1
|
Wang Z, Li Y, Wang Z, Liao Y, Ye Q, Tang S, Wei T, Xiao P, Huang J, Lu W. Edaravone Maintains AQP4 Polarity Via OS/MMP9/β-DG Pathway in an Experimental Intracerebral Hemorrhage Mouse Model. Mol Neurobiol 2024; 61:7639-7658. [PMID: 38421470 DOI: 10.1007/s12035-024-04028-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/05/2023] [Accepted: 02/09/2024] [Indexed: 03/02/2024]
Abstract
Oxidative stress (OS) is the main cause of secondary damage following intracerebral hemorrhage (ICH). The polarity expression of aquaporin-4 (AQP4) has been shown to be important in maintaining the homeostasis of water transport and preventing post-injury brain edema in various neurological disorders. This study primarily aimed to investigate the effect of the oxygen free radical scavenger, edaravone, on AQP4 polarity expression in an ICH mouse model and determine whether it involves in AQP4 polarity expression via the OS/MMP9/β-dystroglycan (β-DG) pathway. The ICH mouse model was established by autologous blood injection into the basal nucleus. Edaravone or the specific inhibitor of matrix metalloproteinase 9 (MMP9), MMP9-IN-1, called MMP9-inh was administered 10 min after ICH via intraperitoneal injection. ELISA detection, neurobehavioral tests, dihydroethidium staining (DHE staining), intracisternal tracer infusion, hematoxylin and eosin (HE) staining, immunofluorescence staining, western blotting, Evans blue (EB) permeability assay, and brain water content test were performed. The results showed that OS was exacerbated, AQP4 polarity was lost, drainage function of brain fluids was damaged, brain injury was aggravated, expression of AQP4, MMP9, and GFAP increased, while the expression of β-DG decreased after ICH. Edaravone reduced OS, restored brain drainage function, reduced brain injury, and downregulated the expression of AQP4, MMP9. Both edaravone and MMP9-inh alleviated brain edema, maintained blood-brain barrier (BBB) integrity, mitigated the loss of AQP4 polarity, downregulated GFAP expression, and upregulated β-DG expression. The current study suggests that edaravone can maintain AQP4 polarity expression by inhibiting the OS /MMP9/β-DG pathway after ICH.
Collapse
Affiliation(s)
- Zhenhua Wang
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, China
- Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China
| | - Yuan Li
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, China
- Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China
| | - Zhixu Wang
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, China
- Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China
| | - Yuhui Liao
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, China
- Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China
- Sichuan University of Arts and Science, Sichuan, China
| | - Qingqing Ye
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, China
- Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China
| | - Shilong Tang
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, China
- Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ting Wei
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, China
- Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China
| | - Pengyu Xiao
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, China
- Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China
| | - Juan Huang
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, China.
- Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China.
| | - Weitian Lu
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, China.
- Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Das TK, Ganesh BP, Fatima-Shad K. Common Signaling Pathways Involved in Alzheimer's Disease and Stroke: Two Faces of the Same Coin. J Alzheimers Dis Rep 2023; 7:381-398. [PMID: 37220617 PMCID: PMC10200243 DOI: 10.3233/adr-220108] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/23/2022] [Accepted: 04/03/2023] [Indexed: 05/25/2023] Open
Abstract
Alzheimer's disease (AD) and stroke are two interrelated neurodegenerative disorders which are the leading cause of death and affect the neurons in the brain and central nervous system. Although amyloid-β aggregation, tau hyperphosphorylation, and inflammation are the hallmarks of AD, the exact cause and origin of AD are still undefined. Recent enormous fundamental discoveries suggest that the amyloid hypothesis of AD has not been proven and anti-amyloid therapies that remove amyloid deposition have not yet slowed cognitive decline. However, stroke, mainly ischemic stroke (IS), is caused by an interruption in the cerebral blood flow. Significant features of both disorders are the disruption of neuronal circuitry at different levels of cellular signaling, leading to the death of neurons and glial cells in the brain. Therefore, it is necessary to find out the common molecular mechanisms of these two diseases to understand their etiological connections. Here, we summarized the most common signaling cascades including autotoxicity, ApoE4, insulin signaling, inflammation, mTOR-autophagy, notch signaling, and microbiota-gut-brain axis, present in both AD and IS. These targeted signaling pathways reveal a better understanding of AD and IS and could provide a distinguished platform to develop improved therapeutics for these diseases.
