151
|
Francesca F, Caitlin A, Sarah L, Robyn GL. Antroquinonol administration in animal preclinical studies for Alzheimer's disease (AD): A new avenue for modifying progression of AD pathophysiology. Brain Behav Immun Health 2022; 21:100435. [PMID: 35252893 PMCID: PMC8892093 DOI: 10.1016/j.bbih.2022.100435] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/28/2022] [Accepted: 02/22/2022] [Indexed: 11/28/2022] Open
Abstract
Despite the rise of Alzheimer's disease (AD) in an ageing population, no cure is currently available for this disorder. This study assessed the role of a natural compound, Antroquinonol, in modifying the progression of AD when administered at the start and/or before appearance of symptoms and when the disease was well established, in a transgenic animal model. Antroquinonol was administered daily for 8 weeks, in 11 week (early stage) and 9 month (late stage) male transgenic mice (3 times Transgenic mice PS1M146V, APPSwe, and tauP301L, 3 Tg XAD) and their respective aged controls. Behavioural testing (including Elevated Plus Maze Watermaze, Recognition object testing and Y maze) was performed at the end of the drug administration. In addition AD biomarkers (Amyloid beta 42 (Aβ42), tau and phospho-tau levels), oxidative stress and inflammatory markers, were assessed in tested mice brains after their sacrifice at the end of the treatment. When administered before the start of symptoms at 11 weeks, Antroquinonol treatment at 34 mg/kg (D2) and more consistently at 75 mg/kg (D3), had a significant effect on reducing systemic inflammatory markers (Interleukin 1, IL-1β and TNF-α) and AD biomarker (Amyloid Beta 42, Aβ42 and tau) levels in the brain. The reduction of behavioural impairment reported for 3TgXAD mice was observed significantly for the D3 drug dose only and for all behavioural tests, when administered at 11 weeks. Similarly, beneficial effects of Antroquinonol (at higher dose D3) were noted in the transgenic mice in terms of AD biomarkers (tau and phosphorylated-tau), systemic inflammatory (IL-1β), brain anti-inflammatory (Nrf2) and oxidative (3-Nitrotyrosine, 3NT) markers. Improvement of memory impairment was also reported when Antroquinonol (D3) was administered at late stage (9 months). Since Antroquinonol has been used without adverse effects in previous successful clinical trials, this drug may offer a new avenue of treatment to modify AD development and progression.
Collapse
Affiliation(s)
- Fernandez Francesca
- School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, 1100 Nudgee Rd, Banyo, Queensland, 4014, Australia
- Centre for Genomics and Personalised Medicine, Genomics Research Centre, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, QLD, 4059, Australia
| | - Aust Caitlin
- School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, 1100 Nudgee Rd, Banyo, Queensland, 4014, Australia
- Centre for Genomics and Personalised Medicine, Genomics Research Centre, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, QLD, 4059, Australia
| | - Lye Sarah
- School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, 1100 Nudgee Rd, Banyo, Queensland, 4014, Australia
- Centre for Genomics and Personalised Medicine, Genomics Research Centre, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, QLD, 4059, Australia
| | - Griffiths Lyn Robyn
- Centre for Genomics and Personalised Medicine, Genomics Research Centre, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, QLD, 4059, Australia
- Corresponding author. Centre for Genomics and Personalised Health Genomics Research Centre, Queensland University of Technology, Australia.
| |
Collapse
|
152
|
Zhou R, Qian S, Cho WCS, Zhou J, Jin C, Zhong Y, Wang J, Zhang X, Xu Z, Tian M, Chan LWC, Zhang H. Microbiota-microglia connections in age-related cognition decline. Aging Cell 2022; 21:e13599. [PMID: 35349746 PMCID: PMC9124309 DOI: 10.1111/acel.13599] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/08/2022] [Accepted: 03/13/2022] [Indexed: 12/13/2022] Open
Abstract
Aging is an inevitable process that all individuals experience, of which the extent differs among individuals. It has been recognized as the risk factor of neurodegenerative diseases by affecting gut microbiota compositions, microglia, and cognition abilities. Aging-induced changes in gut microbiota compositions have a critical role in orchestrating the morphology and functions of microglia through the gut-brain axis. Gut microbiota communicates with microglia by its secreted metabolites and neurotransmitters. This is highly associated with age-related cognitive declines. Here, we review the main composition of microbiota in the aged individuals, outline the changes of the brain in age-related cognitive decline from a neuroinflammation perspective, especially the changes of morphology and functions of microglia, discuss the crosstalk between microbiota and microglia in the aged brain and further highlight the role of microbiota-microglia connections in neurodegenerative diseases (Alzheimer's disease and Parkinson's disease).
Collapse
Affiliation(s)
- Rui Zhou
- Department of Nuclear Medicine and Medical PET CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong SARChina
| | - Shufang Qian
- Department of Nuclear Medicine and Medical PET CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - William C. S. Cho
- Department of Clinical OncologyQueen Elizabeth HospitalHong Kong SARChina
| | - Jinyun Zhou
- Department of Nuclear Medicine and Medical PET CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Chentao Jin
- Department of Nuclear Medicine and Medical PET CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Yan Zhong
- Department of Nuclear Medicine and Medical PET CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Jing Wang
- Department of Nuclear Medicine and Medical PET CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Xiaohui Zhang
- Department of Nuclear Medicine and Medical PET CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Zhoujiao Xu
- Department of Nuclear Medicine and Medical PET CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Mei Tian
- Department of Nuclear Medicine and Medical PET CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Lawrence W. C. Chan
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong SARChina
| | - Hong Zhang
- Department of Nuclear Medicine and Medical PET CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Key Laboratory for Biomedical Engineering of Ministry of EducationZhejiang UniversityHangzhouChina
- The College of Biomedical Engineering and Instrument Science of Zhejiang UniversityHangzhouChina
| |
Collapse
|
153
|
Nicotinic Acetylcholine Receptors and Microglia as Therapeutic and Imaging Targets in Alzheimer's Disease. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27092780. [PMID: 35566132 PMCID: PMC9102429 DOI: 10.3390/molecules27092780] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022]
Abstract
Amyloid-β (Aβ) accumulation and tauopathy are considered the pathological hallmarks of Alzheimer’s disease (AD), but attenuation in choline signaling, including decreased nicotinic acetylcholine receptors (nAChRs), is evident in the early phase of AD. Currently, there are no drugs that can suppress the progression of AD due to a limited understanding of AD pathophysiology. For this, diagnostic methods that can assess disease progression non-invasively before the onset of AD symptoms are essential, and it would be valuable to incorporate the concept of neurotheranostics, which simultaneously enables diagnosis and treatment. The neuroprotective pathways activated by nAChRs are attractive targets as these receptors may regulate microglial-mediated neuroinflammation. Microglia exhibit both pro- and anti-inflammatory functions that could be modulated to mitigate AD pathogenesis. Currently, single-cell analysis is identifying microglial subpopulations that may have specific functions in different stages of AD pathologies. Thus, the ability to image nAChRs and microglia in AD according to the stage of the disease in the living brain may lead to the development of new diagnostic and therapeutic methods. In this review, we summarize and discuss the recent findings on the nAChRs and microglia, as well as their methods for live imaging in the context of diagnosis, prophylaxis, and therapy for AD.
Collapse
|
154
|
Chansawhang A, Phochantachinda S, Temviriyanukul P, Chantong B. Corticosterone potentiates ochratoxin A-induced microglial activation. Biomol Concepts 2022; 13:230-241. [DOI: 10.1515/bmc-2022-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/10/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
Microglial activation in the central nervous system (CNS) has been associated with brain damage and neurodegenerative disorders. Ochratoxin A (OTA) is a mycotoxin that occurs naturally in food and feed and has been associated with neurotoxicity, while corticosteroids are CNS’ physiological function modulators. This study examined how OTA affected microglia activation and how corticosteroids influenced microglial neuroinflammation. Murine microglial cells (BV-2) were stimulated by OTA, and the potentiation effects on OTA-induced inflammation were determined by corticosterone pre-treatment. Expressions of pro-inflammatory mediators including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), and inducible nitric oxide synthase (iNOS) were determined. Phosphorylation of mitogen-activated protein kinases (MAPKs) was analyzed by western blotting. OTA significantly increased the mRNA expression of IL-6, TNF-α, IL-1β, and iNOS and also elevated IL-6 and NO levels. Corticosterone pre-treatment enhanced the neuroinflammatory response to OTA in a mineralocorticoid receptor (MR)-dependent mechanism, which is associated with increases in extracellular signal-regulated kinase (ERK) and p38 MAPK activation. In response to OTA, microglial cells produced pro-inflammatory cytokines and NO, while corticosterone increased OTA-induced ERK and p38 MAPK phosphorylation via MR. Findings indicated the direct role of OTA in microglia activation and neuroinflammatory response and suggested that low corticosterone concentrations in the brain exacerbated neurodegeneration.
Collapse
Affiliation(s)
- Anchana Chansawhang
- The Center for Veterinary Diagnosis, Faculty of Veterinary Science, Mahidol University , Salaya , Phutthamonthon, Nakhon Pathom 73170 , Thailand
| | - Sataporn Phochantachinda
- Prasu-Arthorn Animal Hospital, Faculty of Veterinary Science, Mahidol University , Salaya , Phutthamonthon, Nakhon Pathom 73170 , Thailand
| | - Piya Temviriyanukul
- Institute of Nutrition, Mahidol University , Salaya , Phutthamonthon, Nakhon Pathom 73170 , Thailand
| | - Boonrat Chantong
- Department of Pre-clinical and Applied Animal Science, Faculty of Veterinary Science, Mahidol University , Salaya , Phutthamonthon, Nakhon Pathom 73170 , Thailand
| |
Collapse
|
155
|
Lim JY, Lee JE, Park SA, Park SI, Yon JM, Park JA, Jeun SS, Kim SJ, Lee HJ, Kim SW, Yang SH. Protective Effect of Human-Neural-Crest-Derived Nasal Turbinate Stem Cells against Amyloid-β Neurotoxicity through Inhibition of Osteopontin in a Human Cerebral Organoid Model of Alzheimer’s Disease. Cells 2022; 11:cells11061029. [PMID: 35326480 PMCID: PMC8947560 DOI: 10.3390/cells11061029] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 11/16/2022] Open
Abstract
The aim of this study was to validate the use of human brain organoids (hBOs) to investigate the therapeutic potential and mechanism of human-neural-crest-derived nasal turbinate stem cells (hNTSCs) in models of Alzheimer’s disease (AD). We generated hBOs from human induced pluripotent stem cells, investigated their characteristics according to neuronal markers and electrophysiological features, and then evaluated the protective effect of hNTSCs against amyloid-β peptide (Aβ1–42) neurotoxic activity in vitro in hBOs and in vivo in a mouse model of AD. Treatment of hBOs with Aβ1–42 induced neuronal cell death concomitant with decreased expression of neuronal markers, which was suppressed by hNTSCs cocultured under Aβ1–42 exposure. Cytokine array showed a significantly decreased level of osteopontin (OPN) in hBOs with hNTSC coculture compared with hBOs only in the presence of Aβ1–42. Silencing OPN via siRNA suppressed Aβ-induced neuronal cell death in cell culture. Notably, compared with PBS, hNTSC transplantation significantly enhanced performance on the Morris water maze, with reduced levels of OPN after transplantation in a mouse model of AD. These findings reveal that hBO models are useful to evaluate the therapeutic effect and mechanism of stem cells for application in treating AD.
