151
|
孙 晓, 史 航, 张 磊, 刘 中, 李 克, 钱 玲, 朱 星, 杨 康, 付 强, 丁 华. [Exosomes from ectoderm mesenchymal stem cells inhibits lipopolysaccharide-induced microglial M1 polarization and promotes survival of H 2O 2-exposed PC12 cells by suppressing inflammatory response and oxidative stress]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:119-128. [PMID: 38293983 PMCID: PMC10878899 DOI: 10.12122/j.issn.1673-4254.2024.01.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Indexed: 02/01/2024]
Abstract
OBJECTIVE To investigate the potential value of exosomes derived from rat ectoderm mesenchymal stem cells (EMSCs-exo) for repairing secondary spinal cord injury. METHODS EMSCs-exo were obtained using ultracentrifugation from EMSCs isolated from rat nasal mucosa, identified by transmission electron microscope, nanoparticle tracking analysis (NTA), and Western blotting, and quantified using the BCA method. Neonatal rat microglia purified by differential attachment were induced with 100 μg/L lipopolysaccharide (LPS) and treated with 37.5 or 75 mg/L EMSCs-exo. PC12 cells were exposed to 400 μmol/L H2O2 and treated with EMSCs-exo at 37.5 or 75 mg/L. The protein and mRNA expressions of Arg1 and iNOS in the treated cells were determined with Western blotting and qRT- PCR, and the concentrations of IL- 6, IL-10, and IGF-1 in the supernatants were measured with ELISA. The viability and apoptosis of PC12 cells were detected using CCK-8 assay and flow cytometry. RESULTS The isolated rat EMSCs showed high expressions of nestin, CD44, CD105, and vimentin. The obtained EMSCs-exo had a typical cup-shaped structure under transmission electron microscope with an average particle size of 142 nm and positivity for CD63, CD81, and TSG101 but not vimentin. In LPS-treated microglia, EMSCs-exo treatment at 75 mg/L significantly increased Arg1 protein level and lowered iNOS protein expression (P < 0.05). EMSCs-exo treatment at 75 mg/L, as compared with the lower concentration at 37.5 mg/L, more strongly increased Arg1 mRNA expression and IGF-1 and IL-10 production and decreased iNOS mRNA expression and IL-6 production in LPS-induced microglia, and more effectively promoted cell survival and decreased apoptosis rate of H2O2-induced PC12 cells (P < 0.05). CONCLUSION EMSCs-exo at 75 mg/L can effectively reduce the proportion of M1 microglia and alleviate neuronal apoptosis under oxidative stress to promote neuronal survival, suggesting its potential in controlling secondary spinal cord injury.
Collapse
Affiliation(s)
- 晓鹏 孙
- 江苏大学附属人民医院骨科,江苏 镇江 212000Department of Orthopedics, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212000, China
| | - 航 史
- 江苏大学附属人民医院骨科,江苏 镇江 212000Department of Orthopedics, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212000, China
| | - 磊 张
- 江苏大学附属人民医院骨科,江苏 镇江 212000Department of Orthopedics, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212000, China
| | - 中 刘
- 上海交通大学医学院附属第一人民医院骨科,上海 200080Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - 克威 李
- 上海交通大学医学院附属第一人民医院骨科,上海 200080Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - 玲玲 钱
- 江苏大学医学院,江苏 镇江 212013School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - 星宇 朱
- 江苏大学附属人民医院骨科,江苏 镇江 212000Department of Orthopedics, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212000, China
| | - 康佳 杨
- 江苏大学附属人民医院骨科,江苏 镇江 212000Department of Orthopedics, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212000, China
| | - 强 付
- 上海交通大学医学院附属第一人民医院骨科,上海 200080Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - 华 丁
- 江苏大学附属人民医院骨科,江苏 镇江 212000Department of Orthopedics, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212000, China
| |
Collapse
|
152
|
Cao M, Wang Z, Lan W, Xiang B, Liao W, Zhou J, Liu X, Wang Y, Zhang S, Lu S, Lang J, Zhao Y. The roles of tissue resident macrophages in health and cancer. Exp Hematol Oncol 2024; 13:3. [PMID: 38229178 PMCID: PMC10790434 DOI: 10.1186/s40164-023-00469-0] [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: 10/08/2023] [Accepted: 12/28/2023] [Indexed: 01/18/2024] Open
Abstract
As integral components of the immune microenvironment, tissue resident macrophages (TRMs) represent a self-renewing and long-lived cell population that plays crucial roles in maintaining homeostasis, promoting tissue remodeling after damage, defending against inflammation and even orchestrating cancer progression. However, the exact functions and roles of TRMs in cancer are not yet well understood. TRMs exhibit either pro-tumorigenic or anti-tumorigenic effects by engaging in phagocytosis and secreting diverse cytokines, chemokines, and growth factors to modulate the adaptive immune system. The life-span, turnover kinetics and monocyte replenishment of TRMs vary among different organs, adding to the complexity and controversial findings in TRMs studies. Considering the complexity of tissue associated macrophage origin, macrophages targeting strategy of each ontogeny should be carefully evaluated. Consequently, acquiring a comprehensive understanding of TRMs' origin, function, homeostasis, characteristics, and their roles in cancer for each specific organ holds significant research value. In this review, we aim to provide an outline of homeostasis and characteristics of resident macrophages in the lung, liver, brain, skin and intestinal, as well as their roles in modulating primary and metastatic cancer, which may inform and serve the future design of targeted therapies.
Collapse
Affiliation(s)
- Minmin Cao
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zihao Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wanying Lan
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
- Guixi Community Health Center of the Chengdu High-Tech Zone, Chengdu, China
| | - Binghua Xiang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenjun Liao
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Jie Zhou
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaomeng Liu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yiling Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Shichuan Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Shun Lu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Jinyi Lang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yue Zhao
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
153
|
Zaccai S, Nemirovsky A, Lerner L, Alfahel L, Eremenko E, Israelson A, Monsonego A. CD4 T-cell aging exacerbates neuroinflammation in a late-onset mouse model of amyotrophic lateral sclerosis. J Neuroinflammation 2024; 21:17. [PMID: 38212835 PMCID: PMC10782641 DOI: 10.1186/s12974-023-03007-1] [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: 11/05/2023] [Accepted: 12/26/2023] [Indexed: 01/13/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset progressive neurodegenerative disorder characterized by the loss of upper and lower motor neurons in the brain and spinal cord. Accumulating evidence suggests that ALS is not solely a neuronal cell- or brain tissue-autonomous disease and that neuroinflammation plays a key role in disease progression. Furthermore, whereas both CD4 and CD8 T cells were observed in spinal cords of ALS patients and in mouse models of the disease, their role in the neuroinflammatory process, especially considering their functional changes with age, is not fully explored. In this study, we revealed the structure of the CD4 T-cell compartment during disease progression of early-onset SOD1G93A and late-onset SOD1G37R mouse models of ALS. We show age-related changes in the CD4 T-cell subset organization between these mutant SOD1 mouse models towards increased frequency of effector T cells in spleens of SOD1G37R mice and robust infiltration of CD4 T cells expressing activation markers and the checkpoint molecule PD1 into the spinal cord. The frequency of infiltrating CD4 T cells correlated with the frequency of infiltrating CD8 T cells which displayed a more exhausted phenotype. Moreover, RNA-Seq and immunohistochemistry analyses of spinal cords from SOD1G37R mice with early clinical symptoms demonstrated immunological trajectories reminiscent of a neurotoxic inflammatory response which involved proinflammatory T cells and antigen presentation related pathways. Overall, our findings suggest that age-related changes of the CD4 T cell landscape is indicative of a chronic inflammatory response, which aggravates the disease process and can be therapeutically targeted.
Collapse
Affiliation(s)
- Shir Zaccai
- Department of Physiology and Cell Biology, Faculty of Health Sciences and The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel
| | - Anna Nemirovsky
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, Faculty of Health Sciences and The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel
| | - Livnat Lerner
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, Faculty of Health Sciences and The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel
| | - Leenor Alfahel
- Department of Physiology and Cell Biology, Faculty of Health Sciences and The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel
| | - Ekaterina Eremenko
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, Faculty of Health Sciences and The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel
| | - Adrian Israelson
- Department of Physiology and Cell Biology, Faculty of Health Sciences and The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel.
| | - Alon Monsonego
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, Faculty of Health Sciences and The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel.
| |
Collapse
|
154
|
Gargas J, Janowska J, Gebala P, Maksymiuk W, Sypecka J. Reactive Gliosis in Neonatal Disorders: Friend or Foe for Neuroregeneration? Cells 2024; 13:131. [PMID: 38247822 PMCID: PMC10813898 DOI: 10.3390/cells13020131] [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: 11/17/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/23/2024] Open
Abstract
A developing nervous system is particularly vulnerable to the influence of pathophysiological clues and injuries in the perinatal period. Astrocytes are among the first cells that react to insults against the nervous tissue, the presence of pathogens, misbalance of local tissue homeostasis, and a lack of oxygen and trophic support. Under this background, it remains uncertain if induced astrocyte activation, recognized as astrogliosis, is a friend or foe for progressing neonatal neurodevelopment. Likewise, the state of astrocyte reactivity is considered one of the key factors discriminating between either the initiation of endogenous reparative mechanisms compensating for aberrations in the structures and functions of nervous tissue or the triggering of neurodegeneration. The responses of activated cells are modulated by neighboring neural cells, which exhibit broad immunomodulatory and pro-regenerative properties by secreting a plethora of active compounds (including interleukins and chemokines, neurotrophins, reactive oxygen species, nitric oxide synthase and complement components), which are engaged in cell crosstalk in a paracrine manner. As the developing nervous system is extremely sensitive to the influence of signaling molecules, even subtle changes in the composition or concentration of the cellular secretome can have significant effects on the developing neonatal brain. Thus, modulating the activity of other types of cells and their interactions with overreactive astrocytes might be a promising strategy for controlling neonatal astrogliosis.
Collapse
Affiliation(s)
| | | | | | | | - Joanna Sypecka
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, A. Pawinskiego 5, 02-106 Warsaw, Poland; (J.G.); (J.J.)
| |
Collapse
|
155
|
Chen CA, Li CX, Zhang ZH, Xu WX, Liu SL, Ni WC, Wang XQ, Cheng FF, Wang QG. Qinzhizhudan formula dampens inflammation in microglia polarization of vascular dementia rats by blocking MyD88/NF-κB signaling pathway: Through integrating network pharmacology and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116769. [PMID: 37400007 DOI: 10.1016/j.jep.2023.116769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/24/2023] [Accepted: 06/09/2023] [Indexed: 07/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qinzhizhudan Formula (QZZD) is composed of Scutellaria baicalensis Georgi (Huang Qin) extract, Gardenia jasminoides (Zhizi) extract and Suis Fellis Pulvis (Zhudanfen) (ratio of 4:5:6). This formula is optimized from Qingkailing (QKL) injection. Regarding brain injury, QZZD is protective. However, the mechanism by which QZZD treats vascular dementia (VD) has not been elucidated. AIM OF THE STUDY To ascertain QZZD's effect on the treatment of VD and further investigate the molecular mechanisms. MATERIALS AND METHODS In this study, we screened the possible components and targets of QZZD against VD and microglia polarization using network pharmacology (NP), then an animal model of bilateral common carotid artery ligation method (2VO) was induced. Afterward, The Morris water maze was employed to evaluate cognitive ability, and pathological alterations in the CA1 area of the hippocampus were detected using HE and Nissl staining. To confirm the affect of QZZD on VD and its molecular mechanism, the contents of inflammatory factors IL-1β, TNF-α, IL-4, and IL-10 were performed to detect by ELISA, the phenotype polarization of microglia cells was detected by immunofluorescence staining, and the expressions of MyD88, p-IκBα and p-NF-κB p65 in brain tissue were detected by western blot. RESULTS A total of 112 active compounds and 363 common targets of QZZD, microglia polarization, and VD were identified, according to the NP analysis. 38 hub targets were screened out from the PPI network. GO analysis and KEGG pathway analysis showed that QZZD may regulate microglia polarization through anti-inflammatory mechanism such as Toll-like receptor signaling pathway and NF-κB signaling pathway. The further results showed that QZZD can alleviate the memory impairment induced by 2VO. QZZD profoundly rescued brain hippocampus neuronal damage and increased the number of neurons. These advantageous outcomes were linked to the control of microglia polarization. QZZD decreased M1 phenotypic marker expression while increasing M2 phenotypic marker expression. QZZD may controll the polarization of the M1 microglia by blocking the core part of Toll-like receptor signaling pathway, that is the MyD88/NF-κB signaling pathway, which reduced the neurotoxic effects of the microglia. CONCLUSION Here, we explored the anti-VD microglial polarization characteristic of QZZD for the first time and clarified its mechanisms. These findings will provide valuable clues for the discovery of anti-VD agents.
Collapse
Affiliation(s)
- Cong-Ai Chen
- Dongzhimen Hospital Beijing University of Chinese Medicine, Beijing, 100700, China; Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Chang-Xiang Li
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Ze-Han Zhang
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Wen-Xiu Xu
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Shu-Ling Liu
- Dongzhimen Hospital Beijing University of Chinese Medicine, Beijing, 100700, China; Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Wen-Chao Ni
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Xue-Qian Wang
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Fa-Feng Cheng
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Qing-Guo Wang
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
156
|
Lankhuijzen LM, Ridler T. Opioids, microglia, and temporal lobe epilepsy. Front Neurol 2024; 14:1298489. [PMID: 38249734 PMCID: PMC10796828 DOI: 10.3389/fneur.2023.1298489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/15/2023] [Indexed: 01/23/2024] Open
Abstract
A lack of treatment options for temporal lobe epilepsy (TLE) demands an urgent quest for new therapies to recover neuronal damage and reduce seizures, potentially interrupting the neurotoxic cascades that fuel hyper-excitability. Endogenous opioids, along with their respective receptors, particularly dynorphin and kappa-opioid-receptor, present as attractive candidates for controlling neuronal excitability and therapeutics in epilepsy. We perform a critical review of the literature to evaluate the role of opioids in modulating microglial function and morphology in epilepsy. We find that, in accordance with anticonvulsant effects, acute opioid receptor activation has unique abilities to modulate microglial activation through toll-like 4 receptors, regulating downstream secretion of cytokines. Abnormal activation of microglia is a dominant feature of neuroinflammation, and inflammatory cytokines are found to aggravate TLE, inspiring the challenge to alter microglial activation by opioids to suppress seizures. We further evaluate how opioids can modulate microglial activation in epilepsy to enhance neuroprotection and reduce seizures. With controlled application, opioids may interrupt inflammatory cycles in epilepsy, to protect neuronal function and reduce seizures. Research on opioid-microglia interactions has important implications for epilepsy and healthcare approaches. However, preclinical research on opioid modulation of microglia supports a new therapeutic pathway for TLE.
