1
|
Sabry HA, Ali EHA, Osman AA, Zahra MM. Nanotechnology strategy for inhibition of PARP1 and IL-17A-associated with neurotoxicity in rats exposed to hospital wastewater. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4149-4164. [PMID: 39422747 PMCID: PMC11978713 DOI: 10.1007/s00210-024-03512-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024]
Abstract
Hospital wastewater (HWW) poses a serious hazard to human health security concerning its high susceptibility to neurodegeneration. Water sources and ecosystems are exposed to a complicated pollution load from a variety of refractory organics and pharmaceutical active composites. This study evaluates the treated newly developed nanocomposite (NiFe2O4) HWW on the neural injury induced by HWW action in rats. Three groups of male Wistar rats were distributed, with eight rats in each: group I: tap water served as a control; group II: HWW; and group III: nano-HWW. Each group was intragastrical administrated with each type of water (2.5 ml/100 g b.wt/6 h) for 28 consecutive days. The open field test and Morris Water Maze assessed behavioral activity and spatial learning 2 days before the last day. The research demonstrated that HWW treated with nanocomposite (NiFe2O4) may exert decreased risks of the neural impairment effect of HWW. This improvement was achieved by reducing the neurotoxicity by lowering nitric oxide contents, lipid peroxidation, acetylcholinesterase, interleukin-17A (IL-17A), and poly(ADP-ribose) polymerase1(PARP1) while restoring the antioxidant biomarkers and neurotransmitter levels (β-endorphin, norepinephrine, dopamine, and serotonin) of the treated groups in the cortex and brainstem and enhancement of the histopathology of the cortex as well. In conclusion, this study introduced a newly developed nanotechnology application for treating HWW to protect from neural injury. The findings of this research have significant value for policymakers, Ministry of Health management, and environmental organizations in their selection of suitable techniques and procedures to optimize hospital wastewater treatment efficiency.
Collapse
Affiliation(s)
- Hend A Sabry
- Zoology Department, Faculty of Women for Arts, Science, and Education, Ain Shams University, Cairo, Egypt.
| | - Elham H A Ali
- Zoology Department, Faculty of Women for Arts, Science, and Education, Ain Shams University, Cairo, Egypt
| | - Amany A Osman
- Zoology Department, Faculty of Women for Arts, Science, and Education, Ain Shams University, Cairo, Egypt
| | - Mai M Zahra
- Zoology Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
2
|
Sampath D, Zardeneta ME, Akbari Z, Singer J, Gopalakrishnan B, Hurst DA, Villarreal M, McDaniel EA, Noarbe BP, Obenaus A, Sohrabji F. Loss of white matter tracts and persistent microglial activation in the chronic phase of ischemic stroke in female rats and the effect of miR-20a-3p treatment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.01.636074. [PMID: 39975179 PMCID: PMC11838816 DOI: 10.1101/2025.02.01.636074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Our previous studies showed that intravenous injections of the small non-coding RNA mir-20a-3p is neuroprotective for stroke in the acute phase and attenuates long-term cognitive impairment in middle-aged female rats. In this study, we evaluated postmortem brain pathology at 100+d after stroke in a set of behaviorally characterized animals. This included Sham (no stroke) controls or stroke animals that received either mir20a-3p at 4h, 24h and 70d iv post stroke (MCAo+mir20a-3p) or a scrambled oligo (MCAo+Scr). Brain volumetric features were analyzed with T2 weighted and Diffusion Tensor magnetic resonance imaging (MRI) followed by histological analysis. Principal component analysis of Fractional Anisotropy (FA)-diffusion tensor MRI measures showed that MCAo+Scr and MCAo+mir20a-3p groups differed significantly in the volume of white matter but not gray matter. Weil myelin-stained sections confirmed decreased volume of the corpus callosum, internal capsule and the anterior commissure in the ischemic hemisphere of MCAo+Scr animals compared to the non-ischemic hemisphere, while sham and MCAo+Mir-20a-3p showed no hemispheric asymmetries. The MCAo+Scr group also exhibited asymmetry in hemisphere and lateral ventricle volumes, with ventricular enlargement in the ischemic hemisphere as compared to the non-ischemic hemisphere. The numbers of microglia were significantly elevated in white matter tracts in the MCAo+Scr group, with a trend towards increased myelin phagocytic microglia in these tracts. Regression analysis indicated that performance on an episodic memory test (novel object recognition test; NORT) was associated with decreased white matter volume and increased microglial numbers. These data support the hypothesis that stroke-induced cognitive impairment is accompanied by white matter attrition and persistent microglial activation and is consistent with reports that cognitive deterioration resulting from vascular diseases, such as stroke, is associated with secondary neurodegeneration in regions distal from the initial infarction.
Collapse
|
3
|
Luo Y, Zhu J, Hu Z, Luo W, Du X, Hu H, Peng S. Progress in the Pathogenesis of Diabetic Encephalopathy: The Key Role of Neuroinflammation. Diabetes Metab Res Rev 2024; 40:e3841. [PMID: 39295168 DOI: 10.1002/dmrr.3841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 04/29/2024] [Accepted: 06/27/2024] [Indexed: 09/21/2024]
Abstract
Diabetic encephalopathy (DE) is a severe complication that occurs in the central nervous system (CNS) and leads to cognitive impairment. DE involves various pathophysiological processes, and its pathogenesis is still unclear. This review summarised current research on the pathogenesis of diabetic encephalopathy, which involves neuroinflammation, oxidative stress, iron homoeostasis, blood-brain barrier disruption, altered gut microbiota, insulin resistance, etc. Among these pathological mechanisms, neuroinflammation has been focused on. This paper summarises some of the molecular mechanisms involved in neuroinflammation, including the Mammalian Target of Rapamycin (mTOR), Lipocalin-2 (LCN-2), Pyroptosis, Advanced Glycosylation End Products (AGEs), and some common pro-inflammatory factors. In addition, we discuss recent advances in the study of potential therapeutic targets for the treatment of DE against neuroinflammation. The current research on the pathogenesis of DE is progressing slowly, and more research is needed in the future. Further study of neuroinflammation as a mechanism is conducive to the discovery of more effective treatments for DE in the future.
