1
|
Liang J, Yang F, Li Z, Li Q. Epigenetic regulation of the inflammatory response in stroke. Neural Regen Res 2025; 20:3045-3062. [PMID: 39589183 PMCID: PMC11881735 DOI: 10.4103/nrr.nrr-d-24-00672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/15/2024] [Accepted: 09/20/2024] [Indexed: 11/27/2024] Open
Abstract
Stroke is classified as ischemic or hemorrhagic, and there are few effective treatments for either type. Immunologic mechanisms play a critical role in secondary brain injury following a stroke, which manifests as cytokine release, blood-brain barrier disruption, neuronal cell death, and ultimately behavioral impairment. Suppressing the inflammatory response has been shown to mitigate this cascade of events in experimental stroke models. However, in clinical trials of anti-inflammatory agents, long-term immunosuppression has not demonstrated significant clinical benefits for patients. This may be attributable to the dichotomous roles of inflammation in both tissue injury and repair, as well as the complex pathophysiologic inflammatory processes in stroke. Inhibiting acute harmful inflammatory responses or inducing a phenotypic shift from a pro-inflammatory to an anti-inflammatory state at specific time points after a stroke are alternative and promising therapeutic strategies. Identifying agents that can modulate inflammation requires a detailed understanding of the inflammatory processes of stroke. Furthermore, epigenetic reprogramming plays a crucial role in modulating post-stroke inflammation and can potentially be exploited for stroke management. In this review, we summarize current findings on the epigenetic regulation of the inflammatory response in stroke, focusing on key signaling pathways including nuclear factor-kappa B, Janus kinase/signal transducer and activator of transcription, and mitogen-activated protein kinase as well as inflammasome activation. We also discuss promising molecular targets for stroke treatment. The evidence to date indicates that therapeutic targeting of the epigenetic regulation of inflammation can shift the balance from inflammation-induced tissue injury to repair following stroke, leading to improved post-stroke outcomes.
Collapse
Affiliation(s)
- Jingyi Liang
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Fei Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Zixiao Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Center for Healthcare Quality Management in Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Engineering Research Center of Digital Healthcare for Neurological Diseases, Beijing, China
| | - Qian Li
- Laboratory for Clinical Medicine, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Apostolo D, Ferreira LL, D'Onghia D, Vincenzi F, Vercellino N, Perazzi M, Pirisi M, Cantello R, Minisini R, Mazzini L, Bellan M, De Marchi F. Lower Circulating Gas6 Levels Are Associated with Bulbar Phenotype and Faster Disease Progression in Amyotrophic Lateral Sclerosis Patients. Mol Neurobiol 2025; 62:6273-6282. [PMID: 39762711 DOI: 10.1007/s12035-024-04671-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/16/2024] [Indexed: 03/29/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder that primarily affects the motor neurons in the brain and spinal cord. While the exact cause of ALS is not fully understood, a combination of genetic and environmental factors is believed to contribute to its development. Growth arrest-specific 6 (Gas6), a vitamin K-dependent protein, has been recognized to enhance oligodendrocytes and neurons' survival and is associated with different kinds of (neuro)inflammatory conditions. Therefore, we aimed to determine a possible implication of Gas6 in ALS phenotype and progression by evaluating the value of circulating Gas6 and its soluble receptors (sAxl, sMer, sTyro-3) in ALS patients. We conducted a prospective observational study including 65 ALS patients and measured the circulating serum levels of Gas6, sAxl, sMer, soluble Tyro-3 (sTyro-3), and neurofilaments (NfLs). In our ALS cohort, lower serum levels of Gas6 and concomitantly higher levels of NfLs were associated with a more aggressive disease, expressed with bulbar phenotype (p-value for Gas6 = 0.03) and faster progression (p-value for Gas6 = 0.03). Also, serum Gas6 was able to distinguish (area under the curve, cut-off 13.70 ng/mL, sensitivity 69.57%, specificity 72.72%) between fast and slow progressors. Due to its neuroprotective properties, our data suggest that Gas6 could be an intriguing biomarker in ALS patients.
Collapse
Affiliation(s)
- Daria Apostolo
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Luciana L Ferreira
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Davide D'Onghia
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Federica Vincenzi
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Nicole Vercellino
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Mattia Perazzi
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
- AOU Maggiore Della Carità, Novara, Italy
| | - Mario Pirisi
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
- AOU Maggiore Della Carità, Novara, Italy
| | - Roberto Cantello
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
- AOU Maggiore Della Carità, Novara, Italy
| | - Rosalba Minisini
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Letizia Mazzini
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
- AOU Maggiore Della Carità, Novara, Italy
| | - Mattia Bellan
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.
- AOU Maggiore Della Carità, Novara, Italy.
| | - Fabiola De Marchi
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
- AOU Maggiore Della Carità, Novara, Italy
| |
Collapse
|
3
|
Xu L, Li C, Zhao C, Wang Z, Zhang Z, Shu X, Cao X, Xia S, Bao X, Shao P, Xu Y. Shionone protects cerebral ischemic injury through alleviating microglia-mediated neuroinflammation. Chin J Nat Med 2025; 23:471-479. [PMID: 40274349 DOI: 10.1016/s1875-5364(25)60812-0] [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: 11/11/2024] [Revised: 03/15/2025] [Accepted: 04/03/2025] [Indexed: 04/26/2025]
Abstract
Microglia, the resident immune cells in the central nervous system (CNS), rapidly transition from a resting to an active state in the acute phase of ischemic brain injury. This active state mediates a pro-inflammatory response that can exacerbate the injury. Targeting the pro-inflammatory response of microglia in the semi-dark band during this acute phase may effectively reduce brain injury. Shionone (SH), an active ingredient extracted from the dried roots and rhizomes of the genus Aster (Asteraceae), has been reported to regulate the inflammatory response of macrophages in sepsis-induced acute lung injury. However, its function in post-stroke neuroinflammation, particularly microglia-mediated neuroinflammation, remains uninvestigated. This study found that SH significantly inhibited lipopolysaccharide (LPS)-induced elevation of inflammatory cytokines, including interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and inducible nitric oxide synthase (iNOS), in microglia in vitro. Furthermore, the results demonstrated that SH alleviated infarct volume and improved behavioral performance in middle cerebral artery occlusion (MCAO) mice, which may be attributed to the inhibition of the microglial inflammatory response induced by SH treatment. Mechanistically, SH potently inhibited the phosphorylation of serine-threonine protein kinase B (AKT), mammalian target of rapamycin (mTOR), and signal transducer and activator of transcription 3 (STAT3). These findings suggest that SH may be a potential therapeutic agent for relieving ischemic stroke (IS) by alleviating microglia-associated neuroinflammation.
Collapse
Affiliation(s)
- Lushan Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Chenggang Li
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau 999078, China
| | - ChenChen Zhao
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing 210008, China
| | - Zibu Wang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing 210008, China
| | - Zhi Zhang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing 210008, China
| | - Xin Shu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing 210008, China
| | - Xiang Cao
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing 210008, China; Jiangsu Provincial Key Discipline of Neurology, Nanjing 210008, China
| | - Shengnan Xia
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing 210008, China
| | - Xinyu Bao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing 210008, China
| | - Pengfei Shao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing 210008, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China; School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau 999078, China; Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing 210008, China; Jiangsu Provincial Key Discipline of Neurology, Nanjing 210008, China.
| |
Collapse
|
4
|
Zhang W, Sun M, Liu N, Li X, Sun J, Wang M. Curcumin ameliorates astrocyte inflammation through AXL in cuprizone-induced mice. Toxicol Appl Pharmacol 2025; 494:117170. [PMID: 39586379 DOI: 10.1016/j.taap.2024.117170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/30/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Curcumin has gained global attention owning to its anti-inflammatory, antioxidant, anticancer, and antimicrobial activities. Curcumin has recently been shown to have well-documented effects on neuroinflammation in multiple sclerosis (MS). Astrocytes, the most widely distributed glial cells in the brain, have a significant influence on the regulation of neuroinflammation in MS. However, it is unknown how curcumin exerts neuroprotective effects in astrocytes. To elucidate the mechanism underlying the effects of curcumin on astrocytes, we explored the effect of curcumin on cuprizone (CPZ)-induced mice in vivo and on primary astrocytes in vitro. In this study, we observed that curcumin significantly ameliorated myelin loss and reduced astrocyte activation in the corpus callosum (CC) region in mice induced with CPZ, and in primary astrocytes stimulated with lipopolysaccharide (LPS). Meanwhile, our research indicated that curcumin may exert neuroprotective effects in CPZ-induced mice by downregulating astrocyte-mediated inflammation by AXL. This study provides new insights into possible targeted therapies for MS.
Collapse
Affiliation(s)
- Wenjing Zhang
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, China
| | - Mengjiao Sun
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, China
| | - Ning Liu
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, China
| | - Xiaoling Li
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, China
| | - Jing Sun
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, China
| | - Manxia Wang
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, China.
| |
Collapse
|
5
|
Zhang B, Zou Y, Yuan Z, Jiang K, Zhang Z, Chen S, Zhou X, Wu Q, Zhang X. Efferocytosis: the resolution of inflammation in cardiovascular and cerebrovascular disease. Front Immunol 2024; 15:1485222. [PMID: 39660125 PMCID: PMC11628373 DOI: 10.3389/fimmu.2024.1485222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Cardiovascular and cerebrovascular diseases have surpassed cancer as significant global health challenges, which mainly include atherosclerosis, myocardial infarction, hemorrhagic stroke and ischemia stroke. The inflammatory response immediately following these diseases profoundly impacts patient prognosis and recovery. Efficient resolution of inflammation is crucial not only for halting the inflammatory process but also for restoring tissue homeostasis. Efferocytosis, the phagocytic clearance of dying cells by phagocytes, especially microglia and macrophages, plays a critical role in this resolution process. Upon tissue injury, phagocytes are recruited to the site of damage where they engulf and clear dying cells through efferocytosis. Efferocytosis suppresses the secretion of pro-inflammatory cytokines, stimulates the production of anti-inflammatory cytokines, modulates the phenotype of microglia and macrophages, accelerates the resolution of inflammation, and promotes tissue repair. It involves three main stages: recognition, engulfment, and degradation of dying cells. Optimal removal of apoptotic cargo by phagocytes requires finely tuned machinery and associated modifications. Key molecules in efferocytosis, such as 'Find-me signals', 'Eat-me signals', and 'Don't eat-me signals', have been shown to enhance efferocytosis following cardiovascular and cerebrovascular diseases. Moreover, various additional molecules, pathways, and mitochondrial metabolic processes have been identified to enhance prognosis and outcomes via efferocytosis in diverse experimental models. Impaired efferocytosis can lead to inflammation-associated pathologies and prolonged recovery periods. Therefore, this review consolidates current understanding of efferocytosis mechanisms and its application in cardiovascular and cerebrovascular diseases, proposing future research directions.
Collapse
Affiliation(s)
- Bingtao Zhang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yan Zou
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zixuan Yuan
- Department of Neurosurgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Kun Jiang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhaoxiang Zhang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shujuan Chen
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiaoming Zhou
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qi Wu
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xin Zhang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
6
|
Liao HX, Mao X, Wang L, Wang N, Ocansey DKW, Wang B, Mao F. The role of mesenchymal stem cells in attenuating inflammatory bowel disease through ubiquitination. Front Immunol 2024; 15:1423069. [PMID: 39185411 PMCID: PMC11341407 DOI: 10.3389/fimmu.2024.1423069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024] Open
Abstract
Inflammatory bowel disease (IBD), a condition of the digestive tract and one of the autoimmune diseases, is becoming a disease of significant global public health concern and substantial clinical burden. Various signaling pathways have been documented to modulate IBD, but the exact activation and regulatory mechanisms have not been fully clarified; thus, a need for constant exploration of the molecules and pathways that play key roles in the development of IBD. In recent years, several protein post-translational modification pathways, such as ubiquitination, phosphorylation, methylation, acetylation, and glycolysis, have been implicated in IBD. An aberrant ubiquitination in IBD is often associated with dysregulated immune responses and inflammation. Mesenchymal stem cells (MSCs) play a crucial role in regulating ubiquitination modifications through the ubiquitin-proteasome system, a cellular machinery responsible for protein degradation. Specifically, MSCs have been shown to influence the ubiquitination of key signaling molecules involved in inflammatory pathways. This paper reviews the recent research progress in MSC-regulated ubiquitination in IBD, highlighting their therapeutic potential in treating IBD and offering a promising avenue for developing targeted interventions to modulate the immune system and alleviate inflammatory conditions.
Collapse
Affiliation(s)
- Hong Xi Liao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu, China
| | - Xiaojun Mao
- The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, Jiangsu, China
| | - Lan Wang
- Department of Laboratory Medicine, Danyang Blood Station, Zhenjiang, Jiangsu, China
| | - Naijian Wang
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu, China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Bo Wang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu, China
| |
Collapse
|
7
|
Yuan K, Wu Q, Yao Y, Shao J, Zhu S, Yang J, Sun Q, Zhao J, Xu J, Wu P, Li Y, Shi H. Deacetylase SIRT2 Inhibition Promotes Microglial M2 Polarization Through Axl/PI3K/AKT to Alleviate White Matter Injury After Subarachnoid Hemorrhage. Transl Stroke Res 2024:10.1007/s12975-024-01282-5. [PMID: 39103659 DOI: 10.1007/s12975-024-01282-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/01/2024] [Accepted: 07/22/2024] [Indexed: 08/07/2024]
Abstract
White matter injury (WMI) subsequent to subarachnoid hemorrhage (SAH) frequently leads to an unfavorable patient prognosis. Previous studies have indicated that microglial M1 polarization following SAH results in the accumulation of amyloid precursor protein (APP) and degradation of myelin basic protein (MBP), thereby catalyzing the exacerbation of WMI. Consequently, transitioning microglial polarization towards the M2 phenotype (neuroprotective state) represents a potential therapeutic approach for reversing WMI. The SIRT2 gene is pivotal in neurological disorders such as neurodegeneration and ischemic stroke. However, its function and underlying mechanisms in SAH, particularly how it influences microglial function to ameliorate WMI, remain unclear. Our investigations revealed that in post-SAH, there was a temporal increase in SIRT2 expression, predominantly in the cerebral corpus callosum area, with notable colocalization with microglia. However, following the administration of the SIRT2 inhibitor AK-7, a shift in microglial polarization towards the M2 phenotype and an improvement in both short-term and long-term neuronal functions in rats were observed. Mechanistically, CO-IP experiments confirmed that SIRT2 can interact with the receptor tyrosine kinase Axl within the TAM receptor family and act as a deacetylase to regulate the deacetylation of Axl. Concurrently, the inhibition of SIRT2 by AK-7 can lead to increased expression of Axl and activation of the anti-inflammatory pathway PI3K/Akt signaling pathway, which regulates microglial M2 polarization and consequently reduces WMI. However, when Axl expression was inhibited by the injection of the shAxl virus into the lateral ventricles, the downstream signaling pathways were significantly suppressed. Rescue experiments also confirmed that the neuroprotective effects of AK-7 can be reversed by PI3K inhibitors. These data suggest that SIRT2 influences WMI by affecting microglial polarization through the Axl/PI3K/AKT pathway, and that AK-7 could serve as an effective therapeutic drug for improving neurological functions in SAH patients.