Collapse
Affiliation(s)
- Jung Yeon Lim
- Department of Otolaryngology-Head and Neck Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Jung Eun Lee
- Department of Neurosurgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon 16247, Kyonggi-do, Korea
| | - Soon A Park
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Sang In Park
- Institute of Catholic Integrative Medicine (ICIM), Incheon St. Mary's Hospital, The Catholic University of Korea, Seoul 06591, Korea
| | - Jung-Min Yon
- Department of Otolaryngology-Head and Neck Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Jeong-Ah Park
- Department of Otolaryngology-Head and Neck Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Sin-Soo Jeun
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Seung Joon Kim
- Division of Pulmonology, Critical Care and Allergy, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Hong Jun Lee
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju 28644, Korea
| | - Sung Won Kim
- Department of Otolaryngology-Head and Neck Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Seung Ho Yang
- Department of Neurosurgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon 16247, Kyonggi-do, Korea
| |
Collapse
|
156
|
Xu Y, Zhao H, Wang Z, Gao H, Liu J, Li K, Song Z, Yuan C, Lan X, Pan C, Zhang S. Developmental exposure to environmental levels of cadmium induces neurotoxicity and activates microglia in zebrafish larvae: From the perspectives of neurobehavior and neuroimaging. CHEMOSPHERE 2022; 291:132802. [PMID: 34752834 DOI: 10.1016/j.chemosphere.2021.132802] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/15/2021] [Accepted: 11/04/2021] [Indexed: 06/13/2023]
Abstract
Cadmium (Cd) is a worldwide environmental pollutant that postures serious threats to humans and ecosystems. Over the years, its adverse effects on the central nervous system (CNS) have been concerned, whereas the underlying cellular/molecular mechanisms remain unclear. In this study, taking advantages of zebrafish model in high-throughput imaging and behavioral tests, we have explored the potential developmental neurotoxicity of Cd at environmentally relevant levels, from the perspectives of neurobehavior and neuroimaging. Briefly, Cd2+ exposure resulted in a general impairment of zebrafish early development. Zebrafish neurobehavioral patterns including locomotion and reactivity to environmental signals were significantly perturbed upon Cd2+ exposure. Importantly, a combination of in vivo two-photon neuroimaging, flow cytometry and gene expression analyses revealed notable neurodevelopmental disorders as well as neuroimmune responses induced by Cd2+ exposure. Both cell-cycle arrest and apoptosis contributed jointly to a significant decrease of neuronal density in zebrafish larvae exposed to Cd2+. The dramatic morphological alterations of microglia from multi-branched to amoeboid, the microgliosis, as well as the modulation of gene expression profiles demonstrated a strong activation of microglia and neuroinflammation triggered by environmental levels of Cd2+. Together, our study points to the developmental toxicity of Cd in inducing CNS impairment and neuroinflammation thereby providing visualized etiological evidence of this heavy metal induced neurodevelopmental disorders. It's tempting to speculate that this research model might represent a promising tool not only for understanding the molecular mechanisms of Cd-induced neurotoxicity, but also for developing pharmacotherapies to mitigate the neurological damage resulting from exposure to Cd, and other neurotoxicants.
Collapse
Affiliation(s)
- Yanyi Xu
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, China
| | - Haiyu Zhao
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, China.
| | - Zuo Wang
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, China
| | - Hao Gao
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, China
| | - Junru Liu
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, China
| | - Kemin Li
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, China
| | - Zan Song
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, China
| | - Cong Yuan
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, China
| | - Xianyong Lan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, 712100, Shaanxi Province, China
| | - Chuanying Pan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, 712100, Shaanxi Province, China
| | - Shengxiang Zhang
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
157
|
Hampe CS, Mitoma H. A Breakdown of Immune Tolerance in the Cerebellum. Brain Sci 2022; 12:brainsci12030328. [PMID: 35326284 PMCID: PMC8946792 DOI: 10.3390/brainsci12030328] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 11/21/2022] Open
Abstract
Cerebellar dysfunction can be associated with ataxia, dysarthria, dysmetria, nystagmus and cognitive deficits. While cerebellar dysfunction can be caused by vascular, traumatic, metabolic, genetic, inflammatory, infectious, and neoplastic events, the cerebellum is also a frequent target of autoimmune attacks. The underlying cause for this vulnerability is unclear, but it may be a result of region-specific differences in blood–brain barrier permeability, the high concentration of neurons in the cerebellum and the presence of autoantigens on Purkinje cells. An autoimmune response targeting the cerebellum—or any structure in the CNS—is typically accompanied by an influx of peripheral immune cells to the brain. Under healthy conditions, the brain is protected from the periphery by the blood–brain barrier, blood–CSF barrier, and blood–leptomeningeal barrier. Entry of immune cells to the brain for immune surveillance occurs only at the blood-CSF barrier and is strictly controlled. A breakdown in the barrier permeability allows peripheral immune cells uncontrolled access to the CNS. Often—particularly in infectious diseases—the autoimmune response develops because of molecular mimicry between the trigger and a host protein. In this review, we discuss the immune surveillance of the CNS in health and disease and also discuss specific examples of autoimmunity affecting the cerebellum.
Collapse
Affiliation(s)
- Christiane S. Hampe
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Correspondence: ; Tel.: +1-206-554-9181
| | - Hiroshi Mitoma
- Department of Medical Education, Tokyo Medical University, Tokyo 160-0023, Japan;
| |
Collapse
|
158
|
Mabrouk R. Principal Component Analysis versus Subject’s Residual Profile Analysis for Neuroinflammation Investigation in Parkinson Patients: A PET Brain Imaging Study. J Imaging 2022; 8:jimaging8030056. [PMID: 35324611 PMCID: PMC8954189 DOI: 10.3390/jimaging8030056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 12/10/2022] Open
Abstract
Dysfunction of neurons in the central nervous system is the primary pathological feature of Parkinson’s disease (PD). Despite different triggering, emerging evidence indicates that neuroinflammation revealed through microglia activation is critical for PD. Moreover, recent investigations sought a potential relationship between Lrrk2 genetic mutation and microglia activation. In this paper, neuroinflammation in sporadic PD, Lrrk2-PD and unaffected Lrrk2 mutation carriers were investigated. The principal component analysis (PCA) and the subject’s residual profile (SRP) techniques were performed on multiple groups and regions of interest in 22 brain-regions. The 11C-PBR28 binding profiles were compared in four genotypes depending on groups, i.e., HC, sPD, Lrrk2-PD and UC, using the PCA and SPR scores. The genotype effect was found as a principal feature of group-dependent 11C-PBR28 binding, and preliminary evidence of a MAB-Lrrk2 mutation interaction in manifest Parkinson’s and subjects at risk was found.
Collapse
Affiliation(s)
- Rostom Mabrouk
- Department of Computer Science, Bishop's University, Sherbrooke, QC J1M 1Z7, Canada
| |
Collapse
|
159
|
Lee H, Park JH, Kim H, Woo SK, Choi JY, Lee KH, Choe YS. Synthesis and Evaluation of a 18F-Labeled Ligand for PET Imaging of Colony-Stimulating Factor 1 Receptor. Pharmaceuticals (Basel) 2022; 15:ph15030276. [PMID: 35337075 PMCID: PMC8954204 DOI: 10.3390/ph15030276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 02/01/2023] Open
Abstract
Neuroinflammation involves activation of glial cells in the brain, and activated microglia play a particularly important role in neurodegenerative diseases such as Alzheimer’s disease (AD). In this study, we developed 5-cyano-N-(4-(4-(2-[18F]fluoroethyl)piperazin-1-yl)-2-(piperidin-1-yl)phenyl)furan-2-carboxamide ([18F]1) for PET imaging of colony-stimulating factor 1 receptor (CSF1R), an emerging target for neuroinflammation imaging. Non-radioactive ligand 1 exhibited binding affinity comparable to that of a known CSF1R inhibitor, 5-cyano-N-(4-(4-methylpiperazin-1-yl)-2-(piperidin-1-yl)phenyl)furan-2-carboxamide (CPPC). Therefore, we synthesized radioligand [18F]1 by radiofluorination of chlorine-substituted precursor 7 in 13–15% decay-corrected radiochemical yield. Dynamic PET/CT images showed higher uptake in the lipopolysaccharide (LPS)-treated mouse brain than in control mouse brain. Ex vivo biodistribution study conducted at 45 min after radioligand injection showed that the brain uptake in LPS mice increased by 78% compared to that of control mice and was inhibited by 22% in LPS mice pretreated with CPPC, indicating specificity of [18F]1 for CSF1R. A metabolism study demonstrated that the radioligand underwent little metabolism in the mouse brain. Taken together, these results suggest that [18F]1 may hold promise as a radioligand for CSF1R imaging.
Collapse
Affiliation(s)
- Hyeokjin Lee
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (H.L.); (J.-H.P.); (H.K.); (J.Y.C.); (K.-H.L.)
| | - Ji-Hun Park
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (H.L.); (J.-H.P.); (H.K.); (J.Y.C.); (K.-H.L.)
| | - Hyunjung Kim
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (H.L.); (J.-H.P.); (H.K.); (J.Y.C.); (K.-H.L.)
| | - Sang-keun Woo
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea;
| | - Joon Young Choi
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (H.L.); (J.-H.P.); (H.K.); (J.Y.C.); (K.-H.L.)
| | - Kyung-Han Lee
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (H.L.); (J.-H.P.); (H.K.); (J.Y.C.); (K.-H.L.)
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06355, Korea
| | - Yearn Seong Choe
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (H.L.); (J.-H.P.); (H.K.); (J.Y.C.); (K.-H.L.)
