51
|
Gupta DP, Park SH, Lee YS, Lee S, Lim S, Byun J, Cho IH, Song GJ. Daphne genkwa flower extract promotes the neuroprotective effects of microglia. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154486. [PMID: 36240609 DOI: 10.1016/j.phymed.2022.154486] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Microglia are innate immune cells in the central nervous system that play a crucial role in neuroprotection by releasing neurotrophic factors, removing pathogens through phagocytosis, and regulating brain homeostasis. The constituents extracted from the roots and stems of the Daphne genkwa plant have shown neuroprotective effects in an animal model of Parkinson's disease. However, the effect of Daphne genkwa plant extract on microglia has yet to be demonstrated. PURPOSE To study the anti-inflammatory and neuroprotective effects of Daphne genkwa flower extract (GFE) in microglia and explore the underlying mechanisms. METHODS In-vitro mRNA expression levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), inducible nitric oxide synthase, Arginase1, and brain derived neurotropic factor (BDNF) were analyzed by reverse transcription polymerase chain reaction in microglia cells. Nitric oxide (NO) and TNF-α protein were respectively analyzed by Griess reagent and Enzyme Linked Immunosorbent Assay. Immunoreactivity of Iba-1, Neu-N, and BDNF in mouse brain were analyzed by immunofluorescence staining. Phagocytosis capacity of microglia was examined using fluorescent zymosan-red particles. RESULTS GFE significantly inhibited lipopolysaccharide (LPS)-induced neuroinflammation and promoted neuroprotection both in vitro and in vivo. First, GFE inhibited the LPS-induced inflammatory factors NO, iNOS, and TNF-α in microglial cell lines and primary glial cells, thus demonstrating anti-inflammatory effects. Arginase1 and BDNF mRNA levels were increased in primary glial cells treated with GFE. Phagocytosis was also increased in microglia treated with GFE, suggesting a neuroprotective effect of GFE. In vivo, neuroprotective and anti-neuroinflammatory effects of GFE were also found in the mouse brain, as oral administration of GFE significantly inhibited LPS-induced neuronal loss and inflammatory activation of microglia. CONCLUSION GFE has anti-inflammatory effects and promotes microglial neuroprotective effects. GFE inhibited the pro-inflammatory mediators and enhanced neuroprotective microglia activity by increasing BDNF expression and phagocytosis. These novel findings of the GFE effect on microglia show an innovative approach that can potentially promote neuroprotection for the prevention of neurodegenerative diseases.
Collapse
Affiliation(s)
- Deepak Prasad Gupta
- Department of Medical Science, Catholic Kwandong University College of Medicine, Gangneung, Gangwon-do, Korea
| | - Sung Hee Park
- Department of Medical Science, Catholic Kwandong University College of Medicine, Gangneung, Gangwon-do, Korea
| | - Young-Sun Lee
- Department of Medical Science, Catholic Kwandong University College of Medicine, Gangneung, Gangwon-do, Korea; The Convergence Institute of Healthcare and Medical Science, Catholic Kwandong University, International St. Mary's Hospital, Incheon, Korea
| | - Sanghyun Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong, Korea
| | - Sujin Lim
- Department of Life Science, The Catholic University of Korea, Bucheon, Korea
| | - Jiin Byun
- Department of Life Science, The Catholic University of Korea, Bucheon, Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Korea.
| | - Gyun Jee Song
- Department of Medical Science, Catholic Kwandong University College of Medicine, Gangneung, Gangwon-do, Korea; The Convergence Institute of Healthcare and Medical Science, Catholic Kwandong University, International St. Mary's Hospital, Incheon, Korea.
| |
Collapse
|
52
|
Wang X, Liu X, Chen L, Zhang X. The inflammatory injury in the striatal microglia-dopaminergic-neuron crosstalk involved in Tourette syndrome development. Front Immunol 2023; 14:1178113. [PMID: 37187752 PMCID: PMC10175669 DOI: 10.3389/fimmu.2023.1178113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Background Tourette syndrome (TS) is associated with immunological dysfunction. The DA system is closely related to TS development, or behavioral stereotypes. Previous evidence suggested that hyper-M1-polarized microglia may exist in the brains of TS individuals. However, the role of microglia in TS and their interaction with dopaminergic neurons is unclear. In this study, we applied iminodipropionitrile (IDPN) to establish a TS model and focused on the inflammatory injury in the striatal microglia-dopaminergic-neuron crosstalk. Methods Male Sprague-Dawley rats were intraperitoneally injected with IDPN for seven consecutive days. Stereotypic behavior was observed to verify the TS model. Striatal microglia activation was evaluated based on different markers and expressions of inflammatory factors. The striatal dopaminergic neurons were purified and co-cultured with different microglia groups, and dopamine-associated markers were assessed. Results First, there was pathological damage to striatal dopaminergic neurons in TS rats, as indicated by decreased expression of TH, DAT, and PITX3. Next, the TS group showed a trend of increased Iba-1 positive cells and elevated levels of inflammatory factors TNF-α and IL-6, as well as an enhanced M1-polarization marker (iNOS) and an attenuated M2-polarization marker (Arg-1). Finally, in the co-culture experiment, IL-4-treated microglia could upregulate the expression of TH, DAT, and PITX3 in striatal dopaminergic neurons vs LPS-treated microglia. Similarly, the TS group (microglia from TS rats) caused a decreased expression of TH, DAT, and PITX3 compared with the Sham group (microglia from control rats) in the dopaminergic neurons. Conclusion In the striatum of TS rats, microglia activation is M1 hyperpolarized, which transmits inflammatory injury to striatal dopaminergic neurons and disrupts normal dopamine signaling.
Collapse
Affiliation(s)
- Xueming Wang
- Plastic Surgery Department, Fujian Children’s Hospital, Fuzhou, China
- Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiumei Liu
- Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Developmental and Behavior Pediatrics Department, Fujian Children’s Hospital, Fuzhou, China
- *Correspondence: Xiumei Liu,
| | - Liangliang Chen
- Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Developmental and Behavior Pediatrics Department, Fujian Children’s Hospital, Fuzhou, China
| | - Xiaoling Zhang
- Child Healthcare Department, Fuzhou Maternal and Child Health Hospital, Fuzhou, China
| |
Collapse
|
53
|
Xiao L, Wang M, Shi Y, Xu Y, Gao Y, Zhang W, Wu Y, Deng H, Pan W, Wang W, Sun H. Secondary White Matter Injury Mediated by Neuroinflammation after Intracerebral Hemorrhage and Promising Therapeutic Strategies of Targeting the NLRP3 Inflammasome. Curr Neuropharmacol 2023; 21:669-686. [PMID: 36043798 PMCID: PMC10207923 DOI: 10.2174/1570159x20666220830115018] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 11/22/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a neurological disease with high mortality and disability. Recent studies showed that white matter injury (WMI) plays an important role in motor dysfunction after ICH. WMI includes WMI proximal to the lesion and WMI distal to the lesion, such as corticospinal tract injury located at the cervical enlargement of the spinal cord after ICH. Previous studies have tended to focus only on gray matter (GM) injury after ICH, and fewer studies have paid attention to WMI, which may be one of the reasons for the poor outcome of previous drug treatments. Microglia and astrocyte-mediated neuroinflammation are significant mechanisms responsible for secondary WMI following ICH. The NOD-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome activation, has been shown to exacerbate neuroinflammation and brain injury after ICH. Moreover, NLRP3 inflammasome is activated in microglia and astrocytes and exerts a vital role in microglia and astrocytes-mediated neuroinflammation. We speculate that NLRP3 inflammasome activation is closely related to the polarization of microglia and astrocytes and that NLRP3 inflammasome activation may exacerbate WMI by polarizing microglia and astrocytes to the pro-inflammatory phenotype after ICH, while NLRP3 inflammasome inhibition may attenuate WMI by polarizing microglia and astrocytes to the anti-inflammatory phenotype following ICH. Therefore, NLRP3 inflammasome may act as leveraged regulatory fulcrums for microglia and astrocytes polarization to modulate WMI and WM repair after ICH. This review summarized the possible mechanisms by which neuroinflammation mediated by NLRP3 inflammasome exacerbates secondary WMI after ICH and discussed the potential therapeutic targets.
Collapse
Affiliation(s)
- Linglong Xiao
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Mengqi Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Yifeng Shi
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Yangyang Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Yuan Gao
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Wei Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Yang Wu
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Hao Deng
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Wei Pan
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Wei Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Haitao Sun
- Department of Laboratory Medicine, Clinical Biobank Center, Microbiome Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| |
Collapse
|
54
|
Liu X, Tang M, He TY, Zhao S, Li HZ, Li Z, Guo YX, Wang XL. Resveratrol Improves Paclitaxel-Induced Cognitive Impairment in Mice by Activating SIRT1/PGC-1α Pathway to Regulate Neuronal State and Microglia Cell Polarization. Drug Des Devel Ther 2023; 17:1125-1138. [PMID: 37077409 PMCID: PMC10106825 DOI: 10.2147/dddt.s400936] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/29/2023] [Indexed: 04/21/2023] Open
Abstract
Objective This study aimed to investigate the effect of resveratrol (Res) on paclitaxel (PTX)-induced cognitive impairment and elucidate the underlying molecular mechanisms. Methods Morris Water Maze (MWM) test was used to evaluate the mice's spatial learning and memory abilities. Western blotting was applied to detect protein expression of receptor-interacting protein (RIP3), mixed lineage kinase domain-like protein (MLKL), silencing information regulator 2 related enzyme 1 (SIRT1), peroxisome proliferator activated receptor coactivator-1 (PGC-1α), NADPH oxidase 2 (NOX2), NOX4, postsynaptic density zone 95 (PSD95), arginase-1 (Arg-1) and inducible nitric oxide synthase (iNOS). Immunofluorescence of RIP3, MLKL, Arg-1, Iba-1 and iNOS was conducted to observe the apoptosis of hippocampal cells and the polarization of microglia. qRT-PCR was performed to detect BDNF mRNA expressions. DHE staining was used to assess the level of oxidative stress response. Golgi-Cox staining and dendritic spine counting were applied to visualize synaptic structural plasticity. Postsynaptic density was performed by transmission electron microscope. ELISA was used to detect the contents of tumour necrosis factor alpha (TNF-α), IL-1β, IL-4, and IL-10. Results PTX-induced cognitive impairment model was constructed after the application of PTX, represented as longer latency to platform and less platform crossing times over the whole period in PTX group. After Res treatment, the above indicators were reversed, indicating that cognitive function was improved. Moreover, Res reduced neuronal apoptosis and oxidative stress through SIRT1/PGC-1α pathway in mice, manifesting as down-regulated expression of RIP3, MLKL, NOX2 and NOX4. Meanwhile, Res increased the density of dendritic spines and the expression of PSD95 and BDNF, thereby ameliorating the PTX induced synaptic damage. Besides, M2 microglia was in the majority, eliciting the expression of anti-inflammatory cytokines IL-4 and IL-10 after Res treatment in PTX+Res group, while immunofluorescence images results demonstrated an decrease in the proportion of M2 microglia a following SIRT1 inhibitor EX-527. Conclusion Res improves PTX-induced cognitive impairment in mice by activating SIRT1/PGC-1α pathways to regulate neuronal state and microglia cell polarization.
Collapse
Affiliation(s)
- Xin Liu
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People’s Republic of China
| | - Miao Tang
- Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People’s Republic of China
| | - Tian-Yi He
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People’s Republic of China
| | - Shuang Zhao
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People’s Republic of China
| | - Hui-Zhou Li
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People’s Republic of China
| | - Zhao Li
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People’s Republic of China
| | - Yue-Xian Guo
- Department of Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, People’s Republic of China
| | - Xiu-Li Wang
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People’s Republic of China
- Correspondence: Xiu-Li Wang, Department of Anesthesiology, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, Hebei, 050051, People’s Republic of China, Tel +86-13313019968, Email
| |
Collapse
|
55
|
Role of Zerumbone, a Phytochemical Sesquiterpenoid from Zingiber zerumbet Smith, in Maintaining Macrophage Polarization and Redox Homeostasis. Nutrients 2022; 14:nu14245402. [PMID: 36558562 PMCID: PMC9783216 DOI: 10.3390/nu14245402] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Macrophages and microglia are highly versatile cells that can be polarized into M1 and M2 phenotypes in response to diverse environmental stimuli, thus exhibiting different biological functions. In the central nervous system, activated resident macrophages and microglial cells trigger the production of proinflammatory mediators that contribute to neurodegenerative diseases and psychiatric disorders. Therefore, modulating the activation of macrophages and microglia by optimizing the inflammatory environment is beneficial for disease management. Several naturally occurring compounds have been reported to have anti-inflammatory and neuroprotective properties. Zerumbone is a phytochemical sesquiterpenoid and also a cyclic ketone isolated from Zingiber zerumbet Smith. In this study, we found that zerumbone effectively reduced the expression of lipocalin-2 in macrophages and microglial cell lines. Lipocalin-2, also known as neutrophil gelatinase-associated lipocalin (NGAL), has been characterized as an adipokine/cytokine implicated in inflammation. Moreover, supplement with zerumbone inhibited reactive oxygen species production. Phagocytic activity was decreased following the zerumbone supplement. In addition, the zerumbone supplement remarkably reduced the production of M1-polarization-associated chemokines CXC10 and CCL-2, as well as M1-polarization-associated cytokines interleukin (IL)-6, IL-1β, and tumor necrosis factor-α. Furthermore, the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 and the production of NO were attenuated in macrophages and microglial cells supplemented with zerumbone. Notably, we discovered that zerumbone effectively promoted the production of the endogenous antioxidants heme oxygenase-1, glutamate-cysteine ligase modifier subunit, glutamate-cysteine ligase catalytic subunit, and NAD(P)H quinone oxidoreductase-1 and remarkably enhanced IL-10, a marker of M2 macrophage polarization. Endogenous antioxidant production and M2 macrophage polarization were increased through activation of the AMPK/Akt and Akt/GSK3 signaling pathways. In summary, this study demonstrated the protective role of zerumbone in maintaining M1 and M2 polarization homeostasis by decreasing inflammatory responses and enhancing the production of endogenous antioxidants in both macrophages and microglia cells. This study suggests that zerumbone can be used as a potential therapeutic drug for the supplement of neuroinflammatory diseases.
