1
|
Zhang Y, Yang L, Gan Y, Zhao C, Zhou C, Chen J, Yin Y, Xia S, Yang H, Bao X, Zhang M, Xu Y, Li J. Benzydamine attenuates microglia-mediated neuroinflammation and ischemic brain injury by targeting cathepsin s. Int Immunopharmacol 2025; 146:113824. [PMID: 39700961 DOI: 10.1016/j.intimp.2024.113824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Microglia, the primary immune cells of the central nervous system, play a crucial role in the neuroinflammatory processes following ischemic stroke. Targeting neuroinflammation is a promising strategy to enhance the outcomes of ischemic stroke. Benzydamine (BA), a well-known non-steroidal anti-inflammatory drug, has demonstrated potential in inhibiting pro-inflammatory cytokines across various disease models. However, the potential role of BA in microglial activation and post-stroke neuroinflammation remains unclear. Our study reveals that BA effectively suppresses the lipopolysaccharide (LPS)-stimulated pro-inflammatory responses of primary microglia, with high-dose BA (10 μM) suppressing LPS-induced inflammatory markers by up to 59.1 % in the mRNA levels of IL-1β. Furthermore, BA mitigated ischemic brain injury in experimental stroke mice. BA treatment also significantly attenuated neuroinflammatory responses and attenuates ischemic brain injury in experimental stroke mice. Further investigation revealed that BA reduces the release of the LPS-stimulated pro-inflammatory factors and activation of primary microglia by directly binding to and inhibiting the activity of cathepsin S (CTSS). In conclusion, our study identifies BA as a promising CTSS inhibitor with potential to suppress neuroinflammation following ischemic stroke. Our findings provide a theoretical basis for developing new neuroprotective strategies.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, 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
| | - Lixuan Yang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yonghui Gan
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, 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
| | - Chenchen Zhao
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, 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
| | - Chao Zhou
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Jian Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yanping Yin
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, 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
| | - 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 Disease, Nanjing University, Nanjing 210008, China
| | - Haiyan Yang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 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 Disease, Nanjing University, Nanjing 210008, China
| | - Meijuan Zhang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, 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
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, 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; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Disease, Nanjing University, Nanjing 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China; Nanjing Neurology Clinical Medical Center, Nanjing, 210008, China.
| | - Jingwei Li
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, 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.
| |
Collapse
|
2
|
Lu W, Wang Y, Wen J. The Roles of RhoA/ROCK/NF-κB Pathway in Microglia Polarization Following Ischemic Stroke. J Neuroimmune Pharmacol 2024; 19:19. [PMID: 38753217 DOI: 10.1007/s11481-024-10118-w] [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: 11/18/2023] [Accepted: 04/21/2024] [Indexed: 05/21/2024]
Abstract
Ischemic stroke is the leading cause of death and disability worldwide. Nevertheless, there still lacks the effective therapies for ischemic stroke. Microglia are resident macrophages of the central nervous system (CNS) and can initiate immune responses and monitor the microenvironment. Microglia are activated and polarize into proinflammatory or anti‑inflammatory phenotype in response to various brain injuries, including ischemic stroke. Proinflammatory microglia could generate immunomodulatory mediators, containing cytokines and chemokines, these mediators are closely associated with secondary brain damage following ischemic stroke. On the contrary, anti-inflammatory microglia facilitate recovery following stroke. Regulating the activation and the function of microglia is crucial in exploring the novel treatments for ischemic stroke patients. Accumulating studies have revealed that RhoA/ROCK pathway and NF-κB are famous modulators in the process of microglia activation and polarization. Inhibiting these key modulators can promote the polarization of microglia to anti-inflammatory phenotype. In this review, we aimed to provide a comprehensive overview on the role of RhoA/ROCK pathway and NF-κB in the microglia activation and polarization, reveal the relationship between RhoA/ROCK pathway and NF-κB in the pathological process of ischemic stroke. In addition, we likewise discussed the drug modulators targeting microglia polarization.
Collapse
Affiliation(s)
- Weizhuo Lu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Medical Branch, Hefei Technology College, Hefei, China
| | - Yilin Wang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
3
|
Kang YJ, Hyeon SJ, McQuade A, Lim J, Baek SH, Diep YN, Do KV, Jeon Y, Jo D, Lee CJ, Blurton‐Jones M, Ryu H, Cho H. Neurotoxic Microglial Activation via IFNγ-Induced Nrf2 Reduction Exacerbating Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304357. [PMID: 38482922 PMCID: PMC11132036 DOI: 10.1002/advs.202304357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 01/08/2024] [Indexed: 05/29/2024]
Abstract
Microglial neuroinflammation appears to be neuroprotective in the early pathological stage, yet neurotoxic, which often precedes neurodegeneration in Alzheimer's disease (AD). However, it remains unclear how the microglial activities transit to the neurotoxic state during AD progression, due to complex neuron-glia interactions. Here, the mechanism of detrimental microgliosis in AD by employing 3D human AD mini-brains, brain tissues of AD patients, and 5XFAD mice is explored. In the human and animal AD models, amyloid-beta (Aβ)-overexpressing neurons and reactive astrocytes produce interferon-gamma (IFNγ) and excessive oxidative stress. IFNγ results in the downregulation of mitogen-activated protein kinase (MAPK) and the upregulation of Kelch-like ECH-associated Protein 1 (Keap1) in microglia, which inactivate nuclear factor erythroid-2-related factor 2 (Nrf2) and sensitize microglia to the oxidative stress and induces a proinflammatory microglia via nuclear factor kappa B (NFκB)-axis. The proinflammatory microglia in turn produce neurotoxic nitric oxide and proinflammatory mediators exacerbating synaptic impairment, phosphorylated-tau accumulation, and discernable neuronal loss. Interestingly, recovering Nrf2 in the microglia prevents the activation of proinflammatory microglia and significantly blocks the tauopathy in AD minibrains. Taken together, it is envisioned that IFNγ-driven Nrf2 downregulation in microglia as a key target to ameliorate AD pathology.
Collapse
Affiliation(s)
- You Jung Kang
- Institute of Quantum BiophysicsSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
- Department of BiophysicsSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
| | - Seung Jae Hyeon
- Center for Brain DisordersBrain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Amanda McQuade
- Institute for Neurodegenerative DiseasesUniversity of CaliforniaSan FranciscoCA94158USA
- Department of Neurobiology & BehaviorUniversity of California IrvineIrvineCA92697USA
- Sue and Bill Gross Stem Cell Research CenterUniversity of California IrvineIrvineCA92697USA
- Institute for Memory Impairments and Neurological DisordersUniversity of California IrvineIrvineCA92697USA
| | - Jiwoon Lim
- IBS SchoolUniversity of Science and Technology (UST)Daejeon34114Republic of Korea
- Center for Cognition and SocialityInstitute for Basic Science (IBS)Daejeon34126Republic of Korea
| | - Seung Hyun Baek
- School of PharmacySungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
| | - Yen N. Diep
- Institute of Quantum BiophysicsSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
- Department of BiophysicsSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
- Department of Intelligent Precision Healthcare ConvergenceSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
| | - Khanh V. Do
- Institute of Quantum BiophysicsSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
- Department of Intelligent Precision Healthcare ConvergenceSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
| | - Yeji Jeon
- School of PharmacySungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
| | - Dong‐Gyu Jo
- School of PharmacySungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
- Biomedical Institute for ConvergenceSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
- Samsung Advanced Institute for Health Sciences and TechnologySungkyunkwan UniversitySeoul16419Republic of Korea
| | - C. Justin Lee
- Center for Cognition and SocialityInstitute for Basic Science (IBS)Daejeon34126Republic of Korea
| | - Mathew Blurton‐Jones
- Department of Neurobiology & BehaviorUniversity of California IrvineIrvineCA92697USA
- Sue and Bill Gross Stem Cell Research CenterUniversity of California IrvineIrvineCA92697USA
- Institute for Memory Impairments and Neurological DisordersUniversity of California IrvineIrvineCA92697USA
| | - Hoon Ryu
- Center for Brain DisordersBrain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Hansang Cho
- Institute of Quantum BiophysicsSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
- Department of BiophysicsSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
- Department of Intelligent Precision Healthcare ConvergenceSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
| |
Collapse
|
4
|
Anton PE, Nagpal P, Moreno J, Burchill MA, Chatterjee A, Busquet N, Mesches M, Kovacs EJ, McCullough RL. NF-κB/NLRP3 Translational Inhibition by Nanoligomer Therapy Mitigates Ethanol and Advanced Age-Related Neuroinflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582114. [PMID: 38464118 PMCID: PMC10925165 DOI: 10.1101/2024.02.26.582114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Binge alcohol use is increasing among aged adults (>65 years). Alcohol-related toxicity in aged adults is associated with neurodegeneration, yet the molecular underpinnings of age-related sensitivity to alcohol are not well described. Studies utilizing rodent models of neurodegenerative disease reveal heightened activation of Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and Nod like receptor 3 (NLRP3) mediate microglia activation and associated neuronal injury. Our group, and others, have implicated hippocampal-resident microglia as key producers of inflammatory mediators, yet the link between inflammation and neurodegeneration has not been established in models of binge ethanol exposure and advanced age. Here, we report binge ethanol increased the proportion of NLRP3+ microglia in the hippocampus of aged (18-20 months) female C57BL/6N mice compared to young (3-4 months). In primary microglia, ethanol-induced expression of reactivity markers and NLRP3 inflammasome activation were more pronounced in microglia from aged mice compared to young. Making use of an NLRP3-specific inhibitor (OLT1177) and a novel brain-penetrant Nanoligomer that inhibits NF-κB and NLRP3 translation (SB_NI_112), we find ethanol-induced microglial reactivity can be attenuated by OLT1177 and SB_NI_112 in microglia from aged mice. In a model of intermittent binge ethanol exposure, SB_NI_112 prevented ethanol-mediated microglia reactivity, IL-1β production, and tau hyperphosphorylation in the hippocampus of aged mice. These data suggest early indicators of neurodegeneration occurring with advanced age and binge ethanol exposure are NF-κB- and NLRP3-dependent. Further investigation is warranted to explore the use of targeted immunosuppression via Nanoligomers to attenuate neuroinflammation after alcohol consumption in the aged.
Collapse
Affiliation(s)
- Paige E. Anton
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO
- Alcohol Research Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | | | - Julie Moreno
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - Matthew A. Burchill
- GI and Liver Innate Immune Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
- Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | | | - Nicolas Busquet
- Animal Behavior & In Vivo Neurophysiology Core, NeuroTechnology Center, University of Colorado Anschutz Medical Campus, Aurora, CO
- Department of Neurology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora Colorado
| | - Michael Mesches
- Animal Behavior & In Vivo Neurophysiology Core, NeuroTechnology Center, University of Colorado Anschutz Medical Campus, Aurora, CO
- Department of Neurology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora Colorado
| | - Elizabeth J. Kovacs
- Alcohol Research Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
- Division of GI Trauma and Endocrine Surgery, Department of Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
- Veterans’ Health Administration, Eastern Colorado Health Care System, Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO
| | - Rebecca L. McCullough
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO
- Alcohol Research Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
- GI and Liver Innate Immune Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
5
|
Tian X, Yang W, Jiang W, Zhang Z, Liu J, Tu H. Multi-Omics Profiling Identifies Microglial Annexin A2 as a Key Mediator of NF-κB Pro-inflammatory Signaling in Ischemic Reperfusion Injury. Mol Cell Proteomics 2024; 23:100723. [PMID: 38253182 PMCID: PMC10879806 DOI: 10.1016/j.mcpro.2024.100723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 12/19/2023] [Accepted: 12/23/2023] [Indexed: 01/24/2024] Open
Abstract
Cerebral stroke is one of the leading causes of mortality and disability worldwide. Restoring the cerebral circulation following a period of occlusion and subsequent tissue oxygenation leads to reperfusion injury. Cerebral ischemic reperfusion (I/R) injury triggers immune and inflammatory responses, apoptosis, neuronal damage, and even death. However, the cellular function and molecular mechanisms underlying cerebral I/R-induced neuronal injury are incompletely understood. By integrating proteomic, phosphoproteomic, and transcriptomic profiling in mouse hippocampi after cerebral I/R, we revealed that the differentially expressed genes and proteins mainly fall into several immune inflammatory response-related pathways. We identified that Annexin 2 (Anxa2) was exclusively upregulated in microglial cells in response to cerebral I/R in vivo and oxygen-glucose deprivation and reoxygenation (OGD/R) in vitro. RNA-seq analysis revealed a critical role of Anxa2 in the expression of inflammation-related genes in microglia via the NF-κB signaling. Mechanistically, microglial Anxa2 is required for nuclear translocation of the p65 subunit of NF-κB and its transcriptional activity upon OGD/R in BV2 microglial cells. Anxa2 knockdown inhibited the OGD/R-induced microglia activation and markedly reduced the expression of pro-inflammatory factors, including TNF-α, IL-1β, and IL-6. Interestingly, conditional medium derived from Anxa2-depleted BV2 cell cultures with OGD/R treatment alleviated neuronal death in vitro. Altogether, our findings revealed that microglia Anxa2 plays a critical role in I/R injury by regulating NF-κB inflammatory responses in a non-cell-autonomous manner, which might be a potential target for the neuroprotection against cerebral I/R injury.