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06355, Korea
- Correspondence:
| |
Collapse
|
160
|
Liu L, Zhang X, Wang C, Wu X, Long B. Hypercholesterolemia aggravates sevoflurane-induced cognitive impairment in aged rats by inducing neurological inflammation and apoptosis. J Biochem Mol Toxicol 2022; 36:e23009. [PMID: 35174938 DOI: 10.1002/jbt.23009] [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] [Received: 09/24/2021] [Revised: 12/28/2021] [Accepted: 01/04/2022] [Indexed: 12/30/2022]
Abstract
We aimed to explore the effects of hypercholesterolemia on sevoflurane-induced cognitive impairment in aged rats and the underlying mechanism(s). Aged rats were administrated with high-fat diet, sevoflurane, or both. Thereafter, the plasma levels of total cholesterol (TC), triglyceride (TG), low-density lipoprotein (LDL), and high-density lipoprotein (HDL) were evaluated. The Morris water maze task was performed to evaluate the spatial learning and memory ability of rats. Moreover, Nissl and Evans blue staining were conducted to test nerve damage and detect the blood-brain barrier permeability, respectively. The percentage of apoptotic cells was evaluated by terminal deoxynucleotidyl transferase dUTP nick end labeling assay. The messenger RNA expression of inflammatory factors and protein expression of microglial activation markers and apoptosis-related proteins were tested by real-time polymerase chain reaction, enzyme-linked immunosorbent assay, or western blot analysis, respectively. High-fat diet induced high levels of TC, TG, and LDL but decreased levels of HDL. However, sevoflurane had no effects on these levels. In contrast, sevoflurane significantly induced the impairment of learning and memory, nerve damage, neuroinflammatory damage, and neuronal apoptosis. Hypercholesterolemia exacerbated the sevoflurane-induced impairment in aged rats. These results suggested that hypercholesterolemia aggravates sevoflurane-induced cognitive impairment in aged rats, possibly by inducing neurological inflammation and apoptosis.
Collapse
Affiliation(s)
- Lidan Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xin Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Cong Wang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiuying Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Bo Long
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
161
|
de Guzman AE, Ahmed M, Perrier S, Hammill C, Li YQ, Wong CS, Nieman BJ. Protection from radiation-induced neuroanatomical deficits by CCL2-deficiency is dependent on sex. Int J Radiat Oncol Biol Phys 2022; 113:390-400. [PMID: 35143888 DOI: 10.1016/j.ijrobp.2022.01.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/19/2022] [Accepted: 01/23/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE Cranial radiation therapy for the treatment of paediatric brain tumours results in changes to brain development that are detectable with magnetic resonance imaging (MRI). We have previously demonstrated similar structural changes in both humans and mice. The goal of the current study was to examine the role of inflammation in this response. Since neuroanatomical volume deficits in paediatric survivors are more pronounced in females, we also evaluated possible dependence on sex. EXPERIMENTAL DESIGN Male mice deficient in the C-C chemokine ligand 2 gene (Ccl2; previously Mcp-1) have been shown by others to have a muted neuroinflammatory response after irradiation. We irradiated Ccl2-/- (HOM; females[f]=12, males[m]=13), Ccl2+/- (HET; f=13, m=16), and Ccl2+/+ (WT; f=11, m=13) mice with a whole brain dose of 7 Gy during infancy. Control mice (with approximately equal groups sizes) were anaesthetized but not irradiated. In vivo MR images were acquired at 4 time points up to 3 months following irradiation, and deformation-based morphometry was used to identify volume differences. RESULTS Irradiation of WT mice resulted in a deficit in neuroanatomical growth with limited sex dependence. HOM and HET males were significantly protected from this radiation-induced damage, while HOM and HET females were not. We conclude that interventions aimed at mitigating the effects of cranial radiation therapy in paediatric cancer survivors by modulating inflammatory response will need to consider patient sex.
Collapse
Affiliation(s)
- A Elizabeth de Guzman
- Mouse Imaging Centre, Hospital for Sick Children, 25 Orde Street, Toronto, Ontario, M5T 3H7, Canada; Translational Medicine, Hospital for Sick Children, 555 University Ave, Toronto, Ontario, M5G 1X8, Canada; Department of Medical Biophysics, University of Toronto, 610 University Avenue, Rm 7-411, Toronto, Ontario, M5G 2M9, Canada
| | - Mashal Ahmed
- Mouse Imaging Centre, Hospital for Sick Children, 25 Orde Street, Toronto, Ontario, M5T 3H7, Canada; Translational Medicine, Hospital for Sick Children, 555 University Ave, Toronto, Ontario, M5G 1X8, Canada
| | - Stefanie Perrier
- Mouse Imaging Centre, Hospital for Sick Children, 25 Orde Street, Toronto, Ontario, M5T 3H7, Canada; Translational Medicine, Hospital for Sick Children, 555 University Ave, Toronto, Ontario, M5G 1X8, Canada
| | - Christopher Hammill
- Mouse Imaging Centre, Hospital for Sick Children, 25 Orde Street, Toronto, Ontario, M5T 3H7, Canada
| | - Yu-Qing Li
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Odette Cancer Centre, 2075 Bayview Avenue, Toronto, Ontario, M4N 3M5, Canada
| | - C Shun Wong
- Department of Medical Biophysics, University of Toronto, 610 University Avenue, Rm 7-411, Toronto, Ontario, M5G 2M9, Canada; Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Odette Cancer Centre, 2075 Bayview Avenue, Toronto, Ontario, M4N 3M5, Canada; Department of Radiation Oncology, University of Toronto, 149 College Street - Stewart Building, Suite 504, Toronto, Ontario, M5T 1P5, Canada
| | - Brian J Nieman
- Mouse Imaging Centre, Hospital for Sick Children, 25 Orde Street, Toronto, Ontario, M5T 3H7, Canada; Translational Medicine, Hospital for Sick Children, 555 University Ave, Toronto, Ontario, M5G 1X8, Canada; Department of Medical Biophysics, University of Toronto, 610 University Avenue, Rm 7-411, Toronto, Ontario, M5G 2M9, Canada; Ontario Institute for Cancer Research, Toronto, Ontario, Canada.
| |
Collapse
|
162
|
Kasen A, Houck C, Burmeister AR, Sha Q, Brundin L, Brundin P. Upregulation of α-synuclein following immune activation: Possible trigger of Parkinson's disease. Neurobiol Dis 2022; 166:105654. [DOI: 10.1016/j.nbd.2022.105654] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/31/2022] [Accepted: 02/03/2022] [Indexed: 12/20/2022] Open
|
163
|
4R-cembranoid protects neuronal cells from oxygen-glucose deprivation by modulating microglial cell activation. Brain Res Bull 2022; 179:74-82. [PMID: 34942325 PMCID: PMC8849140 DOI: 10.1016/j.brainresbull.2021.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 02/03/2023]
Abstract
As major immune responsive cells in the central nervous system (CNS), activated microglia can present pro-inflammatory M1 phenotype aggravating the neuronal injury or anti-inflammatory M2 phenotype providing neuroprotection and promoting neuronal survival in neurodegenerative diseases. In this study, we demonstrated that a compound, 4R-cembranoid (4R, 1S, 2E, 4R, 6R,-7E, 11E-2, 7, 11-cembratriene-4, 6-diol cembranoids) promoted M2 phenotype while attenuated M1 phenotype in N9 cells, a microglial cell line. Following Lipopolysaccharides (LPS) or Oxygen-glucose deprivation (OGD) treatment, the N9 cells treated by 1 µM 4R showed an increased Arginase-1 (Arg1, a M2 marker) expression and a reduced inducible nitric oxide synthase (iNOS, M1 marker) expression. In addition, the conditioned medium of 4R-treated post-OGD N9 cells protected neuro2a cells, a neuronal cell line, from OGD-induced injury. The viability of neuro2a cells in OGD condition was increased by 54.5% after treated with the conditioned medium of 4R-treated post-OGD N9 cells. Furthermore, we demonstrated the protective mechanism of 4R was associated with a decreased TNF-α release and an increased IL-10 release from N9 cells. In conclusion, our study demonstrated that the neuroprotective effects of 4R were through the regulation of microglial activation by promoting the protective M2 activation and inhibiting the damaging M1 activation. Therefore, the findings of this study suggest that 4R could be a promising lead structure for the development of drugs for the treatment of ischemic stroke and other neurodegenerative diseases with an inflammatory component involved.
Collapse
|
164
|
Alam Q, Krishnamurthy S. Dihydroquercetin ameliorates LPS-induced neuroinflammation and memory deficit. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 3:100091. [PMID: 35243333 PMCID: PMC8857648 DOI: 10.1016/j.crphar.2022.100091] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 11/23/2022] Open
Abstract
Dihydroquercetin (DHQ) is a pentahydroxyflavanone that has been used as an important suppliment against oxidative stress related inflammation and neuroinflammation. Neuroinflammation, which is the activation of the defense mechanism of the central nervous system, upon exposure to stimuli like amyloid β, Lewy bodies, lipopolysaccharide (LPS) and reactive oxygen species. It is an important pathophysiological mediator of a number of neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, multiple sclerosis and others. The objective of the present study is to evaluate the neuroprotective effect of DHQ, a potent antioxidant molecule, against LPS induced neuroinflammation. On the first day of the experiment (day-1), neuroinflammation was induced through intracerebroventricular injection of LPS (5 μg/5 μl) into each lateral ventricle in the rats. DHQ-0.5, 1 and 2 μg/kg was injected into the tail vein in respective groups from day-2 to day-10. Behavioral studies showed that DHQ attenuated the LPS-induced loss in long-term memory and working memory as evaluated by elevated plus maze and Y-maze test, respectively. Further, the biochemical estimations revealed that DHQ dose-dependently attenuated the LPS-induced decrease in acetylcholine level and increased in the acetylcholine-esterase activity in the hippocampal region. DHQ also increased the catalase activity and decreased nitric oxide and lipid peroxidation altered by LPS injection. DHQ also attenuated interleukin-6 in the brain, which has elevated upon LPS induction. The decrease in IL-6 is attributed to its antioxidant activity. Hence, DHQ could be a potential therapeutic candidate in the management of neuroinflammation and related neurodegenerative disorders. Dihydroquercetin (DHQ) improves LPS induced loss in working memory and long-term memory. DHQ attenuates LPS –induced decrease in cholinergic activity. DHQ shows anti-oxidant properties. DHQ shows anti-neuroinflammatory effect.
Collapse
|
165
|
Anti-neuroinflammatory effect of oxaline, isorhodoptilometrin, and 5-hydroxy-7-(2′-hydroxypropyl)-2-methyl-chromone obtained from the marine fungal strain Penicillium oxalicum CLC-MF05. Arch Pharm Res 2022; 45:90-104. [DOI: 10.1007/s12272-022-01370-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 01/24/2022] [Indexed: 11/02/2022]
|
166
|
Howe AM, Burke S, O'Reilly ME, McGillicuddy FC, Costello DA. Palmitic Acid and Oleic Acid Differently Modulate TLR2-Mediated Inflammatory Responses in Microglia and Macrophages. Mol Neurobiol 2022; 59:2348-2362. [PMID: 35079937 PMCID: PMC9016023 DOI: 10.1007/s12035-022-02756-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/17/2022] [Indexed: 01/09/2023]
Abstract
The relationship between systemic immunity and neuroinflammation is widely recognised. Infiltration of peripheral immune cells to the CNS during certain chronic inflammatory states contributes significantly to neuropathology. Obesity and its co-morbidities are primary risk factors for neuroinflammatory and neurodegenerative conditions, including Alzheimer’s disease (AD). Dietary fats are among the most proinflammatory components of the obesogenic diet and play a prominent role in the low-grade systemic inflammation associated with the obese state. Saturated fatty acid (SFA) is largely implicated in the negative consequences of obesity, while the health benefits of monounsaturated fatty acid (MUFA) are widely acknowledged. The current study sought to explore whether SFA and MUFA differently modulate inflammatory responses in the brain, compared with peripheral immune cells. Moreover, we assessed the neuroinflammatory impact of high-fat-induced obesity and hypothesised that a MUFA-rich diet might mitigate inflammation despite obesogenic conditions. Toll-like receptor (TLR)2 mediates the inflammation associated with both obesity and AD. Using the TLR2 agonist lipoteichoic acid (LTA), we report that pre-exposure to either palmitic acid (PA) or oleic acid (OA) attenuated cytokine secretion from microglia, but heightened sensitivity to nitric oxide (NO) production. The reduction in cytokine secretion was mirrored in LTA-stimulated macrophages following exposure to PA only, while effects on NO were restricted to OA, highlighting important cell-specific differences. An obesogenic diet over 12 weeks did not induce prominent inflammatory changes in either cortex or hippocampus, irrespective of fat composition. However, we reveal a clear disparity in the effects of MUFA under obesogenic and non-obesogenic conditions.