Collapse
Affiliation(s)
| | - Thomas Ridler
- Hatherly Laboratories, Department of Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
157
|
Bou Sader Nehme S, Sanchez-Sarasua S, Adel R, Tuifua M, Ali A, Essawy AE, Abdel Salam S, Hleihel W, Boué-Grabot E, Landry M. P2X4 signalling contributes to hyperactivity but not pain sensitization comorbidity in a mouse model of attention deficit/hyperactivity disorder. Front Pharmacol 2024; 14:1288994. [PMID: 38239187 PMCID: PMC10794506 DOI: 10.3389/fphar.2023.1288994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/12/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction: Attention deficit/hyperactivity disorder (ADHD) is a common neurodevelopmental disorder characterized by hyperactivity, inattention, and impulsivity that often persist until adulthood. Frequent comorbid disorders accompany ADHD and two thirds of children diagnosed with ADHD also suffer from behavioural disorders and from alteration of sensory processing. We recently characterized the comorbidity between ADHD-like symptoms and pain sensitisation in a pharmacological mouse model of ADHD, and we demonstrated the implication of the anterior cingulate cortex and posterior insula. However, few studies have explored the causal mechanisms underlying the interactions between ADHD and pain. The implication of inflammatory mechanisms has been suggested but the signalling pathways involved have not been explored. Methods: We investigated the roles of purinergic signalling, at the crossroad of pain and neuroinflammatory pathways, by using a transgenic mouse line that carries a total deletion of the P2X4 receptor. Results: We demonstrated that P2X4 deletion prevents hyperactivity in the mouse model of ADHD. In contrast, the absence of P2X4 lowered thermal pain thresholds in sham conditions and did not affect pain sensitization in ADHD-like conditions. We further analysed microglia reactivity and the expression of inflammatory markers in wild type and P2X4KO mice. Our results revealed that P2X4 deletion limits microglia reactivity but at the same time exerts proinflammatory effects in the anterior cingulate cortex and posterior insula. Conclusion: This dual role of P2X4 could be responsible for the differential effects noted on ADHD-like symptoms and pain sensitization and calls for further studies to investigate the therapeutic benefit of targeting the P2X4 receptor in ADHD patients.
Collapse
Affiliation(s)
- Sarah Bou Sader Nehme
- University of Bordeaux, CNRS, Institute of Neurodegenerative Diseases, IMN, UMR 5293, Bordeaux, France
- Department of Biology, Faculty of Arts and Sciences, Holy Spirit University of Kaslik, Jounieh, Lebanon
| | - Sandra Sanchez-Sarasua
- University of Bordeaux, CNRS, Institute of Neurodegenerative Diseases, IMN, UMR 5293, Bordeaux, France
- Faculty of Health Sciences, University of Jaume I, Castellon, Spain
| | - Ramy Adel
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Marie Tuifua
- University of Bordeaux, CNRS, Institute of Neurodegenerative Diseases, IMN, UMR 5293, Bordeaux, France
| | - Awatef Ali
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Amina E. Essawy
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Sherine Abdel Salam
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Walid Hleihel
- Department of Biology, Faculty of Arts and Sciences, Holy Spirit University of Kaslik, Jounieh, Lebanon
| | - Eric Boué-Grabot
- University of Bordeaux, CNRS, Institute of Neurodegenerative Diseases, IMN, UMR 5293, Bordeaux, France
| | - Marc Landry
- University of Bordeaux, CNRS, Institute of Neurodegenerative Diseases, IMN, UMR 5293, Bordeaux, France
| |
Collapse
|
158
|
Garcia-Segura ME, Pluchino S, Peruzzotti-Jametti L. Metabolic Control of Microglia. ADVANCES IN NEUROBIOLOGY 2024; 37:607-622. [PMID: 39207716 DOI: 10.1007/978-3-031-55529-9_34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia, immune sentinels of the central nervous system (CNS), play a critical role in maintaining its health and integrity. This chapter delves into the concept of immunometabolism, exploring how microglial metabolism shapes their diverse immune functions. It examines the impact of cell metabolism on microglia during various CNS states, including homeostasis, development, aging, and inflammation. Particularly in CNS inflammation, the chapter discusses how metabolic rewiring in microglia can initiate, resolve, or perpetuate inflammatory responses. The potential of targeting microglial metabolism as a therapeutic strategy for chronic CNS disorders with prominent innate immune cell activation is also explored.
Collapse
Affiliation(s)
- Monica Emili Garcia-Segura
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Stefano Pluchino
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK.
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
159
|
Jucá PM, de Almeida Duque É, Covre LHH, Mariano KAA, Munhoz CD. Microglia and Systemic Immunity. ADVANCES IN NEUROBIOLOGY 2024; 37:287-302. [PMID: 39207698 DOI: 10.1007/978-3-031-55529-9_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia are specialized immune cells that reside in the central nervous system (CNS) and play a crucial role in maintaining the homeostasis of the brain microenvironment. While traditionally regarded as a part of the innate immune system, recent research has highlighted their role in adaptive immunity. The CNS is no longer considered an immune-privileged organ, and increasing evidence suggests bidirectional communication between the immune system and the CNS. Microglia are sensitive to systemic immune signals and can respond to systemic inflammation by producing various inflammatory cytokines and chemokines. This response is mediated by activating pattern recognition receptors (PRRs), which recognize pathogen- and danger-associated molecular patterns in the systemic circulation. The microglial response to systemic inflammation has been implicated in several neurological conditions, including depression, anxiety, and cognitive impairment. Understanding the complex interplay between microglia and systemic immunity is crucial for developing therapeutic interventions to modulate immune responses in the CNS.
Collapse
Affiliation(s)
- Paloma Marinho Jucá
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, São Paulo, Brazil
| | - Érica de Almeida Duque
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, São Paulo, Brazil
| | - Luiza Helena Halas Covre
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, São Paulo, Brazil
| | | | - Carolina Demarchi Munhoz
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, São Paulo, Brazil.
| |
Collapse
|
160
|
Howlader MSI, Prateeksha P, Hansda S, Naidu P, Das M, Barthels D, Das H. Secretory products of DPSC mitigate inflammatory effects in microglial cells by targeting MAPK pathway. Biomed Pharmacother 2024; 170:115971. [PMID: 38039760 DOI: 10.1016/j.biopha.2023.115971] [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: 08/16/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 12/03/2023] Open
Abstract
Activated microglial cells in the central nervous system (CNS) are the main contributors to neurodegenerative disorders such as Alzheimer's disease and Parkinson's disease. Inhibiting their activation will help in reducing inflammation and oxidative stress during pathogenesis, potentially limiting the progression of the diseases. The immunomodulation properties of dental pulp-derived stem cells (DPSC) make it a promising therapy for neurodegenerative disorders. This study aims to determine whether secretory factors of DPSC (DPSC℗) inhibit inflammation and proliferation of microglial cells and define the molecular mechanisms. Our quantitative RT-PCR analysis showed that the DPSC℗ reduced the markers of the inflammation and induced anti-inflammatory molecules in microglial cells. DPSC ℗ reduced the intracellular and mitochondrial reactive oxygen species (ROS) production and mitochondrial membrane potential in microglial cells. In addition, DPSC ℗ decreased the cellular bioenergetics parameters related to oxygen consumption rate (OCAR) and extracellular acidification rate (ECAR). We found that DPSC℗ inhibited microglial cell proliferation by activating a checkpoint molecule, Chk1 leading an arrest at the G1 phase of the cell cycle. To define the mechanism, we performed the western blot analysis and observed that the MAPK P38 pathway was inhibited by DPSC℗. Furthermore, a System biology analysis revealed that the BDNF and GDNF, secretory factors of DPSC, blocked at the phosphorylation site (Tyr 182) of the P38 molecule resulting in the inhibition of downstream signaling of inflammation. These data suggest that the DPSC℗ may be a potential therapeutic agent for neurodegenerative diseases.
Collapse
Affiliation(s)
- Md Sariful Islam Howlader
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Prateeksha Prateeksha
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Surajit Hansda
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Prathyusha Naidu
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Manjusri Das
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Derek Barthels
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Hiranmoy Das
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA.
| |
Collapse
|
161
|
Li Y, Chen X, Zhou M, Feng S, Peng X, Wang Y. Microglial TLR4/NLRP3 Inflammasome Signaling in Alzheimer's Disease. J Alzheimers Dis 2024; 97:75-88. [PMID: 38043010 DOI: 10.3233/jad-230273] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2023]
Abstract
Alzheimer's disease is a pervasive neurodegenerative disease that is estimated to represent approximately 70% of dementia cases worldwide, and the molecular complexity that has been highlighted remains poorly understood. The accumulation of extracellular amyloid-β (Aβ), intracellular neurofibrillary tangles formed by tau hyperphosphorylation, and neuroinflammation are the major pathological features of Alzheimer's disease (AD). Over the years, there has been no apparent breakthrough in drug discovery based on the Aβ and tau hypotheses. Neuroinflammation has gradually become a hot spot in AD treatment research. As the primary cells of innate immunity in the central nervous system, microglia play a key role in neuroinflammation. Toll-like receptor 4 (TLR4) and nucleotide-binding oligomerization domain-like receptor 3 (NLRP3) inflammasomes are vital molecules in neuroinflammation. In the pathological context of AD, the complex interplay between TLR4 and the NLRP3 inflammasomes in microglia influences AD pathology via neuroinflammation. In this review, the effect of the activation and inhibition of TLR4 and NLRP3 in microglia on AD pathology, as well as the cross-talk between TLR4 and the NLRP3 inflammasome, and the influence of essential molecules in the relevant signaling pathway on AD pathology, were expounded. In addition, the feasibility of these factors in representing a potential treatment option for AD has been clarified.
Collapse
Affiliation(s)
- Yunfeng Li
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiongjin Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Mulan Zhou
- Department of Pharmacy, The People's Hospital of Gaozhou, Maoming, China
| | - Sifan Feng
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaoping Peng
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yan Wang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
162
|
Au NPB, Wu T, Kumar G, Jin Y, Li YYT, Chan SL, Lai JHC, Chan KWY, Yu KN, Wang X, Ma CHE. Low-dose ionizing radiation promotes motor recovery and brain rewiring by resolving inflammatory response after brain injury and stroke. Brain Behav Immun 2024; 115:43-63. [PMID: 37774892 DOI: 10.1016/j.bbi.2023.09.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/24/2023] [Accepted: 09/22/2023] [Indexed: 10/01/2023] Open
Abstract
Traumatic brain injury (TBI) and stroke share a common pathophysiology that worsens over time due to secondary tissue injury caused by sustained inflammatory response. However, studies on pharmacological interventions targeting the complex secondary injury cascade have failed to show efficacy. Here, we demonstrated that low-dose ionizing radiation (LDIR) reduced lesion size and reversed motor deficits after TBI and photothrombotic stroke. Magnetic resonance imaging demonstrated significant reduction of infarct volume in LDIR-treated mice after stroke. Systems-level transcriptomic analysis showed that genes upregulated in LDIR-treated stoke mice were enriched in pathways associated with inflammatory and immune response involving microglia. LDIR induced upregulation of anti-inflammatory- and phagocytosis-related genes, and downregulation of key pro-inflammatory cytokine production. These findings were validated by live-cell assays, in which microglia exhibited higher chemotactic and phagocytic capacities after LDIR. We observed substantial microglial clustering at the injury site, glial scar clearance and reversal of motor deficits after stroke. Cortical microglia/macrophages depletion completely abolished the beneficial effect of LDIR on motor function recovery in stroke mice. LDIR promoted axonal projections (brain rewiring) in motor cortex and recovery of brain activity detected by electroencephalography recordings months after stroke. LDIR treatment delayed by 8 h post-injury still maintained full therapeutic effects on motor recovery, indicating that LDIR is a promising therapeutic strategy for TBI and stroke.
Collapse
Affiliation(s)
| | - Tan Wu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China; Department of Surgery, Chinese University of Hong Kong, Hong Kong, China
| | - Gajendra Kumar
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | - Yuting Jin
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | | | - Shun Lam Chan
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | - Joseph Ho Chi Lai
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Kannie Wai Yan Chan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| | - Kwan Ngok Yu
- Department of Physics, City University of Hong Kong, Hong Kong, China
| | - Xin Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China; Department of Surgery, Chinese University of Hong Kong, Hong Kong, China
| | - Chi Him Eddie Ma
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China; City University of Hong Kong Shenzhen Research Institute, Shenzhen, China.
| |
Collapse
|
163
|
Tang J, Yousaf M, Wu YP, Li QQ, Xu YQ, Liu DM. Mechanisms and structure-activity relationships of polysaccharides in the intervention of Alzheimer's disease: A review. Int J Biol Macromol 2024; 254:127553. [PMID: 37865357 DOI: 10.1016/j.ijbiomac.2023.127553] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disease. Despite several decades of research, the development of effective treatments and responses for Alzheimer's disease remains elusive. The utilization of polysaccharides for Alzheimer's disease became more popular due to their beneficial characteristics, notably their multi-target activity and low toxicity. This review mainly focuses on the researches of recent 5 years in the regulation of AD by naturally derived polysaccharides, systematically lists the possible intervention pathways of polysaccharides from different mechanisms, and explores the structure-activity relationship between polysaccharide structural activities, so as to provide references for the intervention and treatment of AD by polysaccharides.