Collapse
Affiliation(s)
- Tushar Kanti Das
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Bhanu Priya Ganesh
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kaneez Fatima-Shad
- School of Life Sciences, University of Technology Sydney, NSW, Australia
- Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia
- School of Behavioral and Health Sciences, Faculty of Health Sciences, Australian Catholic University, NSW, Australia
| |
Collapse
|
3
|
Zhu G, Wang X, Chen L, Lenahan C, Fu Z, Fang Y, Yu W. Crosstalk Between the Oxidative Stress and Glia Cells After Stroke: From Mechanism to Therapies. Front Immunol 2022; 13:852416. [PMID: 35281064 PMCID: PMC8913707 DOI: 10.3389/fimmu.2022.852416] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/11/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
Stroke is the second leading cause of global death and is characterized by high rates of mortality and disability. Oxidative stress is accompanied by other pathological processes that together lead to secondary brain damage in stroke. As the major component of the brain, glial cells play an important role in normal brain development and pathological injury processes. Multiple connections exist in the pathophysiological changes of reactive oxygen species (ROS) metabolism and glia cell activation. Astrocytes and microglia are rapidly activated after stroke, generating large amounts of ROS via mitochondrial and NADPH oxidase pathways, causing oxidative damage to the glial cells themselves and neurons. Meanwhile, ROS cause alterations in glial cell morphology and function, and mediate their role in pathological processes, such as neuroinflammation, excitotoxicity, and blood-brain barrier damage. In contrast, glial cells protect the Central Nervous System (CNS) from oxidative damage by synthesizing antioxidants and regulating the Nuclear factor E2-related factor 2 (Nrf2) pathway, among others. Although numerous previous studies have focused on the immune function of glial cells, little attention has been paid to the role of glial cells in oxidative stress. In this paper, we discuss the adverse consequences of ROS production and oxidative-antioxidant imbalance after stroke. In addition, we further describe the biological role of glial cells in oxidative stress after stroke, and we describe potential therapeutic tools based on glia cells.
Collapse
Affiliation(s)
- Ganggui Zhu
- Department of Neurosurgery, Hangzhou First People's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyu Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Luxi Chen
- Department of Medical Genetics, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cameron Lenahan
- Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, United States.,Department of Biomedical Science, Burrell College of Osteopathic Medicine, Las Cruces, NM, United States
| | - Zaixiang Fu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuanjian Fang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenhua Yu
- Department of Neurosurgery, Hangzhou First People's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
4
|
Shu W, Yang Z, Xu Z, Zhu T, Tian X, Yang Y. Effects of one-dimensional nanomaterial polyaniline nanorods on earthworm biomarkers and soil enzymes. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:35217-35229. [PMID: 35048350 DOI: 10.1007/s11356-021-18260-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 07/29/2021] [Accepted: 12/17/2021] [Indexed: 06/14/2023]
Abstract
Polyaniline nanorods (PANRs) are typical one-dimensional nanomaterials (1D NMs), which are widely used in medicine, batteries and water treatment, etc. Applications of PANRs will eventually enter the soil environment, but their ecotoxicity has been barely reported. Therefore, we measured earthworm biomass, earthworm biomarkers and soil enzymes to investigate the ecotoxicity of PANRs. The result of positive and increasing growth inhibition rates (GIR) showed that PANRs inhibited earthworm growth. As for earthworm biomarkers, PANRs caused a decrease in protein content, indicating that PANRs stress would increase earthworm energy consumption. Except for the 7th day, the activities of SOD, CAT and POD consistently increased, suggesting that PANRs activated the earthworm antioxidant system. The continually augment of MDA content indicated that PANRs stress would cause earthworm lipid damage. Na+-K+-ATPase increased with an excellent dose-time relationship. Differently, cellulase and AChE activities promoted at low concentrations and inhibited at high concentrations. The positive and dose-dependent IBRv2 indicated that the higher the concentrations of PANRs, the greater the ecotoxicity to earthworms. PANRs inhibited the soil enzyme activities such as sucrase, neutral phosphatase, protease and urease, while induced catalase activity in a dose-dependent manner. Earthworm addition reduced catalase activity by 10.74-29.99%, but improved other soil enzymes activities, demonstrating that earthworms played a positive role in regulating soil enzyme activity. GMean and T-SQI consistently increased due to earthworm activity, meaning a higher soil microbial functional diversity. Generally, this study provided data support for future PANRs toxicity studies, but their toxicity mechanisms still need to be further studied.