Collapse
Affiliation(s)
- Anne-Marie Howe
- UCD School of Biomolecular & Biomedical Science, University College Dublin, Dublin 4, Ireland
- UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Sinéad Burke
- UCD School of Biomolecular & Biomedical Science, University College Dublin, Dublin 4, Ireland
- UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Marcella E O'Reilly
- UCD Conway Institute, University College Dublin, Dublin 4, Ireland
- UCD School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Fiona C McGillicuddy
- UCD Conway Institute, University College Dublin, Dublin 4, Ireland
- UCD School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Derek A Costello
- UCD School of Biomolecular & Biomedical Science, University College Dublin, Dublin 4, Ireland.
- UCD Conway Institute, University College Dublin, Dublin 4, Ireland.
| |
Collapse
|
167
|
Lim JS, Oh J, Yun HS, Lee JS, Hahn D, Kim JS. Anti-neuroinflammatory activity of 6,7-dihydroxy-2,4-dimethoxy phenanthrene isolated from Dioscorea batatas Decne partly through suppressing the p38 MAPK/NF-κB pathway in BV2 microglial cells. JOURNAL OF ETHNOPHARMACOLOGY 2022; 282:114633. [PMID: 34520827 DOI: 10.1016/j.jep.2021.114633] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The rhizome of Dioscorea batatas Decne (called Chinses yam) widely distributed in East Asian countries including China, Japan, Korea and Taiwan has long been used in oriental folk medicine owing to its tonic, antitussive, expectorant and anti-ulcerative effects. It has been reported to have anti-inflammatory, antioxidative, cholesterol-lowering, anticholinesterase, growth hormone-releasing, antifungal and immune cell-stimulating activities. AIM OF THE STUDY Neuroinflammation caused by activated microglia contributes to neuronal dysfunction and neurodegeneration. In the present study, the anti-neuroinflammatory activity of 6,7-dihydroxy-2,4-dimethoxy phenanthrene (DHDMP), a phenanthrene compound isolated from Dioscorea batatas Decne, was examined in microglial and neuronal cells. MATERIALS AND METHODS A natural phenanthrene compound, DHDMP, was isolated from the peel of Dioscorea batatas Decne. The anti-neuroinflammatory capability of the compound was examined using the co-culture system of BV2 murine microglial and HT22 murine neuronal cell lines. The expression levels of inflammatory mediators and cytoprotective proteins in the cells were quantified by enzyme-linked immunosorbent assay and Western blot analysis. RESULTS DHDMP at the concentrations of ≤1 μg/mL did not exhibit a cytotoxic effect for BV2 and HT22 cells. Rather DHDMP effectively restored the growth rate of HT22 cells, which was reduced by co-culture with lipopolysaccharide (LPS)-treated BV2 cells. DHDMP significantly decreased the production of proinflammatory mediators, such as nitric oxide, tumor necrosis factor-α, interleukin-6, inducible nitric oxide synthase, and cyclooxygenase-2 in BV2 cells. Moreover, DHDMP strongly inhibited the nuclear translocation of nuclear factor κB (NF-κB) and phosphorylation of p38 mitogen-activated protein kinase (MAPK) in BV2 cells. The compound did not affect the levels and phosphorylation of ERK and JNK. Concurrently, DHDMP increased the expression of heme oxygenase-1 (HO-1), an inducible cytoprotective enzyme, in HT22 cells. CONCLUSIONS Our findings indicate that DHDMP effectively dampened LPS-mediated inflammatory responses in BV2 microglial cells by suppressing transcriptional activity of NF-κB and its downstream mediators and contributed to HT22 neuronal cell survival. This study provides insight into the therapeutic potential of DHDMP for inflammation-related neurological diseases.
Collapse
Affiliation(s)
- Ji Sun Lim
- Institute of Agricultural Science and Technology, Kyungpook National University, Daegu, 41566, South Korea.
| | - Jisun Oh
- Institute of Agricultural Science and Technology, Kyungpook National University, Daegu, 41566, South Korea.
| | - Hyun Seok Yun
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, 41566, South Korea.
| | - Jeong Soon Lee
- Forest Resources Development Institute of Gyeongsangbuk-do, Andong, 36605, South Korea.
| | - Dongyup Hahn
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, 41566, South Korea.
| | - Jong-Sang Kim
- Institute of Agricultural Science and Technology, Kyungpook National University, Daegu, 41566, South Korea; School of Food Science and Biotechnology, Kyungpook National University, Daegu, 41566, South Korea.
| |
Collapse
|
168
|
The Effects of Modified Curcumin Preparations on Glial Morphology in Aging and Neuroinflammation. Neurochem Res 2022; 47:813-824. [PMID: 34988899 DOI: 10.1007/s11064-021-03499-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 11/26/2021] [Accepted: 11/27/2021] [Indexed: 12/14/2022]
Abstract
Neuroinflammation is characterized by reactive microglia and astrocytes (collectively called gliosis) in the central nervous system and is considered as one of the main pathological hallmarks in different neurodegenerative diseases such as Alzheimer's disease, age-related dementia, and multiple sclerosis. Upon activation, glia undergoes structural and morphological changes such as the microglial cells swell in size and astrocytes become bushy, which play both beneficial and detrimental roles. Hence, they are unable to perform the normal physiological role in brain immunity. Curcumin, a cytokine suppressive anti-inflammatory drug, has a high proven pre-clinical potency and efficacy to reverse chronic neuroinflammation by attenuating the activation and morphological changes that occur in the microglia and astrocytes. This review will highlight the recent findings on the tree structure changes of microglia and astrocytes in neuroinflammation and the effects of curcumin against the activation and morphology of glial cells.
Collapse
|
169
|
Craig CF, Filippone RT, Stavely R, Bornstein JC, Apostolopoulos V, Nurgali K. Neuroinflammation as an etiological trigger for depression comorbid with inflammatory bowel disease. J Neuroinflammation 2022; 19:4. [PMID: 34983592 PMCID: PMC8729103 DOI: 10.1186/s12974-021-02354-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/14/2021] [Indexed: 02/06/2023] Open
Abstract
Patients with inflammatory bowel disease (IBD) suffer from depression at higher rates than the general population. An etiological trigger of depressive symptoms is theorised to be inflammation within the central nervous system. It is believed that heightened intestinal inflammation and dysfunction of the enteric nervous system (ENS) contribute to impaired intestinal permeability, which facilitates the translocation of intestinal enterotoxins into the blood circulation. Consequently, these may compromise the immunological and physiological functioning of distant non-intestinal tissues such as the brain. In vivo models of colitis provide evidence of increased blood–brain barrier permeability and enhanced central nervous system (CNS) immune activity triggered by intestinal enterotoxins and blood-borne inflammatory mediators. Understanding the immunological, physiological, and structural changes associated with IBD and neuroinflammation may aid in the development of more tailored and suitable pharmaceutical treatment for IBD-associated depression.
Collapse
Affiliation(s)
- Colin F Craig
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia
| | - Rhiannon T Filippone
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia
| | - Rhian Stavely
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia.,Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Joel C Bornstein
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Australia
| | - Vasso Apostolopoulos
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia.,Immunology Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia
| | - Kulmira Nurgali
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia. .,Department of Medicine Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia. .,Regenerative Medicine and Stem Cells Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia. .,Institute for Health and Sport, Victoria University, Level 4 Research Labs, Western Centre for Health Research and Education, Sunshine Hospital, 176 Furlong Road, St Albans, VIC, 3021, Australia.
| |
Collapse
|
170
|
Abdelhamid M, Zhou C, Ohno K, Kuhara T, Taslima F, Abdullah M, Jung CG, Michikawa M. Probiotic Bifidobacterium breve Prevents Memory Impairment Through the Reduction of Both Amyloid-β Production and Microglia Activation in APP Knock-In Mouse. J Alzheimers Dis 2022; 85:1555-1571. [PMID: 34958017 PMCID: PMC8925106 DOI: 10.3233/jad-215025] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Probiotic supplementation reestablishes microbiome diversity and improves brain function in Alzheimer's disease (AD); their molecular mechanisms, however, have not yet been fully illustrated. OBJECTIVE We investigated the effects of orally supplemented Bifidobacterium breve MCC1274 on cognitive function and AD-like pathologies in AppNL-G-F mice. METHODS Three-month-old AppNL-G-F mice were orally supplemented with B. breve MCC1274 for four months. The short-term memory function was evaluated using a novel object recognition test. Amyloid plaques, amyloid-β (Aβ) levels, Aβ fibril, amyloid-β protein precursor and its processing enzymes, its metabolic products, glial activity, and cell proliferation in the subgranular zone of the dentate gyrus were evaluated by immunohistochemistry, Aβ ELISA, western blotting, and immunofluorescence staining. The mRNA expression levels of pro- and anti-inflammatory cytokines were determined by qRT-PCR analysis. RESULTS We found that the oral B. breve MCC1 274 supplementation prevented memory impairment in AppNL-G-F mice and decreased hippocampal Aβ levels through the enhancement of the a-disintegrin and metalloproteinase 10 (ADAM10) level. Moreover, administration of the probiotic activated the ERK/HIF-1α signaling pathway responsible for increasing the ADAM10 level and also attenuated microglial activation, which in turn led to reduction in the mRNA expression levels of pro-inflammatory cytokines in the brain. In addition, B. breve MCC1274 supplementation increased the level of synaptic proteins in the hippocampus. CONCLUSION Our findings support the possibility that oral B. breve MCC1274 supplementation might be used as a potential preventive therapy for AD progression.