Collapse
Affiliation(s)
- Yifan Luo
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Clinical Medicine, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Jinxi Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Clinical Medicine, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Ziyan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Clinical Medicine, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Wei Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaohong Du
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Haijun Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shengliang Peng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
4
|
Li JQ, Shi YH, Min-Xu, Shi CX, Teng-Wang, Wang TH, Zuo ZF, Liu XZ. Discovery of astragaloside IV against high glucose-induced apoptosis in retinal ganglion cells: Bioinformatics and in vitro studies. Gene 2024; 905:148219. [PMID: 38286267 DOI: 10.1016/j.gene.2024.148219] [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: 12/09/2023] [Revised: 01/22/2024] [Accepted: 01/26/2024] [Indexed: 01/31/2024]
Abstract
OBJECTIVE To examine the therapeutic mechanism of astragaloside IV (AS-IV) in the management of retinal ganglion cell (RGC) injury induced by high glucose (HG), a comprehensive approach involving the integration of network pharmacology and conducting in vitro and in vivo experiments was utilized. METHODS A rat model of diabetic retinopathy (DR) injury was created by administering streptozotocin through intraperitoneal injection. Additionally, a model of RGC injury induced by HG was established using a glucose concentration of 0.3 mmol/mL. Optical coherence tomography (OCT) images were captured 8 weeks after the injection of AS-IV. AS-IV and FBS were added to the culture medium and incubated for 48 h. The viability of cells was assessed using a CCK-8 assay, while the content of reactive oxygen species (ROS) was measured using DCFH-DA. Apoptosis was evaluated using Annexin V-PI. To identify the targets of AS-IV, hyperglycemia, and RGC, publicly available databases were utilized. The Metascape platform was employed for conducting GO and KEGG enrichment analyses. The STRING database in conjunction with Cytoscape 3.7.2 was used to determine common targets of protein-protein interactions (PPIs) and to identify the top 10 core target proteins in the RGC based on the MCC algorithm. qRT-PCR was used to measure the mRNA expression levels of the top10 core target proteins in RGCs. RESULTS OCT detection indicated that the thickness of the outer nucleus, and inner and outer accessory layers of the retina increased in the AS-IV treated retina compared to that in the DM group but decreased compared to that in the CON group. Coculturing RGC cells with AS-IV after HG induction resulted in a significant increase in cell viability and a decrease in ROS and apoptosis, suggesting that AS-IV can reduce damage to RGC cells caused by high glucose levels by inhibiting oxidative stress. There were 14 potential targets of AS-IV in the treatment of RGC damage induced by high glucose levels. The top 10 core target proteins identified by the MCC algorithm were HIF1α, AKT1, CTNNB1, SMAD2, IL6, SMAD3, IL1β, PPARG, TGFβ1, and NOTCH3. qRT-PCR analysis showed that AS-IV could upregulate the mRNA expression levels of SMAD3, TGF-β1, and NOTCH3, and downregulate the mRNA expression levels of HIF1α, AKT1, CTNNB1, SMAD2, SMAD3, and IL-1β in high glucose-induced RGC cells. CONCLUSION The findings of this study validate the efficacy of astragaloside IV in the treatment of DR and shed light on the molecular network involved. Specifically, HIF1α, AKT1, CTNNB1, SMAD2, SMAD3, and IL-1β were identified as the crucial candidate molecules responsible for the protective effects of astragaloside IV on RGCs.
Collapse
Affiliation(s)
- Jun-Qi Li
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou 121000, China
| | - Ya-Hui Shi
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou 121000, China
| | - Min-Xu
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou 121000, China
| | - Cai-Xing Shi
- School of Basic Medicine, Jining Medical University, Jining 272067, China
| | - Teng-Wang
- The First Affiliated Hospital of Jinzhou Medical University, 121000, China
| | - Ting-Hua Wang
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Institute of Neuroscience, Kunming Medical University, Kunming 650500, China.
| | - Zhong-Fu Zuo
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou 121000, China.
| | - Xue-Zheng Liu
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou 121000, China.
| |
Collapse
|
5
|
Mews P, Cunningham AM, Scarpa J, Ramakrishnan A, Hicks EM, Bolnick S, Garamszegi S, Shen L, Mash DC, Nestler EJ. Convergent abnormalities in striatal gene networks in human cocaine use disorder and mouse cocaine administration models. SCIENCE ADVANCES 2023; 9:eadd8946. [PMID: 36763659 PMCID: PMC9916993 DOI: 10.1126/sciadv.add8946] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/06/2023] [Indexed: 06/11/2023]
Abstract
Cocaine use disorder (CUD) is an intractable syndrome, and rising overdose death rates represent a substantial public health crisis that exacts tremendous personal and financial costs on patients and society. Sharp increases in cocaine use drive the urgent need for better mechanistic insight into this chronic relapsing brain disorder that currently lacks effective treatment options. To investigate the transcriptomic changes involved, we conducted RNA sequencing on two striatal brain regions that are heavily implicated in CUD, the nucleus accumbens and caudate nucleus, from men suffering from CUD and matched controls. Weighted gene coexpression analyses identified CUD-specific gene networks enriched in ionotropic receptors and linked to lowered neuroinflammation, contrasting the proinflammatory responses found in opioid use disorder. Integration of comprehensive transcriptomic datasets from mouse cocaine self-administration models revealed evolutionarily conserved gene networks in CUD that implicate especially D1 medium spiny neurons as drivers of cocaine-induced plasticity.
Collapse
Affiliation(s)
- Philipp Mews
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ashley M. Cunningham
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph Scarpa
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily M. Hicks
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah Bolnick
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Susanna Garamszegi
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Li Shen
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Deborah C. Mash
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Eric J. Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
6
|
Singh Gautam A, Kumar Singh R. Therapeutic potential of targeting IL-17 and its receptor signaling in neuroinflammation. Drug Discov Today 2023; 28:103517. [PMID: 36736763 DOI: 10.1016/j.drudis.2023.103517] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 12/26/2022] [Accepted: 01/26/2023] [Indexed: 02/04/2023]
Abstract
T helper 17 cells are thought to significantly contribute to the neuroinflammation process during neurogenerative diseases via their signature cytokine, interleukin (IL)-17. Recently, an emerging key role of IL-17 and its receptors has been documented in inflammatory and autoimmune diseases. The clinical studies conducted on patients with neurodegenerative disease have also shown an increase in IL-17 levels in serum as well as cerebrospinal fluid samples. Therapeutic targeting of either IL-17 receptors or direct IL-17 neutralizing antibodies has shown a promising preclinical and clinical proof of concept for treating chronic autoimmune neurodegenerative diseases such as multiple sclerosis. Thus, IL-17 and its receptors have a central role in regulation of neuroinflammation and can be considered as one of the major therapeutic targets in chronic neuroinflammatory diseases.