Collapse
Affiliation(s)
- Jun Tang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, Guangdong, China
| | - Muhammad Yousaf
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, Guangdong, China
| | - Ya-Ping Wu
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, Guangdong, China
| | - Qin-Qin Li
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, Guangdong, China
| | - Yi-Qian Xu
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, Guangdong, China
| | - Dong-Mei Liu
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, Guangdong, China.
| |
Collapse
|
164
|
von Bernhardi R, Eugenín J. Aging Microglia and Their Impact in the Nervous System. ADVANCES IN NEUROBIOLOGY 2024; 37:379-395. [PMID: 39207703 DOI: 10.1007/978-3-031-55529-9_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Aging is the greatest risk factor for neurodegenerative diseases. Microglia are the resident immune cells in the central nervous system (CNS), playing key roles in its normal functioning, and as mediators for age-dependent changes of the CNS, condition at which they generate a hostile environment for neurons. Transforming Growth Factor β1 (TGFβ1) is a regulatory cytokine involved in immuneregulation and neuroprotection, affecting glial cell inflammatory activation, neuronal survival, and function. TGFβ1 signaling undergoes age-dependent changes affecting the regulation of microglial cells and can contribute to the pathophysiology of neurodegenerative diseases. This chapter focuses on assessing the role of age-related changes on the regulation of microglial cells and their impact on neuroinflammation and neuronal function, for understanding age-dependent changes of the nervous system.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Faculty of Odontology and Rehabilitation Sciences, Universidad San Sebastian, Santiago, Chile.
| | - Jaime Eugenín
- Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
165
|
Long Y, Li XQ, Deng J, Ye QB, Li D, Ma Y, Wu YY, Hu Y, He XF, Wen J, Shi A, Yu S, Shen L, Ye Z, Zheng C, Li N. Modulating the polarization phenotype of microglia - A valuable strategy for central nervous system diseases. Ageing Res Rev 2024; 93:102160. [PMID: 38065225 DOI: 10.1016/j.arr.2023.102160] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023]
Abstract
Central nervous system (CNS) diseases have become one of the leading causes of death in the global population. The pathogenesis of CNS diseases is complicated, so it is important to find the patterns of the disease to improve the treatment strategy. Microglia are considered to be a double-edged sword, playing both harmful and beneficial roles in CNS diseases. Therefore, it is crucial to understand the progression of the disease and the changes in the polar phenotype of microglia to provide guidance in the treatment of CNS diseases. Microglia activation may evolve into different phenotypes: M1 and M2 types. We focused on the roles that M1 and M2 microglia play in regulating intercellular dialogues, pathological reactions and specific diseases in CNS diseases. Importantly, we summarized the strategies used to modulate the polarization phenotype of microglia, including traditional pharmacological modulation, biological therapies, and physical strategies. This review will contribute to the development of potential strategies to modulate microglia polarization phenotypes and provide new alternative therapies for CNS diseases.
Collapse
Affiliation(s)
- Yu Long
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Xiao-Qiu Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Jie Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Qiao-Bo Ye
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Dan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yin Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yuan-Yuan Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yue Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Xiao-Fang He
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Jing Wen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Ai Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Shuang Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Lin Shen
- Second Teaching Hospital of Tianjin University of Traditional Chinese Medine, Tianjin, China.
| | - Zhen Ye
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Chuan Zheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Nan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
166
|
Su WJ, Hu T, Jiang CL. Cool the Inflamed Brain: A Novel Anti-inflammatory Strategy for the Treatment of Major Depressive Disorder. Curr Neuropharmacol 2024; 22:810-842. [PMID: 37559243 PMCID: PMC10845090 DOI: 10.2174/1570159x21666230809112028] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/21/2023] [Accepted: 02/23/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Abundant evidence suggests that inflammatory cytokines contribute to the symptoms of major depressive disorder (MDD) by altering neurotransmission, neuroplasticity, and neuroendocrine processes. Given the unsatisfactory response and remission of monoaminergic antidepressants, anti-inflammatory therapy is proposed as a feasible way to augment the antidepressant effect. Recently, there have been emerging studies investigating the efficiency and efficacy of anti-inflammatory agents in the treatment of MDD and depressive symptoms comorbid with somatic diseases. METHODS In this narrative review, prospective clinical trials focusing on anti-inflammatory treatment for depression have been comprehensively searched and screened. Based on the included studies, we summarize the rationale for the anti-inflammatory therapy of depression and discuss the utilities and confusions regarding the anti-inflammatory strategy for MDD. RESULTS This review included over 45 eligible trials. For ease of discussion, we have grouped them into six categories based on their mechanism of action, and added some other anti-inflammatory modalities, including Chinese herbal medicine and non-drug therapy. Pooled results suggest that anti-inflammatory therapy is effective in improving depressive symptoms, whether used as monotherapy or add-on therapy. However, there remain confusions in the application of anti-inflammatory therapy for MDD. CONCLUSION Based on current clinical evidence, anti-inflammatory therapy is a promisingly effective treatment for depression. This study proposes a novel strategy for clinical diagnosis, disease classification, personalized treatment, and prognostic prediction of depression. Inflammatory biomarkers are recommended to be assessed at the first admission of MDD patients, and anti-inflammatory therapy are recommended to be included in the clinical practice guidelines for diagnosis and treatment. Those patients with high levels of baseline inflammation (e.g., CRP > 3 mg/L) may benefit from adjunctive anti-inflammatory therapy.
Collapse
Affiliation(s)
- Wen-Jun Su
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, Shanghai, 200433, China
| | - Ting Hu
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, Shanghai, 200433, China
| | - Chun-Lei Jiang
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, Shanghai, 200433, China
| |
Collapse
|
167
|
Zhang Y, Yang Y, Li H, Feng Q, Ge W, Xu X. Investigating the Potential Mechanisms and Therapeutic Targets of Inflammatory Cytokines in Post-stroke Depression. Mol Neurobiol 2024; 61:132-147. [PMID: 37592185 DOI: 10.1007/s12035-023-03563-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: 05/05/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023]
Abstract
Post-stroke depression (PSD) affects approximately one-third of stroke survivors, severely impacting general recovery and quality of life. Despite extensive studies, the exact mechanisms underlying PSD remain elusive. However, emerging evidence implicates proinflammatory cytokines, including interleukin-1β, interleukin-6, tumor necrosis factor-alpha, and interleukin-18, play critical roles in PSD development. These cytokines contribute to PSD through various mechanisms, including hypothalamic-pituitary-adrenal (HPA) axis dysfunction, neurotransmitter alterations, neurotrophic factor changes, gut microbiota imbalances, and genetic predispositions. This review is aimed at exploring the role of cytokines in stroke and PSD while identifying their potential as specific therapeutic targets for managing PSD. A more profound understanding of the mechanisms regulating inflammatory cytokine expression and anti-inflammatory cytokines like interleukin-10 in PSD may facilitate the development of innovative interventions to improve outcomes for stroke survivors experiencing depression.
Collapse
Affiliation(s)
- Yutong Zhang
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Yuehua Yang
- Department of Neurology, Suzhou Yongding Hospital, Suzhou, 215028, China
| | - Hao Li
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Qian Feng
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Wei Ge
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221600, China.
| | - Xingshun Xu
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China.
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
168
|
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
|
169
|
Galvis-Montes DS, van Loo KMJ, van Waardenberg AJ, Surges R, Schoch S, Becker AJ, Pitsch J. Highly dynamic inflammatory and excitability transcriptional profiles in hippocampal CA1 following status epilepticus. Sci Rep 2023; 13:22187. [PMID: 38092829 PMCID: PMC10719343 DOI: 10.1038/s41598-023-49310-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023] Open
Abstract
Transient brain insults including status epilepticus (SE) can initiate a process termed 'epileptogenesis' that results in chronic temporal lobe epilepsy. As a consequence, the entire tri-synaptic circuit of the hippocampus is fundamentally impaired. A key role in epileptogenesis has been attributed to the CA1 region as the last relay station in the hippocampal circuit and as site of aberrant plasticity, e.g. mediated by acquired channelopathies. The transcriptional profiles of the distinct hippocampal neurons are highly dynamic during epileptogenesis. Here, we aimed to elucidate the early SE-elicited mRNA signature changes and the respective upstream regulatory cascades in CA1. RNA sequencing of CA1 was performed in the mouse pilocarpine-induced SE model at multiple time points ranging from 6 to 72 h after the initial insult. Bioinformatics was used to decipher altered gene expression, signalling cascades and their corresponding cell type profiles. Robust transcriptomic changes were detected at 6 h after SE and at subsequent time points during early epileptogenesis. Major differentially expressed mRNAs encoded primarily immediate early and excitability-related gene products, as well as genes encoding immune signalling factors. Binding sites for the transcription factors Nfkb1, Spi1, Irf8, and two Runx family members, were enriched within promoters of differentially expressed genes related to major inflammatory processes, whereas the transcriptional repressors Suz12, Nfe2l2 and Rest were associated with hyperexcitability and GABA / glutamate receptor activity. CA1 quickly responds to SE by inducing transcription of genes linked to inflammation and excitation stress. Transcription factors mediating this transcriptomic switch represent targets for new highly selected, cell type and time window-specific anti-epileptogenic strategies.
Collapse
Grants
- SCHO 820/4-1, SCHO 820/6-1, SCHO 820/7-1, SCHO 820/5-2, SPP1757, SFB1089, FOR 2715 Deutsche Forschungsgemeinschaft
- SCHO 820/4-1, SCHO 820/6-1, SCHO 820/7-1, SCHO 820/5-2, SPP1757, SFB1089, FOR 2715 Deutsche Forschungsgemeinschaft
- Promotionskolleg 'NeuroImmunology' Else Kröner-Fresenius-Stiftung
- Promotionskolleg 'NeuroImmunology' Else Kröner-Fresenius-Stiftung
- BONFOR program of the Medical Faculty, University of Bonn
- Rheinische Friedrich-Wilhelms-Universität Bonn (1040)
Collapse
Affiliation(s)
- Daniel S Galvis-Montes
- Department of Epileptology, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Karen M J van Loo
- Department of Epileptology, Neurology, RWTH Aachen University, Aachen, Germany
| | | | - Rainer Surges
- Department of Epileptology, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Susanne Schoch
- Department of Epileptology, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Albert J Becker
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Julika Pitsch
- Department of Epileptology, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
170
|
Lai S, Wang P, Gong J, Zhang S. New insights into the role of GSK-3β in the brain: from neurodegenerative disease to tumorigenesis. PeerJ 2023; 11:e16635. [PMID: 38107562 PMCID: PMC10722984 DOI: 10.7717/peerj.16635] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/18/2023] [Indexed: 12/19/2023] Open
Abstract
Glycogen synthase kinase 3 (GSK-3) is a serine/threonine kinase widely expressed in various tissues and organs. Unlike other kinases, GSK-3 is active under resting conditions and is inactivated upon stimulation. In mammals, GSK-3 includes GSK-3 α and GSK-3β isoforms encoded by two homologous genes, namely, GSK3A and GSK3B. GSK-3β is essential for the control of glucose metabolism, signal transduction, and tissue homeostasis. As more than 100 known proteins have been identified as GSK-3β substrates, it is sometimes referred to as a moonlighting kinase. Previous studies have elucidated the regulation modes of GSK-3β. GSK-3β is involved in almost all aspects of brain functions, such as neuronal morphology, synapse formation, neuroinflammation, and neurological disorders. Recently, several comparatively specific small molecules have facilitated the chemical manipulation of this enzyme within cellular systems, leading to the discovery of novel inhibitors for GSK-3β. Despite these advancements, the therapeutic significance of GSK-3β as a drug target is still complicated by uncertainties surrounding the potential of inhibitors to stimulate tumorigenesis. This review provides a comprehensive overview of the intricate mechanisms of this enzyme and evaluates the existing evidence regarding the therapeutic potential of GSK-3β in brain diseases, including Alzheimer's disease, Parkinson's disease, mood disorders, and glioblastoma.
Collapse
Affiliation(s)
- Shenjin Lai
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Peng Wang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jingru Gong
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Shuaishuai Zhang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| |
Collapse
|
171
|
Malvaso A, Gatti A, Negro G, Calatozzolo C, Medici V, Poloni TE. Microglial Senescence and Activation in Healthy Aging and Alzheimer's Disease: Systematic Review and Neuropathological Scoring. Cells 2023; 12:2824. [PMID: 38132144 PMCID: PMC10742050 DOI: 10.3390/cells12242824] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
The greatest risk factor for neurodegeneration is the aging of the multiple cell types of human CNS, among which microglia are important because they are the "sentinels" of internal and external perturbations and have long lifespans. We aim to emphasize microglial signatures in physiologic brain aging and Alzheimer's disease (AD). A systematic literature search of all published articles about microglial senescence in human healthy aging and AD was performed, searching for PubMed and Scopus online databases. Among 1947 articles screened, a total of 289 articles were assessed for full-text eligibility. Microglial transcriptomic, phenotypic, and neuropathological profiles were analyzed comprising healthy aging and AD. Our review highlights that studies on animal models only partially clarify what happens in humans. Human and mice microglia are hugely heterogeneous. Like a two-sided coin, microglia can be protective or harmful, depending on the context. Brain health depends upon a balance between the actions and reactions of microglia maintaining brain homeostasis in cooperation with other cell types (especially astrocytes and oligodendrocytes). During aging, accumulating oxidative stress and mitochondrial dysfunction weaken microglia leading to dystrophic/senescent, otherwise over-reactive, phenotype-enhancing neurodegenerative phenomena. Microglia are crucial for managing Aβ, pTAU, and damaged synapses, being pivotal in AD pathogenesis.
Collapse
Affiliation(s)
- Antonio Malvaso
- IRCCS “C. Mondino” Foundation, National Neurological Institute, Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (A.M.); (A.G.)
| | - Alberto Gatti
- IRCCS “C. Mondino” Foundation, National Neurological Institute, Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (A.M.); (A.G.)
| | - Giulia Negro
- Department of Neurology, University of Milano Bicocca, 20126 Milan, Italy;
| | - Chiara Calatozzolo
- Department of Neurology and Neuropathology, Golgi-Cenci Foundation, Abbiategrasso, 20081 Milan, Italy;
| | - Valentina Medici
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy;
| | - Tino Emanuele Poloni
- Department of Neurology and Neuropathology, Golgi-Cenci Foundation, Abbiategrasso, 20081 Milan, Italy;
| |
Collapse
|
172
|
Martinez M, Yu W, Menden HL, Lei T, Monaghan-Nichols P, Sampath V. Butyrate suppresses experimental necrotizing enterocolitis-induced brain injury in mice. Front Pediatr 2023; 11:1284085. [PMID: 38130941 PMCID: PMC10733464 DOI: 10.3389/fped.2023.1284085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023] Open
Abstract
Background Necrotizing enterocolitis (NEC) is a devastating disease in premature infants, and 50% of infants with surgical NEC develop neurodevelopmental defects. The mechanisms by which NEC-induced cytokine release and activation of inflammatory cells in the brain mediate neuronal injury, and whether enteral immunotherapy attenuates NEC-associated brain injury remain understudied. Based on our prior work, which demonstrated that experimental NEC-like intestinal injury is attenuated by the short-chain fatty acid, butyrate, in this study, we hypothesize that NEC-induced brain injury would be suppressed by enteral butyrate supplementation. Methods A standardized NEC mouse model [enteral formula feeding, lipopolysaccharide (LPS), and hypoxia] was used. Mice were randomized into the following groups: control, NEC, butyrate pretreated NEC, and butyrate control. NEC scoring (1-4 with 4 representing severe injury) was performed on ileal sections using a validated scoring system. Intestinal and brain lysates were used to assess inflammation, proinflammatory signaling, and apoptosis. Results NEC-induced intestinal injury was attenuated by butyrate supplementation. NEC-induced microglial activation in the cerebral cortex and hippocampus was suppressed with butyrate. NEC increased the number of activated microglial cells but decreased the number of oligodendrocytes. Butyrate pretreatment attenuated these changes. Increased activation of proinflammatory Toll-like receptor signaling, cytokine expression, and induction of GFAP and IBA1 in the cerebral cortex observed with NEC was suppressed with butyrate. Conclusion Experimental NEC induced inflammation and activation of microglia in several regions of the brain, most prominently in the cortex. NEC-induced neuroinflammation was suppressed with butyrate pretreatment. The addition of short-chain fatty acids to diet may be used to attenuate NEC-induced intestinal injury and neuroinflammation in preterm infants.
