on the cognitive and emotional impairments induced by bilateral common carotid artery occlusion (BCCAO) in mice. Using a multi-tiered behavioral testing battery during 21days, we found that BCCAO mice exhibited long-lasting functional deficits reflected by increase in anxiety-like behavior (day 9), memory impairments (days 12-18) and despair-like behavior (day 21). Short-term CBD 10mg/kg treatment prevented the cognitive and emotional impairments, attenuated hippocampal neurodegeneration and white matter (WM) injury, and reduced glial response that were induced by BCCAO. In addition, ischemic mice treated with CBD exhibited an increase in the hippocampal brain derived neurotrophic factor (BDNF) protein levels. CBD also stimulated neurogenesis and promoted dendritic restructuring in the hippocampus of BCCAO animals. Collectively, the present results demonstrate that short-term CBD treatment results in global functional recovery in ischemic mice and impacts multiple and distinct targets involved in the pathophysiology of brain ischemic injury.
Collapse
Affiliation(s)
- Marco Aurélio Mori
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, 5790, 87020-900 Maringá, Paraná, Brazil
| | - Erika Meyer
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, 5790, 87020-900 Maringá, Paraná, Brazil
| | - Ligia Mendes Soares
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, 5790, 87020-900 Maringá, Paraná, Brazil
| | - Humberto Milani
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, 5790, 87020-900 Maringá, Paraná, Brazil
| | - Francisco Silveira Guimarães
- Department of Pharmacology, School of Medicine, USP, Av. Bandeirantes, 14015-000 Ribeirão Preto, São Paulo, Brazil
| | - Rúbia Maria Weffort de Oliveira
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, 5790, 87020-900 Maringá, Paraná, Brazil.
| |
Collapse
|
42
|
Qu Y, Zhang HL, Zhang XP, Jiang HL. Arachidonic acid attenuates brain damage in a rat model of ischemia/reperfusion by inhibiting inflammatory response and oxidative stress. Hum Exp Toxicol 2017; 37:135-141. [PMID: 29233001 DOI: 10.1177/0960327117692134] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The aim of the present study was to study the effects of arachidonic acid (ARA) in a rat brain ischemia/reperfusion model induced by middle cerebral artery occlusion (MCAO). A total of 50 rats were randomly divided into five groups: control group, MCAO group, MCAO + ARA 0.3 g/kg group, MCAO + ARA 1 g/kg group, and MCAO + ARA 3 g/kg group. The MCAO + ARA groups received ARA by intraperitoneal injection daily for 14 consecutive days, while the rats in the control and MCAO groups were given equivalent volume of saline. We detected the Morris water maze test and pathological changes to investigate the ischemia/reperfusion injury. The protein levels of tumor necrosis factor-alpha and interleukin-6 in the hippocampus were detected by enzyme-linked immunosorbent assay kits. In addition, the activities of superoxide dismutase, glutathione peroxidase, and malondialdehyde were assayed in hippocampus homogenates to evaluate the oxidative stress after ischemia/reperfusion. The results indicated that ARA administration decreased biochemical parameters of inflammation and oxidative stress. Morris water maze test and histopathological examination further verified the protective effects of ARA on ischemia/reperfusion injury rats. These findings demonstrated that ARA could protect MCAO-induced brain injury rats by inhibition of inflammation and oxidative stress, suggesting that it may have potential as a therapy for cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Y Qu
- 1 Department of Emergency, Yuhuangding Hospital, Qingdao University, Yantai 264000, China
| | - H-L Zhang
- 2 Department of Neurology, Yuhuangding Hospital, Qingdao University, Yantai, China
| | - X-P Zhang
- 3 Department of Pharmacy, Yuhuangding Hospital, Qingdao University, Yantai, China
| | - H-L Jiang
- 4 Kidney Disease/Blood Purification, Jinan Central Hospital, Jinan, China
| |
Collapse
|
43
|
Daphnetin Protects against Cerebral Ischemia/Reperfusion Injury in Mice via Inhibition of TLR4/NF- κB Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2016; 2016:2816056. [PMID: 28119924 PMCID: PMC5227117 DOI: 10.1155/2016/2816056] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 11/13/2016] [Indexed: 01/09/2023]
Abstract
Growing evidences indicate that immune-mediated mechanisms contribute to the development of cerebral ischemia/reperfusion (I/R) injury. Daphnetin (DAP) is a coumarin derivative extracted from Daphne odora var., which displays anti-inflammatory properties. However, the effect of DAP on cerebral I/R injury is not yet clear. Recent studies have demonstrated that TLR4/NF-κB signaling pathway takes part in the damaging inflammatory process of cerebral I/R injury. The present study aimed to investigate the effect of DAP on cerebral I/R injury in vivo and its possible mechanisms. DAP was administered before middle cerebral artery occlusion and reperfusion in mice. The neurological scores, cerebral infarct sizes, the levels of inflammatory cytokines, apoptotic neural cells, and the levels of TLR4, NF-κB p65, and IκBα were estimated. The results showed that an obvious improvement of neurological scores and infarct sizes was observed in DAP-treated mice after MCAO/R. DAP treatment decreased the overexpression of TNF-α, IL-1β, and IL-6 and attenuated neural cells apoptosis. Moreover, DAP treatment decreased the TLR4 expression, IκB-α degradation, and nuclear translocation of NF-κB. Taken together, our results suggested that DAP exerted neuroprotective and anti-inflammatory effects on cerebral I/R injury. The potential mechanism was involved in the inhibition of TLR4/NF-κB mediated inflammatory signaling pathway.