Collapse
Affiliation(s)
- Avtar Singh Gautam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Transit Campus, Bijnour-sisendi Road, Sarojini Nagar, Lucknow 226002, Uttar Pradesh, India
| | - Rakesh Kumar Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Transit Campus, Bijnour-sisendi Road, Sarojini Nagar, Lucknow 226002, Uttar Pradesh, India.
| |
Collapse
|
7
|
Schiweck C, Aichholzer M, Reif A, Edwin Thanarajah S. Targeting IL-17A signaling in suicidality, promise or the long arm of coincidence? Evidence in psychiatric populations revisited. JOURNAL OF AFFECTIVE DISORDERS REPORTS 2023. [DOI: 10.1016/j.jadr.2022.100454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
8
|
Yang G, Fan X, Mazhar M, Guo W, Zou Y, Dechsupa N, Wang L. Neuroinflammation of microglia polarization in intracerebral hemorrhage and its potential targets for intervention. Front Mol Neurosci 2022; 15:1013706. [PMID: 36304999 PMCID: PMC9592761 DOI: 10.3389/fnmol.2022.1013706] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS) and play a key role in neurological diseases, including intracerebral hemorrhage (ICH). Microglia are activated to acquire either pro-inflammatory or anti-inflammatory phenotypes. After the onset of ICH, pro-inflammatory mediators produced by microglia at the early stages serve as a crucial character in neuroinflammation. Conversely, switching the microglial shift to an anti-inflammatory phenotype could alleviate inflammatory response and incite recovery. This review will elucidate the dynamic profiles of microglia phenotypes and their available shift following ICH. This study can facilitate an understanding of the self-regulatory functions of the immune system involving the shift of microglia phenotypes in ICH. Moreover, suggestions for future preclinical and clinical research and potential intervention strategies are discussed.
Collapse
Affiliation(s)
- Guoqiang Yang
- Research Center for Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Acupuncture and Rehabilitation Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Xuehui Fan
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Maryam Mazhar
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Wubin Guo
- Department of General Surgery, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Yuanxia Zou
- Research Center for Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Nathupakorn Dechsupa
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- *Correspondence: Li Wang Nathupakorn Dechsupa
| | - Li Wang
- Research Center for Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
- *Correspondence: Li Wang Nathupakorn Dechsupa
| |
Collapse
|
9
|
Xu XJ, Ge QQ, Yang MS, Zhuang Y, Zhang B, Dong JQ, Niu F, Li H, Liu BY. Neutrophil-derived interleukin-17A participates in neuroinflammation induced by traumatic brain injury. Neural Regen Res 2022; 18:1046-1051. [PMID: 36254991 PMCID: PMC9827773 DOI: 10.4103/1673-5374.355767] [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: 01/11/2023] Open
Abstract
After brain injury, infiltration and abnormal activation of neutrophils damages brain tissue and worsens inflammation, but the mediators that connect activated neutrophils with neuroinflammation have not yet been fully clarified. To identify regulators of neutrophil-mediated neuroinflammation after traumatic brain injury, a mouse model of traumatic brain injury was established by controlled cortical impact. At 7 days post-injury (sub-acute phase), genome-wide transcriptomic data showed that interleukin 17A-associated signaling pathways were markedly upregulated, suggesting that interleukin 17A may be involved in neuroinflammation. Double immunofluorescence staining showed that interleukin 17A was largely secreted by neutrophils rather than by glial cells and neurons. Furthermore, nuclear factor-kappaB and Stat3, both of which are important effectors in interleukin 17A-mediated proinflammatory responses, were significantly activated. Collectively, our findings suggest that neutrophil-derived interleukin 17A participates in neutrophil-mediated neuroinflammation during the subacute phase of traumatic brain injury. Therefore, interleukin 17A may be a promising therapeutic target for traumatic brain injury.
Collapse
Affiliation(s)
- Xiao-Jian Xu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Qian-Qian Ge
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China,Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Meng-Shi Yang
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China,Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuan Zhuang
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China,Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bin Zhang
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China,Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jin-Qian Dong
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China,Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fei Niu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Hao Li
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China,Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bai-Yun Liu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China,Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China,Nerve Injury and Repair Center of Beijing Institute for Brain Disorders, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China,Correspondence to: Bai-Yun Liu, .
| |
Collapse
|
10
|
An J, Li H, Xia D, Xu B, Wang J, Qiu H, He J. The role of interleukin-17 in epilepsy. Epilepsy Res 2022; 186:107001. [PMID: 35994860 DOI: 10.1016/j.eplepsyres.2022.107001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/26/2022] [Accepted: 08/11/2022] [Indexed: 11/30/2022]
Abstract
Epilepsy is a common neurological disorder that seriously affects human health. It is a chronic central nervous system dysfunction caused by abnormal discharges of neurons. About 50 million patients worldwide are affected by epilepsy. Although epileptic symptoms of most patients are controllable, some patients with refractory epilepsy have no response to antiseizure medications. It is necessary to investigate the pathogenesis of epilepsy and identify new therapeutic targets for refractory epilepsy. Epileptic disorders often accompany cerebral inflammatory reactions. Recently, the role of inflammation in the onset of epilepsy has increasingly attracted attention. The activation of both innate and adaptive immunity plays a significant role in refractory epilepsy. According to several clinical studies, interleukin-17, an essential inflammatory mediator linking innate and adaptive immunity, increased significantly in the body liquid and epileptic focus of patients with epilepsy. Experimental studies also indicated that interleukin-17 participated in epileptogenesis through various mechanisms. This review summarized the current studies about interleukin-17 in epilepsy and aimed at finding new therapeutic targets for refractory epilepsy.