Collapse
|
56
|
Saikarthik J, Saraswathi I, Alarifi A, Al-Atram AA, Mickeymaray S, Paramasivam A, Shaikh S, Jeraud M, Alothaim AS. Role of neuroinflammation mediated potential alterations in adult neurogenesis as a factor for neuropsychiatric symptoms in Post-Acute COVID-19 syndrome-A narrative review. PeerJ 2022; 10:e14227. [PMID: 36353605 PMCID: PMC9639419 DOI: 10.7717/peerj.14227] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/22/2022] [Indexed: 11/06/2022] Open
Abstract
Persistence of symptoms beyond the initial 3 to 4 weeks after infection is defined as post-acute COVID-19 syndrome (PACS). A wide range of neuropsychiatric symptoms like anxiety, depression, post-traumatic stress disorder, sleep disorders and cognitive disturbances have been observed in PACS. The review was conducted based on PRISMA-S guidelines for literature search strategy for systematic reviews. A cytokine storm in COVID-19 may cause a breach in the blood brain barrier leading to cytokine and SARS-CoV-2 entry into the brain. This triggers an immune response in the brain by activating microglia, astrocytes, and other immune cells leading to neuroinflammation. Various inflammatory biomarkers like inflammatory cytokines, chemokines, acute phase proteins and adhesion molecules have been implicated in psychiatric disorders and play a major role in the precipitation of neuropsychiatric symptoms. Impaired adult neurogenesis has been linked with a variety of disorders like depression, anxiety, cognitive decline, and dementia. Persistence of neuroinflammation was observed in COVID-19 survivors 3 months after recovery. Chronic neuroinflammation alters adult neurogenesis with pro-inflammatory cytokines supressing anti-inflammatory cytokines and chemokines favouring adult neurogenesis. Based on the prevalence of neuropsychiatric symptoms/disorders in PACS, there is more possibility for a potential impairment in adult neurogenesis in COVID-19 survivors. This narrative review aims to discuss the various neuroinflammatory processes during PACS and its effect on adult neurogenesis.
Collapse
Affiliation(s)
- Jayakumar Saikarthik
- Department of Basic Medical Sciences, College of Dentistry, Al Zulfi, Majmaah University, Al-Majmaah, Riyadh, Kingdom of Saudi Arabia,Department of Medical Education, College of Dentistry, Al Zulfi, Majmaah University, Al Majmaah, Riyadh, Kingdom of Saudi Arabia
| | - Ilango Saraswathi
- Department of Physiology, Madha Medical College and Research Institute, Chennai, Tamil Nadu, India
| | - Abdulaziz Alarifi
- Department of Basic Sciences, College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia,King Abdullah International Medical Research Centre, Riyadh, Saudi Arabia
| | - Abdulrahman A. Al-Atram
- Department of Psychiatry, College of Medicine, Majmaah University, Al Majmaah, Riyadh, Kingdom of Saudi Arabia
| | - Suresh Mickeymaray
- Department of Biology, College of Science, Al Zulfi, Majmaah University, Al Majmaah, Riyadh, Kingdom of Saudi Arabia
| | - Anand Paramasivam
- Department of Physiology, RVS Dental College and Hospital, Kumaran Kottam Campus, Kannampalayan, Coimbatore, Tamilnadu, India
| | - Saleem Shaikh
- Department of Medical Education, College of Dentistry, Al Zulfi, Majmaah University, Al Majmaah, Riyadh, Kingdom of Saudi Arabia,Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Al Zulfi, Majmaah University, Al Majmaah, Riyadh, Kingdom of Saudi Arabia
| | - Mathew Jeraud
- Department of Physiology, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia
| | - Abdulaziz S. Alothaim
- Department of Biology, College of Science, Al Zulfi, Majmaah University, Al Majmaah, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
57
|
Choi HR, Ha JS, Kim EA, Cho SW, Yang SJ. MiR-30a-5p and miR-153-3p regulate LPS-induced neuroinflammatory response and neuronal apoptosis by targeting NeuroD1. BMB Rep 2022; 55:447-452. [PMID: 35651331 PMCID: PMC9537026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/11/2022] [Accepted: 04/19/2022] [Indexed: 03/08/2024] Open
Abstract
Neurogenic differentiation 1 (NeuroD1) is an essential transcription factor for neuronal differentiation, maturation, and survival, and is associated with inflammation in lipopolysaccharide (LPS)- induced glial cells; however, the concrete mechanisms are still ambiguous. Therefore, we investigated whether NeuroD1-targeting miRNAs affect inflammation and neuronal apoptosis, as well as the underlying mechanism. First, we confirmed that miR-30a-5p and miR-153-3p, which target NeuroD1, reduced NeuroD1 expression in microglia and astrocytes. In LPS-induced microglia, miR-30a-5p and miR-153-3p suppressed pro-inflammatory cytokines, reactive oxygen species, the phosphorylation of c-Jun N-terminal kinase, extracellular-signal-regulated kinase (ERK), and p38, and the expression of cyclooxygenase and inducible nitric oxide synthase (iNOS) via the NF-κB pathway. Moreover, miR-30a-5p and miR-153-3p inhibited the expression of NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasomes, NLRP3, cleaved caspase-1, and IL-1β, which are involved in the innate immune response. In LPS-induced astrocytes, miR-30a-5p and miR-153-3p reduced ERK phosphorylation and iNOS expression via the STAT-3 pathway. Notably, miR-30a-5p exerted greater anti-inflammatory effects than miR-153-3p. Together, these results indicate that miR-30a-5p and miR-153-3p inhibit MAPK/NF-κB pathway in microglia as well as ERK/STAT-3 pathway in astrocytes to reduce LPS-induced neuronal apoptosis. This study highlights the importance of NeuroD1 in microglia and astrocytes neuroinflammation and suggests that it can be regulated by miR-30a-5p and miR-153-3p. [BMB Reports 2022; 55(9): 447-452].
Collapse
Affiliation(s)
- Hye-Rim Choi
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea
| | - Ji Sun Ha
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea
| | - Eun-A Kim
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Seung-Ju Yang
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea
| |
Collapse
|
58
|
Cao LX, Lin SJ, Zhao SS, Wang SQ, Zeng H, Chen WA, Lin ZW, Chen JX, Zhu MM, Zhang YM. Effects of acupuncture on microglial polarization and the TLR4/TRIF/MyD88 pathway in a rat model of traumatic brain injury. Acupunct Med 2022:9645284221108214. [PMID: 36046956 DOI: 10.1177/09645284221108214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Neuroinflammation caused by traumatic brain injury (TBI) can lead to neurological deficits. Acupuncture can inhibit neuroinflammation and promote nerve repair; however, the specific mechanism is still unclear. The purpose of this study was to explore whether acupuncture could modulate the M1 and M2 phenotypic polarization of microglia in a rat model of TBI via the toll-like receptor 4 (TLR4)/intracellular toll-interleukin-1 receptor (TIR) domain-containing adaptor inducing interferon-β (TRIF)/myeloid differentiation factor 88 (MyD88) pathway. METHODS A total of 90 adult male Sprague-Dawley (SD) rats, SPF grade, were randomly divided into a normal group, model group and acupuncture group. Each group was further divided into three subgroups (first, third, and fifth day groups) according to the treatment time (n = 10 rats/subgroup). We used the modified neurological severity score (mNSS) method to quantify neurological deficits before and after modeling. We used Nissl staining to observe the pathological changes in brain tissue, flow cytometry to detect the proportion of M1 and M2 polarized microglia in the injured area on the first, third and fifth day, and co-immunoprecipitation (Co-IP) to examine TLR4/TRIF/MyD88 expression in microglia on the first, third and fifth day, as well as expression of the amount of binding of TLR4 with TRIF and MyD88. RESULTS Compared to the model group, mNSS in the acupuncture group gradually decreased and pathological morphology improved. The proportion of CD11b/CD86 positive cells was decreased, while that of CD11b/CD206 was increased in the acupuncture group. Expression of IP TLR4, IP TRIF and IP MyD88 also decreased in the acupuncture group. CONCLUSION The results of this study demonstrate that one of the mechanisms through which acupuncture mitigates neuroinflammation and promotes nerve repair in TBI rats may be inhibition of M1 phenotypic polarization and promotion of M2 phenotypic polarization through inhibition of the TLR4/TRIF/MyD88 signaling pathway.
Collapse
Affiliation(s)
- Lu-Xi Cao
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China.,Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Shu-Jun Lin
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China.,Clinical Medical College of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Si-Si Zhao
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Shi-Qi Wang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Hai Zeng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Wen-An Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Zhuo-Wen Lin
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Jia-Xu Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China.,Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Ming-Min Zhu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Yi-Min Zhang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
59
|
Usende IL, Olopade JO, Azeez IA, Andrioli A, Bankole MO, Olopade FE, Nafady AA, Bentivoglio M. Neuroecotoxicology: Effects of environmental heavy metal exposure on the brain of African giant rats and the contribution of vanadium to the neuropathology. IBRO Neurosci Rep 2022; 13:215-234. [PMID: 36590095 PMCID: PMC9795313 DOI: 10.1016/j.ibneur.2022.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/26/2022] [Indexed: 01/04/2023] Open
Abstract
Increased exploitation of minerals has led to pollution of confined environments as documented in Nigeria Niger Delta. Information on the effects on brain of such exposure is limited. Due to its exploratory activities, the African giant rat (Cricetomys gambianus) (AGR) provides a unique model for neuroecotoxicological research to determine levels of animal and human exposure to different pollutants. This study aims to unravel neuropathological features of AGR sampled from three agro-ecological zones of Nigeria. Fifteen AGR were sampled according to previously determined data on heavy metal exposure: high vanadium, high lead, and low metals. Eighteen AGR were collected from low metal zone and divided into two groups. Control group received vehicle while SMV exposed group received 3 mg/kg sodium metavanadate (SMV) intraperitoneally for 14days. Brain immunohistochemical analyses were conducted, and ultrastructural changes were studied in experimentally exposed group. Results showed significant loss of tyrosin hydroxylase, parvalbumin, orexin-A and melanin concentration hormone containing neuronal populations in brains obtained from high vanadium and high lead zones and in experimentally intoxicated SMV groups. Similarly, significant decrease numbers of dendritic arborations; extracellular matrix density, perineuronal nets; astrocytes and microglia activations are documented in same groups. Ultrastructural studies revealed mass denudation, cilia loss, disintegration of ependymal layer and intense destructions of myelin sheaths in SMV exposed group. These are the first "neuroecotoxicological" findings in distinct neuronal cells. The implications of these findings are highly relevant for human population living in these areas, not only in Nigeria but also in similarly polluted areas elsewhere in the world.
Collapse
Affiliation(s)
- Ifukibot Levi Usende
- Department of Veterinary Anatomy, University of Abuja, Nigeria,Department of Veterinary Anatomy, University of Ibadan, Nigeria,Corresponding author at: Department of Veterinary Anatomy, University of Abuja, Nigeria.
| | | | | | - Anna Andrioli
- Department of Neurological and Movement Sciences, University of Verona, Italy
| | - Molakun O. Bankole
- Department of Neurological and Movement Sciences, University of Verona, Italy
| | | | - Allam A. Nafady
- Department of Veterinary Pathology, Electron Microscope Unit, Assuit University, Egypt
| | - Marina Bentivoglio
- Department of Neurological and Movement Sciences, University of Verona, Italy
| |
Collapse
|
60
|
Hong Y, Lyu J, Zhu L, Wang X, Peng M, Chen X, Deng Q, Gao J, Yuan Z, Wang D, Xu G, Xu M. High-frequency repetitive transcranial magnetic stimulation (rTMS) protects against ischemic stroke by inhibiting M1 microglia polarization through let-7b-5p/HMGA2/NF-κB signaling pathway. BMC Neurosci 2022; 23:49. [PMID: 35927640 PMCID: PMC9351069 DOI: 10.1186/s12868-022-00735-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 07/28/2022] [Indexed: 11/22/2022] Open
Abstract
Background Microglia assume opposite phenotypes in response to ischemic brain injury, exerting neurotoxic and neuroprotective effects under different ischemic stages. Modulating M1/M2 polarization is a potential therapy for treating ischemic stroke. Repetitive transcranial magnetic stimulation (rTMS) held the capacity to regulate neuroinflammation and astrocytic polarization, but little is known about rTMS effects on microglia. Therefore, the present study aimed to examine the rTMS influence on microglia polarization and the underlying possible molecular mechanisms in ischemic stroke models. Methods Previously reported 10 Hz rTMS protocol that regulated astrocytic polarization was used to stimulate transient middle cerebral artery occlusion (MCAO) rats and oxygen and glucose deprivation/reoxygenation (OGD/R) injured BV2 cells. Specific expression levels of M1 marker iNOS and M2 marker CD206 were measured by western blotting and immunofluorescence. MicroRNA expression changes detected by high-throughput second-generation sequencing were validated by RT-PCR and fluorescence in situ hybridization (FISH) analysis. Dual-luciferase report assay and miRNA knock-down were applied to verify the possible mechanisms regulated by rTMS. Microglia culture medium (MCM) from different groups were collected to measure the TNF-α and IL-10 concentrations, and detect the influence on neuronal survival. Finally, TTC staining and modified Neurological Severity Score (mNSS) were used to determine the effects of MCM on ischemic stroke volume and neurological functions. Results The 10 Hz rTMS inhibited ischemia/reperfusion induced M1 microglia and significantly increased let-7b-5p level in microglia. HMGA2 was predicted and proved to be the target protein of let-7b-5p. HMGA2 and its downstream NF-κB signaling pathway were inhibited by rTMS. Microglia culture medium (MCM) collected from rTMS treated microglia contained lower TNF-α concentration but higher IL-10 concentration than no rTMS treated MCM, reducing ischemic volumes and neurological deficits of MCAO mice. However, knockdown of let-7b-5p by antagomir reversed rTMS effects on microglia phenotype and associated HMGA/NF-κB activation and neurological recovery. Conclusion High-frequency rTMS could alleviate ischemic stroke injury through inhibiting M1 microglia polarization via regulating let-7b-5p/HMGA2/NF-κB signaling pathway in MCAO models. Supplementary Information The online version contains supplementary material available at 10.1186/s12868-022-00735-7.