Collapse
|
44
|
Kim MS, Bang JH, Lee J, Han JS, Baik TG, Jeon WK. Ginkgo biloba L. extract protects against chronic cerebral hypoperfusion by modulating neuroinflammation and the cholinergic system. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:1356-1364. [PMID: 27765355 DOI: 10.1016/j.phymed.2016.07.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 07/10/2016] [Accepted: 07/31/2016] [Indexed: 05/27/2023]
Abstract
BACKGROUND Ginkgo biloba extract (GBE)-a widely used nutraceutical-is reported to have diverse functions, including positive effects on memory and vasodilatory properties. Although numerous studies have assessed the neuroprotective properties of GBE in ischemia, only a few studies have investigated the neuro-pharmacological mechanisms of action of GBE in chronic cerebral hypoperfusion (CCH). PURPOSE In the present study, we sought to determine the effects of GBE on CCH-induced neuroinflammation and cholinergic dysfunction in a rat model of bilateral common carotid artery occlusion (BCCAo). METHODS Chronic BCCAo was induced in adult male Wistar rats to reflect the CCH conditions. On day 21 after BCCAo, the animals were treated orally with saline or GBE (5, 10, 20, and 40mg/kg) daily for 42 days. After the final treatment, brain tissues were isolated for the immunohistochemical analysis of glial markers and choline acetyltransferase (ChAT), as well as for the western blot analysis of proinflammatory cytokines, toll-like receptor (TLR)-related pathway, receptor for advanced glycation end products (RAGE), angiotensin-II (Ang-II), and phosphorylated mitogen-activated protein kinases (MAPKs). RESULTS BCCAo increased glial proliferation in the hippocampus and white matter, whereas proliferation was significantly attenuated by GBE treatment. GBE also attenuated the BCCAo-related increases in the hippocampal expression of proinflammatory cytokines (TNF-α, IL-1β, and IL-6), TLR4, myeloid differentiation primary response gene 88, RAGE, Ang-II, and phosphorylated MAPKs (ERK, p38, and JNK). Furthermore, GBE treatment restored the ChAT expression in the basal forebrain following BCCAo. CONCLUSIONS These findings suggest that GBE has specific neuroprotective effects that may be useful for the treatment of CCH. The pharmacological mechanism of GBE partly involves the modulation of inflammatory mediators and the cholinergic system.
Collapse
Affiliation(s)
- Min-Soo Kim
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, The Republic of Korea; Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul 02792, The Republic of Korea
| | - Ji Hye Bang
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, The Republic of Korea
| | - Jun Lee
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, The Republic of Korea; Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul 02792, The Republic of Korea
| | - Jung-Soo Han
- Department of Biological Science, Konkuk University, Seoul 05029, The Republic of Korea
| | - Tae Gon Baik
- Central Research Center, Yuyu Pharma. Inc., Seoul 04598, The Republic of Korea
| | - Won Kyung Jeon
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, The Republic of Korea; Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul 02792, The Republic of Korea.