Collapse
Affiliation(s)
- Xibin Tian
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China
| | - Wuyan Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China
| | - Wei Jiang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China
| | - Zhen Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China
| | - Junqiang Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China
| | - Haijun Tu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China; Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China.
| |
Collapse
|
6
|
Razavi SM, Arab ZN, Niknejad A, Hosseini Y, Fouladi A, Khales SD, Shahali M, Momtaz S, Butler AE, Sukhorukov VN, Jamialahmadi T, Abdolghaffari AH, Sahebkar A. Therapeutic effects of anti-diabetic drugs on traumatic brain injury. Diabetes Metab Syndr 2024; 18:102949. [PMID: 38308863 DOI: 10.1016/j.dsx.2024.102949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 02/05/2024]
Abstract
AIMS In this narrative review, we have analyzed and synthesized current studies relating to the effects of anti-diabetic drugs on traumatic brain injury (TBI) complications. METHODS Eligible studies were collected from Scopus, Google Scholar, PubMed, and Cochrane Library for clinical, in-vivo, and in-vitro studies published on the impact of anti-diabetic drugs on TBI. RESULTS Traumatic brain injury (TBI) is a serious brain disease that is caused by any type of trauma. The pathophysiology of TBI is not yet fully understood, though physical injury and inflammatory events have been implicated in TBI progression. Several signaling pathways are known to play pivotal roles in TBI injuries, including Nuclear factor erythroid 2-related factor 2 (Nrf2), High mobility group box 1 protein/Nuclear factor kappa B (HMGB1/NF-κB), Adiponectin, Mammalian Target of Rapamycin (mTOR), Toll-Like Receptor (TLR), Wnt/β-catenin, Janus Kinase/Signal Transducers and Activators of Transcription (JAK/STAT), Nod-like receptor protein3 (NLRP3) inflammasome, Phosphoglycerate kinase 1/Kelch-like ECH-associated protein 1 (PGK1/KEAP1)/Nrf2, and Mitogen-activated protein kinase (MAPK) . Recent studies suggest that oral anti-diabetic drugs such as biguanides, thiazolidinediones (TZDs), sulfonylureas (SUs), sodium-glucose cotransporter-2 inhibitors (SGLT2is), dipeptidyl peptidase-4 inhibitors (DPPIs), meglitinides, and alpha-glucosidase inhibitors (AGIs) could have beneficial effects in the management of TBI complications. These drugs may downregulate the inflammatory pathways and induce antioxidant signaling pathways, thus alleviating complications of TBI. CONCLUSION Based on this comprehensive literature review, antidiabetic medications might be considered in the TBI treatment protocol. However, evidence from clinical trials in patients with TBI is still warranted.
Collapse
Affiliation(s)
- Seyed Mehrad Razavi
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Zahra Najafi Arab
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Amirhossein Niknejad
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Yasamin Hosseini
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Abtin Fouladi
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saba Darban Khales
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mostafa Shahali
- School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeideh Momtaz
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran; Department of Toxicology and Pharmacology, School of Pharmacy, and Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland Bahrain, Adliya, Bahrain
| | - Vasily N Sukhorukov
- Institute of General Pathology and Pathophysiology, Moscow, Russia; Institute of Experimental Cardiology Named after Academician V.N. Smirnov, Federal State Budgetary Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Moscow, Russia
| | - Tannaz Jamialahmadi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Palahati A, Luo Y, Qin L, Duan Y, Zhang M, Gan H, Zhai X. TonEBP: A Key Transcription Factor in Microglia Following Intracerebral Hemorrhage Induced-Neuroinflammation. Int J Mol Sci 2024; 25:1438. [PMID: 38338716 PMCID: PMC10855931 DOI: 10.3390/ijms25031438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/15/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Transcription factors within microglia contribute to the inflammatory response following intracerebral hemorrhage (ICH). Therefore, we employed bioinformatics screening to identify the potential transcription factor tonicity-responsive enhancer-binding protein (TonEBP) within microglia. Inflammatory stimuli can provoke an elevated expression of TonEBP in microglia. Nevertheless, the expression and function of microglial TonEBP in ICH-induced neuroinflammation remain ambiguous. In our recent research, we discovered that ICH instigated an increased TonEBP in microglia in both human and mouse peri-hematoma brain tissues. Furthermore, our results indicated that TonEBP knockdown mitigates lipopolysaccharide (LPS)-induced inflammation and the activation of NF-κB signaling in microglia. In order to more deeply comprehend the underlying molecular mechanisms of how TonEBP modulates the inflammatory response, we sequenced the transcriptomes of TonEBP-deficient cells and sought potential downstream target genes of TonEBP, such as Pellino-1 (PELI1). PELI has been previously reported to mediate nuclear factor-κB (NF-κB) signaling. Through the utilization of CUT & RUN, a dual-luciferase reporter, and qPCR, we confirmed that TonEBP is the transcription factor of Peli1, binding to the Peli1 promoter. In summary, TonEBP may enhance the LPS-induced inflammation and activation of NF-κB signaling via PELI1.
Collapse
Affiliation(s)
- Ailiyaer Palahati
- Department of Neurosurgery Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400010, China; (A.P.)
- Center for Neuroscience Research, Chongqing Medical University, Chongqing 400016, China
| | - Yujia Luo
- Department of Neurosurgery Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400010, China; (A.P.)
- Center for Neuroscience Research, Chongqing Medical University, Chongqing 400016, China
| | - Le Qin
- Department of Neurosurgery Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400010, China; (A.P.)
- Center for Neuroscience Research, Chongqing Medical University, Chongqing 400016, China
| | - Yuhao Duan
- Department of Neurosurgery Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400010, China; (A.P.)
- Center for Neuroscience Research, Chongqing Medical University, Chongqing 400016, China
| | - Mi Zhang
- Department of Neurosurgery Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400010, China; (A.P.)
- Center for Neuroscience Research, Chongqing Medical University, Chongqing 400016, China
| | - Hui Gan
- Department of Neurosurgery Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400010, China; (A.P.)
- Center for Neuroscience Research, Chongqing Medical University, Chongqing 400016, China
| | - Xuan Zhai
- Department of Neurosurgery Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400010, China; (A.P.)
- Center for Neuroscience Research, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
8
|
Desmond LW, Holbrook EM, Wright CTO, Zambrano CA, Stamper CE, Bohr AD, Frank MG, Podell BK, Moreno JA, MacDonald AS, Reber SO, Hernández-Pando R, Lowry CA. Effects of Mycobacterium vaccae NCTC 11659 and Lipopolysaccharide Challenge on Polarization of Murine BV-2 Microglial Cells. Int J Mol Sci 2023; 25:474. [PMID: 38203645 PMCID: PMC10779110 DOI: 10.3390/ijms25010474] [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: 10/31/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Previous studies have shown that the in vivo administration of soil-derived bacteria with anti-inflammatory and immunoregulatory properties, such as Mycobacterium vaccae NCTC 11659, can prevent a stress-induced shift toward an inflammatory M1 microglial immunophenotype and microglial priming in the central nervous system (CNS). It remains unclear whether M. vaccae NCTC 11659 can act directly on microglia to mediate these effects. This study was designed to determine the effects of M. vaccae NCTC 11659 on the polarization of naïve BV-2 cells, a murine microglial cell line, and BV-2 cells subsequently challenged with lipopolysaccharide (LPS). Briefly, murine BV-2 cells were exposed to 100 µg/mL whole-cell, heat-killed M. vaccae NCTC 11659 or sterile borate-buffered saline (BBS) vehicle, followed, 24 h later, by exposure to 0.250 µg/mL LPS (Escherichia coli 0111: B4; n = 3) in cell culture media vehicle (CMV) or a CMV control condition. Twenty-four hours after the LPS or CMV challenge, cells were harvested to isolate total RNA. An analysis using the NanoString platform revealed that, by itself, M. vaccae NCTC 11659 had an "adjuvant-like" effect, while exposure to LPS increased the expression of mRNAs encoding proinflammatory cytokines, chemokine ligands, the C3 component of complement, and components of inflammasome signaling such as Nlrp3. Among LPS-challenged cells, M. vaccae NCTC 11659 had limited effects on differential gene expression using a threshold of 1.5-fold change. A subset of genes was assessed using real-time reverse transcription polymerase chain reaction (real-time RT-PCR), including Arg1, Ccl2, Il1b, Il6, Nlrp3, and Tnf. Based on the analysis using real-time RT-PCR, M. vaccae NCTC 11659 by itself again induced "adjuvant-like" effects, increasing the expression of Il1b, Il6, and Tnf while decreasing the expression of Arg1. LPS by itself increased the expression of Ccl2, Il1b, Il6, Nlrp3, and Tnf while decreasing the expression of Arg1. Among LPS-challenged cells, M. vaccae NCTC 11659 enhanced LPS-induced increases in the expression of Nlrp3 and Tnf, consistent with microglial priming. In contrast, among LPS-challenged cells, although M. vaccae NCTC 11659 did not fully prevent the effects of LPS relative to vehicle-treated control conditions, it increased Arg1 mRNA expression, suggesting that M. vaccae NCTC 11659 induces an atypical microglial phenotype. Thus, M. vaccae NCTC 11659 acutely (within 48 h) induced immune-activating and microglial-priming effects when applied directly to murine BV-2 microglial cells, in contrast to its long-term anti-inflammatory and immunoregulatory effects observed on the CNS when whole-cell, heat-killed preparations of M. vaccae NCTC 11659 were given peripherally in vivo.
Collapse
Affiliation(s)
- Luke W. Desmond
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
| | - Evan M. Holbrook
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
| | - Caelan T. O. Wright
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
| | - Cristian A. Zambrano
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
| | - Christopher E. Stamper
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
| | - Adam D. Bohr
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
| | - Matthew G. Frank
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Brendan K. Podell
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO 80523, USA;
| | - Julie A. Moreno
- Prion Research Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA;
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
- Center for Healthy Aging, Colorado State University, Fort Collins, CO 80523, USA
| | - Andrew S. MacDonald
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9NT, UK;
| | - Stefan O. Reber
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, 89081 Ulm, Germany;
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional De Ciencias Médicas Y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico;
| | - Christopher A. Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
- Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO 80309, USA
- Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
9
|
Hua W, Ma S, Pang Y, Liu Q, Wang Y, Liu Z, Zhao N, Ren N, Jin S, Wang B, Song Y, Qi J. Intracerebral Hemorrhage-Induced Brain Injury: the Role of Lysosomal-Associated Transmembrane Protein 5. Mol Neurobiol 2023; 60:7060-7079. [PMID: 37525083 DOI: 10.1007/s12035-023-03484-8] [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: 01/22/2023] [Accepted: 07/05/2023] [Indexed: 08/02/2023]
Abstract
Intracerebral hemorrhage (ICH) is a lethal stroke with high mortality or disability. However, effective therapy for ICH damage is generally lacking. Previous investigations have suggested that lysosomal protein transmembrane 5 (LAPTM5) is involved in various pathological processes, including autophagy, apoptosis, and inflammation. In this study, we aimed to identify the expression and functions of LAPTM5 in collagenase-induced ICH mouse models and hemoglobin-induced cell models. We found that LAPTM5 was highly expressed in brain tissues around the hematoma, and double immunostaining studies showed that LAPTM5 was co-expressed with microglia cells, neurons, and astrocytes. Following ICH, the mice presented increased brain edema, blood-brain barrier permeability, and neurological deficits, while pathological symptoms were alleviated after the LAPTM5 knockdown. Adeno-associated virus 9-mediated downregulation of LAPTM5 also improves ICH-induced secondary cerebral damage, including neuronal degeneration, the polarization of M1-like microglia, and inflammatory cascades. Furthermore, LAPTM5 promoted activation of the nuclear factor kappa-B (NF-κB) pathway in response to neuroinflammation. Further investigations indicated that brain injury improved by LAPTM5 knockdown was further exacerbated after the overexpression of receptor-interacting protein kinase 1 (RIP1), which is revealed to trigger the NF-κB pathway. In vitro experiments demonstrated that LAPTM5 silencing inhibited hemoglobin-induced cell function and confirmed regulation between RIP1 and LAPTM5. In conclusion, the present study indicates that LAPTM5 may act as a positive regulator in the context of ICH by modulating the RIP1/NF-κB pathway. Thus, it may be a candidate gene for further study of molecular or therapeutic targets.
Collapse
Affiliation(s)
- Wei Hua
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Shuainan Ma
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yuxin Pang
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Qi Liu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yueying Wang
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Zhiyi Liu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Nan Zhao
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Naixin Ren
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Sinan Jin
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Benshuai Wang
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yuejia Song
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| | - Jiping Qi
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
10
|
Wang Y, Liu W, Geng P, Du W, Guo C, Wang Q, Zheng GQ, Jin X. Role of Crosstalk between Glial Cells and Immune Cells in Blood-Brain Barrier Damage and Protection after Acute Ischemic Stroke. Aging Dis 2023; 15:2507-2525. [PMID: 37962453 PMCID: PMC11567273 DOI: 10.14336/ad.2023.1010] [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: 06/29/2023] [Accepted: 10/10/2023] [Indexed: 11/15/2023] Open
Abstract
Blood-brain barrier (BBB) damage is the main pathological basis for acute ischemic stroke (AIS)-induced cerebral vasogenic edema and hemorrhagic transformation (HT). Glial cells, including microglia, astrocytes, and oligodendrocyte precursor cells (OPCs)/oligodendrocytes (OLs) play critical roles in BBB damage and protection. Recent evidence indicates that immune cells also have an important role in BBB damage, vasogenic edema and HT. Therefore, regulating the crosstalk between glial cells and immune cells would hold the promise to alleviate AIS-induced BBB damage. In this review, we first introduce the roles of glia cells, pericytes, and crosstalk between glial cells in the damage and protection of BBB after AIS, emphasizing the polarization, inflammatory response and crosstalk between microglia, astrocytes, and other glia cells. We then describe the role of glial cell-derived exosomes in the damage and protection of BBB after AIS. Next, we specifically discuss the crosstalk between glial cells and immune cells after AIS. Finally, we propose that glial cells could be a potential target for alleviating BBB damage after AIS and we discuss some molecular targets and potential strategies to alleviate BBB damage by regulating glial cells after AIS.