Collapse
Affiliation(s)
- Wenjun Shu
- College of Environmental Science and Engineering, Donghua University, Shanghai, China
| | - Zaifu Yang
- College of Environmental Science and Engineering, Donghua University, Shanghai, China.
| | - Zhinan Xu
- College of Environmental Science and Engineering, Donghua University, Shanghai, China
| | - Tong Zhu
- College of Environmental Science and Engineering, Donghua University, Shanghai, China
| | - Xinyue Tian
- College of Environmental Science and Engineering, Donghua University, Shanghai, China
| | - Yiran Yang
- College of Environmental Science and Engineering, Donghua University, Shanghai, China
| |
Collapse
|
5
|
Chrishtop V, Nikonorova V, Gutsalova A, Rumyantseva T, Dukhinova M, Salmina А. Systematic comparison of basic animal models of cerebral hypoperfusion. Tissue Cell 2022; 75:101715. [DOI: 10.1016/j.tice.2021.101715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/05/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 02/07/2023]
|
6
|
Wu L, Hu Y, Jiang L, Liang N, Liu P, Hong H, Yang S, Chen W. Zhuyu Annao decoction promotes angiogenesis in mice with cerebral hemorrhage by inhibiting the activity of PHD3. Hum Exp Toxicol 2021; 40:1867-1879. [PMID: 33896237 DOI: 10.1177/09603271211008523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/15/2022]
Abstract
Some traditional Chinese decoctions, such as Zhuyu Annao, exert favorable therapeutic effects on acute cerebral hemorrhage, hemorrhagic stroke, and other neurological diseases, but the underlying mechanism remains unclear. This study aimed to determine whether Zhuyu Annao decoction (ZYAND) protects the injured brain by promoting angiogenesis following intracerebral hemorrhage (ICH) and elucidate its specific mechanism. The effect of ZYAND on the nervous system of mice after ICH was explored through behavioral experiments, such as the Morris water maze and Rotarod tests, and its effects on oxidative stress were explored by detecting several oxidative stress markers, including malondialdehyde, nitric oxide, glutathione peroxidase, and superoxide dismutase. Real-time quantitative RT-PCR and WB were used to detect the effects of ZYAND on the levels of prolyl hydroxylase domain 3 (PHD3), hypoxia-inducible factor-1α (HIF-1α), and vascular endothelial growth factor (VEGF) in the brain tissues of mice. The effect of ZYAND on the NF-κB signaling pathway was detected using a luciferase reporter gene. A human umbilical cord vascular endothelial cell angiogenesis experiment was performed to determine whether ZYAND promotes angiogenesis. The Morris water maze test and other behavioral experiments verified that ZYAND improved the neurobehavior of mice after ICH. ZYAND activated the PHD3/HIF-1α signaling pathway, inhibiting the oxidative damage caused by ICH. In angiogenesis experiments, it was found that ZYAND promoted VEGF-induced angiogenesis by upregulating the expression of HIF-1α, and NF-κB signaling regulated the expression of HIF-1α by inhibiting PHD3. ZYAND exerts a reparative effect on brain tissue damaged after ICH through the NF-κB/ PHD3/HIF-1α/VEGF signaling axis.