| |
Collapse
|
45
|
Liu B, Zhang Y, Jiang Y, Li L, Li C, Li J. Electrical stimulation of cerebellar fastigial nucleus protects against cerebral ischemic injury by PPARγ upregulation. Neurol Res 2016; 39:23-29. [PMID: 27819182 DOI: 10.1080/01616412.2016.1251710] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Bin Liu
- Department of Neurology, Shandong Provincial Qianfoshan Hospital , Jinan, Shandong, China
| | - Yanhong Zhang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Ying Jiang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Longling Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Changqing Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Jinfang Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University , Chongqing, China
| |
Collapse
|
46
|
Ye X, Kong D, Wang J, Ishrat T, Shi H, Ding X, Cui G, Hua F. MyD88 contributes to neuroinflammatory responses induced by cerebral ischemia/reperfusion in mice. Biochem Biophys Res Commun 2016; 480:69-74. [PMID: 27717824 DOI: 10.1016/j.bbrc.2016.10.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 10/03/2016] [Indexed: 02/06/2023]
Abstract
Myeloid differentiation primary-response protein-88 (MyD88) is one of adaptor proteins mediating Toll-like receptors (TLRs) signaling. Activation of MyD88 results in the activation of nuclear factor kappa B (NFκB) and the increase of inflammatory responses. Evidences have demonstrated that TLRs signaling contributes to cerebral ischemia/reperfusion (I/R) injury. However, the role of MyD88 in this mechanism of action is disputed and needs to be clarified. In the present study, in a mouse model of cerebral I/R, we examined the activities of NFκB and interferon factor-3 (IRF3), and the inflammatory responses in ischemic brain tissue using ELISA, Western blots, and real-time PCR. Neurological function and cerebral infarct size were also evaluated 24 h after cerebral I/R. Our results showed that NFκB activity increased in ischemic brains, but IRF3 was not activated after cerebral I/R, in wild-type (WT) mice. MyD88 deficit inhibited the activation of NFκB, and the expression of interleukin-1β (IL-1β), IL-6, Beclin-1 (BECN1), pellino-1, and cyclooxygenase-2 (COX-2) increased by cerebral I/R compared with WT mice. Interestingly, the expression of interferon Beta 1 (INFB1) and vascular endothelial growth factor (VEGF) increased in MyD88 KO mice. Unexpectedly, although the neurological function improved in the MyD88 knockout (KO) mice, the deficit of MyD88 failed to reduce cerebral infarct size compared to WT mice. We concluded that MyD88-dependent signaling contributes to the inflammatory responses induced by cerebral I/R. MyD88 deficit may inhibit the increased inflammatory response and increase neuroprotective signaling.
Collapse
Affiliation(s)
- Xinchun Ye
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Delian Kong
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Jun Wang
- Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Tauheed Ishrat
- Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Hongjuan Shi
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Xiaohui Ding
- Department of Human Anatomy, Histology and Embryology, Shenyang Medical College, Shenyang, 110000, China
| | - Guiyun Cui
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Fang Hua
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, USA; Key Laboratory of Anesthesiology of Jiangsu Province, Xuzhou 221002, China.
| |
Collapse
|
47
|
Liu Y, Chen L, Shen Y, Tan T, Xie N, Luo M, Li Z, Xie X. Curcumin Ameliorates Ischemia-Induced Limb Injury Through Immunomodulation. Med Sci Monit 2016; 22:2035-42. [PMID: 27302110 PMCID: PMC4913813 DOI: 10.12659/msm.896217] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background The prevalence of peripheral arterial disease (PAD) is increasing worldwide. Currently, there is no effective treatment for PAD. Curcumin is an ingredient of turmeric that has antioxidant, anti-inflammation, and anticancer properties. In the present study we investigated the potential effect of curcumin in protecting against ischemic limb injury. Material/Methods We used an established hindlimb ischemia mouse model in our study. Curcumin was administrated through intraperitoneal (I.P.) injection. Immunohistochemical staining and ELISA assays were performed. Treadmill training was used to evaluate skeletal muscle functions of animals. Results Our experiments using in vivo treadmill training showed that curcumin treatment improved the running capacity of animals after ischemic injury. Histological analysis revealed that curcumin treatment significantly reduced the skeletal muscle damage and fibrosis associated with ischemic injury. In order to determine the cellular and molecular mechanisms underlying curcumin-mediated tissue protection, immunohistochemical staining and ELISA assays were performed. The results showed that curcumin treatment led to less macrophage infiltration and less local inflammatory responses as demonstrated by decreasing TNF-α, IL-1, and IL-6 levels. Further immunofluorescent staining of tissue slides indicated that curcumin treatment inhibited the NF-κB signaling pathway. Finally, curcumin can inhibit NF-κB activation induced by LPS in macrophages. Conclusions Our study results show that curcumin treatment can ameliorate hindlimb injury following ischemic surgery, which suggests that curcumin could be used for PAD treatment.