Collapse
Affiliation(s)
- Kaikun Yuan
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Qiaowei Wu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Yanting Yao
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- Department of Neurosurgery, Beidahuang Group General Hospital, Harbin, 150001, People's Republic of China
| | - Jiang Shao
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Shiyi Zhu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Jinshuo Yang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Qi Sun
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Junjie Zhao
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Jiayi Xu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Pei Wu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Yuchen Li
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Huaizhang Shi
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China.
| |
Collapse
|
8
|
Guo D, Hu L, Xie P, Sun P, Yu W. Seipin is involved in oxygen-glucose deprivation/reoxygenation induced neuroinflammation by regulating the TLR3/TRAF3/NF-κB pathway. Int Immunopharmacol 2024; 134:112182. [PMID: 38703568 DOI: 10.1016/j.intimp.2024.112182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 04/25/2024] [Accepted: 04/27/2024] [Indexed: 05/06/2024]
Abstract
Seipin plays a crucial role in lipid metabolism and is involved in neurological disorders. However, the function and mechanism of action of seipin in acute ischemic stroke have not yet been elucidated. Here, we aimed to investigate the effect of seipin on neuroinflammation induced by oxygen-glucose deprivation/reoxygenation (OGD/R) and further explore the molecular mechanism by functional experiments. Our results revealed a significant decrease in seipin mRNA levels, accompanied by enhanced expression of TNF-α in patients with AIS, and a significant negative correlation between seipin and TNF-α was observed. Additionally, there was a negative correlation between seipin levels and the National Institutes of Health Stroke Scale (NIHSS) score. Furthermore, seipin levels were also decreased in middle cerebral artery occlusion/reperfusion (MCAO/R) mice and OGD/R-treated BV2 cells. RNA sequencing analysis showed that seipin knockdown altered the Toll-like receptor 3 (TLR3) signaling pathway. It was further confirmed in vitro that seipin knockdown caused significantly increased secretion of inflammatory factors including TNF-α, interleukin (IL)-1β, and interferon (IFN)-β. Meanwhile, seipin knockdown activated the Tlr3 signal pathway while this effect could be reversed by Tlr3 inhibitor in OGD/R treated BV2 cells. Furthermore, neuroinflammation induced by OGD/R was significantly reduced by seipin overexpression. Overall, our study demonstrate that seipin deficiency aggravates neuroinflammation by activating the TLR3/TRAF3/NF-κB signaling pathway after OGD/R stimuli, and suggest that seipin may be a potential therapeutic target for AIS.
Collapse
Affiliation(s)
- Dongfen Guo
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, School of Basic Medical Science, Guizhou Medical University, 09 Beijing Road, Guiyang 550004, Guizhou, China
| | - Lele Hu
- Department of Neurology, The Second People's Hospital of Guiyang, Guiyang 550023, Guizhou, China
| | - Peng Xie
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, School of Basic Medical Science, Guizhou Medical University, 09 Beijing Road, Guiyang 550004, Guizhou, China
| | - Ping Sun
- Department of Neurology, The Second People's Hospital of Guiyang, Guiyang 550023, Guizhou, China.
| | - Wenfeng Yu
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, School of Basic Medical Science, Guizhou Medical University, 09 Beijing Road, Guiyang 550004, Guizhou, China; Key Laboratory of Human Brain Bank for Functions and Diseases of Department of Education of Guizhou Province, Guizhou Medical University, Guiyang 550025, Guizhou, China.
| |
Collapse
|
9
|
Islam R, Choudhary H, Rajan R, Vrionis F, Hanafy KA. An overview on microglial origin, distribution, and phenotype in Alzheimer's disease. J Cell Physiol 2024; 239:e30829. [PMID: 35822939 PMCID: PMC9837313 DOI: 10.1002/jcp.30829] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/12/2022] [Accepted: 07/04/2022] [Indexed: 01/17/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease that is responsible for about one-third of dementia cases worldwide. It is believed that AD is initiated with the deposition of Ab plaques in the brain. Genetic studies have shown that a high number of AD risk genes are expressed by microglia, the resident macrophages of brain. Common mode of action by microglia cells is neuroinflammation and phagocytosis. Moreover, it has been discovered that inflammatory marker levels are increased in AD patients. Recent studies advocate that neuroinflammation plays a major role in AD progression. Microglia have different activation profiles depending on the region of brain and stimuli. In different activation, profile microglia can generate either pro-inflammatory or anti-inflammatory responses. Microglia defend brain cells from pathogens and respond to injuries; also, microglia can lead to neuronal death along the way. In this review, we will bring the different roles played by microglia and microglia-related genes in the progression of AD.
Collapse
Affiliation(s)
- Rezwanul Islam
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL
| | - Hadi Choudhary
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL
| | - Robin Rajan
- Marcus Neuroscience Institute, Boca Raton Medical Center, Boca Raton, FL
| | - Frank Vrionis
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL
- Marcus Neuroscience Institute, Boca Raton Medical Center, Boca Raton, FL
| | - Khalid A. Hanafy
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL
- Marcus Neuroscience Institute, Boca Raton Medical Center, Boca Raton, FL
| |
Collapse
|
10
|
Liang Y, Chen L, Huang J, Lan Z, Xia S, Yang H, Bao X, Yu X, Fan Y, Xu Y, Zhu X, Jin J. Neuroprotective effects of Aucubin against cerebral ischemia-reperfusion injury. Int Immunopharmacol 2024; 129:111648. [PMID: 38335656 DOI: 10.1016/j.intimp.2024.111648] [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: 12/01/2023] [Revised: 01/20/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024]
Abstract
AIMS To study the role of Aucubin (AU) in cerebral ischemia-reperfusion injury and investigate the potential mechanisms. METHODS For the in vitro experiment, primary microglia were cultured and stimulated by Lipopolysaccharides (LPS) and treated with AU. Male C57/BL6J mice were used and middle cerebral artery occlusion (MCAO) model was performed to induce cerebral ischemia-reperfusion injury. For the short-term effects, mice administrated with AU (40 mg/kg) for 3 days after MCAO were evaluated for the infarct volume and neurological deficits. The neuroinflammatory factors and microglia activation were determined by Real-time PCR, western blot and immunofluorescence staining. For the long-term effects, MCAO mice were injected daily with AU (5 mg/kg or 10 mg/kg) for 28 days. Behavior tests were used to assess the neurological deficits of MCAO mice, and white matter integrity was determined by myelin basic protein (MBP) staining and black-gold staining. RESULTS AU suppressed LPS-induced activation of microglia and pro-inflammatory cytokines release, and downregulated the NF-κB and MAPK pathways in primary microglia. In addition, AU attenuated ischemic injury and inhibited the neuro-inflammatory response in MCAO mice. Moreover, AU induced prolonged improvements in sensorimotor function and memory function following MCAO, and preserved white matter integrity in the long-term experiments. CONCLUSIONS AU protected against ischemic injury, which might be correlated with the downregulation of NF-κB and MAPK signaling pathways. Furthermore, AU alleviated cognitive impairment after stroke and restored white matter integrity. Our data indicated that AU might be a potential compound for the treatment of stroke and post-stroke cognitive impairment.
Collapse
Affiliation(s)
- Ying Liang
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Liqiu Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Jing Huang
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Zhen Lan
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China
| | - Shengnan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing 210008, China; Jiangsu Provincial Key Discipline of Neurology, Nanjing 210008, China; Nanjing Neurology Medical Center, Nanjing 210008, China
| | - Haiyan Yang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing 210008, China; Jiangsu Provincial Key Discipline of Neurology, Nanjing 210008, China; Nanjing Neurology Medical Center, Nanjing 210008, China
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing 210008, China; Jiangsu Provincial Key Discipline of Neurology, Nanjing 210008, China; Nanjing Neurology Medical Center, Nanjing 210008, China
| | - Xi Yu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yingao Fan
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing 210008, China; Jiangsu Provincial Key Discipline of Neurology, Nanjing 210008, China; Nanjing Neurology Medical Center, Nanjing 210008, China
| | - Xiaolei Zhu
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing 210008, China; Jiangsu Provincial Key Discipline of Neurology, Nanjing 210008, China; Nanjing Neurology Medical Center, Nanjing 210008, China.
| | - Jiali Jin
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing 210008, China; Jiangsu Provincial Key Discipline of Neurology, Nanjing 210008, China; Nanjing Neurology Medical Center, Nanjing 210008, China.
| |
Collapse
|
11
|
Yu J, Li J, Matei N, Wang W, Tang L, Pang J, Li X, Fang L, Tang J, Zhang JH, Yan M. Intranasal administration of recombinant prosaposin attenuates neuronal apoptosis through GPR37/PI3K/Akt/ASK1 pathway in MCAO rats. Exp Neurol 2024; 373:114656. [PMID: 38114054 PMCID: PMC10922973 DOI: 10.1016/j.expneurol.2023.114656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/19/2023] [Accepted: 12/12/2023] [Indexed: 12/21/2023]
Abstract
Studies have reported that Prosaposin (PSAP) is neuroprotective in cerebrovascular diseases. We hypothesized that PSAP would reduce infarct volume by attenuating neuronal apoptosis and promoting cell survival through G protein-coupled receptor 37(GPR37)/PI3K/Akt/ASK1 pathway in middle cerebral artery occlusion (MCAO) rats. Two hundred and thirty-five male and eighteen female Sprague-Dawley rats were used. Recombinant human PSAP (rPSAP) was administered intranasally 1 h (h) after reperfusion. PSAP small interfering ribonucleic acid (siRNA), GPR37 siRNA, and PI3K specific inhibitor LY294002 were administered intracerebroventricularly 48 h before MCAO. Infarct volume, neurological score, immunofluorescence staining, Western blot, Fluoro-Jade C (FJC) and TUNEL staining were examined. The expression of endogenous PSAP and GPR37 were increased after MCAO. Intranasal administration of rPSAP reduced brain infarction, neuronal apoptosis, and improved both short- and long-term neurological function. Knockdown of endogenous PSAP aggravated neurological deficits. Treatment with exogenous rPSAP increased PI3K expression, Akt and ASK1 phosphorylation, and Bcl-2 expression; phosphorylated-JNK and Bax levels were reduced along with the number of FJC and TUNEL positive neurons. GPR37 siRNA and LY294002 abolished the anti-apoptotic effect of rPSAP at 24 h after MCAO. In conclusion, rPSAP attenuated neuronal apoptosis and improved neurological function through GPR37/PI3K/Akt/ASK1 pathway after MCAO in rats. Therefore, further exploration of PSAP as a potential treatment option in ischemic stroke is warranted.
Collapse
Affiliation(s)
- Jing Yu
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Jinlan Li
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Nathanael Matei
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA; Department of Ophthalmology, University of Southern California, Los Angeles, CA 90007, USA
| | - Wenna Wang
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Lihui Tang
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Jinwei Pang
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Xue Li
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Lili Fang
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiping Tang
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - John H Zhang
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA.
| | - Min Yan
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
12
|
Sun D, Ma J, Du L, Liu Q, Yue H, Peng C, Chen H, Wang G, Liu X, Shen Y. Fluid shear stress induced-endothelial phenotypic transition contributes to cerebral ischemia-reperfusion injury and repair. APL Bioeng 2024; 8:016110. [PMID: 38414635 PMCID: PMC10898918 DOI: 10.1063/5.0174825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 01/30/2024] [Indexed: 02/29/2024] Open
Abstract
Long-term ischemia leads to insufficient cerebral microvascular perfusion and dysfunction. Reperfusion restores physiological fluid shear stress (FSS) but leads to serious injury. The mechanism underlying FSS-induced endothelial injury in ischemia-reperfusion injury (IRI) remains poorly understood. In this study, a rat model of middle cerebral artery occlusion was constructed to explore cerebrovascular endothelial function and inflammation in vivo. Additionally, the rat brain microvascular endothelial cells (rBMECs) were exposed to a laminar FSS of 0.5 dyn/cm2 for 6 h and subsequently restored to physiological fluid shear stress level (2 dyn/cm2) for 2 and 12 h, respectively. We found that reperfusion induced endothelial-to-mesenchymal transition (EndMT) in endothelial cells, leading to serious blood-brain barrier dysfunction and endothelial inflammation, accompanied by the nuclear accumulation of Yes-associated protein (YAP). During the later stage of reperfusion, cerebral endothelium was restored to the endothelial phenotype with a distinct change in mesenchymal-to-endothelial transition (MEndT), while YAP was translocated and phosphorylated in the cytoplasm. Knockdown of YAP or inhibition of actin polymerization markedly impaired the EndMT in rBMECs. These findings suggest that ischemia-reperfusion increased intensity of FSS triggered an EndMT process and, thus, led to endothelial inflammation and tissue injury, whereas continuous FSS induced a time-dependent reversal MEndT event contributing to the endothelial repair. This study provides valuable insight for therapeutic strategies targeting IRI.
Collapse
Affiliation(s)
| | - Jia Ma
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Lingyu Du
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Qiao Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Hongyan Yue
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Chengxiu Peng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Hanxiao Chen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | | | | | | |
Collapse
|
13
|
Jia J, Xu S, Hu J, Gan Y, Sun M, Xia S, Bao X, Zhang M, Xu Y. Growth arrest specific protein 6 alleviated white matter injury after experimental ischemic stroke. J Cereb Blood Flow Metab 2024; 44:77-93. [PMID: 37794790 PMCID: PMC10905636 DOI: 10.1177/0271678x231205078] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/09/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023]
Abstract
Ischemic white matter injury leads to long-term neurological deficits and lacks effective medication. Growth arrest specific protein 6 (Gas6) clears myelin debris, which is hypothesized to promote white matter integrity in experimental stroke models. By the middle cerebral artery occlusion (MCAO) stroke model, we observed that Gas6 reduced infarcted volume and behavior deficits 4 weeks after MCAO. Compared with control mice, Gas6-treatment mice represented higher FA values in the ipsilateral external capsules by MRI DTI scan. The SMI32/MBP ratio of the ipsilateral cortex and striatum was profoundly alleviated by Gas6 administration. Gas6-treatment group manifested thicker myelin sheaths than the control group by electron microscopy. We observed that Gas6 mainly promoted OPC maturation, which was closely related to microglia. Mechanically, Gas6 accelerated microglia-mediated myelin debris clearance and cholesterol transport protein expression (abca1, abcg1, apoc1, apoe) in vivo and in vitro, accordingly less myelin debris and lipid deposited in Gas6 treated stroke mice. HX531 (RXR inhibitor) administration mitigated the functions of Gas6 in speeding up debris clearance and cholesterol transport protein expression. Generally, we concluded that Gas6 cleared myelin debris and promoted cholesterol transportation protein expression through activating RXR, which could be one critical mechanism contributing to white matter repair after stroke.