Collapse
Affiliation(s)
- Ye Hong
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, 68# Changle Road, Nanjing, 210029, Jiangsu, China
| | - Jinfeng Lyu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, 68# Changle Road, Nanjing, 210029, Jiangsu, China
| | - Lin Zhu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, 68# Changle Road, Nanjing, 210029, Jiangsu, China
| | - Xixi Wang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, 68# Changle Road, Nanjing, 210029, Jiangsu, China
| | - Mengna Peng
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Xiangliang Chen
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, 68# Changle Road, Nanjing, 210029, Jiangsu, China
| | - Qiwen Deng
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, 68# Changle Road, Nanjing, 210029, Jiangsu, China
| | - Jie Gao
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, 68# Changle Road, Nanjing, 210029, Jiangsu, China
| | - Zhenhua Yuan
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, 68# Changle Road, Nanjing, 210029, Jiangsu, China
| | - Di Wang
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Gelin Xu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Mengyi Xu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, 68# Changle Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
61
|
Zuo C, Cao H, Feng F, Li G, Huang Y, Zhu L, Gu Z, Yang Y, Chen J, Jiang Y, Wang F. Repetitive transcranial magnetic stimulation exerts anti-inflammatory effects via modulating glial activation in mice with chronic unpredictable mild stress-induced depression. Int Immunopharmacol 2022; 109:108788. [DOI: 10.1016/j.intimp.2022.108788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 04/06/2022] [Accepted: 04/17/2022] [Indexed: 12/21/2022]
|
62
|
Gu Y, Zhao P, Feng W, Xia X, Tian X, Yan Y, Wang X, Gao D, Du Y, Li X. Structural brain network measures in elderly patients with cerebral small vessel disease and depressive symptoms. BMC Geriatr 2022; 22:568. [PMID: 35810313 PMCID: PMC9270825 DOI: 10.1186/s12877-022-03245-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 06/27/2022] [Indexed: 12/20/2022] Open
Abstract
Objectives To investigate the relationship between diffusion tensor imaging (DTI) indicators and cerebral small vessel disease (CSVD) with depressive states, and to explore the underlying mechanisms of white matter damage in CSVD with depression. Method A total of 115 elderly subjects were consecutively recruited from the neurology clinic, including 36 CSVD patients with depressive state (CSVD+D), 34 CSVD patients without depressive state (CSVD-D), and 45 controls. A detailed neuropsychological assessment and multimodal magnetic resonance imaging (MRI) were performed. Based on tract-based spatial statistics (TBSS) analysis and structural network analysis, differences between groups were compared, including white matter fiber indicators (fractional anisotropy and mean diffusivity) and structural brain network indicators (global efficiency, local efficiency and network strength), in order to explore the differences and correlations of DTI parameters among the three groups. Results There were no significant differences in terms of CSVD burden scores and conventional imaging findings between the CSVD-D and CSVD+D groups. Group differences were found in DTI indicators (p < 0.05), after adjusting for age, gender, education level, and vascular risk factors (VRF), there were significant correlations between TBSS analysis indicators and depression, including: fractional anisotropy (FA) (r = − 0.291, p < 0.05), mean diffusivity (MD) (r = 0.297, p < 0.05), at the same time, between structural network indicators and depression also show significant correlations, including: local efficiency (ELocal) (r = − 0.278, p < 0.01) and network strength (r = − 0.403, p < 0.001). Conclusions Changes in FA, MD values and structural network indicators in DTI parameters can predict the depressive state of CSVD to a certain extent, providing a more direct structural basis for the hypothesis of abnormal neural circuits in the pathogenesis of vascular-related depression. In addition, abnormal white matter alterations in subcortical neural circuits probably affect the microstructural function of brain connections, which may be a mechanism for the concomitant depressive symptoms in CSVD patients.
Collapse
Affiliation(s)
- Yumeng Gu
- Department of Neurology, Second Hospital of Tianjin Medical University, No.23, Pingjiang Road, Hexi District, Tianjin, 300211, China
| | - Ping Zhao
- Department of Neurology, Second Hospital of Tianjin Medical University, No.23, Pingjiang Road, Hexi District, Tianjin, 300211, China
| | - Wenjun Feng
- Department of Neurology, Second Hospital of Tianjin Medical University, No.23, Pingjiang Road, Hexi District, Tianjin, 300211, China
| | - Xiaoshuang Xia
- Department of Neurology, Second Hospital of Tianjin Medical University, No.23, Pingjiang Road, Hexi District, Tianjin, 300211, China
| | - Xiaolin Tian
- Department of Rehabilitation, Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Yu Yan
- Department of Neurology, Second Hospital of Tianjin Medical University, No.23, Pingjiang Road, Hexi District, Tianjin, 300211, China
| | - Xiaowen Wang
- Department of Neurology, Second Hospital of Tianjin Medical University, No.23, Pingjiang Road, Hexi District, Tianjin, 300211, China
| | - Decheng Gao
- Department of Neurology, Second Hospital of Tianjin Medical University, No.23, Pingjiang Road, Hexi District, Tianjin, 300211, China
| | - Yanfen Du
- Department of Neurology, Second Hospital of Tianjin Medical University, No.23, Pingjiang Road, Hexi District, Tianjin, 300211, China
| | - Xin Li
- Department of Neurology, Second Hospital of Tianjin Medical University, No.23, Pingjiang Road, Hexi District, Tianjin, 300211, China.
| |
Collapse
|
63
|
Targeting Microglia in Alzheimer’s Disease: From Molecular Mechanisms to Potential Therapeutic Targets for Small Molecules. Molecules 2022; 27:molecules27134124. [PMID: 35807370 PMCID: PMC9268715 DOI: 10.3390/molecules27134124] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 02/01/2023] Open
Abstract
Alzheimer’s disease (AD) is a common, progressive, and devastating neurodegenerative disorder that mainly affects the elderly. Microglial dysregulation, amyloid-beta (Aβ) plaques, and intracellular neurofibrillary tangles play crucial roles in the pathogenesis of AD. In the brain, microglia play roles as immune cells to provide protection against virus injuries and diseases. They have significant contributions in the development of the brain, cognition, homeostasis of the brain, and plasticity. Multiple studies have confirmed that uncontrolled microglial function can result in impaired microglial mitophagy, induced Aβ accumulation and tau pathology, and a chronic neuroinflammatory environment. In the brain, most of the genes that are associated with AD risk are highly expressed by microglia. Although it was initially regarded that microglia reaction is incidental and induced by dystrophic neurites and Aβ plaques. Nonetheless, it has been reported by genome-wide association studies that most of the risk loci for AD are located in genes that are occasionally uniquely and highly expressed in microglia. This finding further suggests that microglia play significant roles in early AD stages and they be targeted for the development of novel therapeutics. In this review, we have summarized the molecular pathogenesis of AD, microglial activities in the adult brain, the role of microglia in the aging brain, and the role of microglia in AD. We have also particularly focused on the significance of targeting microglia for the treatment of AD.
Collapse
|
64
|
Modulating Microglia/Macrophage Activation by CDNF Promotes Transplantation of Fetal Ventral Mesencephalic Graft Survival and Function in a Hemiparkinsonian Rat Model. Biomedicines 2022; 10:biomedicines10061446. [PMID: 35740467 PMCID: PMC9221078 DOI: 10.3390/biomedicines10061446] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/08/2022] [Accepted: 06/11/2022] [Indexed: 11/30/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by the loss of dopaminergic neurons in substantia nigra pars compacta, which leads to the motor control deficits. Recently, cell transplantation is a cutting-edge technique for the therapy of PD. Nevertheless, one key bottleneck to realizing such potential is allogenic immune reaction of tissue grafts by recipients. Cerebral dopamine neurotrophic factor (CDNF) was shown to possess immune-modulatory properties that benefit neurodegenerative diseases. We hypothesized that co-administration of CDNF with fetal ventral mesencephalic (VM) tissue can improve the success of VM replacement therapies by attenuating immune responses. Hemiparkinsonian rats were generated by injecting 6-hydroxydopamine (6-OHDA) into the right medial forebrain bundle of Sprague Dawley (SD) rats. The rats were then intrastriatally transplanted with VM tissue from rats, with/without CDNF administration. Recovery of dopaminergic function and survival of the grafts were evaluated using the apomorphine-induced rotation test and small-animal positron emission tomography (PET) coupled with [18F] DOPA or [18F] FE-PE2I, respectively. In addition, transplantation-related inflammatory response was determined by uptake of [18F] FEPPA in the grafted side of striatum. Immunohistochemistry (IHC) examination was used to determine the survival of the grated dopaminergic neurons in the striatum and to investigate immune-modulatory effects of CDNF. The modulation of inflammatory responses caused by CDNF might involve enhancing M2 subset polarization and increasing fractal dimensions of 6-OHDA-treated BV2 microglial cell line. Analysis of CDNF-induced changes to gene expressions of 6-OHDA-stimulated BV2 cells implies that these alternations of the biomarkers and microglial morphology are implicated in the upregulation of protein kinase B signaling as well as regulation of catalytic, transferase, and protein serine/threonine kinase activity. The effects of CDNF on 6-OHDA-induced alternation of the canonical pathway in BV2 microglial cells is highly associated with PI3K-mediated phagosome formation. Our results are the first to show that CDNF administration enhances the survival of the grafted dopaminergic neurons and improves functional recovery in PD animal model. Modulation of the polarization, morphological characteristics, and transcriptional profiles of 6-OHDA-stimualted microglia by CDNF may possess these properties in transplantation-based regenerative therapies.
Collapse
|
65
|
The central nervous system control of energy homeostasis: high fat diet induced hypothalamic microinflammation and obesity. Brain Res Bull 2022; 185:99-106. [PMID: 35525336 DOI: 10.1016/j.brainresbull.2022.04.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/27/2022] [Accepted: 04/30/2022] [Indexed: 11/22/2022]
Abstract
Obesity is believed to arise through the imbalance of energy homeostasis controlled by the central nervous system, where the hypothalamus plays the fundamental role in energy metabolism. In this review, we will provide an overview regarding the functions of POMC neurons and AgRP neurons in acute nucleus of the hypothalamus which mediated the energy metabolism, highlighting their interactions with peripheral organs derived hormones in control of energy homeostasis. Furthermore, the role of high fat diet induced hypothalamic microinflammation in the pathogenesis of obesity will be discussed. We hope this review could help researchers to understand the mechanism of hypothalamus in control of energy metabolism, and design related drugs to block the pathways involving in the impaired metabolism in obese patients.
Collapse
|
66
|
Slater PG, Domínguez-Romero ME, Villarreal M, Eisner V, Larraín J. Mitochondrial function in spinal cord injury and regeneration. Cell Mol Life Sci 2022; 79:239. [PMID: 35416520 PMCID: PMC11072423 DOI: 10.1007/s00018-022-04261-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 12/21/2022]
Abstract
Many people around the world suffer from some form of paralysis caused by spinal cord injury (SCI), which has an impact on quality and life expectancy. The spinal cord is part of the central nervous system (CNS), which in mammals is unable to regenerate, and to date, there is a lack of full functional recovery therapies for SCI. These injuries start with a rapid and mechanical insult, followed by a secondary phase leading progressively to greater damage. This secondary phase can be potentially modifiable through targeted therapies. The growing literature, derived from mammalian and regenerative model studies, supports a leading role for mitochondria in every cellular response after SCI: mitochondrial dysfunction is the common event of different triggers leading to cell death, cellular metabolism regulates the immune response, mitochondrial number and localization correlate with axon regenerative capacity, while mitochondrial abundance and substrate utilization regulate neural stem progenitor cells self-renewal and differentiation. Herein, we present a comprehensive review of the cellular responses during the secondary phase of SCI, the mitochondrial contribution to each of them, as well as evidence of mitochondrial involvement in spinal cord regeneration, suggesting that a more in-depth study of mitochondrial function and regulation is needed to identify potential targets for SCI therapeutic intervention.
Collapse
Affiliation(s)
- Paula G Slater
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile.
| | - Miguel E Domínguez-Romero
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
| | - Maximiliano Villarreal
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
| | - Verónica Eisner
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
| | - Juan Larraín
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
| |
Collapse
|
67
|
Fan Z, Zhang W, Cao Q, Zou L, Fan X, Qi C, Yan Y, Song B, Wu B. JAK2/STAT3 pathway regulates microglia polarization involved in hippocampal inflammatory damage due to acute paraquat exposure. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 234:113372. [PMID: 35248926 DOI: 10.1016/j.ecoenv.2022.113372] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/14/2022] [Accepted: 03/01/2022] [Indexed: 06/14/2023]
Abstract
OBJECTIVE To explore the effects of acute paraquat (PQ) exposure on the phenotypic polarization of hippocampal microglia and its mechanism. METHODS An acute PQ exposure rat model was established. Male SD rats were exposed to 0, 5, 25, and 50 mg/kg PQ, and brain hippocampal tissue was collected after 1, 3, and 7 days of exposure, respectively. Hippocampal pathological changes were examined by H&E staining, and immunohistochemistry (IHC) was used to detect changes in the number of Iba-1-positive cells, the average number of endpoints, and the average process length. The protein expression of Iba-1 was detected by western blotting. BV-2 microglia were treated with 0, 0.01, 0.025, 0.05, or 0.1 μmol/L PQ for 24 h. ELISA and western blotting assays were performed to detect the expression of TNF-α and IL-1β in vivo and in vitro. The M1 microglia marker iNOS, the M2 microglia marker Arg-1, and the p-JAK2 and p-STAT3 protein were detected by western blotting. JAK2/STAT3 pathway activation role in regulating microglia phenotypic polarization was further validated in vivo and in vitro by JAK2-specific inhibitor AG490 administration. RESULTS After acute PQ exposure, hippocampal neurons showed pathological changes such as loose arrangement and nuclear pyknosis, the number of Iba-1 positive cells and the expression of Iba-1 protein increased, and the average number of endpoints and average process length of microglia decreased. Histological examination revealed that compared with the control group, in the 50 mg/kg PQ group on the 3rd and 7th day, the expression of TNF-α, IL-1β, and iNOS significantly increased, while that of Arg-1 significantly decreased. p-JAK2 and p-STAT3 expression significantly increased in the 50 mg/kg PQ group on the 1st, 3rd, and 7th day. In vitro, compared with the control group, the expression of TNF-α, IL-1β, iNOS, p-JAK2, and p-STAT3 significantly increased, while Arg-1 expression was significantly reduced in the 0.025, 0.05, and 0.1 μmol/L PQ groups. After AG490 administration, the expression levels of p-JAK2, p-STAT3, iNOS, TNF-α, and IL-1β in the AG490 +PQ group were significantly inhibited in vivo and in vitro compared with the PQ-only group. On the contrary, Arg-1 expression was significantly increased. CONCLUSION Our results suggest that acute PQ exposure may induce M1-type polarization of hippocampal microglia by activating the JAK2/STAT3 pathway, which in turn releases pro-inflammatory factors such as TNF-α and IL-1β, leading to hippocampal inflammatory damage.