Collapse
Affiliation(s)
- Mona Abdelhamid
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Chunyu Zhou
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Kazuya Ohno
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Tetsuya Kuhara
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Ferdous Taslima
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Mohammad Abdullah
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan.,Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Cha-Gyun Jung
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Makoto Michikawa
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
171
|
Inflammation Subtypes and Translating Inflammation-Related Genetic Findings in Schizophrenia and Related Psychoses: A Perspective on Pathways for Treatment Stratification and Novel Therapies. Harv Rev Psychiatry 2022; 30:59-70. [PMID: 34995036 PMCID: PMC8746916 DOI: 10.1097/hrp.0000000000000321] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dysregulation of immunological and inflammatory processes is frequently observed in psychotic disorders. Numerous studies have examined the complex components of innate and adaptive immune processes in schizophrenia and related psychoses. Elevated inflammation in these conditions is related to neurobiological phenotypes and associated with both genetics and environmental exposures. Recent studies have utilized multivariate cytokine approaches to identify what appears to be a subset of individuals with elevated inflammation. The degree to which these findings represent a general process of dysregulated inflammation or whether there are more refined subtypes remains unclear. Brain-imaging studies have attempted to establish the link between peripheral inflammation and gray matter disruption, white matter abnormalities, and neuropsychological phenotypes. However, the interplay between peripheral inflammation and neuroinflammation, as well as the consequences of this interplay, in the context of psychosis remains unclear and requires further investigation. This Perspectives article reviews the following elements of immune dysregulation and its clinical and therapeutic implications: (1) evidence supporting inflammation and immune dysregulation in schizophrenia and related psychoses; (2) recent advances in approaches to characterizing subgroups of patients with elevated inflammation; (3) relationships between peripheral inflammation and brain-imaging indicators of neuroinflammation; (4) convergence of large-scale genetic findings and peripheral inflammation findings; and (5) therapeutic implications: anti-inflammation interventions leveraging genetic findings for drug discovery and repurposing. We offer perspectives and examples of how multiomics technologies may be useful for constructing and studying immunogenetic signatures. Advancing research in this area will facilitate biomarker discovery, disease subtyping, and the development of etiological treatments for immune dysregulation in psychosis.
Collapse
|
172
|
Kaur G, Singh NK. The Role of Inflammation in Retinal Neurodegeneration and Degenerative Diseases. Int J Mol Sci 2021; 23:ijms23010386. [PMID: 35008812 PMCID: PMC8745623 DOI: 10.3390/ijms23010386] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 12/19/2022] Open
Abstract
Retinal neurodegeneration is predominantly reported as the apoptosis or impaired function of the photoreceptors. Retinal degeneration is a major causative factor of irreversible vision loss leading to blindness. In recent years, retinal degenerative diseases have been investigated and many genes and genetic defects have been elucidated by many of the causative factors. An enormous amount of research has been performed to determine the pathogenesis of retinal degenerative conditions and to formulate the treatment modalities that are the critical requirements in this current scenario. Encouraging results have been obtained using gene therapy. We provide a narrative review of the various studies performed to date on the role of inflammation in human retinal degenerative diseases such as age-related macular degeneration, inherited retinal dystrophies, retinitis pigmentosa, Stargardt macular dystrophy, and Leber congenital amaurosis. In addition, we have highlighted the pivotal role of various inflammatory mechanisms in the progress of retinal degeneration. This review also offers an assessment of various therapeutic approaches, including gene-therapies and stem-cell-based therapies, for degenerative retinal diseases.
Collapse
Affiliation(s)
- Geetika Kaur
- Integrative Biosciences Center, Wayne State University, Detroit, MI 48202, USA;
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Nikhlesh K. Singh
- Integrative Biosciences Center, Wayne State University, Detroit, MI 48202, USA;
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202, USA
- Correspondence:
| |
Collapse
|
173
|
Khatmi A, Eskandarian Boroujeni M, Ezi S, Hamidreza Mirbehbahani S, Aghajanpour F, Soltani R, Hossein Meftahi G, Abdollahifar MA, Hassani Moghaddam M, Toreyhi H, Khodagholi F, Aliaghaei A. Combined molecular, structural and memory data unravel the destructive effect of tramadol on hippocampus. Neurosci Lett 2021; 771:136418. [PMID: 34954113 DOI: 10.1016/j.neulet.2021.136418] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/20/2021] [Accepted: 12/20/2021] [Indexed: 11/28/2022]
Abstract
Tramadol is a synthetic analogue of codeine and stimulates neurodegeneration in several parts of the brain that leads to various behavioral impairments. Despite the leading role of hippocampus in learning and memory as well as decreased function of them under influence of tramadol, there are few studies analyzing the effect of tramadol administration on gene expression profiling and structural consequences in hippocampus region. Thus, we sought to determine the effect of tramadol on both PC12 cell line and hippocampal tissue, from gene expression changes to structural alterations. In this respect, we investigated genome-wide mRNA expression using high throughput RNA-seq technology and confirmatory quantitative real-time PCR, accompanied by stereological analysis of hippocampus and behavioral assessment following tramadol exposure. At the cellular level, PC12 cells were exposed to 600μM tramadol for 48 hrs, followed by the assessments of ROS amount and gene expression levels of neurotoxicity associated with neurodegenerative pathways such as apoptosis and autophagy. Moreover, the structural and functional alteration of the hippocampus under chronic exposure to tramadol was also evaluated. In this regard, rats were treated with tramadol at doses of 50 mg/kg for three consecutive weeks. In vitro data revealed that tramadol provoked ROS production and caused the increase in the expression of autophagic and apoptotic genes in PC12 cells. Furthermore, in-vivo results demonstrated that tramadol not only did induce hippocampal atrophy, but it also triggered microgliosis and microglial activation, causing upregulation of apoptotic and inflammatory markers as well as over-activation of neurodegeneration. Tramadol also interrupted spatial learning and memory function along with long-term potentiation (LTP). Taken all together, our data disclosed the neurotoxic effects of tramadol on both in vitro and in-vivo. Moreover, we proposed a potential correlation between disrupted biochemical cascades and memory deficit under tramadol administration.
Collapse
Affiliation(s)
- Aysan Khatmi
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Cell Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Eskandarian Boroujeni
- Department of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| | - Samira Ezi
- Department of Anatomy, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | | | - Fakhroddin Aghajanpour
- Department of Cell Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Soltani
- Department of Cell Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mohammad-Amin Abdollahifar
- Department of Cell Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Hossein Toreyhi
- Department of Cell Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Aliaghaei
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Cell Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
174
|
Churchill NW, Hutchison MG, Graham SJ, Schweizer TA. Sex differences in acute and long-term brain recovery after concussion. Hum Brain Mapp 2021; 42:5814-5826. [PMID: 34643005 PMCID: PMC8596946 DOI: 10.1002/hbm.25591] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 05/14/2021] [Accepted: 07/01/2021] [Indexed: 12/25/2022] Open
Abstract
Concussion is associated with acute disturbances in brain function and behavior, with potential long‐term effects on brain health. However, it is presently unclear whether there are sex differences in acute and long‐term brain recovery. In this study, magnetic resonance imaging (MRI) was used to scan 61 participants with sport‐related concussion (30 male, 31 female) longitudinally at acute injury, medical clearance to return to play (RTP), and 1‐year post‐RTP. A large cohort of 167 controls (80 male, 87 female) was also imaged. Each MRI session assessed cerebral blood flow (CBF), along with white matter fractional anisotropy (FA) and mean diffusivity (MD). For concussed athletes, the parameters were converted to difference scores relative to matched control subgroups, and partial least squares modeled the main and sex‐specific effects of concussion. Although male and female athletes did not differ in acute symptoms or time to RTP , all MRI measures showed significant sex differences during recovery. Males had greater reductions in occipital‐parietal CBF (mean difference and 95%CI: 9.97 ml/100 g/min, [4.84, 15.12] ml/100 g/min, z = 3.73) and increases in callosal MD (9.07 × 10−5, [−14.14, −3.60] × 10−5, z = −3.46), with greatest effects at 1‐year post‐RTP. In contrast, females had greater reductions in FA of the corona radiata (16.50 × 10−3, [−22.38, −11.08] × 10−3, z = −5.60), with greatest effects at RTP. These findings provide new insights into how the brain recovers after a concussion, showing sex differences in both the acute and chronic phases of injury.
Collapse
Affiliation(s)
- Nathan W Churchill
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada.,Neuroscience Research Program, St. Michael's Hospital, Toronto, ON, Canada
| | - Michael G Hutchison
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada.,Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, ON, Canada
| | - Simon J Graham
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Tom A Schweizer
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada.,Neuroscience Research Program, St. Michael's Hospital, Toronto, ON, Canada.,Faculty of Medicine (Neurosurgery), University of Toronto, Toronto, ON, Canada.,The Institute of Biomaterials and Biomedical Engineering (IBBME) at the University of Toronto, Toronto, ON, Canada
| |
Collapse
|
175
|
Yan ZY, Jiao HY, Chen JB, Zhang KW, Wang XH, Jiang YM, Liu YY, Xue Z, Ma QY, Li XJ, Chen JX. Antidepressant Mechanism of Traditional Chinese Medicine Formula Xiaoyaosan in CUMS-Induced Depressed Mouse Model via RIPK1-RIPK3-MLKL Mediated Necroptosis Based on Network Pharmacology Analysis. Front Pharmacol 2021; 12:773562. [PMID: 34867405 PMCID: PMC8641697 DOI: 10.3389/fphar.2021.773562] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/01/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Depression is a stress-related disorder that seriously threatens people's physical and mental health. Xiaoyaosan is a classical traditional Chinese medicine formula, which has been used to treat mental depression since ancient times. More and more notice has been given to the relationship between the occurrence of necroptosis and the pathogenesis of mental disorders. Objective: The purpose of present study is to explore the potential mechanism of Xiaoyaosan for the treatment of depression using network pharmacology and experimental research, and identify the potential targets of necroptosis underlying the antidepressant mechanism of Xiaoyaosan. Methods: The mice model of depression was induced by chronic unpredictable mild stress (CUMS) for 6 weeks. Adult C57BL/6 mice were randomly divided into five groups, including control group, chronic unpredictable mild stress group, Xiaoyaosan treatment group, necrostatin-1 (Nec-1) group and solvent group. Drug intervention performed from 4th to 6th week of modeling. The mice in Xiaoyaosan treatment group received Xiaoyaosan by intragastric administration (0.254 g/kg/d), and mice in CUMS group received 0.5 ml physiological saline. Meanwhile, the mice in Nec-1 group were injected intraperitoneally (i.p.) with Nec-1 (10 mg/kg/d), and the equivalent volume of DMSO/PBS (8.3%) was injected into solvent group mice. The behavior tests such as sucrose preference test, forced swimming test and novelty-suppressed feeding test were measured to evaluate depressive-like behaviors of model mice. Then, the active ingredients in Xiaoyaosan and the related targets of depression and necroptosis were compiled through appropriate databases, while the "botanical drugs-active ingredients-target genes" network was constructed by network pharmacology analysis. The expressions of RIPK1, RIPK3, MLKL, p-MLKL were detected as critical target genes of necroptosis and the potential therapeutic target compounds of Xiaoyaosan. Furthermore, the levels of neuroinflammation and microglial activation of hippocampus were measured by detecting the expressions of IL-1β, Lipocalin-2 and IBA1, and the hematoxylin and eosin (H&E) stained was used to observe the morphology in hippocampus sections. Results: After 6-weeks of modeling, the behavioral data showed that mice in CUMS group and solvent group had obvious depressive-like behaviors, and the medication of Xiaoyaosan or Nec-1 could improve these behavioral changes. A total of 96 active ingredients in Xiaoyaosan which could regulate the 23 key target genes were selected from databases. Xiaoyaosan could alleviate the core target genes in necroptosis and improve the hippocampal function and neuroinflammation in depressed mice. Conclusion: The activation of necroptosis existed in the hippocampus of CUMS-induced mice, which was closely related to the pathogenesis of depression. The antidepressant mechanism of Xiaoyaosan included the regulation of multiple targets in necroptosis. It also suggested that necroptosis could be a new potential target for the treatment of depression.