Collapse
Affiliation(s)
- L Wu
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, China.,Guangxi Key Laboratory of Chinese Medicine Foundation Research, Guangxi University of Chinese Medicine, China.,Scientific Laboratorial Centre Guangxi University of Chinese Medicine, China.,Both authors contributed equally to this work and should be considered as equal first coauthors
| | - Y Hu
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, China.,Guangxi Key Laboratory of Chinese Medicine Foundation Research, Guangxi University of Chinese Medicine, China.,Both authors contributed equally to this work and should be considered as equal first coauthors
| | - L Jiang
- Graduate College of Guangxi University of Traditional Chinese Medicine, China
| | - N Liang
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, China
| | - P Liu
- Department of Cardiovascular Disease, Traditional Medicine Hospital Affiliated to Southwest Medical University, Luzhou, Sichuan, China
| | - H Hong
- Graduate College of Guangxi University of Traditional Chinese Medicine, China
| | - S Yang
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, China
| | - W Chen
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, China.,Guangxi Key Laboratory of Chinese Medicine Foundation Research, Guangxi University of Chinese Medicine, China
| |
Collapse
|
7
|
Shao A, Lin D, Wang L, Tu S, Lenahan C, Zhang J. Oxidative Stress at the Crossroads of Aging, Stroke and Depression. Aging Dis 2020; 11:1537-1566. [PMID: 33269106 PMCID: PMC7673857 DOI: 10.14336/ad.2020.0225] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/28/2020] [Accepted: 02/25/2020] [Indexed: 12/17/2022] Open
Abstract
Epidemiologic studies have shown that in the aging society, a person dies from stroke every 3 minutes and 42 seconds, and vast numbers of people experience depression around the globe. The high prevalence and disability rates of stroke and depression introduce enormous challenges to public health. Accumulating evidence reveals that stroke is tightly associated with depression, and both diseases are linked to oxidative stress (OS). This review summarizes the mechanisms of OS and OS-mediated pathological processes, such as inflammation, apoptosis, and the microbial-gut-brain axis in stroke and depression. Pathological changes can lead to neuronal cell death, neurological deficits, and brain injury through DNA damage and the oxidation of lipids and proteins, which exacerbate the development of these two disorders. Additionally, aging accelerates the progression of stroke and depression by overactive OS and reduced antioxidant defenses. This review also discusses the efficacy and safety of several antioxidants and antidepressants in stroke and depression. Herein, we propose a crosstalk between OS, aging, stroke, and depression, and provide potential therapeutic strategies for the treatment of stroke and depression.
Collapse
Affiliation(s)
- Anwen Shao
- 1Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Danfeng Lin
- 2Department of Surgical Oncology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Lingling Wang
- 2Department of Surgical Oncology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Sheng Tu
- 3State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China
| | - Cameron Lenahan
- 4Burrell College of Osteopathic Medicine, Las Cruces, USA.,5Center for Neuroscience Research, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Jianmin Zhang
- 1Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China.,6Brain Research Institute, Zhejiang University, Zhejiang, China.,7Collaborative Innovation Center for Brain Science, Zhejiang University, Zhejiang, China
| |
Collapse
|
8
|
Castañeda-Cabral JL, López-Ortega JG, Fajardo-Fregoso BF, Beas-Zárate C, Ureña-Guerrero ME. Glutamate induced neonatal excitotoxicity modifies the expression level of EAAT1 (GLAST) and EAAT2 (GLT-1) proteins in various brain regions of the adult rat. Neurosci Lett 2020; 735:135237. [PMID: 32645399 DOI: 10.1016/j.neulet.2020.135237] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/09/2019] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 02/08/2023]
Abstract
Glutamate-mediated excitatory synaptic signalling is primarily controlled by excitatory amino acid transporters (EAATs), such as EAAT1 and EAAT2, which are located mostly on astrocytes and, together, uptake more than 95 % of extracellular glutamate. Alterations in the functional expression levels of EAATs can lead to excessive extracellular glutamate accumulation, potentially triggering excitotoxicity and seizures, among other neurological disorders. Excitotoxicity induced in early developmental stages can lead to lasting changes in several neurotransmission systems, including the glutamatergic system, which could make the brain more susceptible to a second insult. In this study, the expression levels of EAAT1 (GLAST) and EAAT2 (GLT-1) proteins were assessed in the cerebral motor cortex (CMC), striatum, hippocampus and entorhinal cortex (EC) of male adult rats following the neonatal excitotoxic process triggered by monosodium glutamate (MSG)-treatment (4 g/kg of body weight at postnatal days 1,3,5 and 7, subcutaneously). Western blot analysis showed that neonatal MSG-treatment decreased EAAT1 expression levels in the CMC, striatum and hippocampus, while EAAT2 levels were increased in the striatum and EC and decreased in the CMC. Immunofluorescence staining confirmed the changes in EAAT1 and EAAT2 expression induced by neonatal MSG-treatment, which were accompanied by an increase in the glial fibrillary acidic protein (GFAP) immunofluorescence signalthat was particularly significant in the hippocampus. Our results show that a neonatal excitotoxic processes can induce lasting changes in the expression levels of EAAT1 and EAAT2 proteins and suggest that although astrogliosis occurs, glutamate uptake could be deficient, particularly in the CMC and hippocampus.