Collapse
Affiliation(s)
- Maribel Martinez
- Division of Neonatology, Department of Pediatrics, Children’s Mercy Kansas City, Kansas, MO, United States
- Neonatal Diseases Research Program, Children’s Mercy Research Institute, Children’s Mercy Kansas City, Kansas, MO, United States
| | - Wei Yu
- Division of Neonatology, Department of Pediatrics, Children’s Mercy Kansas City, Kansas, MO, United States
- Neonatal Diseases Research Program, Children’s Mercy Research Institute, Children’s Mercy Kansas City, Kansas, MO, United States
| | - Heather L. Menden
- Division of Neonatology, Department of Pediatrics, Children’s Mercy Kansas City, Kansas, MO, United States
- Neonatal Diseases Research Program, Children’s Mercy Research Institute, Children’s Mercy Kansas City, Kansas, MO, United States
| | - Tianhua Lei
- Department of Biomedical Sciences, University of Missouri Kansas City School of Medicine, Kansas, MO, United States
| | - Paula Monaghan-Nichols
- Department of Biomedical Sciences, University of Missouri Kansas City School of Medicine, Kansas, MO, United States
| | - Venkatesh Sampath
- Division of Neonatology, Department of Pediatrics, Children’s Mercy Kansas City, Kansas, MO, United States
- Neonatal Diseases Research Program, Children’s Mercy Research Institute, Children’s Mercy Kansas City, Kansas, MO, United States
| |
Collapse
|
173
|
Deng C, Li M, Liu Y, Yan C, He Z, Chen ZY, Zhu H. Cholesterol Oxidation Products: Potential Adverse Effect and Prevention of Their Production in Foods. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:18645-18659. [PMID: 38011512 DOI: 10.1021/acs.jafc.3c05158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Cholesterol oxidation products (COPs) are a group of substances formed during food processing. COPs in diet is a health concern because they may affect human health in association with the risk of various diseases including atherosclerosis, Alzheimer's disease, age-related macular degeneration, diabetes, and chronic gastrointestinal inflammatory colitis. Production of COPs in foods can be affected by many factors such as temperature, pH, light, oxygen, water, carbohydrates, fatty acids, proteins, and metal cations. The key issue is preventing its generation in foods. Some COPs can also be produced in vivo by both nonenzymatic and enzymatic-catalyzed oxidation reactions. Currently, a number of natural antioxidants such as catechins, flavonoids, and other polyphenols have been proven to inhibit the generation of COPs. In addition, measures taken during food processing can also minimize the production of COPs, such as the Maillard reaction and marinating food with plant polyphenol-rich seasonings. In conclusion, a comprehensive approach encompassing the suppression on COPs generation and implementation of processing measures is imperative to safeguard human health against the production of COPs in the food chain.
Collapse
Affiliation(s)
- Chuanling Deng
- School of Food Science and Engineering/Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing/National Technical Center (Foshan) for Quality Control of Famous and Special Agricultural Products (CAQS-GAP-KZZX043), Foshan University, Foshan 528000, Guangdong China
| | - Mingxuan Li
- School of Food Science and Engineering/Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing/National Technical Center (Foshan) for Quality Control of Famous and Special Agricultural Products (CAQS-GAP-KZZX043), Foshan University, Foshan 528000, Guangdong China
- School of Life Sciences, South China Agricultural University, Guangzhou 510000, Guangdong China
| | - Yang Liu
- School of Food Science and Engineering/Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing/National Technical Center (Foshan) for Quality Control of Famous and Special Agricultural Products (CAQS-GAP-KZZX043), Foshan University, Foshan 528000, Guangdong China
| | - Chi Yan
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT 999077, Hong Kong China
| | - Zouyan He
- School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi China
| | - Zhen-Yu Chen
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT 999077, Hong Kong China
| | - Hanyue Zhu
- School of Food Science and Engineering/Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing/National Technical Center (Foshan) for Quality Control of Famous and Special Agricultural Products (CAQS-GAP-KZZX043), Foshan University, Foshan 528000, Guangdong China
| |
Collapse
|
174
|
Mohapatra S, Tripathi S, Sharma V, Basu A. Regulation of microglia-mediated inflammation by host lncRNA Gm20559 upon flaviviral infection. Cytokine 2023; 172:156383. [PMID: 37801852 DOI: 10.1016/j.cyto.2023.156383] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/01/2023] [Accepted: 09/25/2023] [Indexed: 10/08/2023]
Abstract
BACKGROUND Japanese Encephalitis Virus (JEV) and West Nile Viruses (WNV) are neurotropic flaviviruses which cause neuronal death and exaggerated glial activation in the central nervous system. Role of host long non coding RNAs in shaping microglial inflammation upon flavivirus infections has been unexplored. This study attempted to decipher the role of lncRNA Gm20559 in regulating microglial inflammatory response in context of flaviviruses. METHODS Antisense oligonucleotide LNA Gapmers designed against lncRNA Gm20559 and non-specific site (negative control) were used for Gm20559 knockdown in JEV and WNV-infected N9 microglial cells. Upon establishing successful Gm20559 knockdown, expression of various proinflammatory cytokines, chemokines, interferon-stimulated genes (ISGs) and RIG-I were checked by qRT-PCR and cytometric bead array. Western Blotting was done to analyse the phosphorylation level of various inflammatory markers and viral non-structural protein expression. Plaque Assays were employed to quantify viral titres in microglial supernatant upon knocking down Gm20559. Effect of microglial supernatant on HT22 neuronal cells was assessed by checking expression of apoptotic protein and viral non-structural protein by Western Blotting. RESULTS Upregulation in Gm20559 expression was observed in BALB/c pup brains, primary microglia as well as N9 microglia cell line upon both JEV and WNV infection. Knockdown of Gm20559 in JEV and WNV-infected N9 cell led to the reduction of major proinflammatory cytokines - IL-1β, IL-6, IP-10 and IFN-β. Inhibition of Gm20559 upon JEV infection in N9 microglia also led to downregulation of RIG-I and OAS-2, which was not the case in WNV-infected N9 microglia. Phosphorylation level of P38 MAPK was reduced in case of JEV-infected N9 microglia and not WNV-infected N9 microglia. Whereas phosphorylation of NF-κB pathway was unchanged upon Gm20559 knockdown in both JEV and WNV-infected N9 microglia. However, treating HT22 cells with JEV and WNV-infected microglial supernatant with and without Gm20559 could not trigger cell death or influence viral replication. CONCLUSION Knockdown studies on lncRNA Gm20559 suggests its pivotal role in maintaining the inflammatory milieu of microglia in flaviviral infection by modulating the expression of various pro-inflammatory cytokines. However, Gm20559-induced increased microglial proinflammatory response upon flavivirus infection fails to trigger neuronal death.
Collapse
Affiliation(s)
- Stuti Mohapatra
- National Brain Research Centre, Manesar, Haryana 122052, India
| | - Shraddha Tripathi
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Telangana 500078, India
| | - Vivek Sharma
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Telangana 500078, India.
| | - Anirban Basu
- National Brain Research Centre, Manesar, Haryana 122052, India.
| |
Collapse
|
175
|
Hao H, Hou Y, Li A, Niu L, Li S, He B, Zhang X, Song H, Cai R, Zhou Y, Yao C, Wang Y, Wang Y. HIF-1α promotes astrocytic production of macrophage migration inhibitory factor following spinal cord injury. CNS Neurosci Ther 2023; 29:3802-3814. [PMID: 37334735 PMCID: PMC10651974 DOI: 10.1111/cns.14300] [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: 03/15/2023] [Revised: 05/16/2023] [Accepted: 05/28/2023] [Indexed: 06/20/2023] Open
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF) is an important mediator of neuropathology in various central nervous system (CNS) diseases. However, little is known about its inducers for production from the nerve cells, as well as the underlying regulatory mechanism. Injury-induced HIF-1α has been shown to exacerbate neuroinflammation by activating multiple downstream target molecules. It is postulated that HIF-1α is involved in the regulation of MIF following spinal cord injury (SCI). METHODS SCI model of Sprague-Dawley rats was established by cord contusion at T8-T10. The dynamic changes of HIF-1α and MIF protein levels at lesion site of rat spinal cord were determined by Western blot. The specific cell types of HIF-1α and MIF expression were examined by immunostaining. Primary astrocytes were isolated from the spinal cord, cultured and stimulated with various agonist or inhibitor of HIF-1α for analysis of HIF-1α-mediated expression of MIF. Luciferase report assay was used to determine the relationship between HIF-1α and MIF. The Basso, Beattie, and Bresnahan (BBB) locomotor scale was used to assess the locomotor function following SCI. RESULTS The protein levels of HIF-1α and MIF at lesion site were significantly elevated by SCI. Immunofluorescence demonstrated that both HIF-1α and MIF were abundantly expressed in the astrocytes of the spinal cord. By using various agonists or inhibitors of HIF-1α, it was shown that HIF-1α sufficiently induced astrocytic production of MIF. Mechanistically, HIF-1α promoted MIF expression through interaction with MIF promoter. Inhibition of HIF-1α activity using specific inhibitor markedly reduced the protein levels of MIF at lesion site following SCI, which in turn favored for the functional recovery. CONCLUSION SCI-induced activation of HIF-1α is able to promote MIF production from astrocytes. Our results have provided new clues for SCI-induced production of DAMPs, which may be helpful for clinical treatment of neuroinflammation.
Collapse
Affiliation(s)
- Huifei Hao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Yuxuan Hou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Aicheng Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Li Niu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Shaolan Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Bingqiang He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Xingyuan Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Honghua Song
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Rixin Cai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Yue Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Chun Yao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Yongjun Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Yingjie Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| |
Collapse
|
176
|
Meleady L, Towriss M, Kim J, Bacarac V, Dang V, Rowland ME, Ciernia AV. Histone deacetylase 3 regulates microglial function through histone deacetylation. Epigenetics 2023; 18:2241008. [PMID: 37506371 PMCID: PMC10392760 DOI: 10.1080/15592294.2023.2241008] [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: 11/25/2022] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
As the primary innate immune cells of the brain, microglia respond to damage and disease through pro-inflammatory release of cytokines and neuroinflammatory molecules. Histone acetylation is an activating transcriptional mark that regulates inflammatory gene expression. Inhibition of histone deacetylase 3 (Hdac3) has been utilized in pre-clinical models of depression, stroke, and spinal cord injury to improve recovery following injury, but the molecular mechanisms underlying Hdac3's regulation of inflammatory gene expression in microglia is not well understood. To address this lack of knowledge, we examined how pharmacological inhibition of Hdac3 in an immortalized microglial cell line (BV2) impacted histone acetylation and gene expression of pro- and anti-inflammatory genes in response to immune challenge with lipopolysaccharide (LPS). Flow cytometry and cleavage under tags & release using nuclease (CUT & RUN) revealed that Hdac3 inhibition increases global and promoter-specific histone acetylation, resulting in the release of gene repression at baseline and enhanced responses to LPS. Hdac3 inhibition enhanced neuroprotective functions of microglia in response to LPS through reduced nitric oxide release and increased phagocytosis. The findings suggest Hdac3 serves as a regulator of microglial inflammation, and that inhibition of Hdac3 facilitates the microglial response to inflammation and its subsequent clearing of debris or damaged cells. Together, this work provides new mechanistic insights into therapeutic applications of Hdac3 inhibition which mediate reduced neuroinflammatory insults through microglial response.
Collapse
Affiliation(s)
- Laura Meleady
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Morgan Towriss
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Jennifer Kim
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | - Vince Bacarac
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Vivien Dang
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Megan E. Rowland
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Annie Vogel Ciernia
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| |
Collapse
|
177
|
Qiu F, Liu Y, Liu Y, Zhao Z, Zhou L, Chen P, Du Y, Wang Y, Sun H, Zeng C, Wang X, Liu Y, Pan H, Ke C. CD137L Inhibition Ameliorates Hippocampal Neuroinflammation and Behavioral Deficits in a Mouse Model of Sepsis-Associated Encephalopathy. Neuromolecular Med 2023; 25:616-631. [PMID: 37796401 PMCID: PMC10721669 DOI: 10.1007/s12017-023-08764-z] [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: 05/17/2023] [Accepted: 09/20/2023] [Indexed: 10/06/2023]
Abstract
Anxiety manifestations and cognitive dysfunction are common sequelae in patients with sepsis-associated encephalopathy (SAE). Microglia-mediated inflammatory signaling is involved in anxiety, depression, and cognitive dysfunction during acute infection with bacterial lipopolysaccharide (LPS). However, the molecular mechanisms underlying microglia activation and behavioral and cognitive deficits in sepsis have not been in fully elucidated. Based on previous research, we speculated that the CD137 receptor/ligand system modulates microglia function during sepsis to mediate classical neurological SAE symptoms. A murine model of SAE was established by injecting male C57BL/6 mice with LPS, and cultured mouse BV2 microglia were used for in vitro assays. RT-qPCR, immunofluorescence staining, flow cytometry, and ELISA were used to assess microglial activation and the expression of CD137L and inflammation-related cytokines in the mouse hippocampus and in cultured BV2 cells. In addition, behavioral tests were conducted in assess cognitive performance and behavioral distress. Immunofluorescence and RT-qPCR analyses showed that hippocampal expression of CD137L was upregulated in activated microglia following LPS treatment. Pre-treatment with the CD137L neutralizing antibody TKS-1 significantly reduced CD137L levels, attenuated the expression of M1 polarization markers in microglia, and inhibited the production of TNF-α, IL-1β, and IL-6 in both LPS-treated mice and BV2 cells. Conversely, stimulation of CD137L signaling by recombinant CD137-Fc fusion protein activated the synthesis and release of pro-inflammatory cytokines in cultures BV2 microglia. Importantly, open field, elevated plus maze, and Y-maze spontaneous alternation test results indicated that TKS-1 administration alleviated anxiety-like behavior and spatial memory decline in mice with LPS-induced SAE. These findings suggest that CD137L upregulation in activated microglia critically contributes to neuroinflammation, anxiety-like behavior, and cognitive dysfunction in the mouse model of LPS-induced sepsis. Therefore, therapeutic modulation of the CD137L/CD137 signaling pathway may represent an effective way to minimize brain damage and prevent cognitive and emotional deficits associated with SAE.