Collapse
Affiliation(s)
- Yihui Wang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Wencao Liu
- Shanxi Provincial People's Hospital, Taiyuan 030001, China.
| | - Panpan Geng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Weihong Du
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Chun Guo
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, UK.
| | - Qian Wang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Guo-qing Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Xinchun Jin
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
11
|
Song X, Ding Q, Wei W, Zhang J, Sun R, Yin L, Liu S, Pu Y. Peptide-Functionalized Prussian Blue Nanomaterial for Antioxidant Stress and NIR Photothermal Therapy against Alzheimer's Disease. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206959. [PMID: 37322406 DOI: 10.1002/smll.202206959] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 05/18/2023] [Indexed: 06/17/2023]
Abstract
Excessive accumulations of reactive oxygen species (ROS) and amyloid-β (Aβ) protein are closely associated with the complex pathogenesis of Alzheimer's disease (AD). Therefore, approaches that synergistically exert elimination of ROS and dissociation of Aβ fibrils are effective therapeutic strategies for correcting the AD microenvironment. Herein, a novel near infrared (NIR) responsive Prussian blue-based nanomaterial (PBK NPs) is established with excellent antioxidant activity and photothermal effect. PBK NPs possess similar activities to multiple antioxidant enzymes, including superoxide dismutase, peroxidase, and catalase, which can eliminate massive ROS and relieve oxidative stress. Under the NIR irradiation, PBK NPs can generate local heat to disaggregate Aβ fibrils efficiently. By modifying CKLVFFAED peptide, PBK NPs display obvious targeting ability for blood-brain barrier penetration and Aβ binding. Furthermore, in vivo studies demonstrate that PBK NPs have outstanding ability to decompose Aβ plaques and alleviate neuroinflammation in AD mouse model. Overall, PBK NPs provide evident neuroprotection by reducing ROS levels and regulating Aβ deposition, and may accelerate the development of multifunctional nanomaterials for delaying the progression of AD.
Collapse
Affiliation(s)
- Xiaolei Song
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| | - Qin Ding
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| | - Wei Wei
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, P. R. China
- State Key Laboratory of Bioelectronics, Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, P. R. China
| | - Juan Zhang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| | - Rongli Sun
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| | - Songqin Liu
- State Key Laboratory of Bioelectronics, Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, P. R. China
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| |
Collapse
|
12
|
Pavlichenko M, Lafrenaye AD. The Central Fluid Percussion Brain Injury in a Gyrencephalic Pig Brain: Scalable Diffuse Injury and Tissue Viability for Glial Cell Immunolabeling following Long-Term Refrigerated Storage. Biomedicines 2023; 11:1682. [PMID: 37371777 PMCID: PMC10295711 DOI: 10.3390/biomedicines11061682] [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: 05/16/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Traumatic brain injury (TBI) affects millions of people annually; however, our knowledge of the diffuse pathologies associated with TBI is limited. As diffuse pathologies, including axonal injury and neuroinflammatory changes, are difficult to visualize in the clinical population, animal models are used. In the current study, we used the central fluid percussion injury (CFPI) model in a micro pig to study the potential scalability of these diffuse pathologies in a gyrencephalic brain of a species with inflammatory systems very similar to humans. We found that both axonal injury and microglia activation within the thalamus and corpus callosum are positively correlated with the weight-normalized pressure pulse, while subtle changes in blood gas and mean arterial blood pressure are not. We also found that the majority of tissue generated up to 10 years previously is viable for immunofluorescent labeling after long-term refrigeration storage. This study indicates that a micro pig CFPI model could allow for specific investigations of various degrees of diffuse pathological burdens following TBI.
Collapse
Affiliation(s)
- Mark Pavlichenko
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298-0709, USA
| | - Audrey D. Lafrenaye
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298-0709, USA
- Richmond Veterans Affairs Medical Center, Richmond, VA 23249-4915, USA
| |
Collapse
|
13
|
Jing S, Li H, Xu H. Mesenchymal Stem Cell Derived Exosomes Therapy in Diabetic Wound Repair. Int J Nanomedicine 2023; 18:2707-2720. [PMID: 37250470 PMCID: PMC10216860 DOI: 10.2147/ijn.s411562] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 05/15/2023] [Indexed: 05/31/2023] Open
Abstract
Nowadays, refractory diabetic wounds cause a worldwide medical burden. Mesenchymal stem cells derived exosomes (MSC-Exos) show promise as a solid alternative to existing therapeutics in the latest researches, since MSC-Exos share similar biologic activity but less immunogenicity when compared with MSCs. To facilitate further understanding and application, it is essential to summarize the current progress and limitations of MSC-Exos in the treatment of diabetic wounds. In this review, we introduce the effects of different MSC-Exos on diabetic wounds according to their origins and contents and discuss the specific experimental conditions, target wound cells/pathways, and specific mechanisms. In addition, this paper focuses on the combination of MSC-Exos and biomaterials, which improves the efficacy and utilization of MSC-Exos therapy. Together, exosome therapy has high clinical value and application prospects, both in its role and in combination with biomaterials, while novel drugs or molecules loaded into exosomes as carriers targeting wound cells will be development trends.
Collapse
Affiliation(s)
- Shengyu Jing
- Department of Vascular Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, People’s Republic of China
| | - Hongjie Li
- Department of Vascular Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, People’s Republic of China
| | - Hongbo Xu
- Department of Vascular Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
14
|
Zhang Y, Jia P, Wang K, Zhang Y, Lv Y, Fan P, Yang L, Zhang S, Wang T, Zhao J, Lv H, Chen X, Liu Y, Wei H, Zhang P. Lactate modulates microglial inflammatory responses after oxygen-glucose deprivation through HIF-1α-mediated inhibition of NF-κB. Brain Res Bull 2023; 195:1-13. [PMID: 36746287 DOI: 10.1016/j.brainresbull.2023.02.002] [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: 05/06/2022] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 02/07/2023]
Abstract
Metabolic adaption drives microglial inflammatory responses, and lactate shapes immunological and inflammatory states. However, whether lactate was involved in the regulation of microglial inflammatory responses after cerebral ischemia remains unclear. In this study, the expression of iNOS, arginase-1, phosphorylated NF-κB p65 and IκB-α, and HIF-1α in BV2 cells after oxygen-glucose deprivation (OGD) were detected by western blotting and immunofluorescence. The mRNA levels of microglial responsive markers and inflammatory factors were assessed by real-time-qPCR. The effect of lactate-treated BV2 cells on the survival of primary neurons was observed using transwell co-culture. The results showed that the protein levels of iNOS and arginase-1, the ratio of mRNA levels of iNOS/CD206, CD86/Ym1, IL-6/IL-10, TNF-α/IL-10 and the mRNA levels of IL-6 and TNF-α, as well as the protein levels of phosphorylated NF-κB p65 and IκB-α, were increased after OGD. Lactate treatment inhibited the OGD-induced increase in the protein levels of iNOS, phosphorylated NF-κB p65 and IκB-α, as well as iNOS/CD206, CD86/Ym1, IL-6/IL-10, TNF-α/IL-10, IL-6 and TNF-α mRNA levels in BV2 cells, while promoted arginase-1 protein expression as well as IL-10 and TGF-β mRNA level. Interestingly, lactate activated HIF-1α and the HIF-1α inhibitor YC-1 reversed the effect of lactate on levels of microglial responsive markers and phosphorylated NF-κB p65 and IκB-α in BV2 cells. Moreover, knockdown of HIF-1α by lentivirus-delivered shRNA also reversed the effect of lactate on phosphorylated NF-κB p65 and IκB-α in BV2 cells after OGD. Finally, and importantly, lactate-treated BV2 microglia increased the viability and decreased the apoptosis of neurons after OGD. These findings revealed that lactate inhibited NF-κB pathway and skewed BV2 microglia toward the protective response through activation of HIF-1α after OGD, thereby improving neuronal survival.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Pengyu Jia
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Kui Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Yan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Yuying Lv
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Pei Fan
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Liufei Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Shuyue Zhang
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University, Xi'an 710061, China
| | - Tianyue Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Jing Zhao
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Haixia Lv
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xinlin Chen
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yong Liu
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University, Xi'an 710061, China
| | - Haidong Wei
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| | - Pengbo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| |
Collapse
|
15
|
Zhao L, Meng F, Li Y, Liu S, Xu M, Chu F, Li C, Yang X, Luo L. Multivalent Nanobody Conjugate with Rigid, Reactive Oxygen Species Scavenging Scaffold for Multi-Target Therapy of Alzheimer's Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210879. [PMID: 36786375 DOI: 10.1002/adma.202210879] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/08/2023] [Indexed: 05/17/2023]
Abstract
Efficient therapeutic strategies that concurrently target both Aβ aggregation and oxidative stress in the Alzheimer's disease (AD) microenvironment emerge as a cutting-edge tool to combat the intricate pathogenesis of AD. Here, a multivalent nanobody conjugate with rigid, reactive oxygen species (ROS) scavenging scaffold is developed to achieve simultaneous Aβ amyloidogenesis mitigation, ROS elimination, and Aβ plaque clearance. Grafting Aβ segment (33-GLMVGGVVIA-42) into the third complementary-determining region of a parent nanobody generates an engineered nanobody NB that can recognize Aβ and inhibit its aggregation through homotypic interactions. NB is further genetically modified with a fragment of human interleukin-1β (163-VQGEESNDK-171), so that the obtained fusion nanobody NBIL can also facilitate the Aβ clearance by microglia. Linking NBIL covalently onto a rigid, ROS scavenging scaffold poly(deca-4,6-diynedioic acid) (PDDA) creates the multivalent nanobody conjugate PNBIL, which not only boosts the binding affinity between NBIL and Aβ aggregates for nearly 100 times but also possesses a long-term capability of oxidative stress alleviation, inflammation reduction, and neuron protection. PNBIL has significantly attenuated symptoms on two AD mouse models through amyloidogenesis inhibition and AD microenvironment modulation, validating that the multivalent nanobody conjugate design based on combinatory nanobody and molecular engineering is a promising approach of multi-target therapeutic strategies.
Collapse
Affiliation(s)
- Liyuan Zhao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Fanling Meng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Yingjie Li
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P. R. China
| | - Sujuan Liu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Mengmeng Xu
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P. R. China
| | - Fan Chu
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P. R. China
| | - Chuanzhou Li
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P. R. China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Liang Luo
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| |
Collapse
|
16
|
Shan XQ, Luo YY, Chang J, Song JJ, Hao N, Zhao L. Immunomodulation: The next target of mesenchymal stem cell-derived exosomes in the context of ischemic stroke. World J Stem Cells 2023; 15:52-70. [PMID: 37007453 PMCID: PMC10052343 DOI: 10.4252/wjsc.v15.i3.52] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/19/2023] [Accepted: 02/28/2023] [Indexed: 03/23/2023] Open
Abstract
Ischemic stroke (IS) is the most prevalent form of brain disease, characterized by high morbidity, disability, and mortality. However, there is still a lack of ideal prevention and treatment measures in clinical practice. Notably, the transplantation therapy of mesenchymal stem cells (MSCs) has been a hot research topic in stroke. Nevertheless, there are risks associated with this cell therapy, including tumor formation, coagulation dysfunction, and vascular occlusion. Also, a growing number of studies suggest that the therapeutic effect after transplantation of MSCs is mainly attributed to MSC-derived exosomes (MSC-Exos). And this cell-free mediated therapy appears to circumvent many risks and difficulties when compared to cell therapy, and it may be the most promising new strategy for treating stroke as stem cell replacement therapy. Studies suggest that suppressing inflammation via modulation of the immune response is an additional treatment option for IS. Intriguingly, MSC-Exos mediates the inflammatory immune response following IS by modulating the central nervous system, the peripheral immune system, and immunomodulatory molecules, thereby promoting neurofunctional recovery after stroke. Thus, this paper reviews the role, potential mechanisms, and therapeutic potential of MSC-Exos in post-IS inflammation in order to identify new research targets.
Collapse
Affiliation(s)
- Xiao-Qian Shan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yong-Yin Luo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Jun Chang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Jing-Jing Song
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Nan Hao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Lan Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| |
Collapse
|
17
|
Mason ER, Soni DM, Chu S. Microglial Phagocytosis/Cell Health High-Content Assay. Curr Protoc 2023; 3:e724. [PMID: 36971657 PMCID: PMC10433541 DOI: 10.1002/cpz1.724] [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] [Indexed: 03/29/2023]
Abstract
We report a microglial phagocytosis/cell health high-content assay that has been used to test small molecule chemical probes and support our drug discovery projects targeting microglia for Alzheimer's disease therapy. The assay measures phagocytosis and cell health (cell count and nuclear intensity) simultaneously in 384-well plates processed with an automatic liquid handler. The mix-and-read live cell imaging assay is highly reproducible with capacity to meet drug discovery research needs. Assay procedures take 4 days including plating cells, treating cells, adding pHrodo-myelin/membrane debris to cells for phagocytosis, staining cell nuclei before performing high-content imaging, and analysis. Three selected parameters are measured from cells: 1) mean total fluorescence intensity per cell of pHrodo-myelin/membrane debris in phagocytosis vesicles to quantify phagocytosis; 2) cell counts per well (measuring compound effects on proliferation and cell death); and 3) average nuclear intensity (measuring compound induced apoptosis). The assay has been used on HMC3 cells (an immortalized human microglial cell line), BV2 cells (an immortalized mouse microglial cell line), and primary microglia isolated from mouse brains. Simultaneous measurements of phagocytosis and cell health allow for the distinction of compound effects on regulation of phagocytosis from cellular stress/toxicity related changes, a distinguishing feature of the assay. The combination of cell counts and nuclear intensity as indicators of cell health is also an effective way to measure cell stress and compound cytotoxicity, which may have broad applications as simultaneous profiling measurements for other phenotypic assays. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol: Microglial phagocytosis/cell health high-content assay protocol Support Protocol: Procedures to isolate myelin/membrane debris from mouse brain and label with pHrodo.