Collapse
Affiliation(s)
- Junqiu Jia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Siyi Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Zhenjiang, China
| | - Jinglong Hu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yonghui Gan
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Min Sun
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shengnan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Meijuan Zhang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Zhenjiang, China
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
- Jiangsu Provincial medical key discipline, Nanjing, China
| |
Collapse
|
14
|
Tang J, Jin Y, Jia F, Lv T, Manaenko A, Zhang LF, Zhang Z, Qi X, Xue Y, Zhao B, Zhang X, Zhang JH, Lu J, Hu Q. Gas6 Promotes Microglia Efferocytosis and Suppresses Inflammation Through Activating Axl/Rac1 Signaling in Subarachnoid Hemorrhage Mice. Transl Stroke Res 2023; 14:955-969. [PMID: 36324028 DOI: 10.1007/s12975-022-01099-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022]
Abstract
Early brain injury (EBI) following subarachnoid hemorrhage (SAH) is characterized by rapid development of neuron apoptosis and dysregulated inflammatory response. Microglia efferocytosis plays a critical role in the clearance of apoptotic cells, attenuation of inflammation, and minimizing brain injury in various pathological conditions. Here, using a mouse SAH model, we aim to investigate whether microglia efferocytosis is involved in post-SAH inflammation and to determine the underlying signaling pathway. We hypothesized that TAM receptors and their ligands regulate this process. To prove our hypothesis, the expression and cellular location of TAM (Tyro3, Axl, and Mertk) receptors and their ligands growth arrest-specific 6 (Gas6) and Protein S (ProS1) were examined by PCR, western blots, and fluorescence immunostaining. Thirty minutes after SAH, mice received an intraventricular injection of recombinant Gas6 (rGas6) or recombinant ProS1 (rPros1) and underwent evaluations of inflammatory mediator expression, neurological deficits, and blood-brain barrier integrity at 24 h. Microglia efferocytosis of apoptotic neurons was analyzed in vivo and in vitro. The potential mechanism was determined by inhibiting or knocking down TAM receptors and Rac1 by specific inhibitors or siRNA. SAH induced upregulation of Axl and its ligand Gas6. The administration of rGas6 but not rPros1 promoted microglia efferocytosis, alleviated inflammation, and ameliorated SAH-induced BBB breakdown and neurological deficits. The beneficial effects of rGas6 were arrogated by inhibiting or knocking down Axl and Rac1. We concluded that rGas6 attenuated the development of early brain injury in mice after SAH by facilitating microglia efferocytosis and preventing inflammatory response, which is partly dependent on activation of Axl and Rac1.
Collapse
Affiliation(s)
- Junjia Tang
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Yichao Jin
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Feng Jia
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Tao Lv
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Anatol Manaenko
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin-Feng Zhang
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Zeyu Zhang
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xin Qi
- Discipline of Neuroscience, Department of Physiology and Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yajun Xue
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Bin Zhao
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xiaohua Zhang
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Jianfei Lu
- Discipline of Neuroscience, Department of Physiology and Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China.
| | - Qin Hu
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
15
|
Chen Y, Chen J, Zhao L, Zhang X, Wu X, Wang X, Zhang Z, Yang Y, Deng C. Ethyl pyruvate attenuates cerebral hypoxia/reoxygenation injury in neuroblastoma cells: Role of GAS6/Axl signaling. Heliyon 2023; 9:e22787. [PMID: 38090008 PMCID: PMC10711176 DOI: 10.1016/j.heliyon.2023.e22787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 11/19/2023] [Accepted: 11/20/2023] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Ischemic stroke, caused neurological dysfunction due to inadequate blood supply to brain, has a high morbidity and mortality. Ethyl pyruvate (EP), a simple aliphatic ester derived from pyruvic acid, has the advantages of safety and stability. Studies have confirmed that EP has anti-oxidative, anti-inflammation, anti-tumor, and other pharmacological effects, and it demonstrates significant therapeutic effects on multiple diseases. GAS6 and its high affinity Axl receptor play an important role in cell adhesion, anti-apoptosis, proliferation and migration by activating downstream signal transduction pathways. Previous studies have demonstrated the neuroprotective effects of the GAS6/Axl axis. METHODS A series of experimental methods were employed to confirm the effect of EP against cerebral hypoxia/reoxygenation (HR) injury. RESULTS In this study, the protective effect and mechanism of EP on HR injury in N2a cells was explored. The results found that treatment with EP could increase HR-injured neuronal viability, improve cell morphology, and reduce LDH release and ROS accumulation, thereby exhibiting a neuroprotective effect. Furthermore, EP treatment restored the down-regulated expression of GAS6, Axl, NQO1, PGC-1α, NRF1, and UCP2 caused by HR injury. Specifically, it was observed that the neuroprotective effect of EP was partially inhibited by GAS6 siRNA. CONCLUSION In conclusion, these results suggest that EP treatment attenuates HR-induced oxidative stress injury in neuroblastoma cells via activating GAS6/Axl signaling.
Collapse
Affiliation(s)
- Ying Chen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Junmin Chen
- Department of Cardiology, Affiliated Hospital, Yan'an University, 43 North Street, Yan'an, 716000, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, 710069, Xi'an, China
| | - Lin Zhao
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Xin Zhang
- Department of Cardiology, Affiliated Hospital, Yan'an University, 43 North Street, Yan'an, 716000, China
| | - Xue Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, 710069, Xi'an, China
| | - Xin Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, 710069, Xi'an, China
| | - Zhe Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, 710069, Xi'an, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, 710069, Xi'an, China
| | - Chao Deng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, 710069, Xi'an, China
| |
Collapse
|
16
|
Wang W, Cheng W, Wang X, Li Z, Gao J. CircFKBP3 absence alleviates oxygen glucose deprivation-induced function loss of human brain microvascular endothelial cells in vitro via governing the miR-766-3p/TRAF3 axis. Int J Neurosci 2023:1-12. [PMID: 37982234 DOI: 10.1080/00207454.2023.2279506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 10/31/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Brain microvascular endothelial cell (BMEC) functions loss is a key event in the development of ischemic stroke, which may be affected by the dysregulation of circular RNAs (circRNAs). We aimed to unveil the role of circRNA FKBP Prolyl Isomerase 3 (circFKBP3) in cell models of ischemic stroke. METHODS Cell models of ischemic stroke were constructed in human BEMCs (HBMECs) with the treatment of oxygen glucose deprivation (OGD). Quantitative real-time PCR (qPCR) and western blotting were conducted for expression analysis of circFKBP3, miR-766-3p and TNF receptor associated factor 3 (TRAF3). CCK-8, transwell, wound healing, flow cytometry, tube formation and ELISA assays were implemented to monitor cell viability, migration, apoptosis, angiogenesis and inflammation production. The putative binding relationship between miR-766-3p and circFKBP3 or TRAF3 was validated by dual-luciferase, RIP and pull-down assays. RESULTS CircFKBP3 expression was elevated in OGD-treated HBMECs. OGD suppressed HBMEC viability, migration, angiogenesis, and provoked cell apoptosis and inflammation production, while knockdown of circFKBP3 attenuated these effects. CircFKBP3 interacted with miR-766-3p, and circFKBP3 absence-repressed HBMEC function loss and inflammation were recovered by miR-766-3p inhibition. CircFKBP3 targeted miR-766-3p to regulate TRAF3 expression. MiR-766-3p enrichment-repressed HBMEC function loss and inflammation were recovered by TRAF3 overexpression. CONCLUSION CircFKBP3 absence alleviated OGD-induced function loss and inflammatory responses of HBMECs via governing the miR-766-3p/TRAF3 axis.
Collapse
Affiliation(s)
- Wenyan Wang
- Department of Neurology, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Wei Cheng
- Department of Neurology, Wuhan Puren Hospital affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Xudong Wang
- Department of Neurology, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Zhixin Li
- Department of Neurology, Wuhan Puren Hospital affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Jinli Gao
- Department of Neurology, Wuhan Puren Hospital affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
17
|
Zheng X, Zhang X, Dong L, Zhao J, Zhang C, Chen R. Neuroprotective mechanism of salvianolic acid B against cerebral ischemia-reperfusion injury in mice through downregulation of TLR4, p-p38MAPK, p-JNK, NF-κB, and IL-1β. Immun Inflamm Dis 2023; 11:e1030. [PMID: 37904689 PMCID: PMC10549825 DOI: 10.1002/iid3.1030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 08/02/2023] [Accepted: 09/13/2023] [Indexed: 11/01/2023] Open
Abstract
OBJECTIVE Tissue injury and inflammation are two potential outcomes of cerebral ischemia-reperfusion (I/R) injury. Salvianolic acid B (Sal B), isolated from the roots of Salvia miltiorrhiza, is one of the major water-soluble compounds with a wide range of pharmacological effects including antioxidant, anti-inflammatory, antiproliferative, and neuroprotective effects. In the present study, we explored the neuroprotective effects and potential mechanisms of Sal B after I/R injury. METHODS We induced cerebral ischemia in male CD-1 mice through transient (60 min) middle cerebral artery occlusion (tMCAO), and then injected Sal B (30 mg/kg) intraperitoneally. Neurological deficits, infarct volumes, and brain edema were assessed at 24 and 72 h after tMCAO. We detected the expression of Toll-like receptor 4 (TLR4), phosphorylated-p38 mitogen-activated protein kinase (P-p38 MAPK), phosphorylated c-Jun amino (N)-terminal kinases (p-JNK), nuclear factor-κB (NF-κB), and interleukin-1β (IL-1β) in the brain tissue. RESULTS Compared with the tMCAO group, Sal B significantly improved neurological deficits, reduced infarct size, attenuated cerebral edema, and downregulated the expression of pro-inflammatory mediators TLR4, p-p38MAPK, p-JNK, nuclear NF-κB, and IL-1β in brain tissue after I/R injury. CONCLUSION We found that Sal B protects brain tissues from I/R injury by activating its anti-inflammatory properties.
Collapse
Affiliation(s)
- Xiu‐fen Zheng
- Department of NeurologySecond Hospital of Hebei Medical UniversityShijiazhuangHebeiPR China
- Department of PediatricsTangshan Central HospitalTangshanHebeiPR China
| | - Xiang‐jian Zhang
- Department of NeurologySecond Hospital of Hebei Medical UniversityShijiazhuangHebeiPR China
- Hebei Collaborative Innovation Center for Cardio‐cerebrovascular DiseaseShijiazhuangHebeiPR China
- Hebei Key Laboratory of Vascular HomeostasisShijiazhuangHebeiPR China
| | - Li‐peng Dong
- Department of NeurologySecond Hospital of Hebei Medical UniversityShijiazhuangHebeiPR China
- Department of NeurologyHebei General HospitalShijiazhuangHebeiPR China
| | - Jing‐ru Zhao
- Department of NeurologySecond Hospital of Hebei Medical UniversityShijiazhuangHebeiPR China
- Department of NeurologyHebei General HospitalShijiazhuangHebeiPR China
| | - Cong Zhang
- Department of NeurologySecond Hospital of Hebei Medical UniversityShijiazhuangHebeiPR China
| | - Rong Chen
- Department of NeurologySecond Hospital of Hebei Medical UniversityShijiazhuangHebeiPR China
- Hebei Collaborative Innovation Center for Cardio‐cerebrovascular DiseaseShijiazhuangHebeiPR China
- Hebei Key Laboratory of Vascular HomeostasisShijiazhuangHebeiPR China
| |
Collapse
|
18
|
ERGON EY, ÇELİK A, DİNİZ G, ÇOLAK R, ÖZDEMİR SA, ÇALKAVUR Ş, YILMAZ O. Evaluation of syringin's neuroprotective effect in a model of neonatal hypoxic-ischemic brain injury. Turk J Med Sci 2023; 53:1312-1320. [PMID: 38813032 PMCID: PMC10763795 DOI: 10.55730/1300-0144.5697] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 10/26/2023] [Accepted: 06/21/2023] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND/AIM A significant cause of mortality and morbidity in the neonatal era is hypoxic-ischemic encephalopathy (HIE). This study examined the histopathological analysis and neuroprotective impact of syringin (SYR) in an experimental HIE rat model. MATERIAL AND METHODS On the 7th postnatal day, 24 Wistar albino rats were evaluated in 3 groups using the HIE model under gas anesthesia. In the experiment, Group A received 10 mg/kg SYR plus dimethyl sulfoxide (DMSO), Group B received DMSO only, and Group C served as a sham group. Immunohistochemical techniques were used to assess apoptotic cell measurement and proinflammatory cytokines (TNF-α and IL-1β primary antibodies). RESULTS Rats suffering from hypoxic-ischemic brain damage had their apoptosis assessed. The SYR and sham groups had statistically fewer cells undergoing apoptosis (p < 0.001). There was no difference between the groups in terms of IL-1β and TNF-α during immunohistochemical staining. Neuronal degeneration was significantly lower in the histological evaluation of the hippocampus in the SYR group (p = 0.01). A statistically significant difference (p = 0.01) was observed between the SYR and the control groups regarding pericellular and perivascular edema. CONCLUSION SYR reduced apoptosis, perivascular and pericellular edema, and neuronal degeneration in rat cerebral tissue. These results raise the possibility that SYR may have a neuroprotective effect on the harm brought on by HIE. This is the first investigation of SYR's function within the HIE paradigm.