Collapse
Affiliation(s)
- Fang Qiu
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, Guangdong, China
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
- Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yueming Liu
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, Guangdong, China
| | - Yang Liu
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, Guangdong, China
| | - Zhuyun Zhao
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, Guangdong, China
| | - Lile Zhou
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, Guangdong, China
| | - Pengfei Chen
- Department of Traumatic Orthopedics, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Yunbo Du
- Department of Critical Care Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| | - Yanmei Wang
- Department of Critical Care Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| | - Huimin Sun
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| | - Xiaokang Wang
- Department of Pharmacy, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| | - Yuqiang Liu
- Department of Anesthesiology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518025, Guangdong, China.
| | - Haobo Pan
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China.
| | - Changneng Ke
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, Guangdong, China.
| |
Collapse
|
178
|
Steinberg N, Galleguillos D, Zaidi A, Horkey M, Sipione S. Naïve Huntington's disease microglia mount a normal response to inflammatory stimuli but display a partially impaired development of innate immune tolerance that can be counteracted by ganglioside GM1. J Neuroinflammation 2023; 20:276. [PMID: 37996924 PMCID: PMC10668379 DOI: 10.1186/s12974-023-02963-y] [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: 03/07/2023] [Accepted: 11/18/2023] [Indexed: 11/25/2023] Open
Abstract
Chronic activation and dysfunction of microglia have been implicated in the pathogenesis and progression of many neurodegenerative disorders, including Huntington's disease (HD). HD is a genetic condition caused by a mutation that affects the folding and function of huntingtin (HTT). Signs of microglia activation have been observed in HD patients even before the onset of symptoms. It is unclear, however, whether pro-inflammatory microglia activation in HD results from cell-autonomous expression of mutant HTT, is the response of microglia to a diseased brain environment, or both. In this study, we used primary microglia isolated from HD knock-in (Q140) and wild-type (Q7) mice to investigate their response to inflammatory conditions in vitro in the absence of confounding effects arising from brain pathology. We show that naïve Q140 microglia do not undergo spontaneous pro-inflammatory activation and respond to inflammatory triggers, including stimulation of TLR4 and TLR2 and exposure to necrotic cells, with similar kinetics of pro-inflammatory gene expression as wild-type microglia. Upon termination of the inflammatory insult, the transcription of pro-inflammatory cytokines is tapered off in Q140 and wild-type microglia with similar kinetics. However, the ability of Q140 microglia to develop tolerance in response to repeated inflammatory stimulations is partially impaired in vitro and in vivo, potentially contributing to the establishment of chronic neuroinflammation in HD. We further show that ganglioside GM1, a glycosphingolipid with anti-inflammatory effects on wild-type microglia, not only decreases the production of pro-inflammatory cytokines and nitric oxide in activated Q140 microglia, but also dramatically dampen microglia response to re-stimulation with LPS in an experimental model of tolerance. These effects are independent from the expression of interleukin 1 receptor associated kinase 3 (Irak-3), a strong modulator of LPS signaling involved in the development of innate immune tolerance and previously shown to be upregulated by immune cell treatment with gangliosides. Altogether, our data suggest that external triggers are required for HD microglia activation, but a cell-autonomous dysfunction that affects the ability of HD microglia to acquire tolerance might contribute to the establishment of neuroinflammation in HD. Administration of GM1 might be beneficial to attenuate chronic microglia activation and neuroinflammation.
Collapse
Affiliation(s)
- Noam Steinberg
- Department of Pharmacology, Neuroscience and Mental Health Institute and Glycomics Institute of Alberta, University of Alberta, Edmonton, AB, Canada
| | - Danny Galleguillos
- Department of Pharmacology, Neuroscience and Mental Health Institute and Glycomics Institute of Alberta, University of Alberta, Edmonton, AB, Canada
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Asifa Zaidi
- Department of Pharmacology, Neuroscience and Mental Health Institute and Glycomics Institute of Alberta, University of Alberta, Edmonton, AB, Canada
| | | | - Simonetta Sipione
- Department of Pharmacology, Neuroscience and Mental Health Institute and Glycomics Institute of Alberta, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
179
|
Zhao Y, Xia Q, Zong H, Wang Y, Dong H, Zhu L, Xia J, Mao Q, Weng Z, Liao W, Xin Z. Bibliometric and visual analysis of spinal cord injury-associated macrophages from 2002 to 2023. Front Neurol 2023; 14:1285908. [PMID: 38073628 PMCID: PMC10703361 DOI: 10.3389/fneur.2023.1285908] [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: 08/30/2023] [Accepted: 10/30/2023] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Spinal cord injury (SCI) triggers motor, sensory, and autonomic impairments that adversely damage patients' quality of life. Its pathophysiological processes include inflammation, oxidative stress, and apoptosis, although existing treatment options have little success. Macrophages have a vital function in controlling inflammation in SCI, with their M1-type and M2-type macrophages dominating early inflammatory effects and late brain tissue repair and regeneration, respectively. However, there is a dearth of rigorous bibliometric study in this sector to explore its dynamics and trends. This study intends to examine the current status and trends of macrophage usage in SCI using bibliometric methodologies, which may drive novel therapeutic options. METHODS In this study, the Web of Science Core Collection (WOSCC) was utilized to collect publications and reviews on macrophages in SCI from 2002 to 2023. Bibliometrics and visualization analyses were performed by VOSviewer, CiteSpace, the R package "bibliometrix", and online analytic platforms. These analyses covered a variety of aspects, including countries and institutions, authors and co-cited authors, journals and co-cited journals, subject categories, co-cited references, and keyword co-occurrences, in order to provide insights into the research trends and hotspots in this field. RESULTS 1,775 papers were included in the study, comprising 1,528 articles and 247 reviews. Our research analysis demonstrates that the number of relevant studies in this sector is expanding, specifically the number of publications in the United States and China has risen dramatically. However, there are fewer collaborations between institutions in different nations, and international cooperation needs to be reinforced. Among them, Popovich PG became the leader in the field, and significant journals include Experimental Neurology, Journal of Neurotrauma, and Journal of Neuroscience. Research hotspots involve macrophage polarization, microglia, astrocytes, signaling, cytokines, inflammation, and neuroprotection. CONCLUSIONS This analysis gives, for the first time, a comprehensive overview of bibliometric studies on macrophages in SCI over the past 20 years. This study not only gives an extensive picture of the knowledge structure but also indicates trends in the subject. The systematic summarization gives a complete and intuitive understanding of the link between spinal cord damage and macrophages and provides a great reference for future related studies.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Qiuqiu Xia
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Hui Zong
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yanyang Wang
- Department of Cell Engineering Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Huaize Dong
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Lu Zhu
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Jiyue Xia
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Qiming Mao
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Zijing Weng
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Wenbo Liao
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Zhijun Xin
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
180
|
Han H, Zhao Y, Du J, Wang S, Yang X, Li W, Song J, Zhang S, Zhang Z, Tan Y, Hatch GM, Zhang M, Chen L. Exercise improves cognitive dysfunction and neuroinflammation in mice through Histone H3 lactylation in microglia. Immun Ageing 2023; 20:63. [PMID: 37978517 PMCID: PMC10655345 DOI: 10.1186/s12979-023-00390-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Exercise is postulated to be a promising non-pharmacological intervention for the improvement of neurodegenerative disease pathology. However, the mechanism of beneficial effects of exercise on the brain remains to be further explored. In this study, we investigated the effect of an exercise-induced metabolite, lactate, on the microglia phenotype and its association with learning and memory. RESULTS Microglia were hyperactivated in the brains of AlCl3/D-gal-treated mice, which was associated with cognitive decline. Running exercise ameliorated the hyperactivation and increased the anti-inflammatory/reparative phenotype of microglia and improved cognition. Mice were injected intraperitoneally with sodium lactate (NaLA) had similar beneficial effects as that of exercise training. Exogenous NaLA addition to cultured BV2 cells promoted their transition from a pro-inflammatory to a reparative phenotype. CONCLUSION The elevated lactate acted as an "accelerator" of the endogenous "lactate timer" in microglia promoting this transition of microglia polarization balance through lactylation. These findings demonstrate that exercise-induced lactate accelerates the phenotypic transition of microglia, which plays a key role in reducing neuroinflammation and improving cognitive function.
Collapse
Affiliation(s)
- Hao Han
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, 130021, Jilin, China
| | - Yawei Zhao
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, 130021, Jilin, China
| | - Junda Du
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Sushan Wang
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, 130021, Jilin, China
| | - Xuehan Yang
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, 130021, Jilin, China
| | - Weijie Li
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, 130021, Jilin, China
| | - Jiayi Song
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, 130021, Jilin, China
| | - Siwei Zhang
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, 130021, Jilin, China
| | - Ziyi Zhang
- The Second Hospital of Jilin University, Changchun, 130041, China
| | - Yongfei Tan
- South China Institute of Collaborative Innovation, Dongguan, 523808, China
| | - Grant M Hatch
- Departments of Pharmacology and Therapeutics, Biochemistry and Medical Genetics, Center for Research and Treatment of Atherosclerosis, DREAM Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, R3E0T6, Canada
| | - Ming Zhang
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China.
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, 130021, Jilin, China.
| | - Li Chen
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China.
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, 130021, Jilin, China.
| |
Collapse
|
181
|
Fleisher-Berkovich S, Battaglia V, Baratta F, Brusa P, Ventura Y, Sharon N, Dahan A, Collino M, Ben-Shabat S. An Emerging Strategy for Neuroinflammation Treatment: Combined Cannabidiol and Angiotensin Receptor Blockers Treatments Effectively Inhibit Glial Nitric Oxide Release. Int J Mol Sci 2023; 24:16254. [PMID: 38003444 PMCID: PMC10671332 DOI: 10.3390/ijms242216254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Cannabidiol (CBD), the major non-psychoactive phytocannabinoid found in cannabis, has anti-neuroinflammatory properties. Despite the increasing use of CBD, little is known about its effect in combination with other substances. Combination therapy has been gaining attention recently, aiming to produce more efficient effects. Angiotensin II activates the angiotensin 1 receptor and regulates neuroinflammation and cognition. Angiotensin receptor 1 blockers (ARBs) were shown to be neuroprotective and prevent cognitive decline. The present study aimed to elucidate the combined role of CBD and ARBs in the modulation of lipopolysaccharide (LPS)-induced glial inflammation. While LPS significantly enhanced nitric oxide synthesis vs. the control, telmisartan and CBD, when administered alone, attenuated this effect by 60% and 36%, respectively. Exposure of LPS-stimulated cells to both compounds resulted in the 95% inhibition of glial nitric oxide release (additive effect). A synergistic inhibitory effect on nitric oxide release was observed when cells were co-treated with losartan (5 μM) and CBD (5 μM) (by 80%) compared to exposure to each compound alone (by 22% and 26%, respectively). Telmisartan and CBD given alone increased TNFα levels by 60% and 40%, respectively. CBD and telmisartan, when given together, attenuated the LPS-induced increase in TNFα levels without statistical significance. LPS-induced IL-17 release was attenuated by CBD with or without telmisartan (by 75%) or telmisartan alone (by 60%). LPS-induced Interferon-γ release was attenuated by 80% when telmisartan was administered in the absence or presence of CBD. Anti-inflammatory effects were recorded when CBD was combined with the known anti-inflammatory agent dimethyl fumarate (DMF)/monomethyl fumarate (MMF). A synergistic inhibitory effect of CBD and MMF on glial release of nitric oxide (by 77%) was observed compared to cells exposed to MMF (by 35%) or CBD (by 12%) alone. Overall, this study highlights the potential of new combinations of CBD (5 μM) with losartan (5 μM) or MMF (1 μM) to synergistically attenuate glial NO synthesis. Additive effects on NO production were observed when telmisartan (5 μM) and CBD (5 μM) were administered together to glial cells.
Collapse
Affiliation(s)
- Sigal Fleisher-Berkovich
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (Y.V.); (N.S.); (A.D.)
| | - Veronica Battaglia
- Department of Drug Science and Technology, University of Torino, 10124 Torino, Italy; (V.B.); (F.B.); (P.B.)
| | - Francesca Baratta
- Department of Drug Science and Technology, University of Torino, 10124 Torino, Italy; (V.B.); (F.B.); (P.B.)
| | - Paola Brusa
- Department of Drug Science and Technology, University of Torino, 10124 Torino, Italy; (V.B.); (F.B.); (P.B.)
| | - Yvonne Ventura
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (Y.V.); (N.S.); (A.D.)
| | - Nitzan Sharon
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (Y.V.); (N.S.); (A.D.)
| | - Arik Dahan
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (Y.V.); (N.S.); (A.D.)
| | - Massimo Collino
- Department of Neuroscience “Rita Levi Montalcini”, University of Torino, 10124 Torino, Italy;
| | - Shimon Ben-Shabat
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (Y.V.); (N.S.); (A.D.)
| |
Collapse
|
182
|
Li Y, Li YJ, Zhu ZQ. To re-examine the intersection of microglial activation and neuroinflammation in neurodegenerative diseases from the perspective of pyroptosis. Front Aging Neurosci 2023; 15:1284214. [PMID: 38020781 PMCID: PMC10665880 DOI: 10.3389/fnagi.2023.1284214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Neurodegenerative diseases (NDs), such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and motor neuron disease, are diseases characterized by neuronal damage and dysfunction. NDs are considered to be a multifactorial disease with diverse etiologies (immune, inflammatory, aging, genetic, etc.) and complex pathophysiological processes. Previous studies have found that neuroinflammation and typical microglial activation are important mechanisms of NDs, leading to neurological dysfunction and disease progression. Pyroptosis is a new mode involved in this process. As a form of programmed cell death, pyroptosis is characterized by the expansion of cells until the cell membrane bursts, resulting in the release of cell contents that activates a strong inflammatory response that promotes NDs by accelerating neuronal dysfunction and abnormal microglial activation. In this case, abnormally activated microglia release various pro-inflammatory factors, leading to the occurrence of neuroinflammation and exacerbating both microglial and neuronal pyroptosis, thus forming a vicious cycle. The recognition of the association between pyroptosis and microglia activation, as well as neuroinflammation, is of significant importance in understanding the pathogenesis of NDs and providing new targets and strategies for their prevention and treatment.