Collapse
Affiliation(s)
- Emily R Mason
- Division of Clinical Pharmacology, Department of Medicine, IUSM-Purdue TREAT-AD Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Disha M Soni
- Department of Radiology & Imaging Sciences, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana
| | - Shaoyou Chu
- Division of Clinical Pharmacology, Department of Medicine, IUSM-Purdue TREAT-AD Center, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
18
|
Aminochrome Induces Neuroinflammation and Dopaminergic Neuronal Loss: A New Preclinical Model to Find Anti-inflammatory and Neuroprotective Drugs for Parkinson's Disease. Cell Mol Neurobiol 2023; 43:265-281. [PMID: 34988761 DOI: 10.1007/s10571-021-01173-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/21/2021] [Indexed: 01/07/2023]
Abstract
Studies have suggested aminochrome as an endogenous neurotoxin responsible for the dopaminergic neuron degeneration in Parkinson's disease (PD). However, neuroinflammation, an important alteration in PD pathogenesis, has been strictly induced in vitro by aminochrome. The aim of this study was to characterize the neuroinflammation induced in vivo by aminochrome. Wistar rats (male, 250-270 g) received a unilateral single dose by stereotaxic injection of saline into three sites in the striatum in the negative control group, or 32 nmol 6-hydroxydopamine (6-OHDA) in the positive control, or 6 nmol aminochrome. After 14 days, histological and molecular analyses were performed. We observed by immunofluorescence that aminochrome, as well as 6-OHDA, induced an increase in the number of Iba-1+ cells and in the number of activated (Iba-1+/ CD68+) microglia. An increase in the number of S100b+ cells and in the GFAP expression were also evidenced in the striatum and the SNpc of animals from aminochrome and positive control group. Dopaminergic neuronal loss was marked by reduction of TH+ cells and confirmed with reduction in the number of Nissl-stained neurons in the SNpc of rats from aminochrome and positive control groups. In addition, we observed by qPCR that aminocrhome induced an increase in the levels of IL-1β, TNF-α, NLRP3, CCL5 and CCR2 mRNA in the SNpc. This work provides the first evidence of microgliosis, astrogliosis and neuroinflammation induced by aminochrome in an in vivo model. Since aminochrome is an endogenous molecule derived from dopamine oxidation present in the targeted neurons in PD, these results reinforce the potential of aminochrome as a useful preclinical model to find anti-inflammatory and neuroprotective drugs for PD. Aminochrome induced dopaminergic neuronal loss, microglial activation, astroglial activation and neuroinflammation marked by an increase in NLRP3, IL1β, TNF-α, CCL2, CCL5 and CCR2.
Collapse
|
19
|
Sun H, Li L, Lao I, Li X, Xu B, Cao Y, Jin W. Single-cell RNA sequencing reveals cellular and molecular reprograming landscape of gliomas and lung cancer brain metastases. Clin Transl Med 2022; 12:e1101. [PMID: 36336787 PMCID: PMC9637666 DOI: 10.1002/ctm2.1101] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Brain malignancies encompass gliomas and brain metastases originating from extracranial tumours including lung cancer. Approximately 50% of patients with lung adenocarcinoma (LUAD) will eventually develop brain metastases. However, the specific characteristics of gliomas and lung-to-brain metastases (LC) are largely unknown. METHODS We applied single-cell RNA sequencing to profile immune and nonimmune cells in 4 glioma and 10 LC samples. RESULTS Our analysis revealed that tumour microenvironment (TME) cells are present in heterogeneous subpopulations. LC reprogramed cells into immune suppressed state, including microglia, macrophages, endothelial cells, and CD8+ T cells, with unique cell proportions and gene signatures. Particularly, we identified that a subset of macrophages was associated with poor prognosis. ROS (reactive oxygen species)-producing neutrophils was found to participant in angiogenesis. Furthermore, endothelial cells participated in active communication with fibroblasts. Metastatic epithelial cells exhibited high heterogeneity in chromosomal instability (CIN) and cell population. CONCLUSIONS Our findings provide a comprehensive understanding of the heterogenicity of the tumor microenvironment and tumour cells and it will be crucial for successful immunotherapy development for brain metastasis of lung cancer.
Collapse
Affiliation(s)
- He‐Fen Sun
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Liang‐Dong Li
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghaiChina
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - I‐Weng Lao
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xuan Li
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Bao‐Jin Xu
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Yi‐Qun Cao
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Wei Jin
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
20
|
Guo Y, Dai W, Zheng Y, Qiao W, Chen W, Peng L, Zhou H, Zhao T, Liu H, Zheng F, Sun P. Mechanism and Regulation of Microglia Polarization in Intracerebral Hemorrhage. Molecules 2022; 27:molecules27207080. [PMID: 36296682 PMCID: PMC9611828 DOI: 10.3390/molecules27207080] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/17/2022] [Accepted: 10/17/2022] [Indexed: 11/24/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is the most lethal subtype of stroke, but effective treatments are lacking, and neuroinflammation plays a key role in the pathogenesis. In the innate immune response to cerebral hemorrhage, microglia first appear around the injured tissue and are involved in the inflammatory cascade response. Microglia respond to acute brain injury by being activated and polarized to either a typical M1-like (pro-inflammatory) or an alternative M2-like (anti-inflammatory) phenotype. These two polarization states produce pro-inflammatory or anti-inflammatory. With the discovery of the molecular mechanisms and key signaling molecules related to the polarization of microglia in the brain, some targets that regulate the polarization of microglia to reduce the inflammatory response are considered a treatment for secondary brain tissue after ICH damage effective strategies. Therefore, how to promote the polarization of microglia to the M2 phenotype after ICH has become the focus of attention in recent years. This article reviews the mechanism of action of microglia’s M1 and M2 phenotypes in secondary brain injury after ICH. Moreover, it discusses compounds and natural pharmaceutical ingredients that can polarize the M1 to the M2 phenotype.
Collapse
Affiliation(s)
- Yuting Guo
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Weibo Dai
- Department of Pharmacy, Zhongshan Hospital of traditional Chinese Medicine, Zhongshan 528401, China
| | - Yan Zheng
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China
| | - Weilin Qiao
- Zhongshan Zhongzhi Pharmaceutical Group Co., Ltd., Zhongshan 528411, China
| | - Weixuan Chen
- Zhongshan Zhongzhi Pharmaceutical Group Co., Ltd., Zhongshan 528411, China
| | - Lihua Peng
- Zhongshan Zhongzhi Pharmaceutical Group Co., Ltd., Zhongshan 528411, China
| | - Hua Zhou
- The Second School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Tingting Zhao
- School of Foreign Languages, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Correspondence: (T.Z.); (H.L.); (F.Z.); (P.S.)
| | - Huimin Liu
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Correspondence: (T.Z.); (H.L.); (F.Z.); (P.S.)
| | - Feng Zheng
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362002, China
- Correspondence: (T.Z.); (H.L.); (F.Z.); (P.S.)
| | - Peng Sun
- Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Correspondence: (T.Z.); (H.L.); (F.Z.); (P.S.)
| |
Collapse
|
21
|
Mo Y, Xu W, Fu K, Chen H, Wen J, Huang Q, Guo F, Mo L, Yan J. The dual function of microglial polarization and its treatment targets in ischemic stroke. Front Neurol 2022; 13:921705. [PMID: 36212660 PMCID: PMC9538667 DOI: 10.3389/fneur.2022.921705] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 07/14/2022] [Indexed: 11/23/2022] Open
Abstract
Stroke is the leading cause of disability and death worldwide, with ischemic stroke occurring in ~5% of the global population every year. Recently, many studies have been conducted on the inflammatory response after stroke. Microglial/macrophage polarization has a dual function and is critical to the pathology of ischemic stroke. Microglial/macrophage activation is important in reducing neuronal apoptosis, enhancing neurogenesis, and promoting functional recovery after ischemic stroke. In this review, we investigate the physiological characteristics and functions of microglia in the brain, the activation and phenotypic polarization of microglia and macrophages after stroke, the signaling mechanisms of polarization states, and the contribution of microglia to brain pathology and repair. We summarize recent advances in stroke-related microglia research, highlighting breakthroughs in therapeutic strategies for microglial responses after stroke, thereby providing new ideas for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yong Mo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Weilin Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kaijing Fu
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Hainan Chen
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jing Wen
- Department of Rheumatism, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qianrong Huang
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Fangzhou Guo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Ligen Mo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Ligen Mo
| | - Jun Yan
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
- *Correspondence: Jun Yan
| |
Collapse
|
22
|
Li YF, Ren X, Zhang L, Wang YH, Chen T. Microglial polarization in TBI: Signaling pathways and influencing pharmaceuticals. Front Aging Neurosci 2022; 14:901117. [PMID: 35978950 PMCID: PMC9376354 DOI: 10.3389/fnagi.2022.901117] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) is a serious disease that threatens life and health of people. It poses a great economic burden on the healthcare system. Thus, seeking effective therapy to cure a patient with TBI is a matter of great urgency. Microglia are macrophages in the central nervous system (CNS) and play an important role in neuroinflammation. When TBI occurs, the human body environment changes dramatically and microglia polarize to one of two different phenotypes: M1 and M2. M1 microglia play a role in promoting the development of inflammation, while M2 microglia play a role in inhibiting inflammation. How to regulate the polarization direction of microglia is of great significance for the treatment of patients with TBI. The polarization of microglia involves many cellular signal transduction pathways, such as the TLR-4/NF-κB, JAK/STAT, HMGB1, MAPK, and PPAR-γ pathways. These provide a theoretical basis for us to seek therapeutic drugs for the patient with TBI. There are several drugs that target these pathways, including fingolimod, minocycline, Tak-242 and erythropoietin (EPO), and CSF-1. In this study, we will review signaling pathways involved in microglial polarization and medications that influence this process.
Collapse
Affiliation(s)
| | | | | | - Yu-Hai Wang
- Department of Neurosurgery, The 904th Hospital of PLA, Medical School of Anhui Medical University, Wuxi, China
| | - Tao Chen
- Department of Neurosurgery, The 904th Hospital of PLA, Medical School of Anhui Medical University, Wuxi, China
| |
Collapse
|
23
|
Zhang X, Tu D, Li S, Li N, Li D, Gao Y, Tian L, Liu J, Zhang X, Hong JS, Hou L, Zhao J, Wang Q. A novel synthetic peptide SVHRSP attenuates dopaminergic neurodegeneration by inhibiting NADPH oxidase-mediated neuroinflammation in experimental models of Parkinson's disease. Free Radic Biol Med 2022; 188:363-374. [PMID: 35760232 DOI: 10.1016/j.freeradbiomed.2022.06.241] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 01/21/2023]
Abstract
Current treatment of Parkinson's disease (PD) ameliorates symptoms but fails to block disease progression. This study was conducted to explore the protective effects of SVHRSP, a synthetic heat-resistant peptide derived from scorpion venom, against dopaminergic neurodegeneration in experimental models of PD. Results showed that SVHRSP dose-dependently reduced the loss of dopaminergic neuron in the nigrostriatal pathway and motor impairments in both rotenone and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine/probenecid (MPTP/p)-induced mouse PD models. Microglial activation and imbalance of M1/M2 polarization were also abrogated by SVHRSP in both models. In rotenone-treated primary midbrain neuron-glial cultures, loss of dopaminergic neuron and microglial activation were mitigated by SVHRSP. Furthermore, lipopolysaccharide (LPS)-elicited microglial activation, M1 polarization and related dopaminergic neurodegeneration in primary cultures were also abrogated by SVHRSP, suggesting that inhibition of microglial activation contributed to SVHRSP-afforded neuroprotection. Mechanistic studies revealed that SVHRSP blocked both LPS- and rotenone-induced microglial NADPH oxidase (NOX2) activation by preventing membrane translocation of cytosolic subunit p47phox. NOX2 knockdown by siRNA markedly attenuated the inhibitory effects of SVHRSP against LPS- and rotenone-induced gene expressions of proinflammatory factors and related neurotoxicity. Altogether, SVHRSP protects dopaminergic neurons by blocking NOX2-mediated microglial activation in experimental PD models, providing experimental basis for the screening of clinical therapeutic drugs for PD.