Collapse
Affiliation(s)
- Zhuo Fan
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, Hebei 050000, China; Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei 050000, China
| | - Wendi Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, Hebei 050000, China; Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei 050000, China
| | - Qi Cao
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, Hebei 050000, China; Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei 050000, China
| | - Lingyun Zou
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, Hebei 050000, China; Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei 050000, China
| | - Xiaobei Fan
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, Hebei 050000, China; Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei 050000, China
| | - Changcun Qi
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, Hebei 050000, China; Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei 050000, China
| | - Yuandong Yan
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, Hebei 050000, China; Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei 050000, China
| | - Bo Song
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, Hebei 050000, China; Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei 050000, China.
| | - Bailin Wu
- Department of Radiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China.
| |
Collapse
|
68
|
Babchenko VY, Belova AS, Bashirzade AA, Tikhonova MA, Demin KA, Zabegalov KN, Petersen EV, Kalueff AV, Amstislavskaya TG. Traumatic Brain Injury Models in Zebrafish (Danio rerio). NEUROSCIENCE AND BEHAVIORAL PHYSIOLOGY 2022; 52:405-414. [DOI: 10.1007/s11055-022-01254-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/21/2021] [Indexed: 01/04/2025]
|
69
|
Zhang W, Fan X, Fan Z, Wu B, Wang M, Duan W, Song B. Acute exposure to paraquat affects the phenotypic differentiation of substantia nigra microglia in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:21339-21347. [PMID: 34761315 DOI: 10.1007/s11356-021-17262-3] [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: 02/09/2021] [Accepted: 10/25/2021] [Indexed: 06/13/2023]
Abstract
The toxicity of the bipyridine cationic herbicide paraquat (PQ) to the lung and kidneys has been widely documented, but the acute toxic effects of PQ on the nervous system have received little attention. This study aimed to explore the changes in the phenotypic differentiation of microglia in rats caused by acute PQ exposure. As results, acute PQ exposure induced pyknosis, edema, and apoptosis in substantia nigra neurons. Immunohistochemistry and western blotting showed that, on day 18, with the increase of exposure dose, the number of Iba-1-positive cells presented an increasing trend with no statistically significant difference among the groups (P > 0.05). Compared with the control group, the process length of Iba-1-positive cells decreased of acute 25 mg/kg PQ exposure on day 18 (P < 0.05). Compared with the control group, on day 39, the number of Iba-1-positive cells in the SN decreased of acute 25 mg/kg PQ exposure, while that increased of acute 45 mg/kg PQ exposure (P < 0.05). The number of endpoints decreased of acute 25 mg/kg PQ exposure (P < 0.05). The process length became shorter both of acute 25 mg/kg and 45 mg/kg PQ exposure (P < 0.05). On day 69, compared with the control group, the number of Iba-1-positive cells in the SN significantly increased of acute 45 mg/kg PQ exposure (P < 0.05). The number of endpoints increased and the process length became longer of acute 25 mg/kg PQ exposure (P < 0.05). Then, the mean fluorescence intensity of inducible nitric oxide synthase (iNOS) and arginine 1 (ARG1) was compared. The number of the M1 phenotype of microglia increased during the early stage of acute 25 mg/kg PQ exposure, whereas the number of the M2 phenotype of microglia increased during the early stage of acute 45 mg/kg PQ exposure (P < 0.05). On day 39, compared with the control group, the expression of iNOS in the SN of acute 45 mg/kg PQ exposure increased than of acute 25 mg/kg exposure. The expression of Arg-1 of 25 mg/kg PQ exposure was significantly increased (P < 0.05). On day 69, the expression of iNOS and ARG1 increased in the 25 and 45 mg/kg PQ exposure groups. In summary, changes in microglia phenotypic differentiation were related to exposure dose and exposure time (P < 0.05).
Collapse
Affiliation(s)
- Wendi Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, 050000, Hebei, China
- Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, 050000, Hebei, China
| | - Xiaobei Fan
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, 050000, Hebei, China
- Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, 050000, Hebei, China
| | - Zhuo Fan
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, 050000, Hebei, China
- Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, 050000, Hebei, China
| | - Bailin Wu
- Department of Radiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Mengchao Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, 050000, Hebei, China
- Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, 050000, Hebei, China
| | - Wanyu Duan
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, 050000, Hebei, China
- Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, 050000, Hebei, China
| | - Bo Song
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, 050000, Hebei, China.
- Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
70
|
Del'Arco AE, Argolo DS, Guillemin G, Costa MDFD, Costa SL, Pinheiro AM. Neurological Infection, Kynurenine Pathway, and Parasitic Infection by Neospora caninum. Front Immunol 2022; 12:714248. [PMID: 35154065 PMCID: PMC8826404 DOI: 10.3389/fimmu.2021.714248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 12/31/2021] [Indexed: 11/30/2022] Open
Abstract
Neuroinflammation is one of the most frequently studied topics of neurosciences as it is a common feature in almost all neurological disorders. Although the primary function of neuroinflammation is to protect the nervous system from an insult, the complex and sequential response of activated glial cells can lead to neurological damage. Depending on the type of insults and the time post-insult, the inflammatory response can be neuroprotective, neurotoxic, or, depending on the glial cell types, both. There are multiple pathways activated and many bioactive intermediates are released during neuroinflammation. One of the most common one is the kynurenine pathway, catabolizing tryptophan, which is involved in immune regulation, neuroprotection, and neurotoxicity. Different models have been used to study the kynurenine pathway metabolites to understand their involvements in the development and maintenance of the inflammatory processes triggered by infections. Among them, the parasitic infection Neospora caninum could be used as a relevant model to study the role of the kynurenine pathway in the neuroinflammatory response and the subset of cells involved.
Collapse
Affiliation(s)
- Ana Elisa Del'Arco
- Laboratory of Biochemistry and Veterinary Immunology, Center of Agrarian, Environmental and Biological Sciences, Federal University of Recôncavo of Bahia (UFRB), Cruz das Almas, Brazil
| | - Deivison Silva Argolo
- Laboratory of Neurochemistry and Cellular Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia (UFBA), Bahia, Brazil
| | - Gilles Guillemin
- Neuroinflammation Group, Macquarie Medicine School, Macquarie University, Sydney, NSW, Australia
| | - Maria de Fátima Dias Costa
- Laboratory of Neurochemistry and Cellular Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia (UFBA), Bahia, Brazil
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cellular Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia (UFBA), Bahia, Brazil
| | - Alexandre Moraes Pinheiro
- Laboratory of Biochemistry and Veterinary Immunology, Center of Agrarian, Environmental and Biological Sciences, Federal University of Recôncavo of Bahia (UFRB), Cruz das Almas, Brazil
| |
Collapse
|
71
|
Guo S, Wang H, Yin Y. Microglia Polarization From M1 to M2 in Neurodegenerative Diseases. Front Aging Neurosci 2022; 14:815347. [PMID: 35250543 PMCID: PMC8888930 DOI: 10.3389/fnagi.2022.815347] [Citation(s) in RCA: 415] [Impact Index Per Article: 138.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/17/2022] [Indexed: 12/11/2022] Open
Abstract
Microglia-mediated neuroinflammation is a common feature of neurodegenerative diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS). Microglia can be categorized into two opposite types: classical (M1) or alternative (M2), though there’s a continuum of different intermediate phenotypes between M1 and M2, and microglia can transit from one phenotype to another. M1 microglia release inflammatory mediators and induce inflammation and neurotoxicity, while M2 microglia release anti-inflammatory mediators and induce anti-inflammatory and neuroprotectivity. Microglia-mediated neuroinflammation is considered as a double-edged sword, performing both harmful and helpful effects in neurodegenerative diseases. Previous studies showed that balancing microglia M1/M2 polarization had a promising therapeutic prospect in neurodegenerative diseases. We suggest that shifting microglia from M1 to M2 may be significant and we focus on the modulation of microglia polarization from M1 to M2, especially by important signal pathways, in neurodegenerative diseases.
Collapse
|
72
|
Honig MG, Del Mar NA, Henderson DL, O'Neal D, Yammanur M, Cox R, Li C, Perry AM, Moore BM, Reiner A. Raloxifene, a cannabinoid type-2 receptor inverse agonist, mitigates visual deficits and pathology and modulates microglia after ocular blast. Exp Eye Res 2022; 218:108966. [PMID: 35143834 DOI: 10.1016/j.exer.2022.108966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/04/2022] [Accepted: 01/24/2022] [Indexed: 11/19/2022]
Abstract
Visual deficits after ocular blast injury (OBI) are common, but pharmacological approaches to improve long-term outcomes have not been identified. Blast forces frequently damage the retina and optic nerves, and work on experimental animals has shown the pro-inflammatory actions of microglia can further exacerbate such injuries. Cannabinoid type-2 receptor (CB2) inverse agonists specifically target activated microglia, biasing them away from the harmful pro-inflammatory M1 state toward the helpful reparative M2 state. We previously found that treating mice with CB2 inverse agonists after traumatic brain injury, produced by either focal cranial air blast or dorsal cranial impact, greatly attenuated the visual deficits and pathology that otherwise resulted. Here we examined the consequences of single and repeat OBI and the benefit provided by raloxifene, an FDA-approved estrogen receptor drug that possesses noteworthy CB2 inverse agonism. After single OBI, although the amplitudes of the A- and B-waves of the electroretinogram and pupil light response appeared to be normal, the mice showed hints of deficits in contrast sensitivity and visual acuity, a trend toward optic nerve axon loss, and significantly increased light aversion, which were reversed by 2 weeks of daily treatment with raloxifene. Mice subjected to repeat OBI (5 blasts spaced 1 min apart), exhibited more severe visual deficits, including decreases in contrast sensitivity, visual acuity, the amplitudes of the A- and B-waves of the electroretinogram, light aversion, and resting pupil diameter (i.e. hyperconstriction), accompanied by the loss of photoreceptor cells and optic nerve axons, nearly all of which were mitigated by raloxifene. Interestingly, optic nerve axon abundance was strongly correlated with contrast sensitivity and visual acuity across all groups of experimental mice in the repeat OBI study, suggesting optic nerve axon loss with rOBI and its attenuation with raloxifene are associated with the extent of these two deficits while photoreceptor abundance was highly correlated with A-wave amplitude and resting pupil size, suggesting a prominent role for photoreceptors in these two deficits. Quantitative PCR (qPCR) showed levels of M1-type microglial markers (e.g. iNOS, IL1β, TNFα, and CD32) in retina, optic nerve, and thalamus were increased 3 days after repeat OBI. With raloxifene treatment, the overall expression of M1 markers was more similar to that in sham mice. Raloxifene treatment was also associated with the elevation of IL10 transcripts in all three tissues compared to repeat OBI alone, but the results for the three other M2 microglial markers we examined were more varied. Taken together, the qPCR results suggest that raloxifene benefit for visual function and pathology was associated with a lessening of the pro-inflammatory actions of microglia. The benefit we find for raloxifene following OBI provides a strong basis for phase-2 efficacy testing in human clinical trials for treating ocular injury.
Collapse
Affiliation(s)
- Marcia G Honig
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Nobel A Del Mar
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Desmond L Henderson
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Dylan O'Neal
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Meghna Yammanur
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Rachel Cox
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Chunyan Li
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Aaron M Perry
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Bob M Moore
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Anton Reiner
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA; Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
73
|
Xiao HX, Song B, Li Q, Shao YM, Zhang YB, Chang XL, Zhou ZJ. Paraquat mediates BV-2 microglia activation by raising intracellular ROS and inhibiting Akt1 phosphorylation. Toxicol Lett 2022; 355:116-126. [PMID: 34863858 DOI: 10.1016/j.toxlet.2021.11.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 11/03/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022]
Abstract
Microglia is the innate immune cell in central nervous system (CNS) and plays an important role in neuroinflammation. Microglia mediated neuroinflammation is the key factor affecting the development of neurodegenerative diseases. Although there was evidence that paraquat (PQ) could induce inflammatory response, its mechanism was not clear. The present study investigated the mechanisms of PQ-induced inflammatory responses in BV-2 microglia cells, and tried to reveal the role of ROS/Akt1 pathway. The results showed that the cell activation markers (iNOS and CD206) of BV-2 cells were increased after PQ treatment, suggesting that BV-2 microglia were activated. PQ induced the reactive oxygen species (ROS) and inhibited the AKT1 phosphorylation in BV-2 cells. Besides, the M1 markers expression (IL-6, TNF-α and IL-1β) were significantly increased after PQ treatment, which suggested that PQ induced the increase of M1 phenotype of BV-2 microglia. Pre-treated with NAC (ROS scavenger), the M1 phenotype was decreased while the p-Akt1 was restored compared to PQ stimulation. Furthermore, we built an Akt1(S473E)-overexpression BV-2 cell line. The Akt1 (S473E) partially attenuated the PQ induced increase in M1 phenotype, while ROS did not significantly change. These results indicated that PQ induced BV-2 microglia activation by increased ROS mediated Akt1 activation inhibition, leading to neuroinflammation.
Collapse
Affiliation(s)
- Hong-Xi Xiao
- School of Public Health, MOE Key Laboratory of Public Health Safety, NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, 200032, China
| | - Bo Song
- School of Public Health, MOE Key Laboratory of Public Health Safety, NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, 200032, China
| | - Qian Li
- School of Public Health, MOE Key Laboratory of Public Health Safety, NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, 200032, China
| | - Yi-Ming Shao
- School of Public Health, MOE Key Laboratory of Public Health Safety, NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, 200032, China
| | - Yu-Bin Zhang
- School of Public Health, MOE Key Laboratory of Public Health Safety, NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, 200032, China
| | - Xiu-Li Chang
- School of Public Health, MOE Key Laboratory of Public Health Safety, NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, 200032, China.
| | - Zhi-Jun Zhou
- School of Public Health, MOE Key Laboratory of Public Health Safety, NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
74
|
Lin YT, Shi QQ, Zhang L, Yue CP, He ZJ, Li XX, He QJ, Liu Q, Du XB. Hydrogen-rich water ameliorates neuropathological impairments in a mouse model of Alzheimer's disease through reducing neuroinflammation and modulating intestinal microbiota. Neural Regen Res 2022; 17:409-417. [PMID: 34269217 PMCID: PMC8464006 DOI: 10.4103/1673-5374.317992] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/19/2020] [Accepted: 03/18/2021] [Indexed: 11/04/2022] Open
Abstract
Hydrogen exhibits the potential to treat Alzheimer's disease. Stereotactic injection has been previously used as an invasive method of administering active hydrogen, but this method has limitations in clinical practice. In this study, triple transgenic (3×Tg) Alzheimer's disease mice were treated with hydrogen-rich water for 7 months. The results showed that hydrogen-rich water prevented synaptic loss and neuronal death, inhibited senile plaques, and reduced hyperphosphorylated tau and neurofibrillary tangles in 3×Tg Alzheimer's disease mice. In addition, hydrogen-rich water improved brain energy metabolism disorders and intestinal flora imbalances and reduced inflammatory reactions. These findings suggest that hydrogen-rich water is an effective hydrogen donor that can treat Alzheimer's disease. This study was approved by the Animal Ethics and Welfare Committee of Shenzhen University, China (approval No. AEWC-20140615-002) on June 15, 2014.