Collapse
Affiliation(s)
- Yuan Li
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- College of Anesthesiology, Zunyi Medical University, Zunyi, China
| | - Ying-Jie Li
- Department of General Surgery, Mianyang Hospital of Traditional Chinese Medicine, Mianyang, China
| | - Zhao-Qiong Zhu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
183
|
Dubowsky M, Theunissen F, Carr JM, Rogers ML. The Molecular Link Between TDP-43, Endogenous Retroviruses and Inflammatory Neurodegeneration in Amyotrophic Lateral Sclerosis: a Potential Target for Triumeq, an Antiretroviral Therapy. Mol Neurobiol 2023; 60:6330-6345. [PMID: 37450244 PMCID: PMC10533598 DOI: 10.1007/s12035-023-03472-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
Amyotrophic lateral sclerosis (ALS), also known as motor neuron disease (MND), is a progressive neurological disorder, characterised by the death of upper and lower motor neurons. The aetiology of ALS remains unknown, and treatment options are limited. Endogenous retroviruses (ERVs), specifically human endogenous retrovirus type K (HERV-K), have been proposed to be involved in the propagation of neurodegeneration in ALS. ERVs are genomic remnants of ancient viral infection events, with most being inactive and not retaining the capacity to encode a fully infectious virus. However, some ERVs retain the ability to be activated and transcribed, and ERV transcripts have been found to be elevated within the brain tissue of MND patients. A hallmark of ALS pathology is altered localisation of the transactive response (TAR) DNA binding protein 43 kDa (TDP-43), which is normally found within the nucleus of neuronal and glial cells and is involved in RNA regulation. In ALS, TDP-43 aggregates within the cytoplasm and facilitates neurodegeneration. The involvement of ERVs in ALS pathology is thought to occur through TDP-43 and neuroinflammatory mediators. In this review, the proposed involvement of TDP-43, HERV-K and immune regulators on the onset and progression of ALS will be discussed. Furthermore, the evidence supporting a therapy based on targeting ERVs in ALS will be reviewed.
Collapse
Affiliation(s)
- Megan Dubowsky
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia.
| | - Frances Theunissen
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, WA, Australia
| | - Jillian M Carr
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia
| | - Mary-Louise Rogers
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia
| |
Collapse
|
184
|
Fullerton JL, Cosgrove CC, Rooney RA, Work LM. Extracellular vesicles and their microRNA cargo in ischaemic stroke. J Physiol 2023; 601:4907-4921. [PMID: 35421904 PMCID: PMC10952288 DOI: 10.1113/jp282050] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/15/2022] [Indexed: 11/08/2022] Open
Abstract
Acute ischaemic stroke (AIS) is a leading cause of death and disability. MicroRNAs (miRNAs) are short non-coding RNAs which hold the potential to act as a novel biomarker in AIS. The majority of circulating miRNAs are actively encapsulated by extracellular vesicles (EVs) produced by many cells and organs endogenously. EVs released by mesenchymal stem cells (MSCs) have been extensively studied for their therapeutic potential. In health and disease, EVs are vital for intercellular communication, as the cargo within EVs can be exchanged between neighbouring cells or transported to distant sites. It is clear here from both current preclinical and clinical studies that AIS is associated with specific EV-derived miRNAs, including those transported via MSC-derived EVs. In addition, current studies provide evidence to show that modulating levels of specific EV-derived miRNAs in AIS provides a novel therapeutic potential of miRNAs in the treatment of stroke. Commonalities exist in altered miRNAs across preclinical and clinical studies. Of those EV-packaged miRNAs, miRNA-124 was described both as an EV-packaged biomarker and as a potential EV-loaded therapeutic in experimental models. Alterations of miRNA-17 family and miRNA-17-92 cluster were identified in preclinical, clinical and MSC-EV-mediated neuroprotection in experimental stroke. Finally, miRNA-30d and -30a were found to mediate therapeutic effect when overexpressed from MSC and implicated as a biomarker clinically. Combined, EV-derived miRNAs will further our understanding of the neuropathological processes triggered by AIS. In addition, this work will help determine the true clinical value of circulating EV-packaged miRNAs as biomarkers of AIS or as novel therapeutics in this setting.
Collapse
Affiliation(s)
- Josie L. Fullerton
- Institute of Cardiovascular and Medical Sciences College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Caitlin C. Cosgrove
- Institute of Cardiovascular and Medical Sciences College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Rebecca A. Rooney
- Institute of Cardiovascular and Medical Sciences College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Lorraine M. Work
- Institute of Cardiovascular and Medical Sciences College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| |
Collapse
|
185
|
Socodato R, Rodrigues-Santos A, Tedim-Moreira J, Almeida TO, Canedo T, Portugal CC, Relvas JB. RhoA balances microglial reactivity and survival during neuroinflammation. Cell Death Dis 2023; 14:690. [PMID: 37863874 PMCID: PMC10589285 DOI: 10.1038/s41419-023-06217-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 09/29/2023] [Accepted: 10/13/2023] [Indexed: 10/22/2023]
Abstract
Microglia are the largest myeloid cell population in the brain. During injury, disease, or inflammation, microglia adopt different functional states primarily involved in restoring brain homeostasis. However, sustained or exacerbated microglia inflammatory reactivity can lead to brain damage. Dynamic cytoskeleton reorganization correlates with alterations of microglial reactivity driven by external cues, and proteins controlling cytoskeletal reorganization, such as the Rho GTPase RhoA, are well positioned to refine or adjust the functional state of the microglia during injury, disease, or inflammation. Here, we use multi-biosensor-based live-cell imaging approaches and tissue-specific conditional gene ablation in mice to understand the role of RhoA in microglial response to inflammation. We found that a decrease in RhoA activity is an absolute requirement for microglial metabolic reprogramming and reactivity to inflammation. However, without RhoA, inflammation disrupts Ca2+ and pH homeostasis, dampening mitochondrial function, worsening microglial necrosis, and triggering microglial apoptosis. Our results suggest that a minimum level of RhoA activity is obligatory to concatenate microglia inflammatory reactivity and survival during neuroinflammation.
Collapse
Affiliation(s)
- Renato Socodato
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal.
| | - Artur Rodrigues-Santos
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - Joana Tedim-Moreira
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
- Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Tiago O Almeida
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
- ICBAS - School of Medicine and Biomedical Sciences, Porto, Portugal
| | - Teresa Canedo
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - Camila C Portugal
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - João B Relvas
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal.
- Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal.
| |
Collapse
|
186
|
Palumbo L, Carinci M, Guarino A, Asth L, Zucchini S, Missiroli S, Rimessi A, Pinton P, Giorgi C. The NLRP3 Inflammasome in Neurodegenerative Disorders: Insights from Epileptic Models. Biomedicines 2023; 11:2825. [PMID: 37893198 PMCID: PMC10604217 DOI: 10.3390/biomedicines11102825] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Neuroinflammation represents a dynamic process of defense and protection against the harmful action of infectious agents or other detrimental stimuli in the central nervous system (CNS). However, the uncontrolled regulation of this physiological process is strongly associated with serious dysfunctional neuronal issues linked to the progression of CNS disorders. Moreover, it has been widely demonstrated that neuroinflammation is linked to epilepsy, one of the most prevalent and serious brain disorders worldwide. Indeed, NLRP3, one of the most well-studied inflammasomes, is involved in the generation of epileptic seizures, events that characterize this pathological condition. In this context, several pieces of evidence have shown that the NLRP3 inflammasome plays a central role in the pathophysiology of mesial temporal lobe epilepsy (mTLE). Based on an extensive review of the literature on the role of NLRP3-dependent inflammation in epilepsy, in this review we discuss our current understanding of the connection between NLRP3 inflammasome activation and progressive neurodegeneration in epilepsy. The goal of the review is to cover as many of the various known epilepsy models as possible, providing a broad overview of the current literature. Lastly, we also propose some of the present therapeutic strategies targeting NLRP3, aiming to provide potential insights for future studies.
Collapse
Affiliation(s)
- Laura Palumbo
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.P.); (M.C.); (S.M.); (A.R.); (P.P.)
| | - Marianna Carinci
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.P.); (M.C.); (S.M.); (A.R.); (P.P.)
| | - Annunziata Guarino
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy; (A.G.); (L.A.); (S.Z.)
| | - Laila Asth
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy; (A.G.); (L.A.); (S.Z.)
| | - Silvia Zucchini
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy; (A.G.); (L.A.); (S.Z.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy
| | - Sonia Missiroli
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.P.); (M.C.); (S.M.); (A.R.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy
| | - Alessandro Rimessi
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.P.); (M.C.); (S.M.); (A.R.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy
- Center of Research for Innovative Therapies in Cystic Fibrosis, University of Ferrara, 44121 Ferrara, Italy
| | - Paolo Pinton
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.P.); (M.C.); (S.M.); (A.R.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy
- Center of Research for Innovative Therapies in Cystic Fibrosis, University of Ferrara, 44121 Ferrara, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.P.); (M.C.); (S.M.); (A.R.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
187
|
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
|
188
|
Ming Y, Luo C, Ji B, Cheng J. ARPC5 acts as a potential prognostic biomarker that is associated with cell proliferation, migration and immune infiltrate in gliomas. BMC Cancer 2023; 23:937. [PMID: 37789267 PMCID: PMC10548738 DOI: 10.1186/s12885-023-11433-w] [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: 03/27/2023] [Accepted: 09/21/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND Gliomas are the most common malignant brain tumors, with powerful invasiveness and an undesirable prognosis. Actin related protein 2/3 complex subunit 5 (ARPC5) encodes a component of the Arp2/3 protein complex, which plays a significant role in regulating the actin cytoskeleton. However, the prognostic values and biological functions of ARPC5 in gliomas remain unclear. METHODS Based on the TCGA, GEO, HPA, and UALCAN database, we determined the expression of ARPC5 in glioma. The results were verified by immunohistochemistry and Western blot analysis of glioma samples. Moreover, Kaplan-Meier curves, ROC curves, Cox regression analyses, and prognostic nomograms were used to observe the correlation between the ARPC5 expression and the prognosis of glioma patients. GO and KEGG enrichment analyses were conducted to identify immune-related pathways involved with the differential expression of ARPC5. Subsequently, the TCGA database was used to estimate the relationship between ARPC5 expression and immunity-related indexes, such as immune scores, infiltrating immune cells, and TMB. The TCIA database was used to assess the correlation between ARPC5 with immunotherapy. The association between ARPC5 and T cells marker CD3 was also evaluated through immunohistochemistry methods. The correlation between ARPC5 and T cell, as well as the prognosis of patients, was also evaluated using immunological methods. Moreover, the effect of ARPC5 on the biological characteristics of LN229 and U251 cells was determined by MTT, clone formation, and transwell migration assay. RESULTS The high degree of ARPC5 was correlated with worse prognosis and unfavorable clinical characteristics of glioma patients. In the analysis of GO and KEGG, it is shown that ARPC5 was strongly correlated with multiple immune-related signaling pathways. The single-cell analysis revealed that ARPC5 expression was increased in astrocytes, monocytes and T cells. In addition, ARPC5 expression was strongly associated with immune scores, infiltrating immune cells, TMB, MSI, immune biomarkers, and immunotherapy. In experimental analysis, we found that ARPC5 was significantly overexpressed in gliomas and closely correlated with patient prognosis and CD3 expression. Functionally, the knockout of ARPC5 significantly reduced the proliferation and invasion of LN229 and U251 cells. CONCLUSIONS Our study revealed that the high expression level of ARPC5 may serve as a promising prognostic biomarker and be associated with tumor immunity in glioma.
Collapse
Affiliation(s)
- Yue Ming
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Networks, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chunyuan Luo
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Networks, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Beihong Ji
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pennsylvania, USA
| | - Jian Cheng
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
189
|
Zheng J, Zhang C, Wu Y, Zhang C, Che Y, Zhang W, Yang Y, Zhu J, Yang L, Wang Y. Controlled Decompression Alleviates Motor Dysfunction by Regulating Microglial Polarization via the HIF-1α Signaling Pathway in Intracranial Hypertension. Mol Neurobiol 2023; 60:5607-5623. [PMID: 37328678 DOI: 10.1007/s12035-023-03416-6] [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: 01/25/2023] [Accepted: 05/24/2023] [Indexed: 06/18/2023]
Abstract
Decompressive craniectomy (DC) is a major form of surgery that is used to reduce intracranial hypertension (IH), the most frequent cause of death and disability following severe traumatic brain injury (sTBI) and stroke. Our previous research showed that controlled decompression (CDC) was more effective than rapid decompression (RDC) with regard to reducing the incidence of complications and improving outcomes after sTBI; however, the specific mechanisms involved have yet to be elucidated. In the present study, we investigated the effects of CDC in regulating inflammation after IH and attempted to identify the mechanisms involved. Analysis showed that CDC was more effective than RDC in alleviating motor dysfunction and neuronal death in a rat model of traumatic intracranial hypertension (TIH) created by epidural balloon pressurization. Moreover, RDC induced M1 microglia polarization and the release of pro-inflammatory cytokines. However, CDC treatment resulted in microglia primarily polarizing into the M2 phenotype and induced the significant release of anti-inflammatory cytokines. Mechanistically, the establishment of the TIH model led to the increased expression of hypoxia-inducible factor-1α (HIF-1α); CDC ameliorated cerebral hypoxia and reduced the expression of HIF-1α. In addition, 2-methoxyestradiol (2-ME2), a specific inhibitor of HIF-1α, significantly attenuated RDC-induced inflammation and improved motor function by promoting M1 to M2 phenotype transformation in microglial and enhancing the release of anti-inflammatory cytokines. However, dimethyloxaloylglycine (DMOG), an agonist of HIF-1α, abrogated the protective effects of CDC treatment by suppressing M2 microglia polarization and the release of anti-inflammatory cytokines. Collectively, our results indicated that CDC effectively alleviated IH-induced inflammation, neuronal death, and motor dysfunction by regulating HIF-1α-mediated microglial phenotype polarization. Our findings provide a better understanding of the mechanisms that underlie the protective effects of CDC and promote clinical translational research for HIF-1α in IH.