Collapse
Affiliation(s)
- Yang Liu
- Division of Geriatrics, Tongji Hospital, Tongji University, School of Medicine, Shanghai, China (mainland)
| | - Lianyu Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China (mainland)
| | - Yi Shen
- Division of Geriatrics, Tongji Hospital, Tongji University, School of Medicine, Shanghai, China (mainland)
| | - Tao Tan
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Nanzi Xie
- Division of Geriatrics, Tongji Hospital, Tongji University, School of Medicine, Shanghai, China (mainland)
| | - Ming Luo
- Division of Geriatrics, Tongji Hospital, Tongji University, School of Medicine, Shanghai, China (mainland)
| | - Zhihong Li
- Division of General Surgery, Pudong New Area District, Zhoupu Hospital, Shanghai, China (mainland)
| | - Xiaoyun Xie
- Division of Geriatrics, Tongji Hospital, Tongji University, School of Medicine, Shanghai, China (mainland)
| |
Collapse
|
48
|
Bai S, Sun Y, Wu L, Wu Z, Fang M. [Tripotolide ameliorates inflammation and apoptosis induced by focal cerebral ischemia/reperfusion in rats]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2016; 45:493-500. [PMID: 28087909 PMCID: PMC10396981 DOI: 10.3785/j.issn.1008-9292.2016.09.07] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/07/2016] [Indexed: 11/15/2022]
Abstract
Objective: To investigate the effects of triptolide on inflammation and apoptosis induced by focal cerebral ischemia/reperfusion in rats. Methods: The rat model of focal cerebral ischemia/reperfusion injury was established according to Longa's method. A total of 80 SD rats were randomly divided into 5 groups:normal control, sham group, DMSO group, middle cerebral artery occlusion (MCAO) group, and MCAO with tripolide treatment group. TTC staining was used to examine the site and volume of cerebral infarction, and Longa score was employed for neurological disorders measurement. Number of astrocytes was measured by fluorescence staining, and neuronal apoptosis was determined by TUNEL staining. The expressions of inducible nitric oxide synthase(iNOS), cyclooxygenase 2(COX-2) and NF-κB proteins were detected by immunohistochemistry, and the expression of iNOS, COX-2 mRNA was detected by real-time PCR. Results: Compared with DMSO group and MCAO group, brain edema was improved (80.03±0.46)% (P<0.05), infarct volume was reduced (8.3±1.4)% (P<0.01), Longa score was decreased (1.38±0.20, P<0.05) in triptolide treatment group. Meanwhile triptolide also dramatically reduced the number of GFAP-positive astrocytes (P<0.05), alleviated protein expression of COX-2 (91.67±1.31), iNOS (95.24±5.07) and NF-κB (75.03±2.06) triggered by MCAO (all P<0.05), and induced a down-regulation of cell apoptosis as showed by TUNEL assay (64.15±3.52, P<0.05). Conclusion: Triptolide can reduce the cerebral infarction volume, attenuate brain edema and ameliorate the neurological deficits induced by cerebral ischemia-reperfusion injury rats, indicating that it might be used as a potential anti-inflammatory agent.
Collapse
Affiliation(s)
- Shi Bai
- School of Medicine, Taizhou University, Taizhou 318000, China; Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yayi Sun
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Lijuan Wu
- School of Medicine, Taizhou University, Taizhou 318000, China
| | - Zhongmin Wu
- School of Medicine, Taizhou University, Taizhou 318000, China
| | - Marong Fang
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
49
|
Zhao B, Zou CJ, Zhou P. Delayed administration IL-1β neutralizing antibody improves cognitive function after transient global ischemia in rats. Behav Brain Res 2016; 303:53-60. [DOI: 10.1016/j.bbr.2016.01.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 01/06/2016] [Accepted: 01/10/2016] [Indexed: 12/23/2022]
|
50
|
Gao S, Mo J, Chen L, Wang Y, Mao X, Shi Y, Zhang X, Yu R, Zhou X. Astrocyte GGTI-mediated Rac1 prenylation upregulates NF-κB expression and promotes neuronal apoptosis following hypoxia/ischemia. Neuropharmacology 2016; 103:44-56. [DOI: 10.1016/j.neuropharm.2015.12.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 11/20/2015] [Accepted: 12/07/2015] [Indexed: 12/14/2022]
|