Collapse
Affiliation(s)
- Yi-Tong Lin
- College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory for Plant Epigenetics, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Qing-Qing Shi
- College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory for Plant Epigenetics, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Lei Zhang
- College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory for Plant Epigenetics, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Cai-Ping Yue
- College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory for Plant Epigenetics, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Zhi-Jun He
- College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory for Plant Epigenetics, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Xue-Xia Li
- College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory for Plant Epigenetics, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Qian-Jun He
- Guangdong Provincial Key Laboratory of Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Qiong Liu
- College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory for Plant Epigenetics, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Xiu-Bo Du
- College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory for Plant Epigenetics, Shenzhen University, Shenzhen, Guangdong Province, China
- Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China
| |
Collapse
|
75
|
Huang W, Tao Y, Zhang X, Zhang X. TGF-β1/SMADs signaling involved in alleviating inflammation induced by nanoparticulate titanium dioxide in BV2 cells. Toxicol In Vitro 2022; 80:105303. [PMID: 34990773 DOI: 10.1016/j.tiv.2021.105303] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/28/2021] [Accepted: 12/29/2021] [Indexed: 10/19/2022]
Abstract
There are increasing safety concerns accompanying the widespread use of nanoparticulate titanium dioxide (nano-TiO2). It has been demonstrated that nano-TiO2 can cross the blood-brain barrier and enter the brain, causing damage to the nervous system, consisting mainly of neuroinflammation and neuronal apoptosis. Several studies have linked the TGF-β1/SMADs signaling to the development of inflammatory response in various organs. However, no studies have connected the induction of microglial inflammation by nano-TiO2 to this signaling. Therefore, this study aimed to investigate the role of TGF-β1/SMADs signaling in microglia inflammatory response induced by nano-TiO2. The results showed that nano-TiO2 increased the secretions of pro-inflammatory cytokines (IL-1α, IL-6, and TNF-α) and decreased the expressions of TGF-β1 and SMAD1/2/3 proteins in BV2 cells. When TGF-β1/SMADs signaling was inhibited, the inflammatory effect induced by nano-TiO2 increased, suggesting a suppressive effect of this signaling on the inflammation. In addition, exogenous TGF-β1 upregulated the expressions of TGF-β1 and SMADs1/2/3 proteins as well as decreased the secretions of pro-inflammatory cytokines (IL-1α, IL-6, and TNF-α) compared to BV2 cells treated with only nano-TiO2. Our results suggest that nano-TiO2 may inhibit the TGF-β1/SMADs signaling by suppressing the intracellular secretion of active TGF-β1, leading to microglial activation and the induction or exacerbation of inflammatory responses.
Collapse
Affiliation(s)
- Wendi Huang
- School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing 210009, Jiangsu, China
| | - Yifan Tao
- School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing 210009, Jiangsu, China
| | - Xiuwen Zhang
- School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing 210009, Jiangsu, China
| | - Xiaoqiang Zhang
- School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing 210009, Jiangsu, China.
| |
Collapse
|
76
|
Abstract
The pathology of fetal alcohol syndrome and the less severe fetal alcohol spectrum disorders includes brain dysmyelination. Recent studies have shed light on the molecular mechanisms underlying these white matter abnormalities. Rodent models of fetal alcohol syndrome and human studies have shown suppressed oligodendrocyte differentiation and apoptosis of oligodendrocyte precursor cells. Ethanol exposure led to reduced expression of myelin basic protein and delayed myelin basic protein expression in rat and mouse models of fetal alcohol syndrome and in human histopathological specimens. Several studies have reported increased expression of many chemokines in dysmyelinating disorders in central nervous system, including multiple sclerosis and fetal alcohol syndrome. Acute ethanol exposure reduced levels of the neuroprotective insulin-like growth factor-1 in fetal and maternal sheep and in human fetal brain tissues, while ethanol increased the expression of tumor necrosis factor α in mouse and human neurons. White matter lesions have been induced in the developing sheep brain by alcohol exposure in early gestation. Rat fetal alcohol syndrome models have shown reduced axon diameters, with thinner myelin sheaths, as well as reduced numbers of oligodendrocytes, which were also morphologically aberrant oligodendrocytes. Expressions of markers for mature myelination, including myelin basic protein, also were reduced. The accumulating knowledge concerning the mechanisms of ethanol-induced dysmyelination could lead to the development of strategies to prevent dysmyelination in children exposed to ethanol during fetal development. Future studies using fetal oligodendrocyte- and oligodendrocyte precursor cell-derived exosomes isolated from the mother’s blood may identify biomarkers for fetal alcohol syndrome and even implicate epigenetic changes in early development that affect oligodendrocyte precursor cell and oligodendrocyte function in adulthood. By combining various imaging modalities with molecular studies, it may be possible to determine which fetuses are at risk and to intervene therapeutically early in the pregnancy.
Collapse
Affiliation(s)
- Nune Darbinian
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Michael E Selzer
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
77
|
Katsuki H. Nuclear receptors of NR1 and NR4 subfamilies in the regulation of microglial functions and pathology. Pharmacol Res Perspect 2021; 9:e00766. [PMID: 34676987 PMCID: PMC8532137 DOI: 10.1002/prp2.766] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/01/2021] [Accepted: 03/15/2021] [Indexed: 12/15/2022] Open
Abstract
This review provides an overview of researches on the NR1 and NR4 nuclear receptors involved in the regulation of microglial functions. Nuclear receptors are attractive candidates for drug targets in the therapies of the central nervous system disorders, because the activation of these receptors is expected to regulate the functions and the phenotypes of microglia, by controlling the expression of specific gene subsets and also by regulating the cellular signaling mechanisms in a nongenomic manner. Several members of NR1 nuclear receptor subfamily have been examined for their ability to regulate microglial functions. For example, stimulation of vitamin D receptor inhibits the production of pro-inflammatory factors and increases the production of anti-inflammatory cytokines. Similar regulatory actions of nuclear receptor ligands on inflammation-related genes have also been reported for other NR1 members such as retinoic acid receptors, peroxisome proliferator-activated receptors (PPARs), and liver X receptors (LXRs). In addition, stimulation of PPARγ and LXRs may also result in increased phagocytic activities of microglia. Consistent with these actions, the agonists at nuclear receptors of NR1 subfamily are shown to produce therapeutic effects on animal models of various neurological disorders such as experimental allergic encephalomyelitis, Alzheimer's disease, Parkinson's disease, and ischemic/hemorrhagic stroke. On the other hand, increasing lines of evidence suggest that the stimulation of NR4 subfamily members of nuclear receptors such as Nur77 and Nurr1 also regulates microglial functions and alleviates neuropathological events in several disease models. Further advancement of these research fields may prove novel therapeutic opportunities.
Collapse
Affiliation(s)
- Hiroshi Katsuki
- Department of Chemico‐Pharmacological SciencesGraduate School of Pharmaceutical SciencesKumamoto UniversityKumamotoJapan
| |
Collapse
|
78
|
Yawoot N, Sengking J, Wicha P, Govitrapong P, Tocharus C, Tocharus J. Melatonin attenuates reactive astrogliosis and glial scar formation following cerebral ischemia and reperfusion injury mediated by GSK-3β and RIP1K. J Cell Physiol 2021; 237:1818-1832. [PMID: 34825376 DOI: 10.1002/jcp.30649] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/26/2021] [Accepted: 11/15/2021] [Indexed: 11/06/2022]
Abstract
Even though astrocytes have been widely reported to support several brain functions, studies have emerged that they exert deleterious effects on the brain after ischemia and reperfusion (I/R) injury. The present study investigated the neuroprotective effects of melatonin on the processes of reactive astrogliosis and glial scar formation, as well as axonal regeneration after transient middle cerebral artery occlusion. Male Wistar rats were randomly divided into four groups: sham-operated, I/R, I/R treated with melatonin, and I/R treated with edaravone. All drugs were administered via intraperitoneal injection at the onset of reperfusion and were continued until the rats were sacrificed on Day 7 or 14 after the surgery. Melatonin presented long-term benefits on cerebral damage after I/R injury, as demonstrated by a decreased infarct volume, histopathological changes, and reduced neuronal cell death. We also found that melatonin attenuated reactive astrogliosis and glial scar formation and, consequently, enhanced axonal regeneration and promoted neurobehavioral recovery. Furthermore, glycogen synthase kinase-3 beta (GSK-3β) and receptor-interacting serine/threonine-protein 1 kinase (RIP1K), which had previously been revealed as proteins involved in astrocyte responses, were significantly reduced after melatonin administration. Taken together, melatonin effectively counteracted the deleterious effects due to astrocyte responses and improved axonal regeneration to promote functional recovery during the chronic phase of cerebral I/R injury by inhibiting GSK-3β and RIP1K activities.
Collapse
Affiliation(s)
- Nuttapong Yawoot
- Department of Physiology, Chiang Mai University, Chiang Mai, Thailand.,Graduate School, Chiang Mai University, Chiang Mai, Thailand
| | - Jirakhamon Sengking
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Piyawadee Wicha
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Chainarong Tocharus
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Chiang Mai University, Chiang Mai, Thailand.,Department of Physiology, Functional Food Research Center for Well-being, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
79
|
Honig MG, Del Mar NA, Henderson DL, O'Neal D, Doty JB, Cox R, Li C, Perry AM, Moore BM, Reiner A. Raloxifene Modulates Microglia and Rescues Visual Deficits and Pathology After Impact Traumatic Brain Injury. Front Neurosci 2021; 15:701317. [PMID: 34776838 PMCID: PMC8585747 DOI: 10.3389/fnins.2021.701317] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 09/07/2021] [Indexed: 11/29/2022] Open
Abstract
Mild traumatic brain injury (TBI) involves widespread axonal injury and activation of microglia, which initiates secondary processes that worsen the TBI outcome. The upregulation of cannabinoid type-2 receptors (CB2) when microglia become activated allows CB2-binding drugs to selectively target microglia. CB2 inverse agonists modulate activated microglia by shifting them away from the harmful pro-inflammatory M1 state toward the helpful reparative M2 state and thus can stem secondary injury cascades. We previously found that treatment with the CB2 inverse agonist SMM-189 after mild TBI in mice produced by focal cranial blast rescues visual deficits and the optic nerve axon loss that would otherwise result. We have further shown that raloxifene, which is Food and Drug Administration (FDA)-approved as an estrogen receptor modulator to treat osteoporosis, but also possesses CB2 inverse agonism, yields similar benefit in this TBI model through its modulation of microglia. As many different traumatic events produce TBI in humans, it is widely acknowledged that diverse animal models must be used in evaluating possible therapies. Here we examine the consequences of TBI created by blunt impact to the mouse head for visual function and associated pathologies and assess raloxifene benefit. We found that mice subjected to impact TBI exhibited decreases in contrast sensitivity and the B-wave of the electroretinogram, increases in light aversion and resting pupil diameter, and optic nerve axon loss, which were rescued by daily injection of raloxifene at 5 or 10 mg/ml for 2 weeks. Raloxifene treatment was associated with reduced M1 activation and/or enhanced M2 activation in retina, optic nerve, and optic tract after impact TBI. Our results suggest that the higher raloxifene dose, in particular, may be therapeutic for the optic nerve by enhancing the phagocytosis of axonal debris that would otherwise promote inflammation, thereby salvaging less damaged axons. Our current work, together with our prior studies, shows that microglial activation drives secondary injury processes after both impact and cranial blast TBI and raloxifene mitigates microglial activation and visual system injury in both cases. The results thus provide a strong basis for phase 2 human clinical trials evaluating raloxifene as a TBI therapy.
Collapse
Affiliation(s)
- Marcia G Honig
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Nobel A Del Mar
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Desmond L Henderson
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Dylan O'Neal
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - John B Doty
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Rachel Cox
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Chunyan Li
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Aaron M Perry
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Bob M Moore
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Anton Reiner
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States.,Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
80
|
Gu C, Chen Y, Chen Y, Liu CF, Zhu Z, Wang M. Role of G Protein-Coupled Receptors in Microglial Activation: Implication in Parkinson's Disease. Front Aging Neurosci 2021; 13:768156. [PMID: 34867296 PMCID: PMC8635063 DOI: 10.3389/fnagi.2021.768156] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/23/2021] [Indexed: 12/26/2022] Open
Abstract
Parkinson's disease (PD) is one of the prevalent neurodegenerative diseases associated with preferential loss of dopaminergic (DA) neurons in the substantia nigra compacta (SNc) and accumulation of α-synuclein in DA neurons. Even though the precise pathogenesis of PD is not clear, a large number of studies have shown that microglia-mediated neuroinflammation plays a vital role in the process of PD development. G protein-coupled receptors (GPCRs) are widely expressed in microglia and several of them act as regulators of microglial activation upon corresponding ligands stimulations. Upon α-synuclein insults, microglia would become excessively activated through some innate immune receptors. Presently, as lack of ideal drugs for treating PD, certain GPCR which is highly expressed in microglia of PD brain and mediates neuroinflammation effectively could be a prospective source for PD therapeutic intervention. Here, six kinds of GPCRs and two types of innate immune receptors were introduced, containing adenosine receptors, purinergic receptors, metabotropic glutamate receptors, adrenergic receptors, cannabinoid receptors, and melatonin receptors and their roles in neuroinflammation; we highlighted the relationship between these six GPCRs and microglial activation in PD. Based on the existing findings, we tried to expound the implication of microglial GPCRs-regulated neuroinflammation to the pathophysiology of PD and their potential to become a new expectation for clinical therapeutics.