Collapse
Affiliation(s)
- Jiayin An
- Emergency Department, Naval Hospital of Eastern Theater, Zhejiang, China.
| | - He Li
- Emergency Department, Naval Hospital of Eastern Theater, Zhejiang, China.
| | - Demeng Xia
- Emergency Department, Naval Hospital of Eastern Theater, Zhejiang, China; Luodian Clinical Drug Research Center, Shanghai Baoshan Luodian Hospital, Shanghai University, Shanghai, China.
| | - Bin Xu
- Emergency Department, Naval Hospital of Eastern Theater, Zhejiang, China.
| | - Jiayan Wang
- Emergency Department, Naval Hospital of Eastern Theater, Zhejiang, China.
| | - Huahui Qiu
- Zhoushan Hospital, Zhejiang University, Zhoushan, Zhejiang, China.
| | - Jiaojiang He
- Department of Neurosurgery, West China Hospital of Sichuan University, Sichuan, China.
| |
Collapse
|
11
|
Freitas FEDA, Batista MAC, Braga DCDA, de Oliveira LB, Antunes VR, Cardoso LM. The gut-brain axis and sodium appetite: Can inflammation-related signaling influence the control of sodium intake? Appetite 2022; 175:106050. [PMID: 35447164 DOI: 10.1016/j.appet.2022.106050] [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: 09/16/2021] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022]
Abstract
Sodium is the main cation present in the extracellular fluid. Sodium and water content in the body are responsible for volume and osmotic homeostasis through mechanisms involving sodium and water excretion and intake. When body sodium content decreases below the homeostatic threshold, a condition termed sodium deficiency, highly motivated sodium seeking, and intake occurs. This is termed sodium appetite. Classically, sodium and water intakes are controlled by a number of neuroendocrine mechanisms that include signaling molecules from the renin-angiotensin-aldosterone system acting in the central nervous system (CNS). However, recent findings have shown that sodium and water intakes can also be influenced by inflammatory agents and mediators acting in the CNS. For instance, central infusion of IL-1β or TNF-α can directly affect sodium and water consumption in animal models. Some dietary conditions, such as high salt intake, have been shown to change the intestinal microbiome composition, stimulating the immune branch of the gut-brain axis through the production of inflammatory cytokines, such as IL-17, which can stimulate the brain immune system. In this review, we address the latest findings supporting the hypothesis that immune signaling in the brain could produce a reduction in thirst and sodium appetite and, therefore, contribute to sodium intake control.
Collapse
Affiliation(s)
| | | | | | | | - Vagner Roberto Antunes
- Dept. of Physiology and Biophysics - ICB, University of São Paulo, São Paulo, SP, Brazil
| | | |
Collapse
|
12
|
Wu Z, Chhun BB, Popova G, Guo SM, Kim CN, Yeh LH, Nowakowski T, Zou J, Mehta SB. DynaMorph: self-supervised learning of morphodynamic states of live cells. Mol Biol Cell 2022; 33:ar59. [PMID: 35138913 PMCID: PMC9265147 DOI: 10.1091/mbc.e21-11-0561] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/24/2022] [Accepted: 02/04/2022] [Indexed: 11/16/2022] Open
Abstract
A cell's shape and motion represent fundamental aspects of cell identity and can be highly predictive of function and pathology. However, automated analysis of the morphodynamic states remains challenging for most cell types, especially primary human cells where genetic labeling may not be feasible. To enable automated and quantitative analysis of morphodynamic states, we developed DynaMorph-a computational framework that combines quantitative live cell imaging with self-supervised learning. To demonstrate the robustness and utility of this approach, we used DynaMorph to annotate morphodynamic states observed with label-free measurements of optical density and anisotropy of live microglia isolated from human brain tissue. These cells show complex behavior and have varied responses to disease-relevant perturbations. DynaMorph generates quantitative morphodynamic representations that can be used to compare the effects of the perturbations. Using DynaMorph, we identify distinct morphodynamic states of microglia polarization and detect rare transition events between states. The concepts and the methods presented here can facilitate automated discovery of functional states of diverse cellular systems.
Collapse
Affiliation(s)
- Zhenqin Wu
- Department of Chemistry, Stanford University, Stanford, CA 94305
| | | | - Galina Popova
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143
| | | | - Chang N. Kim
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143
| | - Li-Hao Yeh
- Chan Zuckerberg Biohub, San Francisco, CA 94158
| | - Tomasz Nowakowski
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143
| | - James Zou
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305
| | | |
Collapse
|
13
|
Yoshimizu A, Kinoshita K, Ichihara Y, Kurauchi Y, Seki T, Katsuki H. Hydroxychloroquine improves motor function and affords neuroprotection without inhibition of inflammation and autophagy in mice after intracerebral hemorrhage. J Neuroimmunol 2022; 362:577786. [PMID: 34920280 DOI: 10.1016/j.jneuroim.2021.577786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/28/2021] [Accepted: 12/06/2021] [Indexed: 01/05/2023]
Abstract
We examined the effect of an immunomodulator hydroxychloroquine, also known as a Nurr1 ligand and an autophagy inhibitor, on a mouse model of intracerebral hemorrhage (ICH). Daily administration of hydroxychloroquine (100 mg/kg, i.p.) from 3 h after induction of ICH alleviated neurological deficits of mice, increased the number of surviving neurons in the hematoma and prevented fragmentation of axon structures in the internal capsule. Unexpectedly, hydroxychloroquine did not inhibit either upregulation of pro-inflammatory mediators or autophagic responses in the brain. Hence, hydroxychloroquine may produce therapeutic effects on ICH primarily via neuroprotection including preservation of the axon tract integrity.
Collapse
Affiliation(s)
- Ayaka Yoshimizu
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Keita Kinoshita
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Yusei Ichihara
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Yuki Kurauchi
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Takahiro Seki
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Hiroshi Katsuki
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan.
| |
Collapse
|
14
|
Bi R, Fang Z, You M, He Q, Hu B. Microglia Phenotype and Intracerebral Hemorrhage: A Balance of Yin and Yang. Front Cell Neurosci 2021; 15:765205. [PMID: 34720885 PMCID: PMC8549831 DOI: 10.3389/fncel.2021.765205] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 09/23/2021] [Indexed: 11/15/2022] Open
Abstract
Intracerebral hemorrhage (ICH) features extremely high rates of morbidity and mortality, with no specific and effective therapy. And local inflammation caused by the over-activated immune cells seriously damages the recovery of neurological function after ICH. Fortunately, immune intervention to microglia has provided new methods and ideas for ICH treatment. Microglia, as the resident immune cells in the brain, play vital roles in both tissue damage and repair processes after ICH. The perihematomal activated microglia not only arouse acute inflammatory responses, oxidative stress, excitotoxicity, and cytotoxicity to cause neuron death, but also show another phenotype that inhibit inflammation, clear hematoma and promote tissue regeneration. The proportion of microglia phenotypes determines the progression of brain tissue damage or repair after ICH. Therefore, microglia may be a promising and imperative therapeutic target for ICH. In this review, we discuss the dual functions of microglia in the brain after an ICH from immunological perspective, elaborate on the activation mechanism of perihematomal microglia, and summarize related therapeutic drugs researches.