Collapse
Affiliation(s)
- Ezgi Yangın ERGON
- Neonatal Intensive Care Unit, Pediatric Division, Dr Behçet Uz Children’s Education and Research Hospital, İzmir,
Turkiye
| | - Aslı ÇELİK
- Department of Laboratory Animal Science, Faculty of Health Sciences, Dokuz Eylül University, İzmir,
Turkiye
| | - Gülden DİNİZ
- Department of Pathology, Medical Faculty, İzmir Democracy University, İzmir,
Turkiye
| | - Rüya ÇOLAK
- Neonatal Intensive Care Unit, Pediatric Division, Medikal Park Florya Hospital, Aydın University Medical Faculty, İstanbul,
Turkiye
| | - Senem Alkan ÖZDEMİR
- Neonatal Intensive Care Unit, Pediatric Division, Dr Behçet Uz Children’s Education and Research Hospital, İzmir,
Turkiye
| | - Şebnem ÇALKAVUR
- Neonatal Intensive Care Unit, Pediatric Division, Dr Behçet Uz Children’s Education and Research Hospital, İzmir,
Turkiye
| | - Osman YILMAZ
- Department of Laboratory Animal Science, Faculty of Health Sciences, Dokuz Eylül University, İzmir,
Turkiye
| |
Collapse
|
19
|
Cheng CH, Liu YC, Yang YCS, Lin KJ, Wu D, Liu YR, Chang CC, Hong CT, Hu CJ. Plasmon-activated water as a therapeutic strategy in Alzheimer's disease by altering gut microbiota. Aging (Albany NY) 2023; 15:3715-3737. [PMID: 37166426 PMCID: PMC10449311 DOI: 10.18632/aging.204706] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/22/2023] [Indexed: 05/12/2023]
Abstract
Gut microbiota (GM) are involved in the pathophysiology of Alzheimer's disease (AD) and might correlate to the machinery of the gut-brain axis. Alteration of the GM profiles becomes a potential therapy strategy in AD. Here, we found that plasmon-activated water (PAW) therapy altered GM profile and reduced AD symptoms in APPswe/PS1dE9 transgenic mice (AD mice). GM profile showed the difference between AD and WT mice. PAW therapy in AD mice altered GM profile and fecal microbiota transplantation (FMT) reproduced GM profile in AD mice. PAW therapy and FMT in AD mice reduced cognitive decline and amyloid accumulation by novel object recognition (NOR) test and amyloid PET imaging. Immunofluorescent staining and western blot analysis of β-amyloid (Aβ) and phosphorylated (p)-tau in the brain of AD mice were reduced in PAW therapy and FMT. The inflammatory markers, interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α and pro-inflammatory indicator of arginase-1/CD86 ratio were also reduced. Furthermore, immunohistochemistry (IHC) analysis of occludin and claudin-5 in the intestine and AXL in the brain were increased to correlate with the abundant GM in PAW therapy and FMT. Our results showed the machinery of gut-brain axis, and PAW might be a potential therapeutic strategy in AD.
Collapse
Affiliation(s)
- Chia-Hsiung Cheng
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Chuan Liu
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Chen S.H. Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei 11031, Taiwan
| | - Kun-Ju Lin
- Department of Nuclear Medicine and Molecular Imaging Center, Linkou Chang Gung Memorial Hospital, Taoyuan City 33302, Taiwan
- Healthy Aging Research Center and Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Dean Wu
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Sleep Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Yun-Ru Liu
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei 11031, Taiwan
| | - Chun-Chao Chang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chien-Tai Hong
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Neurology and Dementia Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Chaur-Jong Hu
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Neurology and Dementia Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- PhD program of Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
20
|
Lu H, Chen S, Nie Q, Xue Q, Fan H, Wang Y, Fan S, Zhu J, Shen H, Li H, Fang Q, Ni J, Chen G. Synaptotagmin-3 interactions with GluA2 mediate brain damage and impair functional recovery in stroke. Cell Rep 2023; 42:112233. [PMID: 36892998 DOI: 10.1016/j.celrep.2023.112233] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 01/20/2023] [Accepted: 02/23/2023] [Indexed: 03/10/2023] Open
Abstract
Synaptotagmin III (Syt3) is a Ca2+-dependent membrane-traffic protein that is highly concentrated in synaptic plasma membranes and affects synaptic plasticity by regulating post-synaptic receptor endocytosis. Here, we show that Syt3 is upregulated in the penumbra after ischemia/reperfusion (I/R) injury. Knockdown of Syt3 protects against I/R injury, promotes recovery of motor function, and inhibits cognitive decline. Overexpression of Syt3 exerts the opposite effects. Mechanistically, I/R injury augments Syt3-GluA2 interactions, decreases GluA2 surface expression, and promotes the formation of Ca2+-permeable AMPA receptors (CP-AMPARs). Using a CP-AMPAR antagonist or dissociating the Syt3-GluA2 complex via TAT-GluA2-3Y peptide promotes recovery from neurological impairments and improves cognitive function. Furthermore, Syt3 knockout mice are resistant to cerebral ischemia because they show high-level expression of surface GluA2 and low-level expression of CP-AMPARs after I/R. Our results indicate that Syt3-GluA2 interactions, which regulate the formation of CP-AMPARs, may be a therapeutic target for ischemic insults.
Collapse
Affiliation(s)
- Haifeng Lu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Shujun Chen
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Qianqian Nie
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Qun Xue
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Hua Fan
- The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471000, Henan, China
| | - Yiqing Wang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Shenghao Fan
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Juehua Zhu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Qi Fang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Jianqiang Ni
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China.
| |
Collapse
|
21
|
Zhou S, Li Y, Zhang Z, Yuan Y. An insight into the TAM system in Alzheimer's disease. Int Immunopharmacol 2023; 116:109791. [PMID: 36738678 DOI: 10.1016/j.intimp.2023.109791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/13/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023]
Abstract
The TAM receptors may help delay the progression of Alzheimer's disease (AD). AD is the most common neurodegenerative disease associated with human aging. The TAM receptors, derived from the first letter of its three constituents -Tyro3, Axl, and Mertk, are associated with immune responses, cellular differentiation and migration, and clearance of apoptotic cells and debris, with the two canonical ligands, Growth Arrest Specific 6 (Gas6) and ProS1. Several kinds of research have indicated the participation of the TAM system in AD pathology. Also, the TAMs regulate multiple features of microglia, the significant sensors of disorder in the central nervous system (CNS). In this review, we describe the biology of the TAM receptors and ligands in the CNS. Then, we discuss the relationship between the TAM system and AD, specially focusing on its functional expression in the microglia. Finally, we also summarize some agents that could interfere with the TAM signaling pathways and discuss potential difficulties and strategies for drug development.
Collapse
Affiliation(s)
- Shiqi Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yanyan Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yuhe Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
22
|
Morrison VE, Bix GJ. The meal Maketh the Microglia: Why studying microglial phagocytosis is critical to stroke research. Neurochem Int 2023; 164:105488. [PMID: 36707032 DOI: 10.1016/j.neuint.2023.105488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/15/2023] [Accepted: 01/15/2023] [Indexed: 01/25/2023]
Affiliation(s)
- Vivianne E Morrison
- Tulane University School of Medicine Center for Clinical Neuroscience Research Center, United States
| | - Gregory J Bix
- Tulane University School of Medicine Center for Clinical Neuroscience Research Center, United States.
| |
Collapse
|
23
|
Zhu L, Liu S, Liao F, Yang J, Liang T, Yang Y, Huang X, Gu L, Su L. Comprehensive Analysis of Blood-Based m6A Methylation in Human Ischemic Stroke. Mol Neurobiol 2023; 60:431-446. [PMID: 36279101 DOI: 10.1007/s12035-022-03064-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 10/03/2022] [Indexed: 01/21/2023]
Abstract
Alterations of N6-methyladenosine (m6A) methylation have been reported in the cerebral cortices of mouse and rat models of ischemic stroke (IS). However, the role of m6A methylation in human IS is still unknown. We assessed m6A levels in peripheral blood from patients with IS and healthy controls. A transient middle cerebral artery occlusion and reperfusion (tMCAO/R) mouse model, and an oxygen-glucose deprivation/reperfusion (OGD/R) model in A172 cells were established to further assess m6A levels. Methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA sequencing were performed in the peripheral blood of patients with IS and healthy controls. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were used to identify underlying biological processes. In this study, we found that global m6A levels were elevated in the peripheral blood of patients with IS, in the cerebral cortex of mice after tMCAO/R treatment and in A172 cells after OGD/R treatment. MeRIP-seq analysis identified 2115 altered m6A peaks in patients with IS, 1052 upregulated and 1063 downregulated. Downregulated methylated mRNAs were enriched in Hippo signaling pathway, cytokine-cytokine receptor interaction, NF-kappa B signaling pathway, etc. Upregulated methylated mRNAs were enriched in calcium signaling pathways, Hedgehog signaling pathway, MAPK signaling pathway, etc. Moreover, a total of 84 differentially expressed mRNAs with altered m6A peaks were identified and enriched in EGFR tyrosine kinase inhibitor, Hematopoietic cell lineage, and cytokine-cytokine receptor interactions. This study is the first to profile the transcriptome-wide m6A methylome of peripheral blood in human IS and uncover increased global m6A levels in the peripheral blood of patients with IS.
Collapse
Affiliation(s)
- Lulu Zhu
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, China
| | - Shengying Liu
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, China
| | - Fangping Liao
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, China
| | - Jialei Yang
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, China
| | - Tian Liang
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, China
| | - Yibing Yang
- Department of Internal Neurology, First Affiliated Hospital, Guangxi University of Chinese Medicine, 89-9 Dongge Road, Nanning, Guangxi, China
| | - Xianli Huang
- Department of Internal Neurology, First Affiliated Hospital, Guangxi University of Chinese Medicine, 89-9 Dongge Road, Nanning, Guangxi, China
| | - Lian Gu
- Department of Internal Neurology, First Affiliated Hospital, Guangxi University of Chinese Medicine, 89-9 Dongge Road, Nanning, Guangxi, China.
| | - Li Su
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, China.
| |
Collapse
|
24
|
Zhang Y, Su Q, Xia W, Jia K, Meng D, Wang X, Ni X, Su Z. MiR-140-3p directly targets Tyro3 to regulate OGD/R-induced neuronal injury through the PI3K/Akt pathway. Brain Res Bull 2023; 192:93-106. [PMID: 36372373 DOI: 10.1016/j.brainresbull.2022.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 10/21/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND AND PURPOSE MicroRNAs (miRNAs) are highly expressed in the central nervous system and play important roles in ischaemic stroke pathogenesis. However, the role of miRNAs in cerebral ischaemia-reperfusion injury remains unclear. Here, we investigated the role of miR-140-3p in regulating oxygen-glucose deprivation/reoxygenation (OGD/R)-induced neuronal injury in vitro to identify a new biomarker for research on ischaemic stroke. METHODS The differential expression of miR-140-3p and Tyro3 in OGD/R-exposed N2a cells was verified by qRT-PCR. N2a cells were transfected with miR-140-3p mimic, miR-140-3p inhibitor, Tyro3 or siTyro3, and qRT-PCR, Western blotting, the Cell counting kit-8 (CCK-8) assay, Hoechst 33342/PI staining and flow cytometry analyses were performed to measure miRNA, mRNA and protein expression; cell viability; and apoptosis. RESULTS OGD/R-exposed N2a cells exhibited increased miR-140-3p expression, decreased viability, reduced Bcl-2 protein expression and increased Bax and Caspase-3 protein expression and apoptosis; the miR-140-3p mimic markedly amplified these changes, exacerbating OGD/R-induced injury to N2a cells, while the miR-140-3p inhibitor reversed these changes and alleviated OGD/R-induced injury. OGD/R-exposed N2a cells expressed less Tyro3, and Tyro3 overexpression increased cell viability and Bcl-2 protein expression, reduced Bax and Caspase-3 protein expression, and alleviated OGD/R-induced injury. However, silencing Tyro3 reversed these changes and exacerbated OGD/R-induced injury. MiR-140-3p directly bound the Tyro3 mRNA 3'UTR. Rescue experiments indicated that the miR-140-3p mimic-induced changes in cell viability and protein expression were alleviated by Tyro3 overexpression and that the miR-140-3p inhibitor-induced changes in cell viability and protein expression were alleviated by silencing Tyro3. Tyro3 overexpression increased cell viability and PI3K and p-Akt protein expression, but these effects were weakened by the addition of LY294002. CONCLUSIONS MiR-140-3p directly targets Tyro3 to regulate cell viability and apoptosis of OGD/R-exposed N2a cells through the PI3K/Akt pathway, suggesting that miR-140-3p is a novel biomarker and therapeutic target for ischaemic stroke.
Collapse
Affiliation(s)
- Yanli Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China; Central Laboratory of the Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Qian Su
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150001, Heilongjiang, China
| | - Wenbo Xia
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150001, Heilongjiang, China
| | - Kejuan Jia
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150001, Heilongjiang, China
| | - Delong Meng
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Xin Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Xunran Ni
- Department of Neurology, Heilongjiang Provincial Hospital, Harbin 150001, China
| | - Zhiqiang Su
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China.
| |
Collapse
|
25
|
Li T, Li L, Peng R, Hao H, Zhang H, Gao Y, Wang C, Li F, Liu X, Chen F, Zhang S, Zhang J. Abrocitinib Attenuates Microglia-Mediated Neuroinflammation after Traumatic Brain Injury via Inhibiting the JAK1/STAT1/NF-κB Pathway. Cells 2022; 11:cells11223588. [PMID: 36429017 PMCID: PMC9688110 DOI: 10.3390/cells11223588] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/30/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Neuroinflammation has been shown to play a critical role in secondary craniocerebral injury, leading to poor outcomes for TBI patients. Abrocitinib, a Janus kinase1 (JAK1) selective inhibitor approved to treat atopic dermatitis (AD) by the Food and Drug Administration (FDA), possesses a novel anti-inflammatory effect. In this study, we investigated whether abrocitinib could ameliorate neuroinflammation and exert a neuroprotective effect in traumatic brain injury (TBI) models. METHODS First, next-generation sequencing (NGS) was used to select genes closely related to neuroinflammation after TBI. Then, magnetic resonance imaging (MRI) was used to dynamically observe the changes in traumatic focus on the 1st, 3rd, and 7th days after the induction of fluid percussion injury (FPI). Moreover, abrocitinib's effects on neurobehaviors were evaluated. A routine peripheral blood test was carried out and Evans blue dye extravasation, cerebral cortical blood flow, the levels of inflammatory cytokines, and changes in the numbers of inflammatory cells were evaluated to investigate the function of abrocitinib on the 1st day post-injury. Furthermore, the JAK1/signal transducer and activator of transcription1 (STAT1)/nuclear factor kappa (NF-κB) pathway was assessed. RESULTS In vivo, abrocitinib treatment was found to shrink the trauma lesions. Compared to the TBI group, the abrocitinib treatment group showed better neurological function, less blood-brain barrier (BBB) leakage, improved intracranial blood flow, relieved inflammatory cell infiltration, and reduced levels of inflammatory cytokines. In vitro, abrocitinib treatment was shown to reduce the pro-inflammatory M1 microglia phenotype and shift microglial polarization toward the anti-inflammatory M2 phenotype. The WB and IHC results showed that abrocitinib played a neuroprotective role by restraining JAK1/STAT1/NF-κB levels after TBI. CONCLUSIONS Collectively, abrocitinib treatment after TBI is accompanied by improvements in neurological function consistent with radiological, histopathological, and biochemical changes. Therefore, abrocitinib can indeed reduce excessive neuroinflammation by restraining the JAK1/STAT1/NF-κB pathway.
Collapse
Affiliation(s)
- Tuo Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
- Department of Neurosurgery, Yantai Yuhuangding Hospital, Yantai 264000, China
| | - Lei Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
| | - Ruilong Peng
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
| | - Hongying Hao
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin 300000, China
- Department of Neurology, Yantai Yuhuangding Hospital, Yantai 264000, China
| | - Hejun Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
- Department of Neurosurgery, First Hospital of Qinhuangdao, Qinhuangdao 066000, China
| | - Yalong Gao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
| | - Cong Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
| | - Fanjian Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
| | - Xilei Liu
- Tianjin Neurological Institute, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
| | - Fanglian Chen
- Tianjin Neurological Institute, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
| | - Shu Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
- Correspondence: (S.Z.); (J.Z.)