Collapse
Affiliation(s)
- José Luis Castañeda-Cabral
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - José Guadalupe López-Ortega
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Blanca Fabiola Fajardo-Fregoso
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Carlos Beas-Zárate
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Mónica E Ureña-Guerrero
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico.
| |
Collapse
|
9
|
Cordeiro JL, Neves JD, Vizuete AF, Aristimunha D, Pedroso TA, Sanches EF, Gonçalves CA, Netto CA. Arundic Acid (ONO-2506), an Inhibitor of S100B Protein Synthesis, Prevents Neurological Deficits and Brain Tissue Damage Following Intracerebral Hemorrhage in Male Wistar Rats. Neuroscience 2020; 440:97-112. [PMID: 32474054 DOI: 10.1016/j.neuroscience.2020.05.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/28/2020] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 01/13/2023]
Abstract
Stroke is one of the leading causes of mortality and neurological morbidity. Intracerebral hemorrhage (ICH) has the poorest prognosis among all stroke subtypes and no treatment has been effective in improving outcomes. Following ICH, the observed high levels of S100B protein have been associated with worsening of injury and neurological deficits. Arundic acid (AA) exerts neuroprotective effects through inhibition of astrocytic synthesis of S100B in some models of experimental brain injury; however, it has not been studied in ICH. The aim of this study was to evaluate the effects of intracerebroventricular (ICV) administration of AA in male Wistar rats submitted to ICH model assessing the following variables: reactive astrogliosis, S100B levels, antioxidant defenses, cell death, lesion extension and neurological function. Firstly, AA was injected at different doses (0.02, 0.2, 2 and 20 μg/μl) in the left lateral ventricle in order to observe which dose would decrease GFAP and S100B striatal levels in non-injured rats. Following determination of the effective dose, ICH damage was induced by IV-S collagenase intrastrial injection and 2 μg/μl AA was injected through ICV route immediately before injury. AA treatment prevented ICH-induced neurological deficits and tissue damage, inhibited excessive astrocytic activation and cellular apoptosis, reduced peripheral and central S100B levels (in striatum, serum and cerebrospinal fluid), improved neuronal survival and enhanced the antioxidant defences after injury. Altogether, these results suggest that S100B is a viable target for treating ICH and highlight AA as an interesting strategy for improving neurological outcome after experimental brain hemorrhage.
Collapse
Affiliation(s)
- J L Cordeiro
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil; Post-graduation Program of Neurosciences, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 90035-190, Brazil.
| | - J D Neves
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - A F Vizuete
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - D Aristimunha
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - T A Pedroso
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - E F Sanches
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil; Post-graduation Program of Phisiology, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 90035-190, Brazil
| | - C A Gonçalves
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - C A Netto
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| |
Collapse
|
10
|
Gao BY, Xu DS, Liu PL, Li C, Du L, Hua Y, Hu J, Hou JY, Bai YL. Modified constraint-induced movement therapy alters synaptic plasticity of rat contralateral hippocampus following middle cerebral artery occlusion. Neural Regen Res 2020; 15:1045-1057. [PMID: 31823884 PMCID: PMC7034265 DOI: 10.4103/1673-5374.270312] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/12/2022] Open
Abstract
Modified constraint-induced movement therapy is an effective treatment for neurological and motor impairments in patients with stroke by increasing the use of their affected limb and limiting the contralateral limb. However, the molecular mechanism underlying its efficacy remains unclear. In this study, a middle cerebral artery occlusion (MCAO) rat model was produced by the suture method. Rats received modified constraint-induced movement therapy 1 hour a day for 14 consecutive days, starting from the 7th day after middle cerebral artery occlusion. Day 1 of treatment lasted for 10 minutes at 2 r/min, day 2 for 20 minutes at 2 r/min, and from day 3 onward for 20 minutes at 4 r/min. CatWalk gait analysis, adhesive removal test, and Y-maze test were used to investigate motor function, sensory function as well as cognitive function in rodent animals from the 1st day before MCAO to the 21st day after MCAO. On the 21st day after MCAO, the neurotransmitter receptor-related genes from both contralateral and ipsilateral hippocampi were tested by micro-array and then verified by western blot assay. The glutamate related receptor was shown by transmission electron microscopy and the glutamate content was determined by high-performance liquid chromatography. The results of behavior tests showed that modified constraint-induced movement therapy promoted motor and sensory functional recovery in the middle cerebral artery-occluded rats, but had no effect on cognitive function. The modified constraint-induced movement therapy upregulated the expression of glutamate ionotropic receptor AMPA type subunit 3 (Gria3) in the hippocampus and downregulated the expression of the beta3-adrenergic receptor gene Adrb3 and arginine vasopressin receptor 1A, Avpr1a in the middle cerebral artery-occluded rats. In the ipsilateral hippocampus, only Adra2a was downregulated, and there was no significant change in Gria3. Transmission electron microscopy revealed a denser distribution the more distribution of postsynaptic glutamate receptor 2/3, which is an α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor, within 240 nm of the postsynaptic density in the contralateral cornu ammonis 3 region. The size and distribution of the synaptic vesicles within 100 nm of the presynaptic active zone were unchanged. Western blot analysis showed that modified constraint-induced movement therapy also increased the expression of glutamate receptor 2/3 and brain-derived neurotrophic factor in the hippocampus of rats with middle cerebral artery occlusion, but had no effect on Synapsin I levels. Besides, we also found modified constraint-induced movement therapy effectively reduced glutamate content in the contralateral hippocampus. This study demonstrated that modified constraint-induced movement therapy is an effective rehabilitation therapy in middle cerebral artery-occluded rats, and suggests that these positive effects occur via the upregulation of the postsynaptic membrane α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor expression. This study was approved by the Institutional Animal Care and Use Committee of Fudan University, China (approval No. 201802173S) on March 3, 2018.