Collapse
Affiliation(s)
- Rentang Bi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi Fang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingfeng You
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Quanwei He
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
B Cells in Neuroinflammation: New Perspectives and Mechanistic Insights. Cells 2021; 10:cells10071605. [PMID: 34206848 PMCID: PMC8305155 DOI: 10.3390/cells10071605] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
In recent years, the role of B cells in neurological disorders has substantially expanded our perspectives on mechanisms of neuroinflammation. The success of B cell-depleting therapies in patients with CNS diseases such as neuromyelitis optica and multiple sclerosis has highlighted the importance of neuroimmune crosstalk in inflammatory processes. While B cells are essential for the adaptive immune system and antibody production, they are also major contributors of pro- and anti-inflammatory cytokine responses in a number of inflammatory diseases. B cells can contribute to neurological diseases through peripheral immune mechanisms, including production of cytokines and antibodies, or through CNS mechanisms following compartmentalization. Emerging evidence suggests that aberrant pro- or anti-inflammatory B cell populations contribute to neurological processes, including glial activation, which has been implicated in the pathogenesis of several neurodegenerative diseases. In this review, we summarize recent findings on B cell involvement in neuroinflammatory diseases and discuss evidence to support pathogenic immunomodulatory functions of B cells in neurological disorders, highlighting the importance of B cell-directed therapies.
Collapse
|
16
|
Liu J, Liu L, Wang X, Jiang R, Bai Q, Wang G. Microglia: A Double-Edged Sword in Intracerebral Hemorrhage From Basic Mechanisms to Clinical Research. Front Immunol 2021; 12:675660. [PMID: 34025674 PMCID: PMC8135095 DOI: 10.3389/fimmu.2021.675660] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS). It is well established that microglia are activated and polarized to acquire different inflammatory phenotypes, either pro-inflammatory or anti-inflammatory phenotypes, which act as a critical component in the neuroinflammation following intracerebral hemorrhage (ICH). Microglia produce pro-inflammatory mediators at the early stages after ICH onset, anti-inflammatory microglia with neuroprotective effects appear to be suppressed. Previous research found that driving microglia towards an anti-inflammatory phenotype could restrict inflammation and engulf cellular debris. The principal objective of this review is to analyze the phenotypes and dynamic profiles of microglia as well as their shift in functional response following ICH. The results may further the understanding of the body's self-regulatory functions involving microglia following ICH. On this basis, suggestions for future clinical development and research are provided.
Collapse
Affiliation(s)
- Jiachen Liu
- Xiangya Medical College of Central South University, Changsha, China
| | - Lirong Liu
- Department of Neurology, Shanxi Medical University, Taiyuan, China
| | - Xiaoyu Wang
- Xiangya Medical College of Central South University, Changsha, China
| | - Rundong Jiang
- Xiangya Medical College of Central South University, Changsha, China
| | - Qinqin Bai
- Department of Neurology, Shanxi Medical University, Taiyuan, China
| | - Gaiqing Wang
- Department of Neurology, Sanya Central Hospital (Hainan Third People's Hospital), Sanya, China
| |
Collapse
|
17
|
Kim J, Suh YH, Chang KA. Interleukin-17 induced by cumulative mild stress promoted depression-like behaviors in young adult mice. Mol Brain 2021; 14:11. [PMID: 33441182 PMCID: PMC7805143 DOI: 10.1186/s13041-020-00726-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/29/2020] [Indexed: 12/23/2022] Open
Abstract
The number of young adult patients with major depression, one of the most common mental disorders, is gradually increasing in modern society. Stressful experiences in early life are considered one of the risk factors for chronic depressive symptoms, along with an abnormal inflammatory response in later life. Although increased inflammatory activity has been identified in patients with depression, the cause of long-lasting depressive states is still unclear. To identify the effects of cumulative mild stress in brain development periods, we generated a young adult depression mouse model exposed to cumulative mild stress (CPMS; cumulative mild prenatal stress, mild maternal separation, and mild social defeat) to mimic early life adversities. CPMS mice exhibited more long-lasting anxiety and depression-like behaviors than groups exposed to single or double combinations of mild stress in young adult age. Using the molecular works, we found that inflammatory cytokines, especially interleukin (IL)-17, upregulated microglial activation in the hippocampus, amygdala, and prefrontal cortex of CPMS mice. In the brains of CPMS mice, we also identified changes in the T helper (Th)-17 cell population as well as differentiation. Finally, anti-IL-17 treatment rescued anxiety and depression-like behavior in CPMS mice. In conclusion, we found that cumulative mild stress promoted long-lasting depressive symptoms in CPMS mice through the upregulation of IL-17. We suggest that the CPMS model may be useful to study young adult depression and expect that IL-17 may be an important therapeutic target for depression in young adults.
Collapse
Affiliation(s)
- Jinho Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21936, Korea
| | - Yoo-Hun Suh
- Neuroscience Research Institute, Gachon University, Incheon, 21565, Korea
| | - Keun-A Chang
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21936, Korea.
- Neuroscience Research Institute, Gachon University, Incheon, 21565, Korea.
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, 21936, Korea.
| |
Collapse
|
18
|
Sasaki T, Tome S, Takei Y. Intraventricular IL-17A administration activates microglia and alters their localization in the mouse embryo cerebral cortex. Mol Brain 2020; 13:93. [PMID: 32546246 PMCID: PMC7298827 DOI: 10.1186/s13041-020-00635-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/09/2020] [Indexed: 12/20/2022] Open
Abstract
Viral infection during pregnancy has been suggested to increase the probability of autism spectrum disorder (ASD) in offspring via the phenomenon of maternal immune activation (MIA). This has been modeled in rodents. Maternal T helper 17 cells and the effector cytokine, interleukin 17A (IL-17A), play a central role in MIA-induced behavioral abnormalities and cortical dysgenesis, termed cortical patch. However, it is unclear how IL-17A acts on fetal brain cells to cause ASD pathologies. To assess the effect of IL-17A on cortical development, we directly administered IL-17A into the lateral ventricles of the fetal mouse brain. We analyzed injected brains focusing on microglia, which express IL-17A receptors. We found that IL-17A activated microglia and altered their localization in the cerebral cortex. Our data indicate that IL-17A activates cortical microglia, which leads to a cascade of ASD-related brain pathologies, including excessive phagocytosis of neural progenitor cells in the ventricular zone.