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
- Correspondence: (S.Z.); (J.Z.)
| |
Collapse
|
26
|
Changes in somatosensory evoked potentials in rats following transient cerebral ischemia. ACTA BIOMEDICA SCIENTIFICA 2022. [DOI: 10.29413/abs.2022-7.4.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background. Cerebral ischemia induced by transient middle cerebral artery occlusion is one of the most popular ischemic stroke models used to evaluate drug candidates with neuroprotective properties. The possibilities of combining this model with neurophysiological techniques (e.g., electroencephalography, electrocorticography, evoked potential registration, etc.) to assess the effectiveness of novel pharmacotherapeutic strategies appear to be of great interest to current biomedical research.The aim. Identifying specific changes in somatosensory evoked potentials occurring after cerebral ischemia induced by middle cerebral artery occlusion in rats.Materials and methods. A total number of 18 white outbred male rats were randomized into 3 groups by 6 animals in each: 1) control (presumably healthy animals); 2) ischemia-30 (30-minute middle cerebral artery occlusion); 3) ischemia-45 (45-minute occlusion). At post-surgery day 7, cortical responses to sequential electrical stimulation of left and right n. ischiadicus were registered. N1, P2, N2, P3, and N3 peak latencies and amplitudes, peak-to-peak interval durations and amplitudes were calculated. Spearman’s rank correlation coefficients were used to assess the relationship between ischemia duration and evoked potential parameters, and the Chaddock scale was used to qualitatively evaluate the strength of correlations.Results. The rats subjected to cerebral ischemia demonstrated a decrease in some of the peak amplitudes of the ipsi- and contralateral somatosensory potentials evoked by n. ischiadicus stimulation. In the injured hemisphere, decreased P2 and N3 peak and P3–N3 interval amplitudes were registered ipsilaterally, and decreased P3 peak amplitudes and N2–P3 interval durations were observed contralaterally.Conclusions. The obtained data suggest that somatosensory evoked potential registration and analysis can be used to evaluate the functional state of central nerve tracts in rats subjected to cerebral ischemia.
Collapse
|
27
|
AXL Inhibits Proinflammatory Factors to Relieve Rheumatoid Arthritis Pain by Regulating the TLR4/NF-κB Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7625739. [PMID: 35983008 PMCID: PMC9381196 DOI: 10.1155/2022/7625739] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/14/2022] [Indexed: 11/30/2022]
Abstract
Objective This study aims to explore the role and mechanism of AXL receptor tyrosine kinase (AXL) in relieving inflammatory pain caused by rheumatoid arthritis (RA). Methods RA mouse model was constructed by collagen antibody induction. RT-qPCR and Western blot were used to detect the level of AXL in RA fibroblast-like synovial cells (RA-FLS) and joint synovium. The levels of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and nitric oxide (NO) were detected by ELISA. The inflammatory infiltration in joints was determined via HE staining. The mechanical abnormal pain and hyperalgesia were detected by the Von Frey microfilament test. The protein levels of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (3COX-2), toll-like receptor 4 (TLR4), p65, and phosphor (p)-p65 were detected by Western blotting. Results The expression of AXL in RA-FLS and RA mice was downregulated, while the expression of iNOS and COX-2 was upregulated. The levels of inflammatory cytokines IL-6, TNF-α, and NO were increased in RA-FLS and RA mice. RA mice presented inflammatory cell infiltration, bone and cartilage destruction, and joint space stenosis. AXL overexpression alleviated inflammatory cell infiltration, inflammatory cytokine secretion, and pathological injury in RA mice. Additionally, AXL overexpression inhibited the expression of TLR4 and p-p65. Conclusion AXL inhibits inflammatory pain in RA mice by suppressing TLR4/NF-κB pathway.
Collapse
|
28
|
Wang Y, Liu X, Zhang W, He S, Zhang Y, Orgah J, Wang Y, Zhu Y. Synergy of "Yiqi" and "Huoxue" components of QishenYiqi formula in ischemic stroke protection via lysosomal/inflammatory mechanisms. JOURNAL OF ETHNOPHARMACOLOGY 2022; 293:115301. [PMID: 35436536 DOI: 10.1016/j.jep.2022.115301] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/27/2022] [Accepted: 04/12/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ischemic stroke is one of the leading causes of mortality and long-term disability worldwide. Currently, approved therapies of intravenous thrombolysis and mechanical thrombectomy are limited only to selected patients with rescuable brain tissue. Chinese medicine that benefits Qi (Yiqi, YQ) and activates blood (Huoxue, HX) is widely used in the clinic for treating stroke, but their mechanisms are not well understood yet. We have previously reported that QishenYiqi (QSYQ) formula exerts cerebral protective effect and promotes post-stroke recovery. AIM OF THE STUDY This study aimed to explore the chemical basis and molecular mechanism of anti-stroke therapy of QSYQ and its YQ and HX components further. MATERIALS AND METHODS Serum pharmacochemistry was performed to identify the bioactive constituents in QSYQ for cerebral protection. The survival rate, mNSS test, open field test, gait analysis, cerebral infarction volume, and blood-brain barrier (BBB) integrity were determined to uncover the synergistic and differential contributions of YQ and HX components in a cerebral ischemia/reperfusion injury (CI/RI) model. Bioinformatic mining of QSYQ proteomics data and experimental validation were executed to access the functional mechanism of YQ and HX components. RESULTS Eleven prototype ingredients and six metabolites were successfully identified or tentatively characterized in rat plasma. Therapeutically, YQ and HX components of QSYQ synergistically boosted the survival rate, improved neurological and motor functions, alleviated cerebral infarction as well as protected BBB integrity in CI/RI model in rats. Individually, YQ component contributed more to ameliorating locomotive ability than that of HX component. Mechanistically, HX component played a more prominent role in the modulation of galectin-3 mediated inflammation whereas YQ component regulated lysosomal-autophagy signaling. CONCLUSIONS This study identifies major prototype ingredients and metabolites of QSYQ in plasma which may contribute to its cerebral protection. YQ and HX components of QSYQ differentially and synergistically protect the brain from CI/RI by regulating galectin-3-mediated inflammation and lysosomal-autophagy signaling. These findings demonstrate that a maximal stroke protection by a component-based Chinese medicine could be attributed to the combination of its individual components via different mechanisms. It may shed new light on our understanding of the TCM principle of tonifying Qi and activating blood, particularly in a setting of ischemic stroke.
Collapse
Affiliation(s)
- Yule Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, XiHu District, Hangzhou, 310058, China
| | - Xinyan Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biomedicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Wen Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Taiping Qiao Street No.27, Xicheng District, Beijing, China
| | - Shuang He
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biomedicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Yiqian Zhang
- State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin Tasly Holding Group Co, Ltd, Tianjin, China
| | - John Orgah
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biomedicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, XiHu District, Hangzhou, 310058, China
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biomedicine, 220 Dongting Road, TEDA, Tianjin, 300457, China.
| |
Collapse
|
29
|
Zhu X, Li H, You W, Yu Z, Wang Z, Shen H, Li X, Yu H, Wang Z, Chen G. Role of Rph3A in brain injury induced by experimental cerebral ischemia-reperfusion model in rats. CNS Neurosci Ther 2022; 28:1124-1138. [PMID: 35467084 PMCID: PMC9160444 DOI: 10.1111/cns.13850] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/09/2022] [Accepted: 04/13/2022] [Indexed: 01/01/2023] Open
Abstract
Aim The aim was to study the role of Rph3A in neuronal injury induced by cerebral ischemia‐reperfusion. Methods The protein and mRNA levels of Rph3A in penumbra were detected by Western blot. The localization of Rph3A in different cell types in penumbra was detected by immunofluorescence. Apoptosis in the brain was detected by TUNEL staining. We tested neurobehavioral evaluation using rotarod test, adhesive‐removal test, and Morris Water maze test. We examined the expression and localization of Rph3A in cultured neurons and astrocytes in vitro by Western blot and ELISA, respectively. Results The mRNA and protein levels of Rph3A had significantly increased in brain penumbra of the rat MCAO/R model. Rph3A was mainly distributed in neurons and astrocytes and was significantly increased by MCAO/R. We downregulated Rph3A and found that it further worsened the cerebral infarct, neuronal death and behavioral, cognitive, and memory impairments in rats after MCAO/R. We also found that ischemia‐reperfusion upregulated the in vitro protein level and secretion of Rph3A in astrocytes but led to a decrease in the protein level of Rph3A in neurons. Conclusion The increase in Rph3A in the brain penumbra may be an endogenous protective mechanism against ischemia‐reperfusion injury, which is mainly dominated by astrocytes.
Collapse
Affiliation(s)
- Xianlong Zhu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Stroke Research, Soochow University, Suzhou, Jiangsu, China.,Department of Neurosurgery, The Second People's Hospital of Lianyungang City, Lianyungang, Jiangsu, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Stroke Research, Soochow University, Suzhou, Jiangsu, China
| | - Wanchun You
- Department of Neurosurgery & Brain and Nerve Research Laboratory, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Stroke Research, Soochow University, Suzhou, Jiangsu, China
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Stroke Research, Soochow University, Suzhou, Jiangsu, China
| | - Zongqi Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Stroke Research, Soochow University, Suzhou, Jiangsu, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Stroke Research, Soochow University, Suzhou, Jiangsu, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Stroke Research, Soochow University, Suzhou, Jiangsu, China
| | - Hao Yu
- Department of Neurosurgery, The First People's Hospital of Nantong city, Nantong, Jiangsu, China
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Stroke Research, Soochow University, Suzhou, Jiangsu, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Stroke Research, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
30
|
Wang Z, Liu D, Yan Q, Liu F, Zhan M, Qi S, Fang Q, Yao L, Wang W, Zhang R, Du J, Chen L. Activated AXL Protects Against Hepatic Ischemia-reperfusion Injury by Upregulating SOCS-1 Expression. Transplantation 2022; 106:1351-1364. [PMID: 35546091 PMCID: PMC9213082 DOI: 10.1097/tp.0000000000004156] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/24/2022] [Accepted: 03/17/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Hepatic ischemia-reperfusion (I/R) injury is the main factor affecting the morbidity and mortality associated with perioperative complications of liver transplantation and major hepatectomy. AXL is a member of the TYRO3, AXL, MERTK family and is involved in immune and apoptosis processes in multiple organs. However, the role of AXL in hepatic I/R injury remains to be elucidated. METHODS Mice pretreated with rmGas6 or R428 and mice tail vein injected with adeno-associated virus knockdown suppressor of cytokine signaling protein-1 (SOCS-1) underwent liver I/R surgery to detect the function of activated AXL in vivo. Primary hepatocytes undergo hypoxic reoxygenation injury in vitro. RESULTS AXL expression was significantly upregulated, and phosphorylated-AXL was substantially downregulated in liver transplantation patients and hepatic I/R surgery mice. A mouse model of hepatic I/R injury showed that AXL activation reduced liver inflammation and liver cells apoptosis. The inhibition of AXL activation (AXL-specific inhibitor R428) aggravated hepatic I/R injury, resulted in larger areas of liver injury, aggravated inflammatory response, and increased apoptosis of liver cells. In addition, activated AXL promotes the expression level of SOCS-1 and inhibits toll-like receptor 4 and its downstream signaling pathways. Finally, SOCS-1 was knocked down with an adeno-associated virus, and activated AXL failed to protect against hepatic I/R injury. CONCLUSIONS AXL activation protects the liver from I/R injury by upregulating SOCS-1 and inhibiting the toll-like receptor 4/myeloid differentiation factor-88/nuclear factor kappa-B signaling axis. Targeting AXL may be a new therapeutic option for ameliorating hepatic I/R injury.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Deng Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Qi Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Fang Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Mengting Zhan
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Shunli Qi
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qi Fang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Lei Yao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Weizhi Wang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Ruixin Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Jian Du
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Infectious Disease Research Center, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Lijian Chen
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| |
Collapse
|
31
|
Chen L, Liu X, Li Z, Wang J, Tian R, Zhang H. Integrated Analysis of Transcriptome mRNA and miRNA Profiles Reveals Self-Protective Mechanism of Bovine MECs Induced by LPS. Front Vet Sci 2022; 9:890043. [PMID: 35812870 PMCID: PMC9260119 DOI: 10.3389/fvets.2022.890043] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 05/04/2022] [Indexed: 12/29/2022] Open
Abstract
Many studies have investigated the molecular crosstalk between mastitis-pathogens and cows by either miRNA or mRNA profiles. Here, we employed both miRNA and mRNA profiles to understand the mechanisms of the response of bovine mammary epithelial cells (bMECs) to lipopolysaccharide (LPS) by RNA-Seq. The total expression level of miRNAs increased while mRNAs reduced after LPS treatment. About 41 differentially expressed mRNAs and 45 differentially expressed miRNAs involved in inflammation were screened out. We found the NFκB-dependent chemokine, CXCL1, CXCL3, CXCL6, IL8, and CX3CL1 to be strongly induced. The anti-apoptosis was active because BCL2A1 and BIRC3 significantly increased with a higher expression. The effects of anti-microbe and inflammation were weakly activated because TNF, IL1, CCL20, CFB, S100A, MMP9, and NOS2A significantly increased but with a low expression, IL6 and β-defensin decreased. These activities were supervised by the NFKBIA to avoid excessive damage to bMECs. The bta-let-7a-5p, bta-miR-30a-5p, bta-miR-125b, and bta-miR-100 were essential to regulate infection process in bMECs after LPS induction. Moreover, the lactation potential of bMECs was undermined due to significantly downregulated SOSTDC1, WNT7B, MSX1, and bta-miR-2425-5p. In summary, bMECs may not be good at going head-to-head with the pathogens; they seem to be mainly charged with sending out signals for help and anti-apoptosis for maintaining lives after LPS induction.