Collapse
Affiliation(s)
- Chao Gu
- Department of Pharmacy, Children’s Hospital of Soochow University, Suzhou, China
| | - Yajing Chen
- Department of Pharmacy, Children’s Hospital of Soochow University, Suzhou, China
| | - Yan Chen
- Department of Child and Adolescent Healthcare, Children’s Hospital of Soochow University, Suzhou, China
| | - Chun-Feng Liu
- Department of Neurology, Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zengyan Zhu
- Department of Pharmacy, Children’s Hospital of Soochow University, Suzhou, China
| | - Mei Wang
- Department of Pharmacy, Children’s Hospital of Soochow University, Suzhou, China
| |
Collapse
|
81
|
Domínguez-Rivas E, Ávila-Muñoz E, Schwarzacher SW, Zepeda A. Adult hippocampal neurogenesis in the context of lipopolysaccharide-induced neuroinflammation: A molecular, cellular and behavioral review. Brain Behav Immun 2021; 97:286-302. [PMID: 34174334 DOI: 10.1016/j.bbi.2021.06.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 06/17/2021] [Accepted: 06/19/2021] [Indexed: 12/17/2022] Open
Abstract
The continuous generation of new neurons occurs in at least two well-defined niches in the adult rodent brain. One of these areas is the subgranular zone of the dentate gyrus (DG) in the hippocampus. While the DG is associated with contextual and spatial learning and memory, hippocampal neurogenesis is necessary for pattern separation. Hippocampal neurogenesis begins with the activation of neural stem cells and culminates with the maturation and functional integration of a portion of the newly generated glutamatergic neurons into the hippocampal circuits. The neurogenic process is continuously modulated by intrinsic factors, one of which is neuroinflammation. The administration of lipopolysaccharide (LPS) has been widely used as a model of neuroinflammation and has yielded a body of evidence for unveiling the detrimental impact of inflammation upon the neurogenic process. This work aims to provide a comprehensive overview of the current knowledge on the effects of the systemic and central administration of LPS upon the different stages of neurogenesis and discuss their effects at the molecular, cellular, and behavioral levels.
Collapse
Affiliation(s)
- Eduardo Domínguez-Rivas
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Evangelina Ávila-Muñoz
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Stephan W Schwarzacher
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Angélica Zepeda
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico; Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
82
|
Abdi M, Pasbakhsh P, Shabani M, Nekoonam S, Sadeghi A, Fathi F, Abouzaripour M, Mohamed W, Zibara K, Kashani IR, Zendedel A. Metformin Therapy Attenuates Pro-inflammatory Microglia by Inhibiting NF-κB in Cuprizone Demyelinating Mouse Model of Multiple Sclerosis. Neurotox Res 2021; 39:1732-1746. [PMID: 34570348 DOI: 10.1007/s12640-021-00417-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/30/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) is a chronic disorder characterized by reactive gliosis, inflammation, and demyelination. Microglia plays a crucial role in the pathogenesis of MS and has the dynamic plasticity to polarize between pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes. Metformin, a glucose-lowering drug, attenuates inflammatory responses by activating adenosine monophosphate protein kinase (AMPK) which suppresses nuclear factor kappa B (NF-κB). In this study, we indirectly investigated whether metformin therapy would regulate microglia activity in the cuprizone (CPZ)-induced demyelination mouse model of MS via measuring the markers associated with pro- and anti-inflammatory microglia. Evaluation of myelin by luxol fast blue staining revealed that metformin treatment (CPZ + Met) diminished demyelination, in comparison to CPZ mice. In addition, metformin therapy significantly alleviated reactive microgliosis and astrogliosis in the corpus callosum, as measured by Iba-1 and GFAP staining. Moreover, metformin treatment significantly downregulated the expression of pro-inflammatory associated genes (iNOS, H2-Aa, and TNF-α) in the corpus callosum, whereas expression of anti-inflammatory markers (Arg1, Mrc1, and IL10) was not promoted, compared to CPZ mice. Furthermore, protein levels of iNOS (pro-inflammatory marker) were significantly decreased in the metformin group, while those of Trem2 (anti-inflammatory marker) were increased. In addition, metformin significantly increased AMPK activation in CPZ mice. Finally, metformin administration significantly reduced the activation level of NF-κB in CPZ mice. In summary, our data revealed that metformin attenuated pro-inflammatory microglia markers through suppressing NF-κB activity. The positive effects of metformin on microglia and remyelination suggest that it could be used as a promising candidate to lessen the incidence of inflammatory neurodegenerative diseases such as MS.
Collapse
Affiliation(s)
- Mahdad Abdi
- Department of Anatomy, school of medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parichehr Pasbakhsh
- Department of Anatomy, school of medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Maryam Shabani
- Department of Clinical Biochemistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Saied Nekoonam
- Department of Anatomy, school of medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Asie Sadeghi
- Department of Clinical Biochemistry, Faculty of medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Fardin Fathi
- Cellular and Molecular Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | | | - Wael Mohamed
- Basic Medical Science Department, International Islamic University Malaysia, Pahang, Malaysia.,Clinical Pharmacology Department, Menoufia Medical School, Menoufia University, Shebin El Kom, Egypt
| | - Kazem Zibara
- PRASE and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon.
| | - Iraj Ragerdi Kashani
- Department of Anatomy, school of medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Adib Zendedel
- Institute of Neuroanatomy, RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
83
|
Liu Y, Deng S, Song Z, Zhang Q, Guo Y, Yu Y, Wang Y, Li T, Megahed FAK, Addissouky TA, Mao J, Zhang Y. MLIF Modulates Microglia Polarization in Ischemic Stroke by Targeting eEF1A1. Front Pharmacol 2021; 12:725268. [PMID: 34557098 PMCID: PMC8452963 DOI: 10.3389/fphar.2021.725268] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/13/2021] [Indexed: 01/19/2023] Open
Abstract
Monocyte locomotion inhibitory factor (MLIF) is a heat-stable pentapeptide from Entamoeba histolytica. Our previous study found that MLIF protects against ischemic stroke in rats and mice and exerts a neuroprotection effect in human neuroblastoma SH-SY5Y cells. Microglia/macrophage polarization has been proven to be vital in the pathology of ischemic stroke. Nevertheless, whether MLIF is able to modulate microglia/macrophage polarization remains unclear. We performed middle cerebral artery occlusion (MCAO) on C57BL/6J male mice and induced cultured BV2 microglia by oxygen-glucose deprivation (OGD), respectively. Immunfluorescence was utilized to detect the M1/2 markers, such as CD206 and CD16/32. qPCR and ELISA were used to detect the signature gene change of M1/2. The MAPK and NF-κB pathway associated proteins were measured by Western blot. To identify the protein target of MLIF, a pull-down assay was performed. We found that MLIF promoted microglia transferring from a “sick” M1 phenotype to a “healthy” M2 phenotype in vivo or in vitro. Furthermore, we proved that eukaryotic elongation factor 1A1 (eEF1A1) was involved in the modulation of microglia/macrophage polarization. Knocking down eEF1A1 by siRNA exhibited the M1 promotion effect and M2 inhibition effect. Taken together, our results demonstrated MLIF modulated microglia/macrophage polarization by targeting eEF1A1 in ischemic stroke.
Collapse
Affiliation(s)
- Yulan Liu
- School of Medicine, Shanghai University, Shanghai, China.,Department of Pharmacy, The Air Force Hospital From Eastern Theater of PLA, Nanjing, China
| | - Shanshan Deng
- School of Medicine, Shanghai University, Shanghai, China
| | - Zhibing Song
- College of Pharmacology, Anhui University of Chinese Medicine, Hefei, China
| | - Qian Zhang
- College of Pharmacology, Anhui University of Chinese Medicine, Hefei, China
| | - Yuchen Guo
- College of Pharmacology, Anhui University of Chinese Medicine, Hefei, China
| | - Yongsheng Yu
- School of Medicine, Shanghai University, Shanghai, China
| | - Yuliang Wang
- Joint International Research Laboratory of Metabolic and Developmental Sciences, Key Laboratory of Urban Agriculture (South) Ministry of Agriculture, Plant Biotechnology Research Center, Fudan-SJTU-Nottingham Plant Biotechnology R&D Center, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Tiejun Li
- School of Medicine, Shanghai University, Shanghai, China
| | - Fayed A K Megahed
- Nucliec Acid Research Departement, Genetic Engineering and Biotechnological Research Institute, City of Scientific Research and Technological Applications, Alexandria, Egypt
| | | | - Junqin Mao
- Department of Clinical Pharmacy, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuefan Zhang
- School of Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
84
|
Jin GL, Hong LM, Liu HP, Yue RC, Shen ZC, Yang J, Xu Y, Huang HH, Li Y, Xiong BJ, Su YP, Yu CX. Koumine modulates spinal microglial M1 polarization and the inflammatory response through the Notch-RBP-Jκ signaling pathway, ameliorating diabetic neuropathic pain in rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 90:153640. [PMID: 34330066 DOI: 10.1016/j.phymed.2021.153640] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 06/05/2021] [Accepted: 06/22/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Diabetic neuropathic pain (DNP), a complication of diabetes, has serious impacts on human health. As the pathogenesis of DNP is very complex, clinical treatments for DNP is limited. Koumine (KM) is an active ingredient extracted from Gelsemium elegans Benth. that exerts an inhibitory effect on neuropathic pain (NP) in several animal models. PURPOSE To clarify the anti-NP effect of KM on rats with DNP and the molecular mechanisms involving the Notch- Jκ recombination signal binding protein (RBP-Jκ) signaling pathway. METHODS Male Sprague-Dawley rats were administered streptozocin (STZ) by intraperitoneal injection to induce DNP. The effect of KM on mechanical hyperalgesia in rats with DNP was evaluated using the Von Frey test. Microglial polarization in the spinal cord was examined using western blotting and quantitative real-time PCR. The Notch-RBP-Jκ signaling pathway was analysed using western blotting. RESULTS KM attenuated DNP during the observation period. In addition, KM alleviated M1 microglial polarization in STZ-induced rats. Subsequent experiments revealed that Notch-RBP-Jκ signaling pathway was activated in the spinal cord of rats with DNP, and the activation of this pathways was decreased by KM. Additionally, KM-mediated analgesia and deactivation of the Notch-RBP-Jκ signaling pathway were inhibited by the Notch signaling agonist jagged 1, indicating that the anti-DNP effect of KM may be regulated by the Notch-RBP-Jκ signaling pathway. CONCLUSIONS KM is a potentially desirable candidate treatment for DNP that may inhibit microglial M1 polarization through the Notch-RBP-Jκ signaling pathway.
Collapse
Affiliation(s)
- Gui-Lin Jin
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China; Fujian Key Laboratory of Natural Medicine Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China.
| | - Li-Mian Hong
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China; Department of Pharmacy, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Hai-Ping Liu
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Rong-Cai Yue
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Zu-Cheng Shen
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Jian Yang
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China; Fujian Key Laboratory of Natural Medicine Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Ying Xu
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China; Fujian Key Laboratory of Natural Medicine Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Hui-Hui Huang
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China; Fujian Key Laboratory of Natural Medicine Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Yi Li
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Bo-Jun Xiong
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Yan-Ping Su
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China; Fujian Key Laboratory of Natural Medicine Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Chang-Xi Yu
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China; Fujian Key Laboratory of Natural Medicine Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China.
| |
Collapse
|
85
|
Unraveling Axon Guidance during Axotomy and Regeneration. Int J Mol Sci 2021; 22:ijms22158344. [PMID: 34361110 PMCID: PMC8347220 DOI: 10.3390/ijms22158344] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023] Open
Abstract
During neuronal development and regeneration axons extend a cytoskeletal-rich structure known as the growth cone, which detects and integrates signals to reach its final destination. The guidance cues “signals” bind their receptors, activating signaling cascades that result in the regulation of the growth cone cytoskeleton, defining growth cone advance, pausing, turning, or collapse. Even though much is known about guidance cues and their isolated mechanisms during nervous system development, there is still a gap in the understanding of the crosstalk between them, and about what happens after nervous system injuries. After neuronal injuries in mammals, only axons in the peripheral nervous system are able to regenerate, while the ones from the central nervous system fail to do so. Therefore, untangling the guidance cues mechanisms, as well as their behavior and characterization after axotomy and regeneration, are of special interest for understanding and treating neuronal injuries. In this review, we present findings on growth cone guidance and canonical guidance cues mechanisms, followed by a description and comparison of growth cone pathfinding mechanisms after axotomy, in regenerative and non-regenerative animal models.
Collapse
|
86
|
Gu M, Li X, Yan L, Zhang Y, Yang L, Li S, Song C. Endogenous ω-3 fatty acids in Fat-1 mice attenuated depression-like behaviors, spatial memory impairment and relevant changes induced by olfactory bulbectomy. Prostaglandins Leukot Essent Fatty Acids 2021; 171:102313. [PMID: 34246927 DOI: 10.1016/j.plefa.2021.102313] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 06/12/2021] [Accepted: 06/17/2021] [Indexed: 11/21/2022]
Abstract
OBJECTIVES Olfactory bulbectomy (OB) induced behaviors, hypercortisolism, inflammation and neurotrophin dysfunctions are similar to those observed in depressed patients. Omega (n)-3 polyunsaturated fatty acids (PUFAs) can effectively treat depression via anti-inflammatory and neuroprotective effects. However, n-3 PUFA purities, caloric contents, and ratios in different diets often cause contradictive results. This study used Fat-1 mice, which can convert n-6 to n-3 PUFAs in the brain, to study the effect of n-3 PUFAs on OB-induced behaviors and related changes. METHODS Fat-1 and wild-type littermates were fed safflower oil for 3 months. Behaviors were tested on day 21 after surgery. Monoamine neurotransmitters were measured by HPLC. Macrophage activity was measured by MTT assay. Astrocyte phenotypes A1 S100β, A2 BDNF and cholesterol level were measured by ELISA and total cholesterol assay kits respectively. PUFA profile and membrane fluidity were detected by GC and DPH fluorescence probe respectively. RESULTS OB significantly induced animal hyperactivity and spatial memory impairment, while decreased sucrose consumption and social contact with decreased 5-HT turnover, increased the macrophage activity and S100β/BDNF ratio. Meanwhile, n-3/n-6 PUFAs ratio and total cholesterol level were reduced in OB mice. Whereas, OB-induced behavioral changes were attenuated, which were associated with increasing 5-HT turnover, decrease macrophage activity, restored S100β/BDNF and n-3/n-6 PUFAs ratios, and total cholesterol concentrations in Fat-1 mice. CONCLUSION The present study for the first time demonstrated that endogenous n-3 PUFAs attenuated OB-induced depression-like behaviors and spatial memory impairment through modulating serotonergic and immune function, balancing the astrocyte A1/A2 phenotypes, and normalizing PUFAs profile and membrane function.