Collapse
Affiliation(s)
- Xiaomeng Zhang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Dezhen Tu
- Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Sheng Li
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Na Li
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Donglai Li
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Yun Gao
- Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Lu Tian
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Jianing Liu
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Xuan Zhang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Jau-Shyong Hong
- Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Liyan Hou
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Jie Zhao
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China.
| | - Qingshan Wang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| |
Collapse
|
24
|
WKYMVm/FPR2 Alleviates Spinal Cord Injury by Attenuating the Inflammatory Response of Microglia. Mediators Inflamm 2022; 2022:4408099. [PMID: 35935810 PMCID: PMC9348919 DOI: 10.1155/2022/4408099] [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: 05/31/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022] Open
Abstract
Spinal cord injury (SCI) is a common traumatic disease of the nervous system. The pathophysiological process of SCI includes primary injury and secondary injuries. An excessive inflammatory response leads to secondary tissue damage, which in turn exacerbates cellular and organ dysfunction. Due to the irreversibility of primary injury, current research on SCI mainly focuses on secondary injury, and the inflammatory response is considered the primary target. Thus, modulating the inflammatory response has been suggested as a new strategy for the treatment of SCI. In this study, microglial cell lines, primary microglia, and a rat SCI model were used, and we found that WKYMVm/FPR2 plays an anti-inflammatory role and reduces tissue damage after SCI by suppressing the extracellular signal-regulated kinases 1 and 2 (ERK1/2) and nuclear factor-κB (NF-κB) signaling pathways. FPR2 was activated by WKYMVm, suppressing the secretion of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β) by inhibiting M1 microglial polarization. Moreover, FPR2 activation by WKYMVm could reduce structural disorders and neuronal loss in SCI rats. Overall, this study illustrated that the activation of FPR2 by WKYMVm repressed M1 microglial polarization by suppressing the ERK1/2 and NF-κB signaling pathways to alleviate tissue damage and locomotor decline after SCI. These findings provide further insight into SCI and help identify novel treatment strategies.
Collapse
|
25
|
Derivatives of Sarcodonin A Isolated from Sarcodon scabrosus Reversed LPS-induced M1 Polarization in Microglia through MAPK/NF-κB Pathway. Bioorg Chem 2022; 125:105854. [DOI: 10.1016/j.bioorg.2022.105854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/15/2022] [Accepted: 05/03/2022] [Indexed: 12/14/2022]
|
26
|
Watson N, Bonsack F, Sukumari-Ramesh S. Intracerebral Hemorrhage: The Effects of Aging on Brain Injury. Front Aging Neurosci 2022; 14:859067. [PMID: 35547620 PMCID: PMC9082316 DOI: 10.3389/fnagi.2022.859067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/01/2022] [Indexed: 12/25/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating subtype of stroke with high rates of mortality and morbidity. ICH patients often suffer devastating and debilitating neurological impairments, from which the majority of victims are unable to fully recover to functional independence. Unfortunately, there is no established medical therapy for ICH, which is partly attributed to the lack of understanding of the complex pathology of the disorder. Despite advanced age being a major risk factor of ICH, most preclinical studies on ICH employed young animal subjects. Due to this discrepancy, the molecular level changes in the aging brain after ICH are largely unknown, limiting the translation of preclinical studies into potential human treatments. The purpose of this review is to highlight the effects of advanced age on ICH- induced brain injury and recovery and to draw attention to current knowledge gaps, which warrant further investigation.
Collapse
|
27
|
The Pivotal Immunoregulatory Functions of Microglia and Macrophages in Glioma Pathogenesis and Therapy. JOURNAL OF ONCOLOGY 2022; 2022:8903482. [PMID: 35419058 PMCID: PMC9001141 DOI: 10.1155/2022/8903482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/24/2022] [Indexed: 12/16/2022]
Abstract
Gliomas are mixed solid tumors composed of both neoplastic and nonneoplastic cells. In glioma microenvironment, the most common nonneoplastic and infiltrating cells are macrophages and microglia. Microglia are the exact phagocytes of the central nervous system, whereas macrophages are myeloid immune cells that are depicted with ardent phagocytosis. Microglia are heterogeneously located in almost all nonoverlapping sections of the brain as well as the spinal cord, while macrophages are derived from circulating monocytes. Microglia and macrophages utilize a variety of receptors for the detection of molecules, particles, and cells that they engulf. Both microglia and peripheral macrophages interact directly with vessels both in the periphery of and within the tumor. In glioma milieu, normal human astrocytes, glioma cells, and microglia all exhibited the ability of phagocytosing glioma cells and precisely apoptotic tumor cells. Also, microglia and macrophages are robustly triggered by the glioma via the expression of chemoattractants such as monocyte chemoattractant protein, stromal-derived factor-1, and macrophage-colony stimulating factor. Glioma-associated microglia and/or macrophages positively correlated with glioma invasiveness, immunosuppression, and patients' poor outcome, making these cells a suitable target for immunotherapeutic schemes.
Collapse
|
28
|
Wan T, Huang Y, Gao X, Wu W, Guo W. Microglia Polarization: A Novel Target of Exosome for Stroke Treatment. Front Cell Dev Biol 2022; 10:842320. [PMID: 35356292 PMCID: PMC8959940 DOI: 10.3389/fcell.2022.842320] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/14/2022] [Indexed: 12/14/2022] Open
Abstract
The vast majority of cells in the human body are capable of secreting exosomes. Exosomes have become an important vehicle for signaling between cells. Exosomes secreted by different cells have some of the structural and functional properties of that cell and thus have different regulatory functions. A large number of recent experimental studies have shown that exosomes from different sources have different regulatory effects on stroke, and the mechanisms still need to be elucidated. Microglia are core members of central intrinsic immune regulatory cells, which play an important regulatory role in the pathogenesis and progression of stroke. M1 microglia cause neuroinflammation and induce neurotoxic effects, while M2 microglia inhibit neuroinflammation and promote neurogenesis, thus exerting a series of neuroprotective effects. It was found that there is a close link between exosomes and microglia polarization, and that exosome inclusions such as microRNAs play a regulatory role in the M1/M2 polarization of microglia. This research reviews the role of exosomes in the regulation of microglia polarization and reveals their potential value in stroke treatment.
Collapse
Affiliation(s)
- Teng Wan
- Hengyang Medical College, University of South China, Hengyang, China.,Sports Medicine Department, Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Yunling Huang
- Hengyang Medical College, University of South China, Hengyang, China
| | - Xiaoyu Gao
- Hengyang Medical College, University of South China, Hengyang, China
| | - Wanpeng Wu
- Shenzhen Futian District Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Weiming Guo
- Sports Medicine Department, Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
29
|
Zheng K, Lv B, Wu L, Wang C, Xu H, Li X, Wu Z, Zhao Y, Zheng Z. Protecting effect of emodin in experimental autoimmune encephalomyelitis mice by inhibiting microglia activation and inflammation via Myd88/PI3K/Akt/NF-κB signalling pathway. Bioengineered 2022; 13:9322-9344. [PMID: 35287559 PMCID: PMC9161934 DOI: 10.1080/21655979.2022.2052671] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is characterised by demyelination of the central nervous system. Emodin is an anthraquinone derivative with comprehensive anti-inflammatory, anti-cancer, and immunomodulatory effects and is widely used in the treatment of inflammatory, tumour, and immune system diseases. However, none of the clinical or experimental studies have explored the therapeutic efficacy of emodin in EAE/multiple sclerosis (MS). Thus, we evaluated the protective effect of emodin on EAE mediated via inhibition of microglia activation and inflammation. Wild-type mice were randomly divided into the normal control, EAE, low-dose emodin, and high-dose emodin groups. Clinical scores and pathological changes were assessed 21 days after immunisation. The network pharmacology approach was used to elucidate underlying mechanisms by using an online database. Molecular docking, polymerase chain reaction tests, western blotting, and immunofluorescence were performed to verify the network pharmacology results. An in vivo experiment showed that high-dose emodin ameliorated clinical symptoms, inflammatory cell infiltration, and myelination. Pharmacological network analysis showed AKT1 was the main target and that emodin played a key role in MS treatment mainly via the PI3K-Akt pathway. Molecular docking showed that emodin bound well with PI3K, AKT1, and NFKB1. Emodin decreased the expression of phosphorylated(p)-PI3K, p-Akt, NF-κB, and myeloid differentiation factor 88 and the levels of markers (CD86 and CD206) in M1- and M2-phenotype microglia in EAE. Thus, emodin inhibited microglial activation and exhibited anti-inflammatory and neuroprotective effects against EAE via the Myd88/PI3K/Akt/NF-κB signalling pathway. In conclusion, emodin has a promising role in EAE/MS treatment, warranting further detailed studies.
Collapse
Affiliation(s)
- Kenan Zheng
- The First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Baojiang Lv
- The First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lulu Wu
- The First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chen Wang
- Department of Traditional Chinese Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haoyou Xu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Xiaojun Li
- The Second Clinical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhibing Wu
- Department of Neurology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuanqi Zhao
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Zequan Zheng
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China.,Doctoral candidates with the same academic level of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
30
|
The Novel Nrf2 Activator Omaveloxolone Regulates Microglia Phenotype and Ameliorates Secondary Brain Injury after Intracerebral Hemorrhage in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4564471. [PMID: 35308167 PMCID: PMC8933082 DOI: 10.1155/2022/4564471] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 12/08/2021] [Accepted: 02/22/2022] [Indexed: 11/17/2022]
Abstract
The polarization of microglia is recognized as a crucial factor in reducing neuroinflammation and promoting hematoma clearance after intracerebral hemorrhage (ICH). Previous studies have revealed that redox components participate in the regulation of microglial polarization. Recently, the novel Nrf2 activator omaveloxolone (Omav) has been validated to improve neurological function in patients with neurodegenerative disorders by regulating antioxidant responses. In this study, we examined the efficacy of Omav in ICH. Omav significantly promoted Nrf2 nuclear accumulation and the expression of HO-1 and NQO1 in BV2 cells. In addition, both in vitro and in vivo experiments showed that Omav treatment inhibited M1-like activation and promoted the activation of the M2-like microglial phenotype. Omav inhibited OxyHb-induced ROS generation and preserved the function of mitochondria in BV2 cells. Intraperitoneal administration of Omav improved sensorimotor function in the ICH mouse model. Importantly, these effects were blocked by pretreatment with ML385, a selective inhibitor of Nrf2. Collectively, Omav modulated microglial polarization by activating Nrf2 and inhibiting ROS generation in ICH models, suggesting that it might be a promising drug candidate for the treatment of ICH.
Collapse
|
31
|
Cai M, Lin W. The Function of NF-Kappa B During Epilepsy, a Potential Therapeutic Target. Front Neurosci 2022; 16:851394. [PMID: 35360161 PMCID: PMC8961383 DOI: 10.3389/fnins.2022.851394] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 02/22/2022] [Indexed: 01/04/2023] Open
Abstract
The transcriptional regulator nuclear factor kappa B (NF-κB) modulates cellular biological activity by binding to promoter regions in the nucleus and transcribing various protein-coding genes. The NF-κB pathway plays a major role in the expressing genes related to inflammation, including chemokines, interleukins, and tumor necrosis factor. It also transcribes genes that can promote neuronal survival or apoptosis. Epilepsy is one of the most common brain disorders and it not only causes death worldwide but also affects the day-to-day life of affected individuals. While epilepsy has diverse treatment options, there remain patients who are not sensitive to the existing treatment methods. Recent studies have implicated the critical role of NF-κB in epilepsy. It is upregulated in neurons, glial cells, and endothelial cells, due to neuronal loss, glial cell proliferation, blood-brain barrier dysfunction, and hippocampal sclerosis through the glutamate and γ-aminobutyric acid imbalance, ion concentration changes, and other mechanisms. In this review, we summarize the functional changes caused by the upregulation of NF-κB in the central nervous system during different periods after seizures. This review is the first to deconvolute the complicated functions of NF-κB, and speculate that the regulation of NF-κB can be a safe and effective treatment strategy for epilepsy.
Collapse
|
32
|
Sesamol Attenuates Neuroinflammation by Regulating the AMPK/SIRT1/NF- κB Signaling Pathway after Spinal Cord Injury in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8010670. [PMID: 35035666 PMCID: PMC8758308 DOI: 10.1155/2022/8010670] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 11/23/2021] [Indexed: 12/11/2022]
Abstract
Inflammation is one of the crucial mechanisms mediating spinal cord injury (SCI) progress. Sesamol, a component of sesame oil, has anti-inflammatory activity, but its mechanism in SCI remains unclear. We investigated if the AMPK/SIRT1/NF-κB pathway participated in anti-inflammation of sesamol in SCI. Sesamol could inhibit neuronal apoptosis, reduce neuroinflammation, enhance M2 phenotype microglial polarization, and improved motor function recovery in mice after SCI. Furthermore, sesamol increased SIRT1 protein expression and p-AMPK/AMPK ratio, while it downregulated the p-p65/p65 ratio, indicating that sesamol treatment upregulated the AMPK/SIRT1 pathway and inhibited NF-κB activation. However, these effects were blocked by compound C which is a specific AMPK inhibitor. Together, the study suggests that sesamol is a potential drug for antineuroinflammation and improving locomotor functional recovery through regulation of the AMPK/SIRT1/NF-κB pathway in SCI.
Collapse
|
33
|
Zhang J, Li Z, Fan M, Jin W. Lipoxins in the Nervous System: Brighter Prospects for Neuroprotection. Front Pharmacol 2022; 13:781889. [PMID: 35153778 PMCID: PMC8826722 DOI: 10.3389/fphar.2022.781889] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/07/2022] [Indexed: 12/28/2022] Open
Abstract
Lipoxins (LXs) are generated from arachidonic acid and are involved in the resolution of inflammation and confer protection in a variety of pathological processes. In the nervous system, LXs exert an array of protective effects against neurological diseases, including ischemic or hemorrhagic stroke, neonatal hypoxia-ischemia encephalopathy, brain and spinal cord injury, Alzheimer's disease, multiple sclerosis, and neuropathic pain. Lipoxin administration is a potential therapeutic strategy in neurological diseases due to its notable efficiency and unique superiority regarding safety. Here, we provide an overview of LXs in terms of their synthesis, signaling pathways and neuroprotective evidence. Overall, we believe that, along with advances in lipoxin-related drug design, LXs will bring brighter prospects for neuroprotection.