Collapse
Affiliation(s)
- Ling Chen
- School of Modern Agriculture and Biotechnology, Ankang University, Ankang, China
| | - Xiaolin Liu
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China
- *Correspondence: Xiaolin Liu
| | - Zhixiong Li
- College of Life Science and Technology, Southwest University for Nationalities, Chengdu, China
| | - Jian Wang
- School of Modern Agriculture and Biotechnology, Ankang University, Ankang, China
| | - Rongfu Tian
- School of Modern Agriculture and Biotechnology, Ankang University, Ankang, China
| | - Huilin Zhang
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| |
Collapse
|
32
|
Enhanced treatment of cerebral ischemia-Reperfusion injury by intelligent nanocarriers through the regulation of neurovascular units. Acta Biomater 2022; 147:314-326. [PMID: 35588994 DOI: 10.1016/j.actbio.2022.05.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/27/2022] [Accepted: 05/10/2022] [Indexed: 12/11/2022]
Abstract
Reperfusion injury is one of the major causes of disability and death caused by ischemic stroke, and drug development focuses mainly on single neuron protection. However, different kinds of cells in the neurovascular units (NVUs), including neurons, microglia and vascular endothelial cells, are pathologically changed after cerebral ischemia-reperfusion injury, resulting in an urgent need to develop a drug delivery system to comprehensively protect the kinds of cells involved in the NVU. Herein, we have constructed a c(RGDyK) peptide modified, NF-κB inhibitor caffeic acid phenethyl ester (CAPE)-loaded and reactive nitrogen species (RNS) stimuli-responsive liposomal nanocarrier (R-Lipo-CAPE) to target ischemic lesions and then remodel the NVU to reduce the progression of cerebral ischemia-reperfusion injury. The R-Lipo-CAPE liposomes were approximately 170 nm with a zeta potential of -30.8 ± 0.2 mV. The in vitro CAPE release behavior from R-Lipo-CAPE showed an RNS-dependent pattern. For in vivo studies, transient middle cerebral artery occlusion/reperfusion (MCAO) model mice treated with R-Lipo-CAPE had the least neurological impairment and decreased brain tissue damage, with an infarct area of 13%, compared with those treated with saline of 53% or free CAPE of 38%. Furthermore, microglia in the ischemic brain were polarized to the tissue-repairing M2 phenotype after R-Lipo-CAPE treatment. In addition, R-Lipo-CAPE-treated mice displayed a prominent down-regulated expression of MMP-9 and restored expression of the tight junction protein claudin-5. This proof-of-concept indicates that R-Lipo-CAPE is a promising nanomedicine for the treatment of cerebral ischemia-reperfusion injury through the regulation of neurovascular units. STATEMENT OF SIGNIFICANCE: Based on the complex mechanism and difficulty in treatment of cerebral ischemia-reperfusion injury, the overall regulation of neurovascular unit has become an extremely important target. However, little nanomedicine has been directed to remodel the neurovascular units in targeted cerebral ischemia-reperfusion injury therapy. Here, c(RGDyK) peptide modified reactive nitrogen species (RNS) stimuli-responsive liposomal nanocarrier loaded with a NF-κB inhibitor (CAPE), was designed to simultaneously regulate various cells in the microenvironment of cerebral ischemia-reperfusion injury to remodel the neurovascular units. Our in vitro and in vivo data showed that the intelligent nanocarrier exerted the ability of pathological signal stimuli-responsive drug release, cerebral ischemia-reperfusion injury site targeting and neurovascular units remodeling through reducing neuron apoptosis, regulating microglia polarization and repairing vascular endothelial cell. Overall, the intelligent liposomal drug delivery system was a promising and safe nanomedicine in the perspective of cerebral ischemia-reperfusion injury treatment.
Collapse
|
33
|
Mollet I, Martins C, Ângelo-Dias M, Carvalho AS, Aloria K, Matthiesen R, Baptista MV, Borrego LM, Vieira HL. Pilot study in human healthy volunteers on the mechanisms underlying remote ischemic conditioning (RIC) – Targeting circulating immune cells and immune-related proteins. J Neuroimmunol 2022; 367:577847. [DOI: 10.1016/j.jneuroim.2022.577847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/03/2022] [Accepted: 03/15/2022] [Indexed: 11/29/2022]
|
34
|
Owlett LD, Karaahmet B, Le L, Belcher EK, Dionisio-Santos D, Olschowka JA, Elliott MR, O'Banion MK. Gas6 induces inflammation and reduces plaque burden but worsens behavior in a sex-dependent manner in the APP/PS1 model of Alzheimer's disease. J Neuroinflammation 2022; 19:38. [PMID: 35130912 PMCID: PMC8822838 DOI: 10.1186/s12974-022-02397-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/20/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Alzheimer's disease is the leading cause of dementia worldwide. TAM receptor tyrosine kinases (Tyro3, Axl, MerTK) are known for their role in engagement of phagocytosis and modulation of inflammation, and recent evidence suggests a complex relationship between Axl, Mer, and microglial phagocytosis of amyloid plaques in AD. Gas6, the primary CNS TAM ligand, reduces neuroinflammation and improves outcomes in murine models of CNS disease. Therefore, we hypothesized that AAV-mediated overexpression of Gas6 would alleviate plaque pathology, reduce neuroinflammation, and improve behavior in the APP/PS1 model of Alzheimer's disease. METHODS Adeno-associated viral vectors were used to overexpress Gas6 in the APP/PS1 model of Alzheimer's disease. Nine-month-old male and female APP/PS1 and nontransgenic littermates received bilateral stereotactic hippocampal injections of AAV-Gas6 or AAV-control, which expresses a non-functional Gas6 protein. One month after injections, mice underwent a battery of behavioral tasks to assess cognitive function and brains were processed for immunohistochemical and transcriptional analyses. RESULTS Gas6 overexpression reduced plaque burden in male APP/PS1 mice. However, contrary to our hypothesis, Gas6 increased pro-inflammatory microglial gene expression and worsened contextual fear conditioning compared to control-treated mice. Gas6 overexpression appeared to have no effect on phagocytic mechanisms in vitro or in vivo as measured by CD68 immunohistochemistry, microglial methoxy-04 uptake, and primary microglial uptake of fluorescent fibrillar amyloid beta. CONCLUSION Our data describes a triad of worsened behavior, reduced plaque number, and an increase in proinflammatory signaling in a sex-specific manner. While Gas6 has historically induced anti-inflammatory signatures in the peripheral nervous system, our data suggest an alternative, proinflammatory role in the context of Alzheimer's disease pathology.
Collapse
Affiliation(s)
- Laura D Owlett
- Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, Rochester, NY, USA
| | - Berke Karaahmet
- Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, Rochester, NY, USA
| | - Linh Le
- Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, Rochester, NY, USA
| | - Elizabeth K Belcher
- Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, Rochester, NY, USA
| | - Dawling Dionisio-Santos
- Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, Rochester, NY, USA
| | - John A Olschowka
- Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, Rochester, NY, USA
| | - Michael R Elliott
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - M Kerry O'Banion
- Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
35
|
Zhu L, Ding S, Xu L, Wu Z. Ozone treatment alleviates brain injury in cerebral ischemic rats by inhibiting the NF-κB signaling pathway and autophagy. Cell Cycle 2022; 21:406-415. [PMID: 34985377 PMCID: PMC8855843 DOI: 10.1080/15384101.2021.2020961] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Stroke is the most frequent cause of disability in developed countries. A common phenomenon of stroke, cerebral ischemia, is threatening many lives worldwide. In addition, ozone treatment was previously reported to exert functions in relieving brain injury. In the current study, the therapeutic effects of ozone on cerebral ischemia are investigated. A rat model of middle cerebral artery occlusion (MCAO) was established. The brain water content was calculated by weighing brain tissues, and the 2, 3, 5-triphenyltetrazolium chloride staining was performed to measure brain infarction volume in rats. A colorimetric assay was conducted to examine expression levels of malondialdehyde, superoxide dismutase, catalase, and glutathione in the rat hippocampus. Reverse transcription quantitative polymerase-chain reaction and Western blot analyses were employed to evaluate expression levels of Beclin1, LC3B, p62, and critical factors implicated in the NF-κB signaling pathway. We found that ozone significantly improved the survival rate of MCAO model rats, reduced the cerebral water content, and decreased the neurological scores of ischemic rats. Ozone markedly reduced cerebral ischemia-induced infarction in ischemic rats. Ozone decreased MDA levels and increased SOD, catalase, and GSH levels in the hippocampus of rats. Ozone significantly inhibited autophagy by decreasing Beclin1 and LC3B expression and increasing p62 expression. The ozone inactivated the NF-κB signaling pathway by decreasing the protein levels of TLR4, p-IKKβ, p-IKBα, and p-p65. We conclude that ozone treatment alleviates the brain injury in ischemic rats by suppressing autophagy and inactivating the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Liang Zhu
- Department of Anesthesiology, The Affiliated Changzhou NO. 2 People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Shengyang Ding
- Department of Anesthesiology, The Affiliated Changzhou NO. 2 People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Lingshan Xu
- Department of Anesthesiology, The Affiliated Changzhou NO. 2 People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Zhouquan Wu
- Department of Anesthesiology, The Affiliated Changzhou NO. 2 People’s Hospital of Nanjing Medical University, Changzhou, China,CONTACT Zhouquan Wu Department of Anesthesiology, The Affiliated Changzhou NO. 2 People’s Hospital of Nanjing Medical University, 68 Gehu Middle Road, Wujin District, Changzhou, Jiangsu, China
| |
Collapse
|
36
|
Jia J, Yang L, Chen Y, Zheng L, Chen Y, Xu Y, Zhang M. The Role of Microglial Phagocytosis in Ischemic Stroke. Front Immunol 2022; 12:790201. [PMID: 35082781 PMCID: PMC8784388 DOI: 10.3389/fimmu.2021.790201] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022] Open
Abstract
Microglia are the resident immune cells of the central nervous system that exert diverse roles in the pathogenesis of ischemic stroke. During the past decades, microglial polarization and chemotactic properties have been well-studied, whereas less attention has been paid to phagocytic phenotypes of microglia in stroke. Generally, whether phagocytosis mediated by microglia plays a beneficial or detrimental role in stroke remains controversial, which calls for further investigations. Most researchers are in favor of the former proposal currently since efficient clearance of tissue debris promotes tissue reconstruction and neuronal network reorganization in part. Other scholars propose that excessively activated microglia engulf live or stressed neuronal cells, which results in neurological deficits and brain atrophy. Upon ischemia challenge, the microglia infiltrate injured brain tissue and engulf live/dead neurons, myelin debris, apoptotic cell debris, endothelial cells, and leukocytes. Cell phagocytosis is provoked by the exposure of "eat-me" signals or the loss of "don't eat-me" signals. We supposed that microglial phagocytosis could be initiated by the specific "eat-me" signal and its corresponding receptor on the specific cell type under pathological circumstances. In this review, we will summarize phagocytic characterizations of microglia after stroke and the potential receptors responsible for this programmed biological progress. Understanding these questions precisely may help to develop appropriate phagocytic regulatory molecules, which are promoting self-limiting inflammation without damaging functional cells.
Collapse
Affiliation(s)
- Junqiu Jia
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Lixuan Yang
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Yan Chen
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Lili Zheng
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Yanting Chen
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Yun Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.,Nanjing Neuropsychiatry Clinic Medical Center, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Meijuan Zhang
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.,Nanjing Neuropsychiatry Clinic Medical Center, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
37
|
Niu X, Cheng Y, Zhang M, Du L, Wu X, Lu C, Li X, Liu S, Zhao A, Zhang S, Wu Z, Ding B, Shi W, Wang C, Yang Y, Tian Y. Neuroprotective Effects of Omentin-1 Against Cerebral Hypoxia/Reoxygenation Injury via Activating GAS6/Axl Signaling Pathway in Neuroblastoma Cells. Front Cell Dev Biol 2022; 9:784035. [PMID: 35141232 PMCID: PMC8818945 DOI: 10.3389/fcell.2021.784035] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/13/2021] [Indexed: 12/04/2022] Open
Abstract
Ischemic stroke is characterized by insufficient blood supply to brain tissue and is associated with increased morbidity and mortality in adults worldwide. Growth arrest-specific protein 6 (GAS6) is a vitamin K-dependent protein and is widely expressed in the central nervous system. The biological functions of GAS6 are mediated by the interaction with TAM (Tyro3, Axl and Mertk) receptors, including cell survival and proliferation, immune regulation and apoptosis. Omentin-1, also known as intelectin-1 (ITLN-1), is a novel adipocytokine that is involved in a variety of biological events, such as insulin resistance, endothelial dysfunction, programmed cell death and metabolic disorders. Our previous study has found that omentin-1 act as a novel regulator of vascular and anti-apoptotic response in cerebral ischemia. However, the specific molecular mechanism of omentin-1’s protective effect on cerebral ischemia-reperfusion injury (IRI) is still unclear. First, the toxicity of recombinant human omentin-1 (rh-omentin) was assessed and a safe concentration was chosen for the next experiments. Then, rh-omentin exerted neuroprotection against hypoxia/reoxygenation (H/R) injury in N2a cells, indicated by increased cell viability, decreased LDH, ROS generation, and cell apoptotic rate. Furthermore, the similar protective effect was observed in omentin-1 overexpression cells constructed by lentivirus transfection. Rh-omentin could also inhibit H/R-induced apoptotic molecules, oxidative stress molecules, and GAS6/Axl signaling molecules which as evidence by increased omentin-1, GAS6, Axl, p-Axl, NQO1, HO-1, Nrf2, Bcl2 and decreased Bax expressions. However, GAS6 siRNA could reverse rh-omentin-induced neuroprotection and the levels of these molecules mentioned above. In conclusion, these findings suggest that omentin-1 treatment exerts neuroprotection against H/R injury partly via activating GAS6/Axl signaling at least. Therefore, these finding may favor omentin-1 a potential neuroprotective drug candidate to alleviate ischemia-reperfusion injury in clinic.