Collapse
Affiliation(s)
- Tetsuya Sasaki
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan. .,PhD Program of Neurosciences, Degree Program of Comprehensive Human Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Saki Tome
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Yosuke Takei
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan. .,PhD Program of Neurosciences, Degree Program of Comprehensive Human Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
| |
Collapse
|
19
|
Bai Q, Xue M, Yong VW. Microglia and macrophage phenotypes in intracerebral haemorrhage injury: therapeutic opportunities. Brain 2020; 143:1297-1314. [PMID: 31919518 DOI: 10.1093/brain/awz393] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/19/2019] [Accepted: 10/20/2019] [Indexed: 01/24/2023] Open
Abstract
Abstract
The prognosis of intracerebral haemorrhage continues to be devastating despite much research into this condition. A prominent feature of intracerebral haemorrhage is neuroinflammation, particularly the excessive representation of pro-inflammatory CNS-intrinsic microglia and monocyte-derived macrophages that infiltrate from the circulation. The pro-inflammatory microglia/macrophages produce injury-enhancing factors, including inflammatory cytokines, matrix metalloproteinases and reactive oxygen species. Conversely, the regulatory microglia/macrophages with potential reparative and anti-inflammatory roles are outcompeted in the early stages after intracerebral haemorrhage, and their beneficial roles appear to be overwhelmed by pro-inflammatory microglia/macrophages. In this review, we describe the activation of microglia/macrophages following intracerebral haemorrhage in animal models and clinical subjects, and consider their multiple mechanisms of cellular injury after haemorrhage. We review strategies and medications aimed at suppressing the pro-inflammatory activities of microglia/macrophages, and those directed at elevating the regulatory properties of these myeloid cells after intracerebral haemorrhage. We consider the translational potential of these medications from preclinical models to clinical use after intracerebral haemorrhage injury, and suggest that several approaches still lack the experimental support necessary for use in humans. Nonetheless, the preclinical data support the use of deactivator or inhibitor of pro-inflammatory microglia/macrophages, whilst enhancing the regulatory phenotype, as part of the therapeutic approach to improve the prognosis of intracerebral haemorrhage.
Collapse
Affiliation(s)
- Qian Bai
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, Henan, China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, Henan, China
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
20
|
Bai YY, Niu JZ. miR‑222 regulates brain injury and inflammation following intracerebral hemorrhage by targeting ITGB8. Mol Med Rep 2019; 21:1145-1153. [PMID: 31894320 PMCID: PMC7003054 DOI: 10.3892/mmr.2019.10903] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 07/30/2019] [Indexed: 12/11/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a disease associated with high mortality and morbidity. MicroRNAs (miRNAs) have been reported to be associated with the pathogenesis of numerous cerebrovascular diseases, including ICH. miR-222 has been revealed to play important roles in various physiological and pathological processes in cardiovascular diseases. However, its role in ICH remains largely unknown. The aim of the present study was to evaluate the potential effect of miR-222 on brain injury in ICH. The results revealed that the expression of miR-222 was significantly increased in ICH, and downregulation of miR-222 significantly reduced erythrocyte lysate-induced cell apoptosis by decreasing the levels of cleaved caspase-3, cleaved caspase-9 and Bax and increasing the level of Bcl-2. In addition, downregulation of miR-222 suppressed the inflammatory responses in erythrocyte lysate-induced microglia, and inhibited inflammation, brain water content and improved neurological functions in ICH mice. Mechanistically, integrin subunit β8 (ITGB8) was identified as a direct target of negative regulation by miR-222 in microglia cells, and up-regulation of ITGB8 led to the attenuation of inflammation and apoptosis. Collectively, the present findings indicated that miR-222 was a crucial regulator of inflammation via targeting of ITGB8, and represented a promising therapeutic strategy for ICH.
Collapse
Affiliation(s)
- Yan-Yan Bai
- Department of Neurology, The First Hospital of Yulin, Yulin, Shaanxi 719000, P.R. China
| | - Jun-Zhi Niu
- Department of Information, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| |
Collapse
|
21
|
Liu SP, Huang L, Flores J, Ding Y, Li P, Peng J, Zuo G, Zhang JH, Lu J, Tang JP. Secukinumab attenuates reactive astrogliosis via IL-17RA/(C/EBPβ)/SIRT1 pathway in a rat model of germinal matrix hemorrhage. CNS Neurosci Ther 2019; 25:1151-1161. [PMID: 31020769 PMCID: PMC6776744 DOI: 10.1111/cns.13144] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 12/28/2022] Open
Abstract
Aims Reactive astrogliosis plays a critical role in neurological deficits after germinal matrix hemorrhage (GMH). It has been reported that interleukin‐17A and IL‐17A receptor IL‐17RA/(C/EBPβ)/SIRT1 signaling pathway enhances reactive astrogliosis after brain injuries. We evaluated the effects of secukinumab on reactive astrogliosis in a rat pup model of GMH. Methods A total of 146 Sprague Dawley P7 rat pups were used. GMH was induced by intraparenchymal injection of collagenase. Secukinumab was administered intranasally 1 hour post‐GMH. C/EBPβ CRISPR or SIRT1 antagonist EX527 was administrated intracerebroventricularly (icv) 48 hours and 1 hour before GMH induction, respectively. Neurobehavior, Western blot, histology, and immunohistochemistry were used to assess treatment regiments in the short term and long term. Results The endogenous IL‐17A, IL‐17RA, C/EBPβ, and GFAP and proliferation marker CyclinD1 were increased, while SIRT1 expression was decreased after GMH. Secukinumab treatment improved neurological deficits, reduced ventriculomegaly, and increased cortical thickness. Additionally, treatment increased SIRT1 expression and lowered proliferation proteins PCNA and CyclinD1 as well as GFAP expression. C/EBPβ CRISPR activation plasmid and EX527 reversed the antireactive astrogliosis effects of secukinumab. Conclusion Secukinumab attenuated reactive astrogliosis and reduced neurological deficits after GMH, partly by regulating IL‐17RA/(C/EBPβ)/SIRT1 pathways. Secukinumab may provide a promising therapeutic strategy for GMH patients.