Collapse
Affiliation(s)
- Jiayu Zhang
- Graduate School of Hebei Medical University, Shijiazhuang, China.,Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Zhe Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Mingyue Fan
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Wei Jin
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
34
|
Li R, Zhou Y, Zhang S, Li J, Zheng Y, Fan X. The natural (poly)phenols as modulators of microglia polarization via TLR4/NF-κB pathway exert anti-inflammatory activity in ischemic stroke. Eur J Pharmacol 2022; 914:174660. [PMID: 34863710 DOI: 10.1016/j.ejphar.2021.174660] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/04/2021] [Accepted: 11/29/2021] [Indexed: 12/24/2022]
Abstract
Increasing evidences suggest that inflammation plays a key role in the pathogenesis of stroke, a devastating disease second only to cardiac ischemia as a cause of death worldwide. Microglia are the first non-neuronal cells on the scene during the innate immune response to acute ischemic stroke. Microglia respond to acute brain injury by activating and developing classic M1-like (pro-inflammatory) or alternative M2-like (anti-inflammatory) phenotypes. M1 microglia produce pro-inflammatory cytokines to exacerbate neural death, astrocyte apoptosis, and blood brain barrier (BBB) disruption, while M2 microglia play the opposite role. NF-κB, a central regulator of the inflammatory response, was responsible for microglia M1 and M2 polarization. NF-κB p65 and p50 form a heterodimer to initiate a pro-inflammatory cytokine response, which enhances M1 activation and impair M2 response of microglia. TLR4, expressed on the surface of microglia, plays an important role in activating NF-κB, ultimately causing the M1 response of microglia. Therefore, modulation of microglial phenotypes via TLR4/NF-κB signaling pathway may be a promising therapeutic approach for ischemic stroke. Dietary (poly)phenols are present in various foods, which have shown promising protective effects on ischemic stroke. In vivo studies strongly suggest that many (poly)phenols have a pronounced impact on ischemic stroke, as demonstrated by lower neuroinflammation. Thus, this review focuses on the anti-inflammatory properties of dietary (poly)phenols and discusses their effects on the polarization of microglia through modulating TLR4/NF-κB signaling pathway in the ischemic stroke.
Collapse
Affiliation(s)
- Ruoqi Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yuan Zhou
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Shanshan Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jieying Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yingyi Zheng
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
35
|
Lan J, Li K, Gresham A, Miao J. Tanshinone IIA sodium sulfonate attenuates inflammation by upregulating circ-Sirt1 and inhibiting the entry of NF-κB into the nucleus. Eur J Pharmacol 2022; 914:174693. [PMID: 34896110 DOI: 10.1016/j.ejphar.2021.174693] [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] [Received: 05/31/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 01/07/2023]
Abstract
Inflammation is a biological process that exists in a large number of diseases. NF-κB has been proven to play a pivotal role in the development of inflammation. New drugs aimed at inhibiting the expression of NF-κB have gained attention from researchers. Sirt1 has an anti-inflammatory function, and the circRNA encoded by the Sirt1 gene may also play roles in the anti-inflammatory reaction of Sirt1. In the present study, LPS-treated RAW264.7 cells were used as an inflammatory cell model, and tanshinone IIA sodium sulfonate (TSS) was used as a therapeutic drug. We found that TSS downregulated LPS-induced TNF-α and IL-1β expression nearly threefold. LPS reduced Circ-sirt1 mRNA expression by one-third, while TSS started this phenomenon. In addition, overexpression/knockdown of Circ-sirt1 neutralized the function of TSS by regulating the translocation of NF-κB. Thus, we proved that TSS has an anti-inflammatory function by upregulating circ-Sirt1 and subsequently inhibiting the translocation of NF-κB. An in vivo experiment was also performed to confirm the protective function of TSS on inflammation. These results indicated that TSS is a potential treatment for inflammation.
Collapse
Affiliation(s)
- Jiao Lan
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Ke Li
- Henan General Hospital,Zhengzhou, China
| | | | - Jifei Miao
- School of Chemical Biology and Biotechnology, Peking University Shenzhen, Graduate School, Shenzhen, China.
| |
Collapse
|
36
|
Liu X, Lin J, Zhang H, Khan NU, Zhang J, Tang X, Cao X, Shen L. Oxidative Stress in Autism Spectrum Disorder-Current Progress of Mechanisms and Biomarkers. Front Psychiatry 2022; 13:813304. [PMID: 35299821 PMCID: PMC8921264 DOI: 10.3389/fpsyt.2022.813304] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/24/2022] [Indexed: 12/11/2022] Open
Abstract
Autism spectrum disorder (ASD) is a type of neurodevelopmental disorder that has been diagnosed in an increasing number of children around the world. Existing data suggest that early diagnosis and intervention can improve ASD outcomes. However, the causes of ASD remain complex and unclear, and there are currently no clinical biomarkers for autism spectrum disorder. More mechanisms and biomarkers of autism have been found with the development of advanced technology such as mass spectrometry. Many recent studies have found a link between ASD and elevated oxidative stress, which may play a role in its development. ASD is caused by oxidative stress in several ways, including protein post-translational changes (e.g., carbonylation), abnormal metabolism (e.g., lipid peroxidation), and toxic buildup [e.g., reactive oxygen species (ROS)]. To detect elevated oxidative stress in ASD, various biomarkers have been developed and employed. This article summarizes recent studies about the mechanisms and biomarkers of oxidative stress. Potential biomarkers identified in this study could be used for early diagnosis and evaluation of ASD intervention, as well as to inform and target ASD pharmacological or nutritional treatment interventions.
Collapse
Affiliation(s)
- Xukun Liu
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China.,Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Shenzhen, China
| | - Jing Lin
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Huajie Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Naseer Ullah Khan
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Jun Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Xiaoxiao Tang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Xueshan Cao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China.,Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Brain Disease and Big Data Research Institute, Shenzhen University, Shenzhen, China
| |
Collapse
|
37
|
Diethyl Succinate Modulates Microglial Polarization and Activation by Reducing Mitochondrial Fission and Cellular ROS. Metabolites 2021; 11:metabo11120854. [PMID: 34940612 PMCID: PMC8705220 DOI: 10.3390/metabo11120854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/29/2021] [Accepted: 12/04/2021] [Indexed: 12/14/2022] Open
Abstract
Succinate is a metabolite in the tricarboxylic acid cycle (TCA) which plays a central role in mitochondrial activity. Excess succinate is known to be transported out of the cytosol, where it activates a succinate receptor (SUCNR1) to enhance inflammation through macrophages in various contexts. In addition, the intracellular role of succinate beyond an intermediate metabolite and prior to its extracellular release is also important to the polarization of macrophages. However, the role of succinate in microglial cells has not been characterized. Lipopolysaccharide (LPS) stimulates the elevation of intracellular succinate levels. To reveal the function of intracellular succinate associated with LPS-stimulated inflammatory response in microglial cells, we assessed the levels of ROS, cytokine production and mitochondrial fission in the primary microglia pretreated with cell-permeable diethyl succinate mimicking increased intracellular succinate. Our results suggest that elevated intracellular succinate exerts a protective role in the primary microglia by preventing their conversion into the pro-inflammatory M1 phenotype induced by LPS. This protective effect is SUCNR1-independent and mediated by reduced mitochondrial fission and cellular ROS production.
Collapse
|
38
|
Bian HJ, Xu SY, Li HQ, Jia JQ, Ye L, Shu S, Xia SN, Gu Y, Zhu X, Xu Y, Cao X. JLX001 ameliorates cerebral ischemia injury by modulating microglial polarization and compromising NLRP3 inflammasome activation via the NF-κB signaling pathway. Int Immunopharmacol 2021; 101:108325. [PMID: 34740080 DOI: 10.1016/j.intimp.2021.108325] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/15/2021] [Accepted: 10/27/2021] [Indexed: 02/07/2023]
Abstract
Ischemic stroke is a devastating disease with high morbidity and mortality rates, and the proinflammatory microglia-mediated inflammatory response directly affects stroke outcome. Previous studies have reported that JLX001, a novel compound with a structure similar to that of cyclovirobuxine D (CVB-D), exerts antiapoptotic, anti-inflammatory and antioxidative effects on ischemia-induced brain injury. However, the role of JLX001 in microglial polarization and nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome regulation after ischemic stroke has not been fully investigated. In this study, we used the middle cerebral artery occlusion (MCAO) method to establish a focal cerebral ischemia model and found that JLX001 attenuated the brain infarct size and improved cerebral damage. Moreover, the expression levels of proinflammatory cytokines (interleukin [IL]-1β and tumor necrosis factor [TNF]-α) were significantly reduced while those of the anti-inflammatory cytokine IL-10 were increased in the JLX001-treated group. Immunofluorescence staining and flow cytometry revealed an increased number of anti-inflammatory phenotypic microglia and a reduced number of proinflammatory phenotypic microglia in JLX001-treated MCAO mice. Western blotting analysis showed that JLX001 inhibited the expression of NLRP3 and proteins related to the NLRP3 inflammasome axis in vivo. Furthermore, JLX001 reduced the number of NLRP3/Iba1 cells in ischemic penumbra tissues. Finally, mechanistic analysis revealed that JLX001 significantly inhibited the expression of proteins related to the NF-κB signaling pathway. Additionally, pyrrolidine dithiocarbamate (PDTC), an NF-κB inhibitor, ameliorated cerebral ischemia-reperfusion injury by suppressing microglial polarization towards the proinflammatory phenotype and NLRP3 activation in vivo, further suggesting that these protective effects of JLX001 were mediated by inhibition of the NF-κB signaling pathway. These results suggest that JLX001 is a promising therapeutic approach for ischemic stroke.
Collapse
Affiliation(s)
- Hui-Jie Bian
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Si-Yi Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China
| | - Hui-Qin Li
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China
| | - Jun-Qiu Jia
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China
| | - Lei Ye
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China
| | - Shu Shu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China
| | - Sheng-Nan Xia
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China
| | - Yue Gu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China
| | - Xiong Zhu
- Jiangsu Jinglixin Pharmaceutical Technology Company Limited, Nanjing 211100, 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 210008, China; Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing 210008, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China.
| | - Xiang Cao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing 210008, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China.
| |
Collapse
|
39
|
Jia Z, Yuan X, Wei JA, Guo X, Gong Y, Li J, Zhou H, Zhang L, Liu J. A Functionalized Octahedral Palladium Nanozyme as a Radical Scavenger for Ameliorating Alzheimer's Disease. ACS APPLIED MATERIALS & INTERFACES 2021; 13:49602-49613. [PMID: 34641674 DOI: 10.1021/acsami.1c06687] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Oxidative stress is always mentioned as a pathologic appearance of Alzheimer's disease (AD). It is attributed to mitochondrial dysfunction closely linked to Aβ deposition and neurofibrillary tangles (NFTs). Octahedral palladium nanoparticles (Pd NPs) exhibited excellent antioxidant enzyme-like activity and outstanding biocompatibility, but the poor blood-brain barrier (BBB) permeability limits their application in the treatment of Alzheimer's disease. Herein, we constructed a borneol (Bor)-modified octahedral palladium (Pd@PEG@Bor) nanozyme platform to eliminate intracellular reactive oxygen species (ROS) and elevate epithelial cell penetrability. Based on in vitro and in vivo studies, we demonstrate that the Pd@PEG@Bor could efficiently reduce ROS and Ca2+ contents, maintain the mitochondrial membrane potential, and further protect the mitochondria in SH-SY5Y cells. Furthermore, the nanozymes could quickly accumulate in the brain of AD mice and alleviate pathological characteristics such as Aβ plaque deposition, neuron loss, and neuroinflammation. The learning ability and memory function of AD mice are also significantly improved. Overall, this work indicates that the Pd@PEG@Bor nanozymes could delay the progression of AD by regulating ROS levels and also provides a new strategy for the treatment of AD.
Collapse
Affiliation(s)
- Zhi Jia
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, P. R. China
| | - Xiaoyu Yuan
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, P. R. China
| | - Ji-An Wei
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, P. R. China
| | - Xian Guo
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, P. R. China
| | - Youcong Gong
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, P. R. China
| | - Jin Li
- Department of Pain Management, the First Affiliated Hospital, Jinan University, Guangzhou 510632, P. R. China
| | - Hui Zhou
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, P. R. China
| | - Li Zhang
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, P. R. China
| | - Jie Liu
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, P. R. China
| |
Collapse
|
40
|
Murphy CE, Walker AK, Weickert CS. Neuroinflammation in schizophrenia: the role of nuclear factor kappa B. Transl Psychiatry 2021; 11:528. [PMID: 34650030 PMCID: PMC8516884 DOI: 10.1038/s41398-021-01607-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/22/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation, particularly in the dorsolateral prefrontal cortex, is well-established in a subset of people with schizophrenia, with significant increases in inflammatory markers including several cytokines. Yet the cause(s) of cortical inflammation in schizophrenia remains unknown. Clues as to potential microenvironmental triggers and/or intracellular deficits in immunoregulation may be gleaned from looking further upstream of effector immune molecules to transcription factors that control inflammatory gene expression. Here, we focus on the 'master immune regulator' nuclear factor kappa B (NF-κB) and review evidence in support of NF-κB dysregulation causing or contributing to neuroinflammation in patients. We discuss the utility of 'immune biotyping' as a tool to analyse immune-related transcripts and proteins in patient tissue, and the insights into cortical NF-κB in schizophrenia revealed by immune biotyping compared to studies treating patients as a single, homogenous group. Though the ubiquitous nature of NF-κB presents several hurdles for drug development, targeting this key immunoregulator with novel or repurposed therapeutics in schizophrenia is a relatively underexplored area that could aid in reducing symptoms of patients with active neuroinflammation.