Collapse
Affiliation(s)
- Xiaochen Niu
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, School of Life Sciences and Medicine, Northwest University, Xi’an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
| | - Ye Cheng
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, School of Life Sciences and Medicine, Northwest University, Xi’an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
| | - Meng Zhang
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, School of Life Sciences and Medicine, Northwest University, Xi’an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
| | - Luyang Du
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, School of Life Sciences and Medicine, Northwest University, Xi’an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
| | - Xue Wu
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, School of Life Sciences and Medicine, Northwest University, Xi’an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
| | - Chenxi Lu
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, School of Life Sciences and Medicine, Northwest University, Xi’an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
| | - Xiyang Li
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, School of Life Sciences and Medicine, Northwest University, Xi’an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
| | - Shuai Liu
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, School of Life Sciences and Medicine, Northwest University, Xi’an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
| | - Aizhen Zhao
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, School of Life Sciences and Medicine, Northwest University, Xi’an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
| | - Shaofei Zhang
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, School of Life Sciences and Medicine, Northwest University, Xi’an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
| | - Zhen Wu
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, School of Life Sciences and Medicine, Northwest University, Xi’an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
| | - Baoping Ding
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, School of Life Sciences and Medicine, Northwest University, Xi’an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
| | - Wenzhen Shi
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, School of Life Sciences and Medicine, Northwest University, Xi’an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
| | - Changyu Wang
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, School of Life Sciences and Medicine, Northwest University, Xi’an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
| | - Yang Yang
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, School of Life Sciences and Medicine, Northwest University, Xi’an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
- *Correspondence: Yang Yang, ; Ye Tian,
| | - Ye Tian
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, School of Life Sciences and Medicine, Northwest University, Xi’an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
- *Correspondence: Yang Yang, ; Ye Tian,
| |
Collapse
|
38
|
Zhang Y, Ding J, Wang Y, Feng X, Du M, Liu P. Guanxinkang Decoction Attenuates the Inflammation in Atherosclerosis by Regulating Efferocytosis and MAPKs Signaling Pathway in LDLR -/- Mice and RAW264.7 Cells. Front Pharmacol 2021; 12:731769. [PMID: 34950025 PMCID: PMC8688952 DOI: 10.3389/fphar.2021.731769] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 11/17/2021] [Indexed: 01/02/2023] Open
Abstract
Guanxinkang decoction (GXK), a traditional Chinese medicinal drug, is used to treat cardiovascular disease. The aim of the study was to investigate the effects of GXK on inflammation in LDLR−/− mice and RAW264.7 cells. Fed with high fat diet for 12 weeks, the mice were randomly divided into six groups, then administered with oral 0.9% saline or GXK (7.24, 14.48, and 28.96 g/kg) or Atorvastatin (1.3 mg/kg) for 12 weeks. RAW 264.7 cells were induced with ox-LDL or ox-LDL plus different concentrations of GXK (1.25, 2.5, and 5 μg/ml), or ox-LDL plus GXK plus MAPKs activators. Serum lipid profiles and inflammatory cytokines were detected by ELISA, gene expression by RT-qPCR, plaque sizes by Oil Red O, α-SMA, caspase 3, NF-κB p65 and TNF-α production by immunofluorescence staining, and protein expression by Western Blot. The phagocytic ability of cells was determined by neutral red uptake assay. Efferocytosis-related proteins (AML, MERTK, TYRO3 and MFGE8) and MAPKs pathways were detected by Western Blot. Compared to mice fed with high fat diet, the mice with GXK showed lower cholesterol, triglyceride, low-density lipoprotein cholesterol, IL-1β, IL-6, and TNF-α, smaller plaque sizes, higher α-SMA, and lower caspase 3 and NF-κB p65 in aortic roots. RAW264.7 cells treated with ox-LDL plus GXK had lower IL-1β, IL-6, and TNF-α. GXK also increased the phagocytic ability of cells. High levels of AML, MERTK, TYRO3 and MFGE8, and decreased levels of iNOS, VCAM-1, LOX-1 and MCP-1, and phosphorylation of ERK1/2, JNK, p38, and NF-κB were detected in GXK-treated group. MAPKs activators reversed the effects of GXK in repressing inflammation and promoting phagocytosis. These results suggested that GXK could attenuate atherosclerosis and resolve inflammation via efferocytosis and MAPKs signaling pathways in LDLR−/− mice and RAW264.7 cells.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Ding
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiru Wang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoteng Feng
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Min Du
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Liu
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
39
|
Activation of AdipoR1 with rCTRP9 Preserves BBB Integrity through the APPL1/AMPK/Nrf2 Signaling Pathway in ICH Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2801263. [PMID: 34925690 PMCID: PMC8674037 DOI: 10.1155/2021/2801263] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 11/06/2021] [Indexed: 12/26/2022]
Abstract
Background The disruption of the blood brain barrier (BBB) is the key factor leading to neurological impairment after intracerebral hemorrhage (ICH) injury. Adiponectin receptor 1 (AdipoR1) has an important effect contributing to the integrity of BBB. As a homologue of adiponectin, recombinant C1q/TNF-related protein 9 (rCTRP9) has neuroprotective effect in cerebrovascular diseases. The aim of this study was to investigate the protective effect of AdipoR1 activation with rCTRP9 on BBB integrity after ICH injury and the potential mechanisms. Methods 177 male mice were subjected in this study. ICH was induced by injecting collagenase into the right basal ganglia. rCTRP9 was treated intranasally at 1 hour after ICH. Selective siRNA was administered prior to ICH. Western blot, immunofluorescence staining, neurobehavioral tests, and BBB permeability were evaluated. Results ICH increased the expression of endogenous AdipoR1 and CTRP9. Administration of rCTRP9 ameliorated neurological deficits and reduced the BBB permeability at 24 hours in ICH mice. Furthermore, rCTRP9 promoted the expression of AdipoR1, APPL1, p-AMPK, Nrf2, and tight junctional proteins. The intervention of specific siRNA of AdipoR1, APPL1, and p-AMPK reversed the protective effects of rCTRP9. Conclusions Activation of AdipoR1 with rCTRP9 improved neurological functions and preserved BBB integrity through the APPL1/AMPK/Nrf2 signaling pathway in ICH mice. Therefore, CTRP9 could serve as a promising therapeutic method to alleviate BBB injury following ICH in patients.
Collapse
|
40
|
Var SR, Shetty AV, Grande AW, Low WC, Cheeran MC. Microglia and Macrophages in Neuroprotection, Neurogenesis, and Emerging Therapies for Stroke. Cells 2021; 10:3555. [PMID: 34944064 PMCID: PMC8700390 DOI: 10.3390/cells10123555] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 12/20/2022] Open
Abstract
Stroke remains the number one cause of morbidity in the United States. Within weeks to months after an ischemic event, there is a resolution of inflammation and evidence of neurogenesis; however, years following a stroke, there is evidence of chronic inflammation in the central nervous system, possibly by the persistence of an autoimmune response to brain antigens as a result of ischemia. The mechanisms underlying the involvement of macrophage and microglial activation after stroke are widely acknowledged as having a role in ischemic stroke pathology; thus, modulating inflammation and neurological recovery is a hopeful strategy for treating the long-term outcomes after ischemic injury. Current treatments fail to provide neuroprotective or neurorestorative benefits after stroke; therefore, to ameliorate brain injury-induced deficits, therapies must alter both the initial response to injury and the subsequent inflammatory process. This review will address differences in macrophage and microglia nomenclature and summarize recent work in elucidating the mechanisms of macrophage and microglial participation in antigen presentation, neuroprotection, angiogenesis, neurogenesis, synaptic remodeling, and immune modulating strategies for treating the long-term outcomes after ischemic injury.
Collapse
Affiliation(s)
- Susanna R. Var
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (S.R.V.); (A.W.G.)
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN 55108, USA
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Anala V. Shetty
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
- Department of Biological Sciences, University of Minnesota Medical School, Minneapolis, MN 55108, USA
| | - Andrew W. Grande
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (S.R.V.); (A.W.G.)
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Walter C. Low
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (S.R.V.); (A.W.G.)
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Maxim C. Cheeran
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN 55108, USA
| |
Collapse
|
41
|
Gas6/TAM Signalling Negatively Regulates Inflammatory Induction of GM-CSF in Mouse Brain Microglia. Cells 2021; 10:cells10123281. [PMID: 34943789 PMCID: PMC8699038 DOI: 10.3390/cells10123281] [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: 09/16/2021] [Revised: 11/17/2021] [Accepted: 11/21/2021] [Indexed: 12/16/2022] Open
Abstract
Microglia and astrocytes are the main CNS glial cells responsible for the neuroinflammatory response, where they release a plethora of cytokines into the CNS inflammatory milieu. The TAM (Tyro3, Axl, Mer) receptors and their main ligand Gas6 are regulators of this response, however, the underlying mechanisms remain to be determined. We investigated the ability of Gas6 to modulate the CNS glial inflammatory response to lipopolysaccharide (LPS), a strong pro-inflammatory agent, through a qPCR array that explored Toll-like receptor signalling pathway-associated genes in primary cultured mouse microglia. We identified the Csf2 gene, encoding granulocyte-macrophage colony-stimulating factor (GM-CSF), as a major Gas6 target gene whose induction by LPS was markedly blunted by Gas6. Both the Csf2 gene induction and the suppressive effect of Gas6 on this were emulated through measurement of GM-CSF protein release by cells. We found distinct profiles of GM-CSF induction in different glial cell types, with microglia being most responsive during inflammation. Also, Gas6 markedly inhibited the LPS-stimulated nuclear translocation of NF-κB p65 protein in microglia. These results illustrate microglia as a major resident CNS cellular source of GM-CSF as part of the neuroinflammatory response, and that Gas6/TAM signalling inhibits this response through suppression of NF-κB signalling.
Collapse
|
42
|
TLR Signaling in Brain Immunity. Handb Exp Pharmacol 2021; 276:213-237. [PMID: 34761292 DOI: 10.1007/164_2021_542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Toll-like receptors (TLRs) comprise a group of transmembrane proteins with crucial roles in pathogen recognition, immune responses, and signal transduction. This family represented the first line of immune homeostasis in an evolutionarily conserved manner. Extensive researches in the past two decades had emphasized their structural and functional characteristics under both healthy and pathological conditions. In this review, we summarized the current understanding of TLR signaling in the central nervous system (CNS), which had been viewed as a previously "immune-privileged" but now "immune-specialized" area, with major implications for further investigation of pathological nature as well as potential therapeutic manipulation of TLR signaling in various neurological disorders.
Collapse
|
43
|
Guo ZN, Liu J, Chang J, Zhang P, Jin H, Sun X, Yang Y. GAS6/Axl Signaling Modulates Blood-Brain Barrier Function Following Intravenous Thrombolysis in Acute Ischemic Stroke. Front Immunol 2021; 12:742359. [PMID: 34733281 PMCID: PMC8558492 DOI: 10.3389/fimmu.2021.742359] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/30/2021] [Indexed: 01/27/2023] Open
Abstract
Background and Purpose Recent studies have shown that several proteins, including Axl, are related to hemorrhagic transformation (HT) following intravenous thrombolysis by affecting blood-brain barrier (BBB) function. However, the effects of these proteins on BBB function have been studied primarily in animal models. In this study, we aimed to identify serum protein markers that predict HT following intravenous thrombolysis in patients with acute ischemic stroke (AIS) and verify whether these serum proteins regulate BBB function and HT in animal stroke models. Methods First, 118 AIS patients were enrolled in this study, including 52 HT patients and 66 non-HT patients. In Step 1, baseline serum levels of Axl, angiopoietin-like 4, C-reactive protein, ferritin, hypoxia-inducible factor-1 alpha, HTRA2, Lipocalin2, matrix metallopeptidase 9, platelet-derived growth factor-BB, and tumor necrosis factor alpha were measured using a quantitative cytokine chip. Next, sequence mutations and variations in genes encoding the differentially expressed proteins identified in Step 1 and subsequent function-related proteins were detected. Finally, we verified whether manipulation of differentially expressed proteins affected BBB function and HT in a hyperglycemia-induced rat stroke model. Results Serum Axl levels were significantly lower in the HT group than in the non-HT group; none of the other protein markers differed significantly between the two groups. Genetic testing revealed that sequence variations of GAS6 (the gene encoding the Axl ligand)-derived long non-coding RNA, GAS6-AS1, were significantly correlated with an increased risk of HT after intravenous thrombolysis. In animal studies, administration of recombinant GAS6 significantly reduced brain infarction and neurological deficits and attenuated BBB disruption and HT. Conclusions Lower serum Axl levels, which may result from sequence variations in GAS6-AS1, are correlated with an increased risk of HT after intravenous thrombolysis in stroke patients. Activation of the Axl signaling pathway by the GAS6 protein may serve as a therapeutic strategy to reduce HT in AIS patients.
Collapse
Affiliation(s)
- Zhen-Ni Guo
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Clinical Trial and Research Center for Stroke, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Jie Liu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Junlei Chang
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Peng Zhang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Clinical Trial and Research Center for Stroke, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Hang Jin
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Xin Sun
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yi Yang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Clinical Trial and Research Center for Stroke, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
44
|
Hu SQ, Liu DL, Li CR, Xu YH, Hu K, Cui LD, Guo J. Wuzi-Yanzong prescription alleviates spermatogenesis disorder induced by heat stress dependent on Akt, NF-κB signaling pathway. Sci Rep 2021; 11:18824. [PMID: 34552120 PMCID: PMC8458393 DOI: 10.1038/s41598-021-98036-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 08/25/2021] [Indexed: 02/08/2023] Open
Abstract
Akt and nuclear factor kappa B (NF-κB) signaling pathways are involved in germ cell apoptosis and inflammation after testicular heat stress (THS). We observed that after THS induced by the exposure of rat testes to 43 °C for 20 min, their weight decreased, the fraction of apoptotic testicular germ cells significantly increased, and the proliferation of germ cells was inhibited. In addition, THS lowered serum testosterone (T) level, whereas the levels of follicle stimulating hormone and luteinizing hormone were not significantly changed. The ultrastructure of the seminiferous tubules became abnormal after THS, the structure of the blood-testis barrier (BTB) became loose, and the Sertoli cells showed a trend of differentiation. The level of phosphorylated Akt was reduced, whereas the amount of phosphorylated NF-κB p65 was augmented by THS. Wuzi-Yanzong (WZYZ), a classic Chinese medicine prescription for the treatment of male reproductive dysfunctions, alleviated the changes induced by THS. In order to determine the mechanism of action of WZYZ, we investigated how this preparation modulated the levels of T, androgen receptor (AR), erythropoietin (EPO), EPO receptor, and Tyro-3, Axl, and Mer (TAM) family of tyrosine kinase receptors. We found that WZYZ activated the Akt pathway, inhibited the Toll-like receptor/MyD88/NF-κB pathway, and repaired the structure of BTB by regulating the levels of T, AR, TAM receptors, and EPO. In conclusion, these results suggest that WZYZ activates the Akt pathway and inhibits the NF-κB pathway by acting on the upstream regulators, thereby improving spermatogenesis deficit induced by THS.
Collapse
Affiliation(s)
- Su-Qin Hu
- grid.24695.3c0000 0001 1431 9176Department of Physiology, College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, No. 11, East Beisanhuan Road, Chaoyang District, Beijing, China
| | - Dian-Long Liu
- grid.24695.3c0000 0001 1431 9176Department of Physiology, College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, No. 11, East Beisanhuan Road, Chaoyang District, Beijing, China
| | - Chun-Rui Li
- grid.24695.3c0000 0001 1431 9176Department of Physiology, College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, No. 11, East Beisanhuan Road, Chaoyang District, Beijing, China
| | - Ya-Hui Xu
- grid.24695.3c0000 0001 1431 9176Department of Physiology, College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, No. 11, East Beisanhuan Road, Chaoyang District, Beijing, China
| | - Ke Hu
- grid.24695.3c0000 0001 1431 9176Department of Physiology, College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, No. 11, East Beisanhuan Road, Chaoyang District, Beijing, China
| | - Li-Dan Cui
- grid.24695.3c0000 0001 1431 9176Department of Physiology, College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, No. 11, East Beisanhuan Road, Chaoyang District, Beijing, China
| | - Jian Guo
- grid.24695.3c0000 0001 1431 9176Department of Physiology, College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, No. 11, East Beisanhuan Road, Chaoyang District, Beijing, China
| |
Collapse
|
45
|
Sun YW, Zhang LY, Gong SJ, Hu YY, Zhang JG, Xian XH, Li WB, Zhang M. The p38 MAPK/NF-κB pathway mediates GLT-1 up-regulation during cerebral ischemic preconditioning-induced brain ischemic tolerance in rats. Brain Res Bull 2021; 175:224-233. [PMID: 34343641 DOI: 10.1016/j.brainresbull.2021.07.029] [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: 06/02/2021] [Revised: 07/25/2021] [Accepted: 07/29/2021] [Indexed: 10/20/2022]
Abstract
Our previous finding suggests that p38 MAPK contributes to the GLT-1 upregulation during induction of brain ischemic tolerance by cerebral ischemic preconditioning (CIP), however, the underlying mechanism is still unclear. Here, we investigated the molecular mechanisms underlying the CIP-induced GLT-1 upregulation by using Western blotting, Co-immunoprecipitation (Co-IP), electrophoretic mobility shift assay (EMSA) and thionin staining in rat hippocampus CA1 subset. We found that application of BAY11-7082 (an inhibitor of NF-κB), or dihydrokainate (an inhibitor of GLT-1), or SB203580 (an inhibitor of p38 MAPK) could attenuate the CIP-induced neuronal protection in hippocampus CA1 region of rats. Moreover, CIP caused rapid activation of NF-κB, as evidenced by nuclear translocation of NF-κB p50 protein, which led to active p50/p65 dimer formation and increased DNA binding activity. GLT-1 was also increased after CIP. Pretreatment with BAY11-7082 blocked the CIP-induced GLT-1 upregulation. The above results suggest that NF-κB participates in GLT-1 up-regulation during the induction of brain ischemic tolerance by CIP. We also found that pretreatment with SB203580 caused significant reduction of NF-κB p50 protein in nucleus, NF-κB p50/p65 dimer nuclear translocation and DNA binding activity of NF-κB. Together, we conclude that p38 MAPK/NF-κB pathway participates in the mediation of GLT-1 up-regulation during the induction of brain ischemic tolerance induced by CIP.