Collapse
Affiliation(s)
- Jie Zheng
- Department of Neurosurgery, The 904th Hospital of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, 214044, Jiangsu, China
| | - Chenxu Zhang
- Department of Neurosurgery, The 904th Hospital of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, 214044, Jiangsu, China
| | - Yonghui Wu
- Department of Neurosurgery, The 904th Hospital of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, 214044, Jiangsu, China
| | - Chonghui Zhang
- Department of Neurosurgery, The 904th Hospital of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, 214044, Jiangsu, China
| | - Yuanyuan Che
- Department of Neurosurgery, The 904th Hospital of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, 214044, Jiangsu, China
| | - Wang Zhang
- Department of Neurosurgery, The 904th Hospital of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, 214044, Jiangsu, China
| | - Yang Yang
- Department of Neurosurgery, The 904th Hospital of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, 214044, Jiangsu, China
| | - Jie Zhu
- Department of Neurosurgery, The 904th Hospital of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, 214044, Jiangsu, China.
| | - Likun Yang
- Department of Neurosurgery, The 904th Hospital of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, 214044, Jiangsu, China.
| | - Yuhai Wang
- Department of Neurosurgery, The 904th Hospital of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, 214044, Jiangsu, China.
| |
Collapse
|
190
|
Hossein Geranmayeh M, Farokhi-Sisakht F, Sadigh-Eteghad S, Rahbarghazi R, Mahmoudi J, Farhoudi M. Simultaneous Pericytes and M2 Microglia Transplantation Improve Cognitive Function in Mice Model of mPFC Ischemia. Neuroscience 2023; 529:62-72. [PMID: 37591334 DOI: 10.1016/j.neuroscience.2023.08.010] [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: 05/14/2023] [Revised: 07/27/2023] [Accepted: 08/04/2023] [Indexed: 08/19/2023]
Abstract
Cerebral ischemia is one of the major problems threatening global health. Many of the cerebral ischemia survivors would suffer from the physical and cognitive disabilities for their whole lifetime. Cell based-therapies have been introduced as a therapeutic approach for alleviating ischemia-enforced limitations. Photothrombotic stroke model was applied on the left medial prefrontal cortex (mPFC) of adult male BALB/c mice. Then, pericytes isolated from brain microvessels of adult male BALB/c mice, microglia isolated from brain cortices of the neonatal male BALB/c mice, and M2 phenotype shifted microglia by IL-4 treatment were used for transplantation into the injured area after 24 h of ischemia induction. The behavioural outcomes evaluated by social interaction and Barnes tests and the levels of growth associated protein (GAP)-43 and inflammatory cytokine interleukin (IL)-1 protein were assessed by western blotting 7 days after cell transplantation. Animals in both of the microglia + pericytes and microglia M2 + pericytes transplanted groups showed better performance in social memory as well as enhanced spatial learning and memory compared to ischemic controls. Also, improved escape latency was only observed in microglia M2 + pericytes (p < 0.01) group compared to ischemic controls. GAP-43 showed significant protein expression in microglia + pericytes and microglia M2 + pericytes groups compared to the control group. Conversely, IL-1 levels diminished in all of the pericytes microglia + pericytes, and microglia M2 + pericytes groups compared to the ischemic controls. Current study highlights efficiency of M2 microglia and pericytes combinatory transplantation therapeutic role on relieving ischemic stroke outcomes.
Collapse
Affiliation(s)
- Mohammad Hossein Geranmayeh
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cells Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Saeed Sadigh-Eteghad
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Farhoudi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
191
|
Lindblad C, Rostami E, Helmy A. Interleukin-1 Receptor Antagonist as Therapy for Traumatic Brain Injury. Neurotherapeutics 2023; 20:1508-1528. [PMID: 37610701 PMCID: PMC10684479 DOI: 10.1007/s13311-023-01421-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2023] [Indexed: 08/24/2023] Open
Abstract
Traumatic brain injury is a common type of acquired brain injury of varying severity carrying potentially deleterious consequences for the afflicted individuals, families, and society. Following the initial, traumatically induced insult, cellular injury processes ensue. These are believed to be amenable to treatment. Among such injuries, neuroinflammation has gained interest and has become a specific focus for both experimental and clinical researchers. Neuroinflammation is elicited almost immediately following trauma, and extend for a long time, possibly for years, after the primary injury. In the acute phase, the inflammatory response is characterized by innate mechanisms such as the activation of microglia which among else mediates cytokine production. Among the earliest cytokines to emerge are the interleukin- (IL-) 1 family members, comprising, for example, the agonist IL-1β and its competitive antagonist, IL-1 receptor antagonist (IL-1ra). Because of its early emergence following trauma and its increased concentrations also after human TBI, IL-1 has been hypothesized to be a tractable treatment target following TBI. Ample experimental data supports this, and demonstrates restored neurological behavior, diminished lesion zones, and an attenuated inflammatory response following IL-1 modulation either through IL-1 knock-out experiments, IL-1β inhibition, or IL-1ra treatment. Of these, IL-1ra treatment is likely the most physiological. In addition, recombinant human IL-1ra (anakinra) is already approved for utilization across a few rheumatologic disorders. As of today, one randomized clinical controlled trial has utilized IL-1ra inhibition as an intervention and demonstrated its safety. Further clinical trials powered for patient outcome are needed in order to demonstrate efficacy. In this review, we summarize IL-1 biology in relation to acute neuroinflammatory processes following TBI with a particular focus on current evidence for IL-1ra treatment both in the experimental and clinical context.
Collapse
Affiliation(s)
- Caroline Lindblad
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
- Department of Neurosurgery, Uppsala University Hospital, entrance 85 floor 2, Akademiska Sjukhuset, 751 85, Uppsala, Sweden.
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Elham Rostami
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Department of Neurosurgery, Uppsala University Hospital, entrance 85 floor 2, Akademiska Sjukhuset, 751 85, Uppsala, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Adel Helmy
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
192
|
Radpour M, Khoshkroodian B, Asgari T, Pourbadie HG, Sayyah M. Interleukin 4 Reduces Brain Hyperexcitability after Traumatic Injury by Downregulating TNF-α, Upregulating IL-10/TGF-β, and Potential Directing Macrophage/Microglia to the M2 Anti-inflammatory Phenotype. Inflammation 2023; 46:1810-1831. [PMID: 37259014 DOI: 10.1007/s10753-023-01843-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/14/2023] [Accepted: 05/16/2023] [Indexed: 06/02/2023]
Abstract
Macrophage/microglia are activated after Traumatic brain injury (TBI), transform to inflammatory phenotype (M1) and trigger neuroinflammation, which provokes epileptogenesis. Interleukin-4 (IL-4) is a well-known drive of macrophage/microglia to the anti-inflammatory phenotype (M2). We tested effect of IL-4 on speed of epileptogenesis, brain expression of inflammatory and anti-inflammatory cytokines, and lesion size in TBI-injured male rats. Rats underwent TBI by Controlled Cortical Impact. Then 100 ng IL-4 was injected into cerebral ventricles. One day after TBI, pentylenetetrazole (PTZ) kindling started and development of generalized seizures was recorded. The lesion size, cell survival rate, TNF-α, TGF-β, IL-10, and Arginase1 (Arg1) was measured in the brain 6 h, 12 h, 24 h, 48 h, and 5 days after TBI. Astrocytes and macrophage/microglia activation/polarization was assessed by GFAP/Arg1 and Iba1/Arg1 immunostaining. TBI-injured rats were kindled by 50% less PTZ injections than control and sham-operated rats. IL-4 did not change kindling rate in sham-operated rats but inhibited acceleration of kindling rate in the TBI-injured rats. IL-4 decreased damage volume and number of destroyed neurons. IL-4 stopped TNF-α whereas upregulated TGF-β, IL-10, and Arg1 expressions. Iba1/Arg1 positive macrophage/microglia was notably increased 48 h after IL-4 administration. IL-4 suppresses TBI-induced acceleration of epileptogenesis in rats by directing TBI neuroinflammation toward an anti-inflammatory tone and inhibition of cell death.
Collapse
Affiliation(s)
- Mozhdeh Radpour
- Department of Physiology and Pharmacology, Pasteur Institute of Iran , Tehran, Iran
| | - Bahar Khoshkroodian
- Department of Physiology and Pharmacology, Pasteur Institute of Iran , Tehran, Iran
| | - Tara Asgari
- Department of Physiology and Pharmacology, Pasteur Institute of Iran , Tehran, Iran
| | | | - Mohammad Sayyah
- Department of Physiology and Pharmacology, Pasteur Institute of Iran , Tehran, Iran.
| |
Collapse
|
193
|
Fan PL, Wang SS, Chu SF, Chen NH. Time-dependent dual effect of microglia in ischemic stroke. Neurochem Int 2023; 169:105584. [PMID: 37454817 DOI: 10.1016/j.neuint.2023.105584] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Stroke, the third leading cause of death and disability worldwide, is classified into ischemic or hemorrhagic, in which approximately 85% of strokes are ischemic. Ischemic stroke occurs as a result of arterial occlusion due to embolus or thrombus, with ischemia in the perfusion territory supplied by the occluded artery. The traditional concept that ischemic stroke is solely a vascular occlusion disorder has been expanded to include the dynamic interaction between microglia, astrocytes, neurons, vascular cells, and matrix components forming the "neurovascular unit." Acute ischemic stroke triggers a wide spectrum of neurovascular disturbances, glial activation, and secondary neuroinflammation that promotes further injury, ultimately resulting in neuronal death. Microglia, as the resident macrophages in the central nervous system, is one of the first responders to ischemic injury and plays a significant role in post-ischemic neuroinflammation. In this review, we reviewed the mechanisms of microglia in multiple stages of post-ischemic neuroinflammation development, including acute, sub-acute and chronic phases of stroke. A comprehensive understanding of the dynamic variation and the time-dependent role of microglia in post-stroke neuroinflammation could aid in the search for more effective therapeutics and diagnostic strategies for ischemic stroke.
Collapse
Affiliation(s)
- Ping-Long Fan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Sha-Sha Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Shi-Feng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Nai-Hong Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
194
|
Miao J, Chen L, Pan X, Li L, Zhao B, Lan J. Microglial Metabolic Reprogramming: Emerging Insights and Therapeutic Strategies in Neurodegenerative Diseases. Cell Mol Neurobiol 2023; 43:3191-3210. [PMID: 37341833 PMCID: PMC11410021 DOI: 10.1007/s10571-023-01376-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/14/2023] [Indexed: 06/22/2023]
Abstract
Microglia, the resident immune cells of the central nervous system, play a critical role in maintaining brain homeostasis. However, in neurodegenerative conditions, microglial cells undergo metabolic reprogramming in response to pathological stimuli, including Aβ plaques, Tau tangles, and α-synuclein aggregates. This metabolic shift is characterized by a transition from oxidative phosphorylation (OXPHOS) to glycolysis, increased glucose uptake, enhanced production of lactate, lipids, and succinate, and upregulation of glycolytic enzymes. These metabolic adaptations result in altered microglial functions, such as amplified inflammatory responses and diminished phagocytic capacity, which exacerbate neurodegeneration. This review highlights recent advances in understanding the molecular mechanisms underlying microglial metabolic reprogramming in neurodegenerative diseases and discusses potential therapeutic strategies targeting microglial metabolism to mitigate neuroinflammation and promote brain health. Microglial Metabolic Reprogramming in Neurodegenerative Diseases This graphical abstract illustrates the metabolic shift in microglial cells in response to pathological stimuli and highlights potential therapeutic strategies targeting microglial metabolism for improved brain health.
Collapse
Affiliation(s)
- Jifei Miao
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, China
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Lihua Chen
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Xiaojin Pan
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Liqing Li
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Beibei Zhao
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, China.
| | - Jiao Lan
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, China.
| |
Collapse
|
195
|
Zhang M, Hao Z, Wu J, Teng Z, Qiu W, Cheng J. Curcumin ameliorates traumatic brain injury via C1ql3-mediated microglia M2 polarization. Tissue Cell 2023; 84:102164. [PMID: 37478644 DOI: 10.1016/j.tice.2023.102164] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/17/2023] [Accepted: 07/11/2023] [Indexed: 07/23/2023]
Abstract
PURPOSE Curcumin can regulate the polarization of microglia and alleviate traumatic brain injury (TBI). However, its detailed action mechanism on downregulating Complement 1q-like-3 protein (C1ql3) in TBI is less reported. The purpose of this study is to explore the role and mechanism of curcumin-regulated C1ql3 in TBI. METHOD GSE23639 dataset was used to acquire gene data for microglia. C57BL/6 J wild-type (WT) mice were subjected to establish a controlled cortical impact model of TBI. The effects of curcumin (200 mg/kg) on the brain injury, inflammatory cytokine levels, microglia polarization, and C1ql3 protein expression in mice and BV-2 cells were detected by H&E staining, qRT-PCR, immunofluorescence, and Western blot, respectively. The effects of curcumin (5, 10, 20 μmol/L) and lipopolysaccharides (LPS, 1 µg/mL) on the viability of BV-2 cells were determined by MTT assay. After the transfection of C1ql3 overexpression plasmid, C1ql3 expression, IL-1β and IL-6 levels, and the number of CD16+/32+ and CD206+ cells were determined by qRT-PCR, ELISA and flow cytometry, respectively. RESULT C1ql3 expression was down-regulated in microglia after the curcumin treatment. Curcumin treatment could alleviate the TBI-induced brain injury in mice, reduce IL-1β and IL-6 levels, promote M2 polarization of microglia, and decrease C1ql3 protein expression. For BV-2 cells, curcumin treatment had no significant toxic effect on cell viability, but reversed the effect of LPS on cells, while C1ql3 overexpression counteracted the effect of curcumin. CONCLUSION Curcumin induces M2 microglia polarization through down-regulating C1ql3 expression, which may become a new treatment method for TBI. AVAILABILITY OF DATA AND MATERIALS The analyzed data sets generated during the study are available from the corresponding author on reasonable request.