Collapse
Affiliation(s)
- Caitlin E. Murphy
- grid.250407.40000 0000 8900 8842Neuroscience Research Australia, Randwick, NSW 2031 Australia
| | - Adam K. Walker
- grid.250407.40000 0000 8900 8842Neuroscience Research Australia, Randwick, NSW 2031 Australia ,grid.1005.40000 0004 4902 0432School of Psychiatry, University of New South Wales, Randwick, NSW 2031 Australia ,grid.1002.30000 0004 1936 7857Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052 Australia
| | - Cynthia Shannon Weickert
- Neuroscience Research Australia, Randwick, NSW, 2031, Australia. .,School of Psychiatry, University of New South Wales, Randwick, NSW, 2031, Australia. .,Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
41
|
Zhang F, Zhao K, Tang T, Deng Y, Zhang Y, Feng S, Feng P, Guo M, Li X, Cen J. Bisindole compound 4ae ameliorated cognitive impairment in rats with vascular dementia by anti-inflammation effect via microglia cells. Eur J Pharmacol 2021; 908:174357. [PMID: 34284012 DOI: 10.1016/j.ejphar.2021.174357] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 11/21/2022]
Abstract
Neuroinflammation is considered as an important mechanism of vascular dementia (VaD). Our primary study showed that the bisindole analogue (2-(2-(bis(5-chloro-1H-indol-3-yl)methyl)phenoxy)aniline, compound 4ae) had great anti-inflammation in zebrafish. Rat model of permanent occlusion of the bilateral common carotid arteries (2-vessel occlusion, 2VO) was utilized to evaluate the neuroprotective effect of 4ae. Our results showed that 4ae treatment effectively reduced Iba-1 positive microglia cells in cerebral cortex and hippocampus after cerebral ischemia. Compared with the model group, neuroinflammation characterized by Interleukin (IL)-6 and tumor necrosis factor (TNF)-α, oxidative stress characterized by reactive oxygen species (ROS) and superoxide dismutase (SOD) were both improved significantly after treatment with 4ae. Moreover, 4ae treatment significantly reversed ischemia-induced ACE enhancement, while notably increased the level of ACE2. To further elucidate the role of 4ae on neuroinflammation, we investigated the effects of 4ae on lipopolysaccharide (LPS)-induced inflammation in BV2 microglia cells, a kind of innate immune cells in central nervous system. The results demonstrated that the expressions of CD11b, TNFα and IL-6 and the level of ROS were up-regulated after treatment with LPS. More importantly, 4ae was able to block the activation of BV2 by reducing ROS production and the expression of inflammatory cytokines. In addition, our results suggested that 4ae inhibited the inflammatory response mediated by microglia cells by inhibiting NF-κB. This anti-inflammatory effect on microglia may be a potential mechanism for the neuroprotective effect of 4ae in VaD.
Collapse
Affiliation(s)
- Feng Zhang
- School of Pharmacy, Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, 475004, PR China
| | - Keqing Zhao
- School of Pharmacy, Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, 475004, PR China
| | - Tao Tang
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Hong Kong, 999077, PR China
| | - Yan Deng
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Hong Kong, 999077, PR China
| | - Yun Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250014, PR China
| | - Shuo Feng
- School of Pharmacy, Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, 475004, PR China
| | - Pu Feng
- School of Pharmacy, Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, 475004, PR China
| | - Mengyuan Guo
- School of Pharmacy, Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, 475004, PR China
| | - Xiaohui Li
- The First Affiliated Hospital of Henan University, Kaifeng, 475000, PR China.
| | - Juan Cen
- School of Pharmacy, Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, 475004, PR China.
| |
Collapse
|
42
|
Qiu X, Wang Q, Hou L, Zhang C, Wang Q, Zhao X. Inhibition of NLRP3 inflammasome by glibenclamide attenuated dopaminergic neurodegeneration and motor deficits in paraquat and maneb-induced mouse Parkinson's disease model. Toxicol Lett 2021; 349:1-11. [PMID: 34052309 DOI: 10.1016/j.toxlet.2021.05.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 04/26/2021] [Accepted: 05/25/2021] [Indexed: 01/24/2023]
Abstract
Pesticides exposure can lead to damage of dopaminergic neurons, which are associated with increased risk of Parkinson's disease (PD). However, the etiology of PD remains poorly understood and no therapeutic strategy is available. Previous studies suggested the involvement of NLRP3 inflammasome in the onset of PD. This study was designed to investigate whether glibenclamide, an inhibitor of NLRP3 inflammasome, could offer a reliable protective strategy for PD in a mouse PD model induced by paraquat and maneb. We found that glibenclamide exerted potent neuroprotection against paraquat and maneb-induced upregulation of α-synuclein, dopaminergic neurodegeneration and motor impairment in brain of mice. Mechanistically, glibenclamide treatment blocked NLRP3 inflammasome activation evidenced by reduced expressions of NLRP3, activated caspase-1 and mature interleukin-1β in glibenclamide co-treated mice compared with those in paraquat and maneb group mice. Furthermore, glibenclamide treatment mitigated paraquat and maneb-induced microglial M1 proinflammatory response and nuclear factor-κB activation in mice. Finally, the increased superoxide production, lipid peroxidation, protein levels of NADPH oxidase 2 (NOX2) and inducible nitric oxide synthase (iNOS) induced by paraquat and maneb were all attenuated by glibenclamide. Overall, our findings demonstrated that glibenclamide protected dopaminergic neurons in a mouse PD model induced by combined exposures of paraquat and maneb through suppression of NLRP3 inflammasome activation, microglial M1 polarization and oxidative stress.
Collapse
Affiliation(s)
- Xiaofei Qiu
- Qingdao Municipal Center for Disease Control & Prevention, Qingdao Institute of Preventive Medicine, Qingdao, 266033, China; School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Qinghui Wang
- Department of Anesthesiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116023, China
| | - Liyan Hou
- School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Cuili Zhang
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Qingshan Wang
- School of Public Health, Dalian Medical University, Dalian, 116044, China.
| | - Xiulan Zhao
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
43
|
Ferreira Junior NC, dos Santos Pereira M, Francis N, Ramirez P, Martorell P, González-Lizarraga F, Figadère B, Chehin R, Del Bel E, Raisman-Vozari R, Michel PP. The Chemically-Modified Tetracycline COL-3 and Its Parent Compound Doxycycline Prevent Microglial Inflammatory Responses by Reducing Glucose-Mediated Oxidative Stress. Cells 2021; 10:cells10082163. [PMID: 34440932 PMCID: PMC8392055 DOI: 10.3390/cells10082163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 12/31/2022] Open
Abstract
We used mouse microglial cells in culture activated by lipopolysaccharide (LPS) or α-synuclein amyloid aggregates (αSa) to study the anti-inflammatory effects of COL-3, a tetracycline derivative without antimicrobial activity. Under LPS or αSa stimulation, COL-3 (10, 20 µM) efficiently repressed the induction of the microglial activation marker protein Iba-1 and the stimulated-release of the pro-inflammatory cytokine TNF-α. COL-3′s inhibitory effects on TNF-α were reproduced by the tetracycline antibiotic doxycycline (DOX; 50 µM), the glucocorticoid dexamethasone, and apocynin (APO), an inhibitor of the superoxide-producing enzyme NADPH oxidase. This last observation suggested that COL-3 and DOX might also operate themselves by restraining oxidative stress-mediated signaling events. Quantitative measurement of intracellular reactive oxygen species (ROS) levels revealed that COL-3 and DOX were indeed as effective as APO in reducing oxidative stress and TNF-α release in activated microglia. ROS inhibition with COL-3 or DOX occurred together with a reduction of microglial glucose accumulation and NADPH synthesis. This suggested that COL-3 and DOX might reduce microglial oxidative burst activity by limiting the glucose-dependent synthesis of NADPH, the requisite substrate for NADPH oxidase. Coherent with this possibility, the glycolysis inhibitor 2-deoxy-D-glucose reproduced the immunosuppressive action of COL-3 and DOX in activated microglia. Overall, we propose that COL-3 and its parent compound DOX exert anti-inflammatory effects in microglial cells by inhibiting glucose-dependent ROS production. These effects might be strengthened by the intrinsic antioxidant properties of DOX and COL-3 in a self-reinforcing manner.
Collapse
Affiliation(s)
- Nilson Carlos Ferreira Junior
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Av. Café, s/no, Ribeirão Preto 14040-904, Brazil;
- USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), São Paulo 05508-220, Brazil
| | - Maurício dos Santos Pereira
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Av. Café, s/no, Ribeirão Preto 14040-904, Brazil;
- USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), São Paulo 05508-220, Brazil
| | - Nour Francis
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
| | - Paola Ramirez
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
| | - Paula Martorell
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
| | - Florencia González-Lizarraga
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), CP 4000 Tucumán, Argentina; (F.G.-L.); (R.C.)
| | - Bruno Figadère
- BioCIS, CNRS, Université Paris-Saclay, 92290 Châtenay-Malabry, France;
| | - Rosana Chehin
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), CP 4000 Tucumán, Argentina; (F.G.-L.); (R.C.)
| | - Elaine Del Bel
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Av. Café, s/no, Ribeirão Preto 14040-904, Brazil;
- USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), São Paulo 05508-220, Brazil
| | - Rita Raisman-Vozari
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
- Correspondence: (R.R.-V.); (P.P.M.); Tel.: +33-(0)157274550 (R.R.-V.); +33-(0)157274534 (P.P.M.)
| | - Patrick Pierre Michel
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
- Correspondence: (R.R.-V.); (P.P.M.); Tel.: +33-(0)157274550 (R.R.-V.); +33-(0)157274534 (P.P.M.)
| |
Collapse
|
44
|
Circulating HMGB1 is elevated in veterans with Gulf War Illness and triggers the persistent pro-inflammatory microglia phenotype in male C57Bl/6J mice. Transl Psychiatry 2021; 11:390. [PMID: 34253711 PMCID: PMC8275600 DOI: 10.1038/s41398-021-01517-1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 06/16/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
Gulf War Illness (GWI) is a chronic, multi-symptom peripheral and CNS condition with persistent microglial dysregulation, but the mechanisms driving the continuous neuroimmune pathology are poorly understood. The alarmin HMGB1 is an autocrine and paracrine pro-inflammatory signal, but the role of circulating HMGB1 in persistent neuroinflammation and GWI remains largely unknown. Using the LPS model of the persistent microglial pro-inflammatory response, male C57Bl/6J mice injected with LPS (5 mg/kg IP) exhibited persistent changes in microglia morphology and elevated pro-inflammatory markers in the hippocampus, cortex, and midbrain 7 days after LPS injection, while the peripheral immune response had resolved. Ex vivo serum analysis revealed an augmented pro-inflammatory response to LPS when microglia cells were cultured with the 7-day LPS serum, indicating the presence of bioactive circulating factors that prime the microglial pro-inflammatory response. Elevated circulating HMGB1 levels were identified in the mouse serum 7 days after LPS administration and in the serum of veterans with GWI. Tail vein injection of rHMGB1 in male C57Bl/6 J mice elevated TNFα mRNA levels in the liver, hippocampus, and cortex, demonstrating HMGB1-induced peripheral and CNS effects. Microglia isolated at 7 days after LPS injection revealed a unique transcriptional profile of 17 genes when compared to the acute 3 H LPS response, 6 of which were also upregulated in the midbrain by rHMGB1, highlighting a distinct signature of the persistent pro-inflammatory microglia phenotype. These findings indicate that circulating HMGB1 is elevated in GWI, regulates the microglial neuroimmune response, and drives chronic neuroinflammation that persists long after the initial instigating peripheral stimulus.
Collapse
|
45
|
Li S, Cao W, Zhou S, Ma M, Zhang W, Li F, Li C. Expression of Cntn1 is regulated by stress and associated with anxiety and depression phenotypes. Brain Behav Immun 2021; 95:142-153. [PMID: 33737174 DOI: 10.1016/j.bbi.2021.03.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 03/07/2021] [Accepted: 03/11/2021] [Indexed: 12/13/2022] Open
Abstract
In recent years, our understanding of neural circuits associated with depression has increased. Although inherited factors are known to influence individual differences in the risk for this disorder, it has been difficult to identify specific genes that moderate circuit functions affecting depression. Genome-wide association studies have identified genetic variants of Cntn1 that are linked to major depressive disorders. Cntn1, a subset of the neural cell adhesion protein and immunoglobulin supergene family, participates in cell contact formation and axonal growth control and plays a role in degenerative and inflammatory disorders. However, neuronal substrates that mediate Cntn1 action on depression-like phenotypes and involved mechanisms are unclear. Here, we exploited chronic unpredictable stress (CUS) exposure and found that CUS treatment significantly increased hippocampal Cntn1 messenger RNA and protein expression in both mice and rats, but not in the medial prefrontal cortex, which presented a region-specific regulation. Using an adeno-associated virus-based approach to directly overexpress Cntn1 via stereotactic injection, we demonstrated that Cntn1 overexpression in the hippocampus triggered anxiety- and depression-like phenotypes in addition to microglia activation or phagocytosis in the hippocampus, resulting in upregulation of pro-inflammatory cytokine (IL1α, IL6, and Ccl2) mRNA expression and downregulation of anti-inflammatory cytokine (IL4 and CD206) mRNA expression, determined using real-time quantitative PCR, thus impairing hippocampal immature neurons in the dentate gyrus, determined using immunohistochemical staining for doublecortin, a specific marker for immature neurons. Collectively, our results identified Cntn1 as a novel risk gene involved in regulating anxiety and depression via functional actions in the hippocampus that is correlated with microglial activation or phagocytosis and reduced hippocampal immature neurons. These results may provide a better understanding of the pathophysiological mechanisms underlying the risk of depression-related disorders.