Collapse
Affiliation(s)
- Sheng-Peng Liu
- Department of Pediatrics, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China.,Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Lei Huang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California.,Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, California
| | - Jerry Flores
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Yan Ding
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Peng Li
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Jun Peng
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Gang Zuo
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - John H Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California.,Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, California.,Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Jun Lu
- Department of Pediatrics, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Ji-Ping Tang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| |
Collapse
|
22
|
Zhu H, Wang Z, Yu J, Yang X, He F, Liu Z, Che F, Chen X, Ren H, Hong M, Wang J. Role and mechanisms of cytokines in the secondary brain injury after intracerebral hemorrhage. Prog Neurobiol 2019; 178:101610. [PMID: 30923023 DOI: 10.1016/j.pneurobio.2019.03.003] [Citation(s) in RCA: 215] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 03/07/2019] [Accepted: 03/16/2019] [Indexed: 12/18/2022]
Abstract
Intracerebral hemorrhage (ICH) is a common and severe cerebrovascular disease that has high mortality. Few survivors achieve self-care. Currently, patients receive only symptomatic treatment for ICH and benefit poorly from this regimen. Inflammatory cytokines are important participants in secondary injury after ICH. Increases in proinflammatory cytokines may aggravate the tissue injury, whereas increases in anti-inflammatory cytokines might be protective in the ICH brain. Inflammatory cytokines have been studied as therapeutic targets in a variety of acute and chronic brain diseases; however, studies on ICH are limited. This review summarizes the roles and functions of various pro- and anti-inflammatory cytokines in secondary brain injury after ICH and discusses pathogenic mechanisms and emerging therapeutic strategies and directions for treatment of ICH.
Collapse
Affiliation(s)
- Huimin Zhu
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China
| | - Zhiqiang Wang
- Central laboratory, Linyi People's Hospital, Linyi, Shandong 276003, China
| | - Jixu Yu
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China; Central laboratory, Linyi People's Hospital, Linyi, Shandong 276003, China; Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Xiuli Yang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Feng He
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China
| | - Zhenchuan Liu
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China.
| | - Fengyuan Che
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China; Central laboratory, Linyi People's Hospital, Linyi, Shandong 276003, China.
| | - Xuemei Chen
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Honglei Ren
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael Hong
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
23
|
Chen C, Zhou F, Zeng L, Jiang Z, Hu Z. Methylene blue offers neuroprotection after intracerebral hemorrhage in rats through the PI3K/Akt/GSK3β signaling pathway. J Cell Physiol 2018; 234:5304-5318. [PMID: 30216439 DOI: 10.1002/jcp.27339] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 08/10/2018] [Indexed: 12/18/2022]
Abstract
Inflammation and apoptosis are two key factors contributing to secondary brain injury after intracerebral hemorrhage (ICH). In the present study, we explored the neuroprotective role of methylene blue (MB) in ICH rats and studied the potential mechanisms involved. Rats were subjected to local injection of collagenase IV in the striatum or sham surgery. We observed that MB treatment could exert a neuroprotective effect on ICH by promoting neurological scores, decreasing the brain water content, alleviating brain-blood barrier disruption, and improving the histological damages in the perihematomal areas. Furthermore, we demonstrated that the various mechanisms underlying MB's neuroprotective effects linked to inhibited apoptosis and inhibited neuroinflammation. In addition, wortmannin, a selective inhibitor of phosphoinositide 3-kinase (PI3K), could reverse the antiapoptotic and anti-inflammatory effects of MB, which suggested that the PI3K-Akt pathway played an important role. In conclusion, these data suggested that MB could inhibit apoptosis and ameliorate neuroinflammation after ICH, and its neuroprotective effects might be exerted via the activation of the PI3K/Akt/GSK3β pathway.
Collapse
Affiliation(s)
- Chunli Chen
- Department of Neurology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Fangfang Zhou
- Department of Neurology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Liuwang Zeng
- Department of Neurology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zheng Jiang
- Department of Neurology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhiping Hu
- Department of Neurology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
24
|
Weinstock LD, Furness AM, Herron SS, Smith SS, Sankar SB, DeRosa SG, Gao D, Mepyans ME, Scotto Rosato A, Medina DL, Vardi A, Ferreira NS, Cho SM, Futerman AH, Slaugenhaupt SA, Wood LB, Grishchuk Y. Fingolimod phosphate inhibits astrocyte inflammatory activity in mucolipidosis IV. Hum Mol Genet 2018; 27:2725-2738. [PMID: 29771310 PMCID: PMC6915831 DOI: 10.1093/hmg/ddy182] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/09/2018] [Accepted: 05/08/2018] [Indexed: 12/25/2022] Open
Abstract
Mucolipidosis IV (MLIV) is an orphan neurodevelopmental disease that causes severe neurologic dysfunction and loss of vision. Currently there is no therapy for MLIV. It is caused by loss of function of the lysosomal channel mucolipin-1, also known as TRPML1. Knockout of the Mcoln1 gene in a mouse model mirrors clinical and neuropathologic signs in humans. Using this model, we previously observed robust activation of microglia and astrocytes in early symptomatic stages of disease. Here we investigate the consequence of mucolipin-1 loss on astrocyte inflammatory activation in vivo and in vitro and apply a pharmacologic approach to restore Mcoln1-/- astrocyte homeostasis using a clinically approved immunomodulator, fingolimod. We found that Mcoln1-/- mice over-express numerous pro-inflammatory cytokines, some of which were also over-expressed in astrocyte cultures. Changes in the cytokine profile in Mcoln1-/- astrocytes are concomitant with changes in phospho-protein signaling, including activation of PI3K/Akt and MAPK pathways. Fingolimod promotes cytokine homeostasis, down-regulates signaling within the PI3K/Akt and MAPK pathways and restores the lysosomal compartment in Mcoln1-/- astrocytes. These data suggest that fingolimod is a promising candidate for preclinical evaluation in our MLIV mouse model, which, in case of success, can be rapidly translated into clinical trial.