Collapse
Affiliation(s)
- Zhi-Yi Yan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Hai-Yan Jiao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jian-Bei Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Kai-Wen Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xi-Hong Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - You-Ming Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yue-Yun Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhe Xue
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qing-Yu Ma
- Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiao-Juan Li
- Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Jia-Xu Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
176
|
Tastan B, Arioz BI, Tufekci KU, Tarakcioglu E, Gonul CP, Genc K, Genc S. Dimethyl Fumarate Alleviates NLRP3 Inflammasome Activation in Microglia and Sickness Behavior in LPS-Challenged Mice. Front Immunol 2021; 12:737065. [PMID: 34858398 PMCID: PMC8631454 DOI: 10.3389/fimmu.2021.737065] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/25/2021] [Indexed: 12/30/2022] Open
Abstract
NLRP3 inflammasome activation contributes to several pathogenic conditions, including lipopolysaccharide (LPS)-induced sickness behavior characterized by reduced mobility and depressive behaviors. Dimethyl fumarate (DMF) is an immunomodulatory and anti-oxidative molecule commonly used for the symptomatic treatment of multiple sclerosis and psoriasis. In this study, we investigated the potential use of DMF against microglial NLRP3 inflammasome activation both in vitro and in vivo. For in vitro studies, LPS- and ATP-stimulated N9 microglial cells were used to induce NLRP3 inflammasome activation. DMF’s effects on inflammasome markers, pyroptotic cell death, ROS formation, and Nrf2/NF-κB pathways were assessed. For in vivo studies, 12–14 weeks-old male BALB/c mice were treated with LPS, DMF + LPS and ML385 + DMF + LPS. Behavioral tests including open field, forced swim test, and tail suspension test were carried out to see changes in lipopolysaccharide-induced sickness behavior. Furthermore, NLRP3 and Caspase-1 expression in isolated microglia were determined by immunostaining. Here we demonstrated that DMF ameliorated LPS and ATP-induced NLRP3 inflammasome activation by reducing IL-1β, IL-18, caspase-1, and NLRP3 levels, reactive oxygen species formation and damage, and inhibiting pyroptotic cell death in N9 murine microglia via Nrf2/NF-κB pathways. DMF also improved LPS-induced sickness behavior in male mice and decreased caspase-1/NLRP3 levels via Nrf2 activation. Additionally, we showed that DMF pretreatment decreased miR-146a and miR-155 both in vivo and in vitro. Our results proved the effectiveness of DMF on the amelioration of microglial NLRP3 inflammasome activation. We anticipate that this study will provide the foundation consideration for further studies aiming to suppress NLRP3 inflammasome activation associated with in many diseases and a better understanding of its underlying mechanisms.
Collapse
Affiliation(s)
- Bora Tastan
- Genc Laboratory, Izmir Biomedicine and Genome Center, Izmir, Turkey.,Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Burak I Arioz
- Genc Laboratory, Izmir Biomedicine and Genome Center, Izmir, Turkey.,Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Kemal Ugur Tufekci
- Genc Laboratory, Izmir Biomedicine and Genome Center, Izmir, Turkey.,Department of Healthcare Services, Vocational School of Health Services, Izmir Democracy University, Izmir, Turkey
| | - Emre Tarakcioglu
- Genc Laboratory, Izmir Biomedicine and Genome Center, Izmir, Turkey.,Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Ceren Perihan Gonul
- Genc Laboratory, Izmir Biomedicine and Genome Center, Izmir, Turkey.,Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Kursad Genc
- Department of Neuroscience, Health Sciences Institute, Dokuz Eylul University, Izmir, Turkey
| | - Sermin Genc
- Genc Laboratory, Izmir Biomedicine and Genome Center, Izmir, Turkey.,Department of Neuroscience, Health Sciences Institute, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
177
|
Deng Z, Dong Y, Zhou X, Lu JH, Yue Z. Pharmacological modulation of autophagy for Alzheimer’s disease therapy: Opportunities and obstacles. Acta Pharm Sin B 2021; 12:1688-1706. [PMID: 35847516 PMCID: PMC9279633 DOI: 10.1016/j.apsb.2021.12.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 11/04/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a prevalent and deleterious neurodegenerative disorder characterized by an irreversible and progressive impairment of cognitive abilities as well as the formation of amyloid β (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain. By far, the precise mechanisms of AD are not fully understood and no interventions are available to effectively slow down progression of the disease. Autophagy is a conserved degradation pathway that is crucial to maintain cellular homeostasis by targeting damaged organelles, pathogens, and disease-prone protein aggregates to lysosome for degradation. Emerging evidence suggests dysfunctional autophagy clearance pathway as a potential cellular mechanism underlying the pathogenesis of AD in affected neurons. Here we summarize the current evidence for autophagy dysfunction in the pathophysiology of AD and discuss the role of autophagy in the regulation of AD-related protein degradation and neuroinflammation in neurons and glial cells. Finally, we review the autophagy modulators reported in the treatment of AD models and discuss the obstacles and opportunities for potential clinical application of the novel autophagy activators for AD therapy.
Collapse
Affiliation(s)
- Zhiqiang Deng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China
| | - Yu Dong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China
| | - Xiaoting Zhou
- Department of Neurology, the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jia-Hong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China
- Corresponding authors.
| | - Zhenyu Yue
- Department of Neurology, the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Corresponding authors.
| |
Collapse
|
178
|
Feng L, Li J, Zhang R. Current research status of blood biomarkers in Alzheimer's disease: Diagnosis and prognosis. Ageing Res Rev 2021; 72:101492. [PMID: 34673262 DOI: 10.1016/j.arr.2021.101492] [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: 04/11/2021] [Revised: 08/04/2021] [Accepted: 10/15/2021] [Indexed: 01/10/2023]
Abstract
Alzheimer's disease (AD), which mainly occurs in the elderly, is a neurodegenerative disease with a hidden onset, which leads to progressive cognitive and behavioral changes. The annually increasing prevalence rate and number of patients with AD exert great pressure on the society. No effective disease-modifying drug treatments are available; thus, there is no cure yet. The disease progression can only be delayed through early detection and drug assistance. Therefore, the importance of exploring associated biomarkers for the early diagnosis and prediction of the disease progress is highlighted. The National Institute on Aging- Alzheimer's Association (NIA-AA) proposed A/T/N diagnostic criteria in 2018, including Aβ42, p-tau, t-tau in cerebrospinal fluid (CSF), and positron emission tomography (PET). However, the invasiveness of lumbar puncture for CSF assessment and non-popularity of PET have prompted researchers to look for minimally invasive, easy to collect, and cost-effective biomarkers. Therefore, studies have largely focused on some novel molecules in the peripheral blood. This is an emerging research field, facing many obstacles and challenges while achieving some promising results.
Collapse
|
179
|
Kundu S, Saadi F, Sengupta S, Antony GR, Raveendran VA, Kumar R, Kamble MA, Sarkar L, Burrows A, Pal D, Sen GC, Sarma JD. DJ-1-Nrf2 axis is activated upon murine β-coronavirus infection in the CNS. BRAIN DISORDERS 2021; 4:100021. [PMID: 34514445 PMCID: PMC8418700 DOI: 10.1016/j.dscb.2021.100021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 07/03/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Coronaviruses have emerged as alarming pathogens owing to their inherent ability of genetic variation and cross-species transmission. Coronavirus infection burdens the endoplasmic reticulum (ER.), causes reactive oxygen species production and induces host stress responses, including unfolded protein response (UPR) and antioxidant system. In this study, we have employed a neurotropic murine β-coronavirus (M-CoV) infection in the Central Nervous System (CNS) of experimental mice model to study the role of host stress responses mediated by interplay of DJ-1 and XBP1. DJ-1 is an antioxidant molecule with established functions in neurodegeneration. However, its regulation in virus-induced cellular stress response is less explored. Our study showed that M-CoV infection activated the glial cells and induced antioxidant and UPR genes during the acute stage when the viral titer peaks. As the virus particles decreased and acute neuroinflammation diminished at day ten p.i., a significant up-regulation in UPR responsive XBP1, antioxidant DJ-1, and downstream signaling molecules, including Nrf2, was recorded in the brain tissues. Additionally, preliminary in silico analysis of the binding between the DJ-1 promoter and a positively charged groove of XBP1 is also investigated, thus hinting at a mechanism behind the upregulation of DJ-1 during MHV-infection. The current study thus attempts to elucidate a novel interplay between the antioxidant system and UPR in the outcome of coronavirus infection.
Collapse
Affiliation(s)
- Soumya Kundu
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Fareeha Saadi
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Sourodip Sengupta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Gisha Rose Antony
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Vineeth A Raveendran
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Rahul Kumar
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Mithila Ashok Kamble
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Lucky Sarkar
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Amy Burrows
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio, USA
| | - Debnath Pal
- Department of Computational and Data Sciences, Indian Institute of Science, Bangalore, India
| | - Ganes C Sen
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio, USA
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| |
Collapse
|
180
|
New Insights into the Role of Cysteine Cathepsins in Neuroinflammation. Biomolecules 2021; 11:biom11121796. [PMID: 34944440 PMCID: PMC8698589 DOI: 10.3390/biom11121796] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/25/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation, which is mediated by microglia and astrocytes, is associated with the progression of neurodegenerative diseases. Increasing evidence shows that activated microglia induce the expression and secretion of various lysosomal cathepsins, particularly during the early stage of neuroinflammation. This trigger signaling cascade that aggravate neurodegeneration. To date, most research on neuroinflammation has focused on the role of cysteine cathepsins, the largest cathepsin family. Cysteine cathepsins are primarily responsible for protein degradation in lysosomes; however, they also play a role in regulating a number of other important physiological and pathological processes. This review focuses on the functional roles of cysteine cathepsins in the central nervous system during neuroinflammation, with an emphasis on their roles in the polarization of microglia and neuroinflammation signaling, which in turn causes neuronal death and thus neurodegeneration.