Collapse
Affiliation(s)
- Songji Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province 410013, China
| | - Wenyu Cao
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, Hengyang, Hunan Province 421001, China
| | - Shifen Zhou
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province 410013, China
| | - Minhui Ma
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province 410013, China
| | - Wenjuan Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province 410013, China
| | - Fang Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province 410013, China
| | - Changqi Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province 410013, China.
| |
Collapse
|
46
|
Morris G, Walder K, Kloiber S, Amminger P, Berk M, Bortolasci CC, Maes M, Puri BK, Carvalho AF. The endocannabinoidome in neuropsychiatry: Opportunities and potential risks. Pharmacol Res 2021; 170:105729. [PMID: 34119623 DOI: 10.1016/j.phrs.2021.105729] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 02/08/2023]
Abstract
The endocannabinoid system (ECS) comprises two cognate endocannabinoid receptors referred to as CB1R and CB2R. ECS dysregulation is apparent in neurodegenerative/neuro-psychiatric disorders including but not limited to schizophrenia, major depressive disorder and potentially bipolar disorder. The aim of this paper is to review mechanisms whereby both receptors may interact with neuro-immune and neuro-oxidative pathways, which play a pathophysiological role in these disorders. CB1R is located in the presynaptic terminals of GABAergic, glutamatergic, cholinergic, noradrenergic and serotonergic neurons where it regulates the retrograde suppression of neurotransmission. CB1R plays a key role in long-term depression, and, to a lesser extent, long-term potentiation, thereby modulating synaptic transmission and mediating learning and memory. Optimal CB1R activity plays an essential neuroprotective role by providing a defense against the development of glutamate-mediated excitotoxicity, which is achieved, at least in part, by impeding AMPA-mediated increase in intracellular calcium overload and oxidative stress. Moreover, CB1R activity enables optimal neuron-glial communication and the function of the neurovascular unit. CB2R receptors are detected in peripheral immune cells and also in central nervous system regions including the striatum, basal ganglia, frontal cortex, hippocampus, amygdala as well as the ventral tegmental area. CB2R upregulation inhibits the presynaptic release of glutamate in several brain regions. CB2R activation also decreases neuroinflammation partly by mediating the transition from a predominantly neurotoxic "M1" microglial phenotype to a more neuroprotective "M2" phenotype. CB1R and CB2R are thus novel drug targets for the treatment of neuro-immune and neuro-oxidative disorders including schizophrenia and affective disorders.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Ken Walder
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Australia
| | - Stefan Kloiber
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 33 Ursula Franklin Street, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Paul Amminger
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Centre for Youth Mental Health, Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, The University of Melbourne, Melbourne, Australia
| | - Chiara C Bortolasci
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Michael Maes
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| | | | - Andre F Carvalho
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia.
| |
Collapse
|
47
|
Song Z, Feng J, Zhang Q, Deng S, Yu D, Zhang Y, Li T. Tanshinone IIA Protects Against Cerebral Ischemia Reperfusion Injury by Regulating Microglial Activation and Polarization via NF-κB Pathway. Front Pharmacol 2021; 12:641848. [PMID: 33953677 PMCID: PMC8090935 DOI: 10.3389/fphar.2021.641848] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/05/2021] [Indexed: 12/16/2022] Open
Abstract
Tanshinone IIA, a fat-soluble diterpenoid isolated from Salvia miltiorrhiza Bunge, has been shown to attenuate the cerebral ischemic injury. The aim of this study was to examine the effects on neuroprotection and microglia activation of Tanshinone IIA. Male Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO). We found that Tanshinone IIA significantly reduced infarction volume, alleviated neuronal injuries, reduced the release of TNF-α, IL-1β, and IL-6, increased SOD activity, and decrease the content of MDA in MCAO rats. Hematoxylin and eosin staining, Nissl staining, TUNEL staining and immunofluorescence staining showed that Tanshinone IIA improved the distribution and morphology of neurons in brain tissues and reduced apoptosis. In addition, Co-immunofluorescence staining of rat brain tissues and the mRNA expression levels of CD11b, CD32, iNOS, and Arg-1, CD206, IL-10 in BV2 cells indicated that Tanshinone IIA can downregulate M1 microglia and upregulate M2 microglia in MCAO rats. Further, BV2 microglial cells were subjected to oxygen-glucose deprivation, the protein expression levels were detected by western blot. Tanshinone IIA inhibited the expression levels of NF-κB signaling pathway related proteins. Taken together, this study suggested that Tanshinone IIA modulated microglial M1/M2 polarization via the NF-κB signaling pathway to confer anti-neuroinflammatory effects.
Collapse
Affiliation(s)
- Zhibing Song
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai, China.,College of Pharmacology, Anhui University of Chinese Medicine, Hefei, China
| | - Jingjing Feng
- College of Pharmacology, Anhui University of Chinese Medicine, Hefei, China
| | - Qian Zhang
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai, China
| | - Shanshan Deng
- School of Medicine, Shanghai University, Shanghai, China
| | | | - Yuefan Zhang
- School of Medicine, Shanghai University, Shanghai, China
| | - Tiejun Li
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai, China
| |
Collapse
|
48
|
Endothelial Cell-Derived SO 2 Controls Endothelial Cell Inflammation, Smooth Muscle Cell Proliferation, and Collagen Synthesis to Inhibit Hypoxic Pulmonary Vascular Remodelling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5577634. [PMID: 33953829 PMCID: PMC8068783 DOI: 10.1155/2021/5577634] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/27/2021] [Accepted: 04/05/2021] [Indexed: 02/08/2023]
Abstract
Hypoxic pulmonary vascular remodelling (PVR) is the major pathological basis of aging-related chronic obstructive pulmonary disease and obstructive sleep apnea syndrome. The pulmonary artery endothelial cell (PAEC) inflammation, and pulmonary artery smooth muscle cell (PASMC) proliferation, hypertrophy and collagen remodelling are the important pathophysiological components of PVR. Endogenous sulfur dioxide (SO2) was found to be a novel gasotransmitter in the cardiovascular system with its unique biological properties. The study was aimed to investigate the role of endothelial cell- (EC-) derived SO2 in the progression of PAEC inflammation, PASMC proliferation, hypertrophy and collagen remodelling in PVR and the possible mechanisms. EC-specific aspartic aminotransferase 1 transgenic (EC-AAT1-Tg) mice were constructed in vivo. Pulmonary hypertension was induced by hypoxia. Right heart catheterization and echocardiography were used to detect mouse hemodynamic changes. Pathologic analysis was performed in the pulmonary arteries. High-performance liquid chromatography was employed to detect the SO2 content. Human PAECs (HPAECs) with lentiviruses containing AAT1 cDNA or shRNA and cocultured human PASMCs (HPASMCs) were applied in vitro. SO2 probe and enzyme-linked immunosorbent assay were used to detect the SO2 content and determine p50 activity, respectively. Hypoxia caused a significant reduction in SO2 content in the mouse lung and HPAECs and increases in right ventricular systolic pressure, pulmonary artery wall thickness, muscularization, and the expression of PAEC ICAM-1 and MCP-1 and of PASMC Ki-67, collagen I, and α-SMA (p < 0.05). However, EC-AAT1-Tg with sufficient SO2 content prevented the above increases induced by hypoxia (p < 0.05). Mechanistically, EC-derived SO2 deficiency promoted HPAEC ICAM-1 and MCP-1 and the cocultured HPASMC Ki-67 and collagen I expression, which was abolished by andrographolide, an inhibitor of p50 (p < 0.05). Meanwhile, EC-derived SO2 deficiency increased the expression of cocultured HPASMC α-SMA (p < 0.05). Taken together, these findings revealed that EC-derived SO2 inhibited p50 activation to control PAEC inflammation in an autocrine manner and PASMC proliferation, hypertrophy, and collagen synthesis in a paracrine manner, thereby inhibiting hypoxic PVR.
Collapse
|
49
|
Xue Y, Nie D, Wang LJ, Qiu HC, Ma L, Dong MX, Tu WJ, Zhao J. Microglial Polarization: Novel Therapeutic Strategy against Ischemic Stroke. Aging Dis 2021; 12:466-479. [PMID: 33815877 PMCID: PMC7990355 DOI: 10.14336/ad.2020.0701] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 07/01/2020] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke, which is the second highest cause of death and the leading cause of disability, represents ~71% of all strokes globally. Some studies have found that the key elements of the pathobiology of stroke is immunity and inflammation. Microglia are the first line of defense in the nervous system. After stroke, the activated microglia become a double-edged sword, with distinct phenotypic changes to the deleterious M1 types and neuroprotective M2 types. Therefore, ways to promote microglial polarization toward M2 phenotype after stroke have become the focus of attention in recent years. In this review, we discuss the process of microglial polarization, summarize the alternation of signaling pathways and epigenetic regulation that control microglial polarization in ischemic stroke, aiming to find the potential mechanisms by which microglia can be transformed into the M2 polarized phenotype.
Collapse
Affiliation(s)
- Yimeng Xue
- 1Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,2Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Ding Nie
- 1Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lin-Jian Wang
- 1Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,2Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Han-Cheng Qiu
- 1Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Long Ma
- 1Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ming-Xin Dong
- 3Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Wen-Jun Tu
- 1Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,3Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Jizong Zhao
- 1Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,2Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.,4China National Clinical Research Center for Neurological Diseases, Beijing, China.,5Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,6Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| |
Collapse
|
50
|
Childers GM, Perry CA, Blachut B, Martin N, Bortner CD, Sieber S, Li JL, Fessler MB, Harry GJ. Assessing the Association of Mitochondrial Function and Inflammasome Activation in Murine Macrophages Exposed to Select Mitotoxic Tri-Organotin Compounds. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:47015. [PMID: 33929904 PMCID: PMC8086801 DOI: 10.1289/ehp8314] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
BACKGROUND Mitochondrial function is implicated as a target of environmental toxicants and found in disease or injury models, contributing to acute and chronic inflammation. One mechanism by which mitochondrial damage can propagate inflammation is via activation of the nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family, pyrin domain-containing receptor (NLRP)3 inflammasome, a protein complex that processes mature interleukin (IL)-1β. IL-1β plays an important role in the innate immune response and dysregulation is associated with autoinflammatory disorders. OBJECTIVE The objective was to evaluate whether mitochondrial toxicants recruit inflammasome activation and IL-1β processing. METHOD Murine macrophages (RAW 264.7) exposed to tri-organotins (triethyltin bromide (TETBr), trimethyltin hydroxide (TMTOH), triphenyltin hydroxide (TPTOH), bis(tributyltin)oxide) [Bis(TBT)Ox] were examined for pro-inflammatory cytokine induction. TMTOH and TETBr were examined in RAW 264.7 and bone marrow-derived macrophages for mitochondrial bioenergetics, reactive oxygen species (ROS) production, and inflammasome activation via visualization of aggregate formation, caspase-1 flow cytometry, IL-1β enzyme-linked immunosorbent assay and Western blots, and microRNA (miRNA) and mRNA arrays. RESULTS TETBr and TMTOH induced inflammasome aggregate formation and IL-1β release in lipopolysaccharide (LPS)-primed macrophages. Mitochondrial bioenergetics and mitochondrial ROS were suppressed. Il1a and Il1b induction with LPS or LPS+ATP challenge was diminished. Differential miRNA and mRNA profiles were observed. Lower miR-151-3p targeted cyclic adenosine monophosphate (cAMP)-mediated and AMP-activated protein kinase signaling pathways; higher miR-6909-5p, miR-7044-5p, and miR-7686-5p targeted Wnt beta-catenin signaling, retinoic acid receptor activation, apoptosis, signal transducer and activator of transcription 3, IL-22, IL-12, and IL-10 signaling. Functional enrichment analysis identified apoptosis and cell survival canonical pathways. CONCLUSION Select mitotoxic tri-organotins disrupted murine macrophage transcriptional response to LPS, yet triggered inflammasome activation. The differential response pattern suggested unique functional changes in the inflammatory response that may translate to suppressed host defense or prolong inflammation. We posit a framework to examine immune cell effects of environmental mitotoxic compounds for adverse health outcomes. https://doi.org/10.1289/EHP8314.
Collapse
Affiliation(s)
- Gabrielle M. Childers
- Molecular Toxicology Branch, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Caroline A. Perry
- Molecular Toxicology Branch, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Barbara Blachut
- Molecular Toxicology Branch, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Negin Martin
- Laboratory of Neurobiology, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Carl D. Bortner
- Signal Transduction Laboratory, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Stella Sieber
- Molecular Genomics Core Laboratory, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Jian-Liang Li
- Integrative Bioinformatics Support Group, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Michael B. Fessler
- Immunity, Inflammation, and Disease Laboratory, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - G. Jean Harry
- Molecular Toxicology Branch, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| |
Collapse
|