Collapse
Affiliation(s)
- Mei Zhang
- Department of Neurosurgery, The Affiliated Hospital of Hangzhou Normal University, Gongshu, Hangzhou City, Zhejiang 310015, PR China
| | - Zelin Hao
- Department of Neurosurgery, The Affiliated Hospital of Hangzhou Normal University, Gongshu, Hangzhou City, Zhejiang 310015, PR China
| | - Jianyue Wu
- Department of Neurosurgery, The Affiliated Hospital of Hangzhou Normal University, Gongshu, Hangzhou City, Zhejiang 310015, PR China
| | - Zhenfei Teng
- Department of Neurosurgery, The Affiliated Hospital of Hangzhou Normal University, Gongshu, Hangzhou City, Zhejiang 310015, PR China
| | - Wusi Qiu
- Department of Neurosurgery, The Affiliated Hospital of Hangzhou Normal University, Gongshu, Hangzhou City, Zhejiang 310015, PR China
| | - Jun Cheng
- Department of Neurosurgery, The Affiliated Hospital of Hangzhou Normal University, Gongshu, Hangzhou City, Zhejiang 310015, PR China.
| |
Collapse
|
196
|
Chi B, Öztürk MM, Paraggio CL, Leonard CE, Sanita ME, Dastpak M, O’Connell JD, Coady JA, Zhang J, Gygi SP, Lopez-Gonzalez R, Yin S, Reed R. Causal ALS genes impact the MHC class II antigen presentation pathway. Proc Natl Acad Sci U S A 2023; 120:e2305756120. [PMID: 37722062 PMCID: PMC10523463 DOI: 10.1073/pnas.2305756120] [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: 04/10/2023] [Accepted: 08/18/2023] [Indexed: 09/20/2023] Open
Abstract
Mutations in RNA/DNA-binding proteins cause amyotrophic lateral sclerosis (ALS), but the underlying disease mechanisms remain unclear. Here, we report that a set of ALS-associated proteins, namely FUS, EWSR1, TAF15, and MATR3, impact the expression of genes encoding the major histocompatibility complex II (MHC II) antigen presentation pathway. Both subunits of the MHC II heterodimer, HLA-DR, are down-regulated in ALS gene knockouts/knockdown in HeLa and human microglial cells, due to loss of the MHC II transcription factor CIITA. Importantly, hematopoietic progenitor cells (HPCs) derived from human embryonic stem cells bearing the FUSR495X mutation and HPCs derived from C9ORF72 ALS patient induced pluripotent stem cells also exhibit disrupted MHC II expression. Given that HPCs give rise to numerous immune cells, our data raise the possibility that loss of the MHC II pathway results in global failure of the immune system to protect motor neurons from damage that leads to ALS.
Collapse
Affiliation(s)
- Binkai Chi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Muhammet M. Öztürk
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Christina L. Paraggio
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Claudia E. Leonard
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Maria E. Sanita
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Mahtab Dastpak
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Jeremy D. O’Connell
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Jordan A. Coady
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Jiuchun Zhang
- Harvard Medical School Cell Biology Initiative for Genome Editing and Neurodegeneration, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Steven P. Gygi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Rodrigo Lopez-Gonzalez
- Department of Neurosciences Lerner Research Institute, Cleveland Clinic, Cleveland, OH44196
| | - Shanye Yin
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY10461
| | - Robin Reed
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| |
Collapse
|
197
|
Tonev D, Momchilova A. Therapeutic Plasma Exchange and Multiple Sclerosis Dysregulations: Focus on the Removal of Pathogenic Circulatory Factors and Altering Nerve Growth Factor and Sphingosine-1-Phosphate Plasma Levels. Curr Issues Mol Biol 2023; 45:7749-7774. [PMID: 37886933 PMCID: PMC10605592 DOI: 10.3390/cimb45100489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/21/2023] [Accepted: 09/23/2023] [Indexed: 10/28/2023] Open
Abstract
Multiple sclerosis (MS) is predominantly an immune-mediated disease of the central nervous system (CNS) of unknown etiology with a possible genetic predisposition and effect of certain environmental factors. It is generally accepted that the disease begins with an autoimmune inflammatory reaction targeting oligodendrocytes followed by a rapid depletion of their regenerative capacity with subsequent permanent neurodegenerative changes and disability. Recent research highlights the central role of B lymphocytes and the corresponding IgG and IgM autoantibodies in newly forming MS lesions. Thus, their removal along with the modulation of certain bioactive molecules to improve neuroprotection using therapeutic plasma exchange (TPE) becomes of utmost importance. Recently, it has been proposed to determine the levels and precise effects of both beneficial and harmful components in the serum of MS patients undergoing TPE to serve as markers for appropriate TPE protocols. In this review we discuss some relevant examples, focusing on the removal of pathogenic circulating factors and altering the plasma levels of nerve growth factor and sphingosine-1-phosphate by TPE. Altered plasma levels of the reviewed molecular compounds in response to TPE reflect a successful reduction of the pro-inflammatory burden at the expense of an increase in anti-inflammatory potential in the circulatory and CNS compartments.
Collapse
Affiliation(s)
- Dimitar Tonev
- Department of Anesthesiology and Intensive Care, University Hospital “Tzaritza Yoanna—ISUL”, Medical University of Sofia, 1527 Sofia, Bulgaria
| | - Albena Momchilova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Science, 1113 Sofia, Bulgaria;
| |
Collapse
|
198
|
Zhang Y, Park YS, Kim IB. A Distinct Microglial Cell Population Expressing Both CD86 and CD206 Constitutes a Dominant Type and Executes Phagocytosis in Two Mouse Models of Retinal Degeneration. Int J Mol Sci 2023; 24:14236. [PMID: 37762541 PMCID: PMC10532260 DOI: 10.3390/ijms241814236] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/10/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Microglial cells are the key regulators of inflammation during retinal degeneration (RD) and are conventionally classified as M1 or M2. However, whether the M1/M2 classification exactly reflects the functional classification of microglial cells in the retina remains debatable. We examined the spatiotemporal changes of microglial cells in the blue-LED and NaIO3-induced RD mice models using M1/M2 markers and functional genes. TUNEL assay was performed to detect photoreceptor cell death, and microglial cells were labeled with anti-IBA1, P2RY12, CD86, and CD206 antibodies. FACS was used to isolate microglial cells with anti-CD206 and CD86 antibodies, and qRT-PCR was performed to evaluate Il-10, Il-6, Trem-2, Apoe, and Lyz2 expression. TUNEL-positive cells were detected in the outer nuclear layer (ONL) from 24 h to 72 h post-RD induction. At 24 h, P2RY12 was decreased and CD86 was increased, and CD86/CD206 double-labeled cells occupied the dominant population at 72 h. And CD86/CD206 double-labeled cells showed a significant increase in Apoe, Trem2, and Lyz2 levels but not in those of Il-6 and Il-10. Our results demonstrate that microglial cells in active RD cannot be classified as M1 or M2, and the majority of microglia express both CD86 and CD206, which are involved in phagocytosis rather than inflammation.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (Y.Z.); (Y.S.P.)
- Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Yong Soo Park
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (Y.Z.); (Y.S.P.)
- Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - In-Beom Kim
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (Y.Z.); (Y.S.P.)
- Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Catholic Institute for Applied Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
199
|
Kim HY, Ashim J, Park S, Kim W, Ji S, Lee SW, Jung YR, Jeong SW, Lee SG, Kim HC, Lee YJ, Kwon MK, Hwang JS, Shin JM, Lee SJ, Yu W, Park JK, Choi SK. A preliminary study about the potential risks of the UV-weathered microplastic: The proteome-level changes in the brain in response to polystyrene derived weathered microplastics. ENVIRONMENTAL RESEARCH 2023; 233:116411. [PMID: 37354929 DOI: 10.1016/j.envres.2023.116411] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/08/2023] [Accepted: 06/11/2023] [Indexed: 06/26/2023]
Abstract
The growing use of plastic materials has resulted in a constant increase in the risk associated with microplastics (MPs). Ultra-violet (UV) light and wind break down modify MPs in the environment into smaller particles known as weathered MPs (WMPs) and these processes increase the risk of MP toxicity. The neurotoxicity of weathered polystyrene-MPs remains unclear. Therefore, it is important to understand the risks posed by WMPs. We evaluated the chemical changes of WMPs generated under laboratory-synchronized environmentally mimetic conditions and compared them with virgin MPs (VMPs). We found that WMP had a rough surface, slight yellow color, reduced molecular weight, and structural alteration compared with those of VMP. Next, 2 μg of ∼100 μm in size of WMP and VMP were orally administered once a day for one week to C57BL/6 male mice. Proteomic analysis revealed that the WMP group had significantly increased activation of immune and neurodegeneration-related pathways compared with that of the VMP group. Consistently, in in vitro experiments, the human brain-derived microglial cell line (HMC-3) also exhibited a more severe inflammatory response to WMP than to VMP. These results show that WMP is a more profound inflammatory factor than VMP. In summary, our findings demonstrate the toxicity of WMPs and provide theoretical insights into their potential risks to biological systems and even humans in the ecosystem.
Collapse
Affiliation(s)
- Hee-Yeon Kim
- Core Protein Resources Center, DGIST, Daegu, Republic of Korea; College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Janbolat Ashim
- Department of Brain Sciences, DGIST, Daegu, Republic of Korea
| | - Song Park
- Core Protein Resources Center, DGIST, Daegu, Republic of Korea; Department of Brain Sciences, DGIST, Daegu, Republic of Korea
| | - Wansoo Kim
- School of Life Science, BK21 FOUR KNU Creative Bioresearch Group, Kyungpook National University, Daegu, Republic of Korea; Division of Biotechnology, DGIST, Daegu, Republic of Korea
| | - Sangho Ji
- Department of Brain Sciences, DGIST, Daegu, Republic of Korea
| | - Seoung-Woo Lee
- Core Protein Resources Center, DGIST, Daegu, Republic of Korea; Division of Biotechnology, DGIST, Daegu, Republic of Korea
| | - Yi-Rang Jung
- Department of Companion Animal Health Management, Daegu Health College, Daegu, Republic of Korea
| | - Sang Won Jeong
- Division of Biotechnology, DGIST, Daegu, Republic of Korea
| | - Se-Guen Lee
- Division of Biotechnology, DGIST, Daegu, Republic of Korea
| | - Hyun-Chul Kim
- Division of Biotechnology, DGIST, Daegu, Republic of Korea
| | - Young-Jae Lee
- Division of Biotechnology, DGIST, Daegu, Republic of Korea
| | - Mi Kyung Kwon
- Division of Biotechnology, DGIST, Daegu, Republic of Korea
| | | | - Jung Min Shin
- Division of Biotechnology, DGIST, Daegu, Republic of Korea
| | - Sung-Jun Lee
- Division of Biotechnology, DGIST, Daegu, Republic of Korea.
| | - Wookyung Yu
- Core Protein Resources Center, DGIST, Daegu, Republic of Korea; Department of Brain Sciences, DGIST, Daegu, Republic of Korea.
| | - Jin-Kyu Park
- College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea.
| | - Seong-Kyoon Choi
- Core Protein Resources Center, DGIST, Daegu, Republic of Korea; Division of Biotechnology, DGIST, Daegu, Republic of Korea.
| |
Collapse
|
200
|
Yang Y, Chen R, Che Y, Yao X, Fang M, Wang Y, Zhou D, Li N, Hou Y. Isoamericanin A improves lipopolysaccharide-induced memory impairment in mice through suppression of the nicotinamide adenine dinucleotide phosphateoxidase-dependent nuclear factor kappa B signaling pathway. Phytother Res 2023; 37:3982-4001. [PMID: 37209001 DOI: 10.1002/ptr.7858] [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: 10/15/2022] [Revised: 04/08/2023] [Accepted: 04/15/2023] [Indexed: 05/21/2023]
Abstract
Alzheimer's disease (AD) is the most frequent cause of dementia in the elderly. Isoamericanin A (ISOA) is a natural lignan possessing great potential for AD treatment. This study investigated the efficacy of ISOA on memory impairments in the mice intrahippocampal injected with lipopolysaccharide (LPS) and the underlying mechanism. Y-maze and Morris Water Maze data suggested that ISOA (5 and 10 mg/kg) ameliorated short- and long-term memory impairments, and attenuated neuronal loss and lactate dehydrogenase activity. ISOA exerted anti-inflammatory effect demonstrating by the reduction of ionized calcium-binding adapter molecule 1 positive cells and suppression of marker protein and pro-inflammation cytokines expressions induced by LPS. ISOA suppressed the nuclear factor kappa B (NF-κB) signaling pathway by inhibiting IκBα phosphorylation and NF-κB p65 phosphorylation and nuclear translocation. ISOA inhibited superoxide and intracellular reactive oxygen species accumulation by reducing nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activation, demonstrating by suppressing NADP+ and NADPH contents, gp91phox expression, and p47phox expression and membrane translocation. These effects were enhanced in combination with NADPH oxidase inhibitor apocynin. The neuroprotective effect of ISOA was further proved in the in vitro models. Overall, our data revealed a novel pharmacological activity of ISOA: ameliorating memory impairment in AD via inhibiting neuroinflammation.
Collapse
Affiliation(s)
- Yanqiu Yang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Ru Chen
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Yue Che
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Xiaohu Yao
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Mingxia Fang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Yingjie Wang
- School of Traditional Chinese Materia Medica, Key Laboratory of Innovative Traditional Chinese Medicine for Major Chronic Diseases of Liaoning province, Key Laboratory for TCM Material Basis Study and Innovative Drug Development of Shenyang City, Shenyang Pharmaceutical University, Shenyang, China
| | - Di Zhou
- School of Traditional Chinese Materia Medica, Key Laboratory of Innovative Traditional Chinese Medicine for Major Chronic Diseases of Liaoning province, Key Laboratory for TCM Material Basis Study and Innovative Drug Development of Shenyang City, Shenyang Pharmaceutical University, Shenyang, China
| | - Ning Li
- School of Traditional Chinese Materia Medica, Key Laboratory of Innovative Traditional Chinese Medicine for Major Chronic Diseases of Liaoning province, Key Laboratory for TCM Material Basis Study and Innovative Drug Development of Shenyang City, Shenyang Pharmaceutical University, Shenyang, China
| | - Yue Hou
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| |
Collapse
|