Collapse
|
181
|
Chu E, Mychasiuk R, Hibbs ML, Semple BD. Dysregulated phosphoinositide 3-kinase signaling in microglia: shaping chronic neuroinflammation. J Neuroinflammation 2021; 18:276. [PMID: 34838047 PMCID: PMC8627624 DOI: 10.1186/s12974-021-02325-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 11/15/2021] [Indexed: 12/15/2022] Open
Abstract
Microglia are integral mediators of innate immunity within the mammalian central nervous system. Typical microglial responses are transient, intending to restore homeostasis by orchestrating the removal of pathogens and debris and the regeneration of damaged neurons. However, prolonged and persistent microglial activation can drive chronic neuroinflammation and is associated with neurodegenerative disease. Recent evidence has revealed that abnormalities in microglial signaling pathways involving phosphatidylinositol 3-kinase (PI3K) and protein kinase B (AKT) may contribute to altered microglial activity and exacerbated neuroimmune responses. In this scoping review, the known and suspected roles of PI3K-AKT signaling in microglia, both during health and pathological states, will be examined, and the key microglial receptors that induce PI3K-AKT signaling in microglia will be described. Since aberrant signaling is correlated with neurodegenerative disease onset, the relationship between maladapted PI3K-AKT signaling and the development of neurodegenerative disease will also be explored. Finally, studies in which microglial PI3K-AKT signaling has been modulated will be highlighted, as this may prove to be a promising therapeutic approach for the future treatment of a range of neuroinflammatory conditions.
Collapse
Affiliation(s)
- Erskine Chu
- Department of Immunology and Pathology, Central Clinical School, Monash University, Level 6, 89 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Neurology, Alfred Health, Prahran, VIC, 3181, Australia
| | - Margaret L Hibbs
- Department of Immunology and Pathology, Central Clinical School, Monash University, Level 6, 89 Commercial Road, Melbourne, VIC, 3004, Australia.
| | - Bridgette D Semple
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, 99 Commercial Road, Melbourne, VIC, 3004, Australia.
- Department of Neurology, Alfred Health, Prahran, VIC, 3181, Australia.
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, 3050, Australia.
| |
Collapse
|
182
|
Su R, Zhou T. Alpha-Synuclein Induced Immune Cells Activation and Associated Therapy in Parkinson's Disease. Front Aging Neurosci 2021; 13:769506. [PMID: 34803660 PMCID: PMC8602361 DOI: 10.3389/fnagi.2021.769506] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/04/2021] [Indexed: 12/30/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder closely related to immunity. An important aspect of the pathogenesis of PD is the interaction between α-synuclein and a series of immune cells. Studies have shown that accumulation of α-synuclein can induce an autoimmune response that accelerates the progression of PD. This study discusses the mechanisms underlying the interaction between α-synuclein and the immune system. During the development of PD, abnormally accumulated α-synuclein becomes an autoimmune antigen that binds to Toll-like receptors (TLRs) that activate microglia, which differentiate into the microglia type 1 (M1) subtype. The microglia activate intracellular inflammatory pathways, induce the release of proinflammatory cytokines, and promote the differentiation of cluster of differentiation 4 + (CD4 +) T cells into proinflammatory T helper type 1 (Th1) and T helper type 17 (Th17) subtypes. Given the important role of α-synuclein in the immune system of the patients with PD, identifying potential targets of immunotherapy related to α-synuclein is critical for slowing disease progression. An enhanced understanding of immune-associated mechanisms in PD can guide the development of associated therapeutic strategies in the future.
Collapse
Affiliation(s)
- Ruichen Su
- Queen Mary School of Nanchang University, Nanchang University, Nanchang, China
| | - Tian Zhou
- School of Basic Medical Science, Nanchang University, Nanchang, China
| |
Collapse
|
183
|
Mechanosensing and the Hippo Pathway in Microglia: A Potential Link to Alzheimer's Disease Pathogenesis? Cells 2021; 10:cells10113144. [PMID: 34831369 PMCID: PMC8622675 DOI: 10.3390/cells10113144] [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: 08/25/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 01/01/2023] Open
Abstract
The activation of microglia, the inflammatory cells of the central nervous system (CNS), has been linked to the pathogenesis of Alzheimer’s disease and other neurodegenerative diseases. How microglia sense the changing brain environment, in order to respond appropriately, is still being elucidated. Microglia are able to sense and respond to the mechanical properties of their microenvironment, and the physical and molecular pathways underlying this mechanosensing/mechanotransduction in microglia have recently been investigated. The Hippo pathway functions through mechanosensing and subsequent protein kinase cascades, and is critical for neuronal development and many other cellular processes. In this review, we examine evidence for the potential involvement of Hippo pathway components specifically in microglia in the pathogenesis of Alzheimer’s disease. We suggest that the Hippo pathway is worth investigating as a mechanosensing pathway in microglia, and could be one potential therapeutic target pathway for preventing microglial-induced neurodegeneration in AD.
Collapse
|
184
|
Nicolazzo JA, Pan Y, Di Stefano I, Choy KHC, Reddiar SB, Low YL, Wai DCC, Norton RS, Jin L. Blockade of Microglial Kv1.3 Potassium Channels by the Peptide HsTX1[R14A] Attenuates Lipopolysaccharide-mediated Neuroinflammation. J Pharm Sci 2021; 111:638-647. [PMID: 34767826 DOI: 10.1016/j.xphs.2021.11.003] [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: 08/31/2021] [Revised: 11/03/2021] [Accepted: 11/03/2021] [Indexed: 12/19/2022]
Abstract
The expression of voltage-gated potassium Kv1.3 channels is increased in activated microglia, with non-selective blockade reported to attenuate microglial-mediated neuroinflammation. In this study, we evaluated the impact of a potent and selective peptidic blocker of Kv1.3 channels, HsTX1[R14A], on microglial-mediated neuroinflammation in vitro and in vivo. Treatment with both 0.1 and 1 µg/mL lipopolysaccharide (LPS) significantly (p < 0.05) increased Kv1.3 abundance on the surface of BV-2 microglia in association with increased levels of mRNA for tumour necrosis factor-α (TNF-α) and interleukin-6 (IL-6). The increased transcription of TNF-α and IL-6 was significantly attenuated (by 24.9 and 20.2%, respectively) by HsTX1[R14A] (100 nM). The concomitant increase in TNF-α and IL-6 release from BV-2 microglia was significantly attenuated by HsTX1[R14A] by 10.7 and 12.6%, respectively. In LPS-treated primary mouse microglia, the levels of TNF-α and nitric oxide were also attenuated by HsTX1[R14A] (26.1 and 20.4%, respectively). In an LPS-induced mouse model of neuroinflammation, both an immediate and delayed subcutaneous dose of HsTX1[R14A] (2 mg/kg) significantly reduced plasma and brain levels of the pro-inflammatory mediators TNF-α, IL-1β and IL-6, with no impact on the anti-inflammatory IL-10. These results demonstrate that HsTX1[R14A] is a promising therapeutic candidate for the treatment of diseases with a neuroinflammatory component.
Collapse
Affiliation(s)
- Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Yijun Pan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Ilenia Di Stefano
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Kwok H C Choy
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Sanjeevini Babu Reddiar
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Yi Ling Low
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Dorothy C C Wai
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Raymond S Norton
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia; ARC Centre for Fragment-Based Design, Monash University, Parkville, Victoria 3052, Australia
| | - Liang Jin
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
185
|
Saadi F, Pal D, Sarma JD. Spike Glycoprotein Is Central to Coronavirus Pathogenesis-Parallel Between m-CoV and SARS-CoV-2. Ann Neurosci 2021; 28:201-218. [PMID: 35341224 PMCID: PMC8948335 DOI: 10.1177/09727531211023755] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 03/24/2021] [Indexed: 01/04/2023] Open
Abstract
Coronaviruses (CoVs) are single-stranded, polyadenylated, enveloped RNA of positive polarity with a unique potential to alter host tropism. This has been exceptionally demonstrated by the emergence of deadly virus outbreaks of the past: Severe Acute Respiratory Syndrome (SARS-CoV) in 2003 and Middle East Respiratory Syndrome (MERS-CoV) in 2012. The 2019 outbreak by the new cross-species transmission of SARS-CoV-2 has put the world on alert. CoV infection is triggered by receptor recognition, membrane fusion, and successive viral entry mediated by the surface Spike (S) glycoprotein. S protein is one of the major antigenic determinants and the target for neutralizing antibodies. It is a valuable target in antiviral therapies because of its central role in cell-cell fusion, viral antigen spread, and host immune responses leading to immunopathogenesis. The receptor-binding domain of S protein has received greater attention as it initiates host attachment and contains major antigenic determinants. However, investigating the therapeutic potential of fusion peptide as a part of the fusion core complex assembled by the heptad repeats 1 and 2 (HR1 and HR2) is also warranted. Along with receptor attachment and entry, fusion mechanisms should also be explored for designing inhibitors as a therapeutic intervention. In this article, we review the S protein function and its role in mediating membrane fusion, spread, tropism, and its associated pathogenesis with notable therapeutic strategies focusing on results obtained from studies on a murine β-Coronavirus (m-CoV) and its associated disease process.
Collapse
Affiliation(s)
- Fareeha Saadi
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Kolkata, West Bengal, India
| | - Debnath Pal
- Department of Computational and Data Sciences, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Kolkata, West Bengal, India
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
186
|
Xiao H, Han S, Baigude H. Regulation of microglia polarization via mannose receptor-mediated delivery of siRNA by ligand-functionalized DoGo LNP. RSC Adv 2021; 11:32549-32558. [PMID: 35493551 PMCID: PMC9041768 DOI: 10.1039/d1ra04293a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 09/23/2021] [Indexed: 12/19/2022] Open
Abstract
The pro-inflammatory polarization of microglia after stroke is one of the major causes of secondary brain injury. Downregulation of the gene involved in canonical inflammatory pathways in glial cells can exert neuroprotective effects via inhibiting the release of pro-inflammatory factors. In this study, we functionalized DoGo lipids with mannose, the ligand of the mannose receptor (MR) that is expressed in microglia, and evaluated the MR-mediated cellular internalization of DoGo lipid nanoparticles (denote M3) carrying siRNA against TLR4 in BV2 cells in vitro. We confirmed that siTLR4/M3 complexes were specifically internalized by BV2 cells in a MR-dependent manner, and the treatment of oxygen glucose deprivation (OGD)-treated BV2 cells with siTLR4/M3 complexes resulted in remarkable silencing of TLR4, and induced downregulated M1 polarization and upregulated M2 polarization markers. Collectively, our data suggest that the M3 lipoplex is a promising microglia-targeting siRNA delivery agent. Mannose functionalized DoGo lipid nanoparticles (denote M3) can effectively deliver siRNA to microglia via receptor-mediated internalization, knockdown target gene and induce neuroprotective M2 polarization.![]()
Collapse
Affiliation(s)
- Hai Xiao
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University Hohhot Inner Mongolia 010020 P. R. China +86 471 4992511 +86 471 4992511
| | - Shuqin Han
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University Hohhot Inner Mongolia 010020 P. R. China +86 471 4992511 +86 471 4992511
| | - Huricha Baigude
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University Hohhot Inner Mongolia 010020 P. R. China +86 471 4992511 +86 471 4992511
| |
Collapse
|
187
|
Sridharan B, Lee MJ. Ketogenic diet: A promising neuroprotective composition for managing Alzheimer's diseases and its pathological mechanisms. Curr Mol Med 2021; 22:640-656. [PMID: 34607541 DOI: 10.2174/1566524021666211004104703] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 11/22/2022]
Abstract
Ketogenic diet and ketone bodies gained significant attention in recent years due to their ability to influence the specific energy metabolism and restoration of mitochondrial homeostasis that can help in hindering the progression of many metabolic diseases including diabetes and neurodegenerative diseases. Ketogenic diet consists of high fat and low carbohydrate contents which makes the body glucose deprived and rely on alternative sources (ketone bodies) for energy. It has been initially designed and supplemented for the treatment of epilepsy and later its influence on many energy-deriving biochemical pathways made it a highly sorted food supplement for many metabolic diseases and even by healthy individuals for body building and calorie restriction. Among the reported therapeutic action over a range of diseases, neurodegenerative disorders especially Alzheimer's disease gained the attention of many researchers and clinicians because of its potency and its easier supplementation as a food additive. Complex pathology and multiple influencing factors of Alzheimer's disease make exploration of its therapeutic strategies a demanding task. It was a common phenomenon that energy deprivation in neurological disorders including Alzheimer's disease, to progress rapidly. The ability of ketone bodies to stabilize the mitochondrial energy metabolism makes it a suitable intervening agent. In this review, we will discuss various research progress made with regards to ketone bodies/ketogenic diet for management of Alzheimer's disease and elaborate in detail about the mechanisms that are influenced during their therapeutic action.