Collapse
Affiliation(s)
- Minqing Gu
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Xiaohong Li
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Ling Yan
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China; Institute of Biomedicine and Translation Medicine, Department of Physiology, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Yongping Zhang
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China; Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
| | - Longen Yang
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Shurui Li
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Cai Song
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China; Shenzhen Institute of Guangdong Ocean University, Shenzhen, China.
| |
Collapse
|
87
|
Zheng C, Wu A, Zhai X, Ji H, Chen Z, Chen X, Yu X. The cellular immunotherapy of integrated photothermal anti-oxidation Pd-Se nanoparticles in inhibition of the macrophage inflammatory response in rheumatoid arthritis. Acta Pharm Sin B 2021; 11:1993-2003. [PMID: 34386333 PMCID: PMC8343190 DOI: 10.1016/j.apsb.2021.02.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/25/2021] [Accepted: 02/02/2021] [Indexed: 12/17/2022] Open
Abstract
Reducing the inflammatory response is a major goal in the therapy of rheumatoid arthritis (RA). Herein, we integrated palladium nanoparticles (Pd NPs) with selenium nanoparticles (Se NPs) and obtained a multiple nanosystem (Pd@Se-HA NPs) that could simultaneously scavenge hydroxyl radicals (⋅OH) and provide a photothermal effect. The Pd@Se-HA NPs were constructed by a simple self-assembly method in which Se NPs were electrostatically bonded to Pd NPs; hyaluronic acid (HA) was linked to the NPs by ester bonding to provide macrophage targeting ability. The experiments show that the combined therapy of eliminating ⋅OH with Se NPs and utilizing PTT with Pd NPs could effectively reduce the inflammatory response in macrophages more effectively than either individual NP treatment. In addition, the outer layer of HA could specifically target the CD44 receptor to enhance the accumulation of Pd@Se NPs at the lesion, further enhancing the therapeutic effect. After treatment for 15 days, the Pd@Se-HA NPs nearly eliminated the inflammatory response in the joints of mice in an induced RA model, and prevented joint damage and degradation.
Collapse
|
88
|
Hajinejad M, Sahab-Negah S. Neuroinflammation: The next target of exosomal microRNAs derived from mesenchymal stem cells in the context of neurological disorders. J Cell Physiol 2021; 236:8070-8081. [PMID: 34189724 DOI: 10.1002/jcp.30495] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/28/2021] [Accepted: 06/17/2021] [Indexed: 12/16/2022]
Abstract
Among different types of mechanisms involved in neurological disorders, neuroinflammation links initial insults to secondary injuries and triggers some chronic outcomes, for example, neurodegenerative disorders. Thus, anti-inflammatory substances can be targeted as a novel therapeutic option for translational and clinical research to improve brain disease outcomes. In this review, we propose to introduce a new insight into the anti-inflammatory effects of mesenchymal stem cells (MSCs) as the most frequent source for stem cell therapy in neurological diseases. Our insight incorporates a bystander effect of these stem cells in modulating inflammation and microglia/macrophage polarization through exosomes. Exosomes are nano-sized membrane vesicles that carry cell-specific constituents, including protein, lipid, DNA, and RNA. microRNAs (miRNAs) have recently been detected in exosomes that can be taken up by other cells and affect the behavior of recipient cells. In this article, we outline and highlight the potential use of exosomal miRNAs derived from MSCs for inflammatory pathways in the context of neurological disorders. Furthermore, we suggest that focusing on exosomal miRNAs derived from MSCs in the course of neuroinflammatory pathways in the future could reveal their functions for diverse neurological diseases, including brain injuries and neurodegenerative diseases. It is hoped that this study will contribute to a deep understanding of stem cell bystander effects through exosomal miRNAs.
Collapse
Affiliation(s)
- Mehrdad Hajinejad
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Anatomy and Cell Biology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sajad Sahab-Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.,Department of Neuroscience, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
89
|
Fu X, Zhou G, Zhuang J, Xu C, Zhou H, Peng Y, Cao Y, Zeng H, Li J, Yan F, Wang L, Chen G. White Matter Injury After Intracerebral Hemorrhage. Front Neurol 2021; 12:562090. [PMID: 34177751 PMCID: PMC8222731 DOI: 10.3389/fneur.2021.562090] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 05/05/2021] [Indexed: 11/13/2022] Open
Abstract
Spontaneous intracerebral hemorrhage (ICH) accounts for 15% of all stroke cases. ICH is a devastating form of stroke associated with high morbidity, mortality, and disability. Preclinical studies have explored the mechanisms of neuronal death and gray matter damage after ICH. However, few studies have examined the development of white matter injury (WMI) following ICH. Research on WMI indicates that its pathophysiological presentation involves axonal damage, demyelination, and mature oligodendrocyte loss. However, the detailed relationship and mechanism between WMI and ICH remain unclear. Studies of other acute brain insults have indicated that WMI is strongly correlated with cognitive deficits, neurological deficits, and depression. The degree of WMI determines the short- and long-term prognosis of patients with ICH. This review demonstrates the structure and functions of the white matter in the healthy brain and discusses the pathophysiological mechanism of WMI following ICH. Our review reveals that the development of WMI after ICH is complex; therefore, comprehensive treatment is essential. Understanding the relationship between WMI and other brain cells may reveal therapeutic targets for the treatment of ICH.
Collapse
Affiliation(s)
- Xiongjie Fu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Guoyang Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianfeng Zhuang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chaoran Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hang Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yucong Peng
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Cao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hanhai Zeng
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianru Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Yan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lin Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
90
|
Naseh M, Vatanparast J, Rafati A, Bayat M, Haghani M. The emerging role of FTY720 as a sphingosine 1-phosphate analog for the treatment of ischemic stroke: The cellular and molecular mechanisms. Brain Behav 2021; 11:e02179. [PMID: 33969931 PMCID: PMC8213944 DOI: 10.1002/brb3.2179] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 04/23/2021] [Accepted: 04/25/2021] [Indexed: 12/28/2022] Open
Abstract
Finding novel and effective drugs for the treatment of ischemic stroke is warranted because there is not a definitive treatment for this prevalent disease. Due to the relevance between the sphingosine 1-phosphate (S1P) receptor and several neurological diseases including ischemic stroke, it seems that fingolimod (FTY720), as an agonist of S1P receptor, can be a useful therapeutic strategy in these patients. FTY720 is the first oral drug approved by the US food and drug administration for the treatment of multiple sclerosis. Three important mechanisms for neuroprotective effects of FTY720 have been described. First, the functional antagonistic mechanism that is associated with lymphopenia and reduced lymphocytic inflammation. This effect results from the down-regulation and degradation of lymphocytes' S1P receptors, which inhibits lymph node lymphocytes from entering the bloodstream. Second, a functional agonistic activity that is mediated through direct effects via targeting S1P receptors on the membrane of various cells including neurons, microglia, oligodendrocytes, astrocytes, and endothelial cells of blood vessels in the central nervous system (CNS), and the third, receptor-independent mechanisms that are displayed by binding to specific cellular proteins that modulate intracellular signaling pathways or affect epigenetic transcriptions. Therefore, we review these mechanisms in more detail and describe the animal model and in clinical trial studies that support these three mechanisms for the neuroprotective action of FTY720 in ischemic stroke.
Collapse
Affiliation(s)
- Maryam Naseh
- Histomorphometry and Stereology Research CentreShiraz University of Medical SciencesShirazIran
| | | | - Ali Rafati
- Histomorphometry and Stereology Research CentreShiraz University of Medical SciencesShirazIran
- Department of PhysiologyShiraz University of Medical SciencesShirazIran
| | - Mahnaz Bayat
- Clinical Neurology Research CenterShiraz University of Medical SciencesShirazIran
| | - Masoud Haghani
- Histomorphometry and Stereology Research CentreShiraz University of Medical SciencesShirazIran
- Department of PhysiologyShiraz University of Medical SciencesShirazIran
| |
Collapse
|
91
|
A high-fat diet, but not haloperidol or olanzapine administration, increases activated microglial expression in the rat brain. Neurosci Lett 2021; 757:135976. [PMID: 34023409 DOI: 10.1016/j.neulet.2021.135976] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 04/28/2021] [Accepted: 05/17/2021] [Indexed: 11/20/2022]
Abstract
This study examined the effects of chronic treatment of the antipsychotic drugs, haloperidol and olanzapine, on microglial activation in the brain. In addition, we explored the interaction of these antipsychotic drugs with normal and high-fat diet. In order to measure activated microglial expression, we used [3H] PK11195 in vitro autoradiography. Male Sprague Dawley rats were given a diet of either regular chow diet or a high-fat diet, and assigned either water, haloperidol drinking solution (1.5 mg/kg), or olanzapine drinking solution (10 mg/kg) for four weeks. Following treatment, rats were euthanized and brains extracted for [3H] PK11195 autoradiography. Rats on 4 weeks of a high-fat diet showed increased [3H] PK11195 binding compared to rats on a normal diet in the temporal association cortex (19 %), ectorhinal cortex (17 %), entorhinal cortex (18 %), and perirhinal cortex (18 %), irrespective of drug treatment. These are regions associated with memory, sensory, and visual processing. Rats treated with either haloperidol or olanzapine showed no differences in [3H] PK11195 binding compared to the control group. However, there were differences between the 2 different antipsychotic medications themselves. Haloperidol increased [3H] PK11195 binding in the amygdala (23 %), ectorhinal cortex (24 %), and perihinal cortex (29 %), compared to olanzapine. These results corroborate a known role of a high-fat diet and central inflammatory changes but suggest no role of these antipsychotic drugs in promoting neuroinflammation across 4 weeks compared to normal control rats.
Collapse
|
92
|
Uhelski ML, Li Y, Fonseca MM, Romero-Snadoval EA, Dougherty PM. Role of innate immunity in chemotherapy-induced peripheral neuropathy. Neurosci Lett 2021; 755:135941. [PMID: 33961945 DOI: 10.1016/j.neulet.2021.135941] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 01/18/2023]
Abstract
It has become increasingly clear that the innate immune system plays an essential role in the generation of many types of neuropathic pain including that which accompanies cancer treatment. In this article we review current findings of the role of the innate immune system in contributing to cancer treatment pain at the distal endings of peripheral nerve, in the nerve trunk, in the dorsal root ganglion and in the spinal dorsal horn.
Collapse
Affiliation(s)
- Megan L Uhelski
- The Department of Pain Medicine Research, The Division of Anesthesiology, Critical Care and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, United States
| | - Yan Li
- The Department of Pain Medicine Research, The Division of Anesthesiology, Critical Care and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, United States
| | - Miriam M Fonseca
- The Department of Anesthesiology, Wake Forest School of Medicine, United States
| | | | - Patrick M Dougherty
- The Department of Pain Medicine Research, The Division of Anesthesiology, Critical Care and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, United States.
| |
Collapse
|
93
|
Huang Z, Ng TK, Chen W, Sun X, Huang D, Zheng D, Yi J, Xu Y, Zhuang X, Chen S. Nattokinase Attenuates Retinal Neovascularization Via Modulation of Nrf2/HO-1 and Glial Activation. Invest Ophthalmol Vis Sci 2021; 62:25. [PMID: 34036312 PMCID: PMC8164371 DOI: 10.1167/iovs.62.6.25] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 05/04/2021] [Indexed: 02/05/2023] Open
Abstract
PURPOSE Nattokinase (NK), an active ingredient extracted from traditional food Natto, has been studied for prevention and treatment of cardiovascular diseases due to various vasoprotective effects, including fibrinolytic, antihypertensive, anti-atherosclerotic, antiplatelet, and anti-inflammatory activities. Here, we reported an antineovascular effect of NK against experimental retinal neovascularization. METHODS The inhibitory effect of NK against retinal neovascularization was evaluated using an oxygen-induced retinopathy murine model. Expressions of Nrf2/HO-1 signaling and glial activation in the NK-treated retinae were measured. We also investigated cell proliferation and migration of human umbilical vein endothelial cells (HUVECs) after NK administration. RESULTS NK treatment significantly attenuated retinal neovascularization in the OIR retinae. Consistently, NK suppressed VEGF-induced cell proliferation and migration in a concentration-dependent manner in cultured vascular endothelial cells. NK ameliorated ischemic retinopathy partially via activating Nrf2/HO-1. In addition, NK orchestrated reactive gliosis and promoted microglial activation toward a reparative phenotype in ischemic retina. Treatment of NK exhibited no cell toxicity or anti-angiogenic effects in the normal retina. CONCLUSIONS Our results revealed the anti-angiogenic effect of NK against retinal neovascularization via modulating Nrf2/HO-1, glial activation and neuroinflammation, suggesting a promising alternative treatment strategy for retinal neovascularization.
Collapse
Affiliation(s)
- Zijing Huang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Tsz Kin Ng
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Weiqi Chen
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Xiaowei Sun
- Department of Ophthalmology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Dingguo Huang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Dezhi Zheng
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Jingsheng Yi
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Yanxuan Xu
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Xi Zhuang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Shaolang Chen
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
| |
Collapse
|
94
|
Schober ME, Requena DF, Ohde JW, Maves S, Pauly JR. Docosahexaenoic acid decreased inflammatory gene expression, but not 18-kDa translocator protein binding, in rat pup brain after controlled cortical impact. J Trauma Acute Care Surg 2021; 90:866-873. [PMID: 33728886 PMCID: PMC8068600 DOI: 10.1097/ta.0000000000003084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Traumatic brain injury is the leading cause of acquired neurologic disability in children. In our model of pediatric traumatic brain injury, controlled cortical impact (CCI) in rat pups, docosahexaenoic acid (DHA) improved lesion volume and cognitive testing as late as postinjury day (PID) 50. Docosahexaenoic acid decreased proinflammatory messenger RNA (mRNA) in microglia and macrophages at PIDs 3 and 7, but not 30. We hypothesized that DHA affected inflammatory markers differentially relative to impact proximity, early and persistently after CCI. METHODS To provide a temporal snapshot of regional neuroinflammation, we measured 18-kDa translocator protein (TSPO) binding using whole brain autoradiography at PIDs 3, 7, 30, and 50. Guided by TSPO results, we measured mRNA levels in contused cortex and underlying hippocampus for genes associated with proinflammatory and inflammation-resolving states at PIDs 2 and 3. RESULTS Controlled cortical impact increased TSPO binding at all time points, most markedly at PID 3 and in regions closest to impact, not blunted by DHA. Controlled cortical impact increased cortical and hippocampal mRNA proinflammatory markers, blunted by DHA at PID 2 in hippocampus. CONCLUSION Controlled cortical impact increased TSPO binding in the immature brain in a persistent manner more intensely with more severe injury, not altered by DHA. Controlled cortical impact increased PIDs 2 and 3 mRNA levels of proinflammatory and inflammation-resolving genes. Docosahexaenoic acid decreased proinflammatory markers associated with inflammasome activation at PID 2. We speculate that DHA's salutary effects on long-term outcomes result from early effects on the inflammasome. Future studies will examine functional effects of DHA on microglia both early and late after CCI.