Collapse
Affiliation(s)
- Ya-Wei Sun
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Xing Tai People's Hospital, 16 Hong Xing Road, Xing Tai, 054001, People's Republic of China
| | - Ling-Yan Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, People's Republic of China
| | - Shu-Juan Gong
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Yu-Yan Hu
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, People's Republic of China
| | - Jing-Ge Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, People's Republic of China
| | - Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, People's Republic of China
| | - Wen-Bin Li
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, People's Republic of China.
| |
Collapse
|
46
|
Zhou H, Hu L, Li J, Ruan W, Cao Y, Zhuang J, Xu H, Peng Y, Zhang Z, Xu C, Yu Q, Li Y, Dou Z, Hu J, Wu X, Yu X, Gu C, Cao S, Yan F, Chen G. AXL kinase-mediated astrocytic phagocytosis modulates outcomes of traumatic brain injury. J Neuroinflammation 2021; 18:154. [PMID: 34233703 PMCID: PMC8264993 DOI: 10.1186/s12974-021-02201-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/22/2021] [Indexed: 11/23/2022] Open
Abstract
Background Complex changes in the brain microenvironment following traumatic brain injury (TBI) can cause neurological impairments for which there are few efficacious therapeutic interventions. The reactivity of astrocytes is one of the keys to microenvironmental changes, such as neuroinflammation, but its role and the molecular mechanisms that underpin it remain unclear. Methods Male C57BL/6J mice were subjected to the controlled cortical impact (CCI) to develop a TBI model. The specific ligand of AXL receptor tyrosine kinase (AXL), recombinant mouse growth arrest-specific 6 (rmGas6) was intracerebroventricularly administered, and selective AXL antagonist R428 was intraperitoneally applied at 30 min post-modeling separately. Post-TBI assessments included neurobehavioral assessments, transmission electron microscopy, immunohistochemistry, and western blotting. Real-time polymerase chain reaction (RT-PCR), siRNA transfection, and flow cytometry were performed for mechanism assessments in primary cultured astrocytes. Results AXL is upregulated mainly in astrocytes after TBI and promotes astrocytes switching to a phenotype that exhibits the capability of ingesting degenerated neurons or debris. As a result, this astrocytic transformation promotes the limitation of neuroinflammation and recovery of neurological dysfunction. Pharmacological inhibition of AXL in astrocytes significantly decreased astrocytic phagocytosis both in vivo and in primary astrocyte cultures, in contrast to the effect of treatment with the rmGas6. AXL activates the signal transducer and activator of the transcription 1 (STAT1) pathway thereby further upregulating ATP-binding cassette transporter 1 (ABCA1). Moreover, the supernatant from GAS6-depleted BV2 cells induced limited enhancement of astrocytic phagocytosis in vitro. Conclusion Our work establishes the role of AXL in the transformation of astrocytes to a phagocytic phenotype via the AXL/STAT1/ABCA1 pathway which contributes to the separation of healthy brain tissue from injury-induced cell debris, further ameliorating neuroinflammation and neurological impairments after TBI. Collectively, our findings provide a potential therapeutic target for TBI. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02201-3.
Collapse
Affiliation(s)
- Hang Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Libin Hu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Jianru Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Wu Ruan
- Department of Burn and Plastic Surgery, Children's Hospital, Zhejiang University School of Medicine, No. 3333 Binsheng Road, Zhejiang, 310052, Hangzhou, China
| | - Yang Cao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Jianfeng Zhuang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Hangzhe Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Yucong Peng
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Zhongyuan Zhang
- Department of Neurosurgery, Children's Hospital, Zhejiang University School of Medicine, No. 3333 Binsheng Road, Zhejiang, 310052, Hangzhou, China
| | - Chaoran Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Qian Yu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Yin Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Zhangqi Dou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Junwen Hu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Xinyan Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Xiaobo Yu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Chi Gu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Shenglong Cao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Feng Yan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China.
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China.
| |
Collapse
|
47
|
Dienel A, Veettil RA, Matsumura K, Savarraj JPJ, Choi HA, Kumar T P, Aronowski J, Dash P, Blackburn SL, McBride DW. α 7-Acetylcholine Receptor Signaling Reduces Neuroinflammation After Subarachnoid Hemorrhage in Mice. Neurotherapeutics 2021; 18:1891-1904. [PMID: 33970466 PMCID: PMC8609090 DOI: 10.1007/s13311-021-01052-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2021] [Indexed: 02/04/2023] Open
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) causes a robust inflammatory response which leads worse brain injury and poor outcomes. We investigated if stimulation of nicotinic acetylcholine α7 receptors (α7-AChR) (receptors shown to have anti-inflammatory effects) would reduce inflammation and improve outcomes. To investigate the level of peripheral inflammation after aSAH, inflammatory markers were measured in plasma samples collected in a cohort of aSAH patients. To study the effect of α7-AChR stimulation, SAH was induced in adult mice which were then treated with a α7-AChR agonist, galantamine, or vehicle. A battery of motor and cognitive tests were performed 24 h after subarachnoid hemorrhage. Mice were euthanized and tissue collected for analysis of markers of inflammation or activation of α7-AChR-mediated transduction cascades. A separate cohort of mice was allowed to survive for 28 days to assess long-term neurological deficits and histological outcome. Microglia cell culture subjected to hemoglobin toxicity was used to assess the effects of α7-AChR agonism. Analysis of eighty-two patient plasma samples confirmed enhanced systemic inflammation after aSAH. α7-AChR agonism reduced neuroinflammation at 24 h after SAH in male and female mice, which was associated with improved outcomes. This coincided with JAK2/STAT3 and IRAK-M activity modulations and a robust improvement in neurological/cognitive status that was effectively reversed by interfering with various components of these signaling pathways. Pharmacologic inhibition partially reversed the α7-AChR agonist's benefits, supporting α7-AChR as a target of the agonist's therapeutic effect. The cell culture experiment showed that α7-AChR agonism is directly beneficial to microglia. Our results demonstrate that activation of α7-AChR represents an attractive target for treatment of SAH. Our findings suggest that α7-AChR agonists, and specifically galantamine, might provide therapeutic benefit to aSAH patients.
Collapse
Affiliation(s)
- Ari Dienel
- The Vivian L Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center At Houston, Houston, TX, USA
| | - Remya A Veettil
- The Vivian L Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center At Houston, Houston, TX, USA
| | - Kanako Matsumura
- The Vivian L Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center At Houston, Houston, TX, USA
| | - Jude P J Savarraj
- The Vivian L Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center At Houston, Houston, TX, USA
| | - H Alex Choi
- The Vivian L Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center At Houston, Houston, TX, USA
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center At Houston, Houston, TX, USA
| | - Peeyush Kumar T
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA
| | | | - Pramod Dash
- Department of Neurobiology and Anatomy, McGovern Medical School, The University of Texas Health Science Center At Houston, Houston, TX, USA
| | - Spiros L Blackburn
- The Vivian L Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center At Houston, Houston, TX, USA
| | - Devin W McBride
- The Vivian L Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center At Houston, Houston, TX, USA.
| |
Collapse
|
48
|
Agonism of the α 7-acetylcholine receptor/PI3K/Akt pathway promotes neuronal survival after subarachnoid hemorrhage in mice. Exp Neurol 2021; 344:113792. [PMID: 34181928 DOI: 10.1016/j.expneurol.2021.113792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/14/2021] [Accepted: 06/19/2021] [Indexed: 11/22/2022]
Abstract
Subarachnoid hemorrhage (SAH) results in severe neuronal dysfunction and degeneration. Since the nicotinic acetylcholine α7 receptors (α7-AChR) are involved in neuronal function and survival, we investigated if stimulation of α7-AChR would promote neuronal survival and improve behavioral outcome following SAH in mice. Male mice subjected to SAH were treated with either galantamine (α7-AChR agonist) or vehicle. Neurobehavioral testing was performed 24 h after SAH, and mice were euthanized for analysis of neuronal cell death or a cell survival (PI3K/Akt) signaling pathway. Neuron cell cultures were subjected to hemoglobin toxicity to assess the direct effects of α7-AChR agonism independent of other cells. Treatment with the α7-AChR agonist promoted neuronal survival and improved functional outcomes 24 h post-SAH. The improved outcomes corresponded with increased PI3K/Akt activity. Antagonism of α7-AChR or PI3K effectively reversed galantamine's beneficial effects. Tissue from α7-AChR knockout mice confirmed α7-AChR's role in neuronal survival after SAH. Data from the neuronal cell culture experiment supported a direct effect of α7-AChR agonism in promoting cell survival. Our findings indicate that α7-AChR is a therapeutic target following SAH which can promote neuronal survival, thereby improving neurobehavioral outcome. Thus, the clinically relevant α7-AChR agonist, galantamine, might be a potential candidate for human use to improve outcome after SAH.
Collapse
|
49
|
Hirschi KM, Tsai KYF, Davis T, Clark JC, Knowlton MN, Bikman BT, Reynolds PR, Arroyo JA. Growth arrest-specific protein-6/AXL signaling induces preeclampsia in rats†. Biol Reprod 2021; 102:199-210. [PMID: 31347670 DOI: 10.1093/biolre/ioz140] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 07/06/2019] [Accepted: 07/21/2019] [Indexed: 12/19/2022] Open
Abstract
Preeclampsia (PE) is a complicated obstetric complication characterized by increased blood pressure, decreased trophoblast invasion, and inflammation. The growth arrest-specific 6 (Gas6) protein is known to induce dynamic cellular responses and is elevated in PE. Gas6 binds to the AXL tyrosine kinase receptor and AXL-mediated signaling is implicated in proliferation and migration observed in several tissues. Our laboratory utilized Gas6 to induce preeclamptic-like conditions in pregnant rats. Our objective was to determine the role of Gas6/AXL signaling as a possible model of PE. Briefly, pregnant rats were divided into three groups that received daily intraperitoneal injections (from gestational day 7.5 to 17.5) of phosphate buffered saline (PBS), Gas6, or Gas6 + R428 (an AXL inhibitor administered from gestational day 13.5 to 17.5). Animals dispensed Gas6 experienced elevated blood pressure, increased proteinuria, augmented caspase-3-mediated placental apoptosis, and diminished trophoblast invasion. Gas6 also enhanced expression of several PE-related genes and a number of inflammatory mediators. Gas6 further enhanced placental oxidative stress and impaired mitochondrial respiration. Each of these PE-related characteristics was ameliorated in dams and/or their placentae when AXL inhibition by R428 occurred in tandem with Gas6 treatment. We conclude that Gas6 signaling is capable of inducing PE and that inhibition of AXL prevents disease progression in pregnant rats. These results provide insight into pathways associated with PE that could be useful in the clarification of potential therapeutic approaches.
Collapse
Affiliation(s)
- Kelsey M Hirschi
- Lung and Placenta Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, USA
| | - Kary Y F Tsai
- Lung and Placenta Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, USA
| | - Taylor Davis
- Lung and Placenta Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, USA
| | - J Christian Clark
- Lung and Placenta Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, USA
| | - M Nekel Knowlton
- Lung and Placenta Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, USA
| | - Benjamin T Bikman
- Laboratory of Obesity and Metabolism, Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, USA
| | - Paul R Reynolds
- Lung and Placenta Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, USA
| | - Juan A Arroyo
- Lung and Placenta Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, USA
| |
Collapse
|
50
|
Zhang R, Liu C, Li Y, Chen L, Xiang J. Tenacissoside H promotes neurological recovery of cerebral ischaemia/reperfusion injury in mice by modulating inflammation and oxidative stress via TrkB pathway. Clin Exp Pharmacol Physiol 2021; 48:757-769. [PMID: 32799328 DOI: 10.1111/1440-1681.13398] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 12/11/2022]
Abstract
Cerebral ischaemia/reperfusion (I/R)-induced acute brain injury remains a troublesome condition in clinical practice. The present study aimed to investigate the protective effect of tenacissoside H (TH) on I/R-induced cerebral injury in mice. Here, a mouse model of middle cerebral artery occlusion (MCAO) was established by an improved Longa-Zea method. TH was given by intraperitoneal injection once a day within 1 week before establishing the mouse MCAO model. The neurological functions of mice were evaluated and the apoptosis of neurons was also detected by the TUNEL method and Nissl's staining. ELISA and western blot were used to detect the expression of inflammatory factors, oxidation factors and proteins in the cerebral ischaemic cortex. The results revealed that TH dose-dependently reduced neurological impairment, neuron apoptosis and brain oedema induced by MCAO. Furthermore, TH attenuated the expression of pro-inflammatory cytokines (including interleukin (IL)-1β, IL-6 and tumour necrosis factor (TNF)-α), iNOS and nuclear factor (NF)-κB while increased production of anti-inflammatory cytokines (IL-4, IL-10 and BDNF) and proteins of tropomyosin-related kinase receptor B (TrkB) and PPARγ. Nevertheless, after the addition of TrkB inhibitor, the effects of TH above were mostly restrained. In conclusion, TH can protect mice against I/R-induced neurological impairments via modulating inflammation and oxidative stress through TrkB signalling.
Collapse
Affiliation(s)
- Rui Zhang
- Department of NICU, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cui Liu
- Department of Cardiovascular Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yang Li
- Department of NICU, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liang Chen
- Interventional Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus (Shanghai Fengxian District Central Hospital), Shanghai, China
| | - Jianfeng Xiang
- Interventional Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus (Shanghai Fengxian District Central Hospital), Shanghai, China
| |
Collapse
|