Collapse
Affiliation(s)
- Bei-Yao Gao
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Dong-Sheng Xu
- Rehabilitation Section, Department of Spine Surgery, Tongji Hospital of Tongji University; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University) Ministry of Education, Shanghai, China
| | - Pei-Le Liu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ce Li
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Liang Du
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yan Hua
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jian Hu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jia-Yun Hou
- Zhongshan Hospital Institute of Clinical Science, Fudan University, Shanghai, China
| | - Yu-Long Bai
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Tiburcio-Félix R, Cisneros B, Hernández-Kelly LCR, Hernández-Contreras MA, Luna-Herrera J, Rea-Hernández I, Jiménez-Aguilar R, Olivares-Bañuelos TN, Ortega A. Neuronal Nitric Oxide Synthase in Cultured Cerebellar Bergmann Glia: Glutamate-Dependent Regulation. ACS Chem Neurosci 2019; 10:2668-2675. [PMID: 31091406 DOI: 10.1021/acschemneuro.8b00656] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/01/2023] Open
Abstract
Glutamate exerts its actions through the activation of membrane receptors expressed in neurons and glia cells. The signaling properties of glutamate transporters have been characterized recently, suggesting a complex array of signaling transactions triggered by presynaptic released glutamate. In the cerebellar molecular layer, glutamatergic synapses are surrounded by Bergmann glia cells, compulsory participants of glutamate turnover and supply to neurons. Since a glutamate-dependent increase in cGMP levels has been described in these cells and the nitric oxide-cGMP signaling cascade increases their glutamate uptake activity, we describe here the Bergmann glia expression of neuronal nitric oxide synthetase. An augmentation of neuronal nitric oxide synthase was found upon glutamate exposure. This effect is mediated by glutamate transporters and is related to an increase in the stability of the enzyme. These results strengthen the notion of a complex regulation of glial glutamate uptake that supports neuronal glutamate signaling.
Collapse
Affiliation(s)
- Reynaldo Tiburcio-Félix
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Apartado Postal 14-740, Ciudad de México 07360, Mexico
| | - Bulmaro Cisneros
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Apartado Postal 14-740, Ciudad de México 07360, Mexico
| | - Luisa C. R. Hernández-Kelly
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Apartado Postal 14-740, Ciudad de México 07360, Mexico
| | - María A. Hernández-Contreras
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México 11340, México
| | - Julieta Luna-Herrera
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México 11340, México
| | - Ismael Rea-Hernández
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Apartado Postal 14-740, Ciudad de México 07360, Mexico
| | - Rosalinda Jiménez-Aguilar
- Unidad de Cuidados Intensivos Pediátricos, Hospital General La Raza Gaudencio González Garza, Unidad de Alta Especialidad Médica (UMAE), Instituto Mexicano del Seguro Social, Ciudad de México 02990, México
| | - Tatiana N. Olivares-Bañuelos
- Instituto de Investigaciones Oceanológicas, Universidad Autónoma de Baja California, Ensenada, Baja California 22860, México
| | - Arturo Ortega
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Apartado Postal 14-740, Ciudad de México 07360, Mexico
| |
Collapse
|