Collapse
Affiliation(s)
- Michelle Elena Schober
- From the Primary Children's Hospital (M.E.S.), and Division of Critical Care, Department of Pediatrics (M.E.S., D.F.R., S.M.), University of Utah, Salt Lake City, Utah; and Department of Pharmaceutical Sciences (J.W.O., J.K.P.), College of Pharmacy, University of Kentucky, Lexington, Kentucky
| | | | | | | | | |
Collapse
|
95
|
Leite Dantas R, Freff J, Ambrée O, Beins EC, Forstner AJ, Dannlowski U, Baune BT, Scheu S, Alferink J. Dendritic Cells: Neglected Modulators of Peripheral Immune Responses and Neuroinflammation in Mood Disorders? Cells 2021; 10:941. [PMID: 33921690 PMCID: PMC8072712 DOI: 10.3390/cells10040941] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/25/2021] [Accepted: 04/07/2021] [Indexed: 12/11/2022] Open
Abstract
Affective disorders (AD) including major depressive disorder (MDD) and bipolar disorder (BD) are common mood disorders associated with increased disability and poor health outcomes. Altered immune responses characterized by increased serum levels of pro-inflammatory cytokines and neuroinflammation are common findings in patients with AD and in corresponding animal models. Dendritic cells (DCs) represent a heterogeneous population of myeloid cells that orchestrate innate and adaptive immune responses and self-tolerance. Upon sensing exogenous and endogenous danger signals, mature DCs secrete proinflammatory factors, acquire migratory and antigen presenting capacities and thus contribute to neuroinflammation in trauma, autoimmunity, and neurodegenerative diseases. However, little is known about the involvement of DCs in the pathogenesis of AD. In this review, we summarize the current knowledge on DCs in peripheral immune responses and neuroinflammation in MDD and BD. In addition, we consider the impact of DCs on neuroinflammation and behavior in animal models of AD. Finally, we will discuss therapeutic perspectives targeting DCs and their effector molecules in mood disorders.
Collapse
Affiliation(s)
- Rafael Leite Dantas
- Department of Mental Health, University of Münster, 48149 Münster, Germany; (R.L.D.); (J.F.); (U.D.); (B.T.B.)
- Cells in Motion Interfaculty Centre, University of Münster, 48149 Münster, Germany
| | - Jana Freff
- Department of Mental Health, University of Münster, 48149 Münster, Germany; (R.L.D.); (J.F.); (U.D.); (B.T.B.)
- Cells in Motion Interfaculty Centre, University of Münster, 48149 Münster, Germany
| | - Oliver Ambrée
- Department of Behavioural Biology, University of Osnabrück, 49076 Osnabrück, Germany;
- Center of Cellular Nanoanalytics, University of Osnabrück, 49076 Osnabrück, Germany
| | - Eva C. Beins
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, 53127 Bonn, Germany; (E.C.B.); (A.J.F.)
| | - Andreas J. Forstner
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, 53127 Bonn, Germany; (E.C.B.); (A.J.F.)
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, 52428 Jülich, Germany
| | - Udo Dannlowski
- Department of Mental Health, University of Münster, 48149 Münster, Germany; (R.L.D.); (J.F.); (U.D.); (B.T.B.)
| | - Bernhard T. Baune
- Department of Mental Health, University of Münster, 48149 Münster, Germany; (R.L.D.); (J.F.); (U.D.); (B.T.B.)
- Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Parkville, VIC 3010, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Stefanie Scheu
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, 40225 Düsseldorf, Germany;
| | - Judith Alferink
- Department of Mental Health, University of Münster, 48149 Münster, Germany; (R.L.D.); (J.F.); (U.D.); (B.T.B.)
- Cells in Motion Interfaculty Centre, University of Münster, 48149 Münster, Germany
| |
Collapse
|
96
|
Auditory Brainstem Deficits from Early Treatment with a CSF1R Inhibitor Largely Recover with Microglial Repopulation. eNeuro 2021; 8:ENEURO.0318-20.2021. [PMID: 33558268 PMCID: PMC8009669 DOI: 10.1523/eneuro.0318-20.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 12/10/2020] [Accepted: 01/13/2021] [Indexed: 12/20/2022] Open
Abstract
Signaling between neurons and glia is necessary for the formation of functional neural circuits. A role for microglia in the maturation of connections in the medial nucleus of the trapezoid body (MNTB) was previously demonstrated by postnatal microglial elimination using a colony stimulating factor 1 receptor (CSF1R). Defective pruning of calyces of Held and significant reduction of the mature astrocyte marker glial fibrillary acidic protein (GFAP) were observed after hearing onset. Here, we investigated the time course required for microglia to populate the mouse MNTB after cessation of CSF1R inhibitor treatment. We then examined whether defects seen after microglial depletion were rectified by microglial repopulation. We found that microglia returned to control levels at four weeks of age (18 d postcessation of treatment). Calyceal innervation of MNTB neurons was comparable to control levels at four weeks and GFAP expression recovered by seven weeks. We further investigated the effects of microglia elimination and repopulation on auditory function using auditory brainstem recordings (ABRs). Temporary microglial depletion significantly elevated auditory thresholds in response to 4. 8, and 12 kHz at four weeks. Treatment significantly affected latencies, interpeak latencies, and amplitudes of all the ABR peaks in response to many of the frequencies tested. These effects largely recovered by seven weeks. These findings highlight the functions of microglia in the formation of auditory neural circuits early in development. Further, the results suggest that microglia retain their developmental functions beyond the period of circuit refinement.
Collapse
|
97
|
Beaulieu J, Costa G, Renaud J, Moitié A, Glémet H, Sergi D, Martinoli MG. The Neuroinflammatory and Neurotoxic Potential of Palmitic Acid Is Mitigated by Oleic Acid in Microglial Cells and Microglial-Neuronal Co-cultures. Mol Neurobiol 2021; 58:3000-3014. [PMID: 33604780 DOI: 10.1007/s12035-021-02328-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 02/10/2021] [Indexed: 12/19/2022]
Abstract
Neuroinflammation has been implicated in the pathogenesis of neurodegeneration and is now accepted as a common molecular feature underpinning neuronal damage and death. Palmitic acid (PA) may represent one of the links between diet and neuroinflammation. The aims of this study were to assess whether PA induced toxicity in neuronal cells by modulating microglial inflammatory responses and/or by directly targeting neurons. We also determined the potential of oleic acid (OA), a monounsaturated fatty acid, to counteract inflammation and promote neuroprotection. We measured the ability of PA to induce the secretion of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), the induction of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signalling pathways, as well as the phosphorylation of c-Jun, and the expression of inducible nitric oxide synthase (iNOS). Finally, to determine whether PA exerted an indirect neurotoxic effect on neuronal cells, we employed a microglia-neuron co-culture paradigm where microglial cells communicate with neuronal cells in a paracrine fashion. Herein, we demonstrate that PA induces the activation of the NF-κB signalling pathway and c-Jun phosphorylation in N9 microglia cells, in the absence of increased cytokine secretion. Moreover, our data illustrate that PA exerts an indirect as well as a direct neurotoxic role on neuronal PC12 cells and these effects are partially prevented by OA. These results are important to establish that PA interferes with neuronal homeostasis and suggest that dietary PA, when consumed in excess, may induce neuroinflammation and possibly concurs in the development of neurodegeneration.
Collapse
Affiliation(s)
- Jimmy Beaulieu
- Department of Medical Biology, Université du Québec à Trois-Rivières, 3351 boul. des Forges, G9A 5H7, Trois-Rivières, QC, Canada
| | - Giulia Costa
- Department of Biomedical Sciences, Section of Neurosciences, University of Cagliari, Cagliari, Italy
| | - Justine Renaud
- Department of Medical Biology, Université du Québec à Trois-Rivières, 3351 boul. des Forges, G9A 5H7, Trois-Rivières, QC, Canada
| | - Amélie Moitié
- Department of Medical Biology, Université du Québec à Trois-Rivières, 3351 boul. des Forges, G9A 5H7, Trois-Rivières, QC, Canada
| | - Hélène Glémet
- Department of Biological and Ecological Sciences, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | - Domenico Sergi
- Nutrition & Health Substantiation Group, Nutrition and Health Program, Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Maria-Grazia Martinoli
- Department of Medical Biology, Université du Québec à Trois-Rivières, 3351 boul. des Forges, G9A 5H7, Trois-Rivières, QC, Canada. .,Department of Psychiatry & Neurosciences, Université Laval and CHU Research Center, Québec, Canada.
| |
Collapse
|
98
|
Cao L, Ali S, Queen NJ. Hypothalamic gene transfer of BDNF promotes healthy aging. VITAMINS AND HORMONES 2021; 115:39-66. [PMID: 33706955 DOI: 10.1016/bs.vh.2020.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The aging process and age-related diseases all involve metabolic decline and impaired ability to cope with adversity. Environmental enrichment (EE)-a housing environment which recapitulates aspects of active lifestyle-exerts a wide range of health benefits in laboratory rodents. Brain-derived neurotrophic factor (BDNF) in the hypothalamus orchestrates autonomic and neuroendocrine processes, serving as one key brain mediator of EE-induced resistance to obesity, cancer, and autoimmunity. Recombinant adeno-associated virus (AAV)-mediated hypothalamic BDNF gene transfer alleviates obesity, diabetes, and metabolic syndromes in both diet-induced and genetic models. One recent study by our lab demonstrates the efficacy and safety of a built-in autoregulatory system to control transgene BDNF expression, mimicking the body's natural feedback systems in middle-age mice. Twelve-month old mice were treated with autoregulatory BDNF vector and monitored for 7months. BDNF gene transfer prevented age-associated metabolic decline by: reducing adiposity, preventing the decline of brown fat activity, increasing adiponectin while reducing leptin and insulin in circulation, improving glucose tolerance, increasing energy expenditure, alleviating hepatic steatosis, and suppressing inflammatory genes in the hypothalamus and adipose tissues. Furthermore, BDNF treatment reduced anxiety-like and depression-like behaviors. This chapter summarizes this work and discusses potential roles that hypothalamic BDNF might play in promoting healthy aging.
Collapse
Affiliation(s)
- Lei Cao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States; The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States.
| | - Seemaab Ali
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States; The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Nicholas J Queen
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States; The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| |
Collapse
|
99
|
Najafian SA, Farbood Y, Sarkaki A, Ghafouri S. FTY720 administration following hypoxia-induced neonatal seizure reverse cognitive impairments and severity of seizures in male and female adult rats: The role of inflammation. Neurosci Lett 2021; 748:135675. [PMID: 33516800 DOI: 10.1016/j.neulet.2021.135675] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/05/2020] [Accepted: 01/21/2021] [Indexed: 11/15/2022]
Abstract
Hypoxia-induced neonatal seizure mainly leads to deleterious effects on brain function, especially cognitive impairments and increased susceptibility to epilepsy later in life. Early inflammation plays an important role in the pathology of these consequences. Therefore, we explored the long-term outcomes of Fingolimod treatment as an anti-inflammatory and neuroprotective agent in a rat model of HINS. Seizures were induced in rats (postnatal day 10) by 5% O2 exposure for 15 min. Sixty minutes after the onset of hypoxia, pups received FTY720 (0.3 mg.kg-1) or normal saline for 12 consecutive days (lactation period), and they were used at P60-P63 for behavioral tests, ELISA and Pentylenetetrazole kindling model. The results of open field, novel object recognition and elevated plus maze tasks showed that Fingolimod prevents hippocampal memory dysfunction and anxiety-like behavior in both male and female hypoxic groups, which was accompanied with decreased TNF-α level in hippocampus. In addition, FTY720 postponed epileptogenesis just in female hypoxic + FTY group and decreased severity of seizures in both genders. Our results suggest that, FTY720 treatment in immature rats, which were previously subjected to HINS, prevented the long-lasting deficits, like cognitive impairments, decreased the severity of seizures and related inflammation. In addition, FTY720 did not show significant interaction with gender in most of the experiments, except the average day to reach fully kindled state. Taken together, FTY720 has therapeutic potential for long lasting effects of HINS in both male and female animals at puberty.
Collapse
Affiliation(s)
- Seyed Ahmad Najafian
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Yaghoob Farbood
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Sarkaki
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Samireh Ghafouri
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
100
|
Li L, Gan H, Jin H, Fang Y, Yang Y, Zhang J, Hu X, Chu L. Astragaloside IV promotes microglia/macrophages M2 polarization and enhances neurogenesis and angiogenesis through PPARγ pathway after cerebral ischemia/reperfusion injury in rats. Int Immunopharmacol 2021; 92:107335. [PMID: 33429332 DOI: 10.1016/j.intimp.2020.107335] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/10/2020] [Accepted: 12/22/2020] [Indexed: 12/16/2022]
Abstract
Microglia/macrophages play a dual role in brain injury and repair following cerebral ischemia/reperfusion. Promoting microglia/macrophage polarization from pro-inflammatory M1 to anti-inflammatory M2 phenotype has been considered as a potential treatment for ischemic stroke. Astragaloside IV (AS-IV) is a primary active ingredient of Chinese herb Radix Astragali, which protects against acute cerebral ischemic/reperfusion injury through its antioxidant, anti-inflammatory, and anti-apoptotic properties. However, it remains unknown whether AS-IV improves ischemic brain tissue repair and its underlying mechanism. A transient middle cerebral artery occlusion (tMCAO) rat model was used in this study. The results showed that AS-IV significantly improved long-term brain injury, reduced the expression of M1 microglia/macrophage markers and increased the expression of M2 microglia/macrophage markers 14 days after cerebral ischemia/reperfusion. AS-IV also increased peroxisome proliferator-activated receptor γ (PPARγ) mRNA and protein expression. Moreover, AS-IV promoted neurogenesis and angiogenesis, and increased the protein expression of brain-derived growth factor (BDNF), insulin-like growth factor-1 (IGF-1) and vascular endothelial growth factor (VEGF). However, these beneficial effects were greatly blocked by PPARγ antagonist T0070907. These results together suggest that AS-IV could enhance neurogenesis, angiogenesis and neurological functional recovery, which may be partially through transforming microglia/macrophage from M1 to M2 phenotype in a PPARγ-dependent manner after cerebral ischemia/reperfusion injury. Therefore, AS-IV can be considered as a promising therapeutic agent for ischemic stroke.
Collapse
Affiliation(s)
- Lin Li
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Haiyan Gan
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Huaqian Jin
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Fang
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Yang
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianping Zhang
- Department of Anatomy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaowei Hu
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lisheng Chu
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|