Collapse
Affiliation(s)
- Laura D Weinstock
- George W. Woodruff School of Mechanical Engineering, Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory, Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Dr., Atlanta, GA, USA
| | - Amanda M Furness
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA, USA
| | - Shawn S Herron
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA, USA
| | - Sierra S Smith
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA, USA
| | - Sitara B Sankar
- George W. Woodruff School of Mechanical Engineering, Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory, Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Dr., Atlanta, GA, USA
| | - Samantha G DeRosa
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA, USA
| | - Dadi Gao
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA, USA
| | - Molly E Mepyans
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA, USA
| | - Anna Scotto Rosato
- Telethon Institute of Genetics and Medicine (TIGEM), via Campi Flegrei 34, Pozzuoli (NA), Italy
| | - Diego L Medina
- Telethon Institute of Genetics and Medicine (TIGEM), via Campi Flegrei 34, Pozzuoli (NA), Italy
| | - Ayelet Vardi
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Natalia S Ferreira
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, Zurich, Switzerland
| | - Soo Min Cho
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Anthony H Futerman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Susan A Slaugenhaupt
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA, USA
| | - Levi B Wood
- George W. Woodruff School of Mechanical Engineering, Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory, Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Dr., Atlanta, GA, USA
| | - Yulia Grishchuk
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA, USA
| |
Collapse
|
25
|
Quercetin attenuates AZT-induced neuroinflammation in the CNS. Sci Rep 2018; 8:6194. [PMID: 29670213 PMCID: PMC5906611 DOI: 10.1038/s41598-018-24618-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 04/04/2018] [Indexed: 02/06/2023] Open
Abstract
Highly active anti-retroviral therapy (HAART) is very effective in suppressing HIV-1 replication in patients. However, continuous HAART is required to prevent viral rebound, which may have detrimental effects in various tissues, including persistent neuroinflammation in the central nervous system (CNS). Here, we show that quercetin (3,5,7,3’,4’-pentahydroxy flavones), a natural antioxidant used in Chinese traditional medicines, suppresses the neuroinflammation that is induced by chronic exposure to Zidovudine (azidothymidine, AZT), a nucleoside reverse transcriptase inhibitor (NRTI) that is commonly part of HAART regimens. We found that the up-regulation of pro-inflammatory cytokines and microglial and astrocytic markers induced by AZT (100 mg/kg/day; 8 days) was significantly inhibited by co-administration of quercetin (50 mg/kg/day) in the mouse cortex, hippocampus and spinal cord. We further showed that quercetin attenuated AZT-induced up-regulation of Wnt5a, a key regulator of neuroinflammation. These results suggest that quercetin has an inhibitory effect on AZT-induced neuroinflammation in the CNS, and Wnt5a signaling may play an important role in this process. Our results may further our understanding of the mechanisms of HAART-related neurotoxicity and help in the development of effective adjuvant therapy.
Collapse
|
26
|
Intracranial IL-17A overexpression decreases cerebral amyloid angiopathy by upregulation of ABCA1 in an animal model of Alzheimer's disease. Brain Behav Immun 2017; 65:262-273. [PMID: 28526436 PMCID: PMC5537015 DOI: 10.1016/j.bbi.2017.05.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/15/2017] [Accepted: 05/16/2017] [Indexed: 01/18/2023] Open
Abstract
Neuroinflammation is a pervasive feature of Alzheimer's disease (AD) and characterized by activated microglia, increased proinflammatory cytokines and/or infiltrating immune cells. T helper 17 (Th17) cells are found in AD brain parenchyma and interleukin-17A (IL-17A) is identified around deposits of aggregated amyloid β protein (Aβ). However, the role of IL-17A in AD pathogenesis remains elusive. We overexpressed IL-17A in an AD mouse model via recombinant adeno-associated virus serotype 5 (rAAV5)-mediated intracranial gene delivery. AD model mice subjected to injection of a vehicle (PBS) or rAAV5 carrying the lacZ gene served as controls. IL-17A did not exacerbate neuroinflammation in IL-17A-overexpressing mice. We found that IL-17A overexpression markedly improved glucose metabolism, decreased soluble Aβ levels in the hippocampus and cerebrospinal fluid, drastically reduced cerebral amyloid angiopathy, and modestly but significantly improved anxiety and learning deficits. Moreover, the ATP-binding cassette subfamily A member 1 (ABCA1), which can transport Aβ from the brain into the blood circulation, significantly increased in IL-17A-overexpressing mice. In vitro treatment of brain endothelial bEnd.3 cells with IL-17A induced a dose-dependent increase in protein expression of ABCA1 through ERK activation. Our study suggests that IL-17A may decrease Aβ levels in the brain by upregulating ABCA1 in blood-brain barrier endothelial cells.
Collapse
|
27
|
MiR-124 contributes to M2 polarization of microglia and confers brain inflammatory protection via the C/EBP-α pathway in intracerebral hemorrhage. Immunol Lett 2016; 182:1-11. [PMID: 28025043 DOI: 10.1016/j.imlet.2016.12.003] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/16/2016] [Accepted: 12/22/2016] [Indexed: 11/22/2022]
Abstract
Microglia mediated inflammation contributes to intracerebral hemorrhage (ICH) induced secondary injury. Activated microglia has dual functions as pro-inflammatory (M1) and anti-inflammatory (M2) factors in brain injury and repair. MiR-124 is a potent anti-inflammatory agent which affects microglia after brain injury. However, the potential of modulating the M1/M2 polarization of microglia after ICH has not been reported. In this experiment, we detected the effect of miR-124 on the M1/M2 polarization state. In addition, the ability miR-124 to subsequently impacted neurological deficit and cerebral water content of ICH mice were studied. Furthermore, the relationship between miR-124 and C/EBP-α target was detected. We found that miR-124 significantly increased in M2-polarized microglia. Transduction of miR-124 mimics decreased proinflammatory cytokine levels. A coculture model of microglia and neuron indicated that M2-polarized microglia protected neuron damage. Furthermore, miR-124 banded to the 3-untranslated region of C/EBP-α and downregulated its protein levels. In vivo, infusion of miR-124 decreased brain levels of C/EBP-α and significantly reduced brain injury in ICH mice. Thus, miR-124 ameliorated ICH-induced inflammatory injury by modulating microglia polarization toward the M2 phenotype via C/EBP-α. MiR-124 regulatory mechanisms also might represent new therapeutic strategy in ICH.
Collapse
|