Collapse
Affiliation(s)
- Badrinathan Sridharan
- Department of Applied Chemistry, Chaoyang University of Technology, 168 Jifeng East Road, Taichung. Taiwan
| | - Meng-Jen Lee
- Department of Applied Chemistry, Chaoyang University of Technology, 168 Jifeng East Road, Taichung. Taiwan
| |
Collapse
|
188
|
Saputra WD, Shono H, Ohsaki Y, Sultana H, Komai M, Shirakawa H. Geranylgeraniol Inhibits Lipopolysaccharide-Induced Inflammation in Mouse-Derived MG6 Microglial Cells via NF-κB Signaling Modulation. Int J Mol Sci 2021; 22:ijms221910543. [PMID: 34638882 PMCID: PMC8508820 DOI: 10.3390/ijms221910543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/25/2021] [Accepted: 09/25/2021] [Indexed: 12/18/2022] Open
Abstract
Persistent inflammatory reactions in microglial cells are strongly associated with neurodegenerative pathogenesis. Additionally, geranylgeraniol (GGOH), a plant-derived isoprenoid, has been found to improve inflammatory conditions in several animal models. It has also been observed that its chemical structure is similar to that of the side chain of menaquinone-4, which is a vitamin K2 sub-type that suppresses inflammation in mouse-derived microglial cells. In this study, we investigated whether GGOH has a similar anti-inflammatory effect in activated microglial cells. Particularly, mouse-derived MG6 cells pre-treated with GGOH were exposed to lipopolysaccharide (LPS). Thereafter, the mRNA levels of pro-inflammatory cytokines were determined via qRT-PCR, while protein expression levels, especially the expression of NF-κB signaling cascade-related proteins, were determined via Western blot analysis. The distribution of NF-κB p65 protein was also analyzed via fluorescence microscopy. Thus, it was observed that GGOH dose-dependently suppressed the LPS-induced increase in the mRNA levels of Il-1β, Tnf-α, Il-6, and Cox-2. Furthermore, GGOH inhibited the phosphorylation of TAK1, IKKα/β, and NF-κB p65 proteins as well as NF-κB nuclear translocation induced by LPS while maintaining IκBα expression. We showed that GGOH, similar to menaquinone-4, could alleviate LPS-induced microglial inflammation by targeting the NF-kB signaling pathway.
Collapse
Affiliation(s)
- Wahyu Dwi Saputra
- Laboratory of Nutrition, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki Aza Aoba, Aoba-ku, Sendai 980-8572, Japan; (W.D.S.); (H.S.); (Y.O.); (H.S.); (M.K.)
| | - Hiroki Shono
- Laboratory of Nutrition, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki Aza Aoba, Aoba-ku, Sendai 980-8572, Japan; (W.D.S.); (H.S.); (Y.O.); (H.S.); (M.K.)
| | - Yusuke Ohsaki
- Laboratory of Nutrition, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki Aza Aoba, Aoba-ku, Sendai 980-8572, Japan; (W.D.S.); (H.S.); (Y.O.); (H.S.); (M.K.)
- International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki Aza Aoba, Aoba-ku, Sendai 980-8572, Japan
| | - Halima Sultana
- Laboratory of Nutrition, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki Aza Aoba, Aoba-ku, Sendai 980-8572, Japan; (W.D.S.); (H.S.); (Y.O.); (H.S.); (M.K.)
| | - Michio Komai
- Laboratory of Nutrition, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki Aza Aoba, Aoba-ku, Sendai 980-8572, Japan; (W.D.S.); (H.S.); (Y.O.); (H.S.); (M.K.)
| | - Hitoshi Shirakawa
- Laboratory of Nutrition, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki Aza Aoba, Aoba-ku, Sendai 980-8572, Japan; (W.D.S.); (H.S.); (Y.O.); (H.S.); (M.K.)
- International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki Aza Aoba, Aoba-ku, Sendai 980-8572, Japan
- Correspondence: ; Tel.: +81-22-757-4402
| |
Collapse
|
189
|
Borgonetti V, Coppi E, Galeotti N. Targeting the RNA-Binding Protein HuR as Potential Thera-Peutic Approach for Neurological Disorders: Focus on Amyo-Trophic Lateral Sclerosis (ALS), Spinal Muscle Atrophy (SMA) and Multiple Sclerosis. Int J Mol Sci 2021; 22:ijms221910394. [PMID: 34638733 PMCID: PMC8508990 DOI: 10.3390/ijms221910394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 01/03/2023] Open
Abstract
The importance of precise co- and post-transcriptional processing of RNA in the regulation of gene expression has become increasingly clear. RNA-binding proteins (RBPs) are a class of proteins that bind single- or double-chain RNA, with different affinities and selectivity, thus regulating the various functions of RNA and the fate of the cells themselves. ELAV (embryonic lethal/abnormal visual system)/Hu proteins represent an important family of RBPs and play a key role in the fate of newly transcribed mRNA. ELAV proteins bind AU-rich element (ARE)-containing transcripts, which are usually present on the mRNA of proteins such as cytokines, growth factors, and other proteins involved in neuronal differentiation and maintenance. In this review, we focused on a member of ELAV/Hu proteins, HuR, and its role in the development of neurodegenerative disorders, with a particular focus on demyelinating diseases.
Collapse
|
190
|
Merelli A, Repetto M, Lazarowski A, Auzmendi J. Hypoxia, Oxidative Stress, and Inflammation: Three Faces of Neurodegenerative Diseases. J Alzheimers Dis 2021; 82:S109-S126. [PMID: 33325385 DOI: 10.3233/jad-201074] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The cerebral hypoxia-ischemia can induce a wide spectrum of biologic responses that include depolarization, excitotoxicity, oxidative stress, inflammation, and apoptosis, and result in neurodegeneration. Several adaptive and survival endogenous mechanisms can also be activated giving an opportunity for the affected cells to remain alive, waiting for helper signals that avoid apoptosis. These signals appear to help cells, depending on intensity, chronicity, and proximity to the central hypoxic area of the affected tissue. These mechanisms are present not only in a large list of brain pathologies affecting commonly older individuals, but also in other pathologies such as refractory epilepsies, encephalopathies, or brain trauma, where neurodegenerative features such as cognitive and/or motor deficits sequelae can be developed. The hypoxia inducible factor 1α (HIF-1α) is a master transcription factor driving a wide spectrum cellular response. HIF-1α may induce erythropoietin (EPO) receptor overexpression, which provides the therapeutic opportunity to administer pharmacological doses of EPO to rescue and/or repair affected brain tissue. Intranasal administration of EPO combined with other antioxidant and anti-inflammatory compounds could become an effective therapeutic alternative, to avoid and/or slow down neurodegenerative deterioration without producing adverse peripheral effects.
Collapse
Affiliation(s)
- Amalia Merelli
- Universidad de Buenos Aires, Facultad de Farmacia y Bioqummica, Departamento de Bioquímica Clínica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Argentina
| | - Marisa Repetto
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica; Instituto de Bioquímica y Medicina Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas (IBIMOL, UBA-CONICET), Argentina
| | - Alberto Lazarowski
- Universidad de Buenos Aires, Facultad de Farmacia y Bioqummica, Departamento de Bioquímica Clínica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Argentina
| | - Jerónimo Auzmendi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioqummica, Departamento de Bioquímica Clínica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| |
Collapse
|
191
|
Churchill NW, Hutchison MG, Graham SJ, Schweizer TA. Acute and Chronic Effects of Multiple Concussions on Midline Brain Structures. Neurology 2021; 97:e1170-e1181. [PMID: 34433678 PMCID: PMC8480483 DOI: 10.1212/wnl.0000000000012580] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 06/22/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES To test the hypothesis that a history of concussion (HOC) causes greater disturbances in cerebral blood flow (CBF) and white matter microstructure of midline brain structures after subsequent concussions, during the acute and chronic phases of recovery. METHODS In this longitudinal MRI study, 61 athletes with uncomplicated concussion (36 with HOC) were imaged at the acute phase of injury (1-7 days after injury), the subacute phase (8-14 days), medical clearance to return to play (RTP), 1 month after RTP, and 1 year after RTP. A normative group of 167 controls (73 with HOC) were also imaged. Each session assessed CBF of the cingulate cortex, along with fractional anisotropy (FA) and mean diffusivity (MD) of the corpus callosum. Linear mixed models tested for interactions of HOC with time since injury. The Sport Concussion Assessment Tool (SCAT) was also used to evaluate effects of HOC on symptoms, cognition, and balance. RESULTS Athletes with HOC had significantly greater declines in midcingulate CBF subacutely (z = -3.29, p = 0.002) and greater declines in posterior cingulate CBF at 1 year after RTP (z = -2.42, p = 0.007). No significant effects of HOC were seen for FA, whereas athletes with HOC had higher MD of the splenium at RTP (z = 2.54, p = 0.008). These effects were seen in the absence of significant differences in SCAT domains (|z| ≤ 1.14, p ≥ 0.256) or time to RTP (z = 0.23, p = 0.818). DISCUSSION Results indicate subacute and chronic effects of HOC on cingulate CBF and callosal microstructure in the absence of differences in clinical indices. These findings provide new insights into physiologic brain recovery after concussion, with cumulative effects of repeated injury detected among young, healthy athletes.
Collapse
Affiliation(s)
- Nathan W Churchill
- From the Keenan Research Centre for Biomedical Science (N.W.C., M.G.H., T.A.S.) and Neuroscience Research Program (N.W.C., T.A.S.), St. Michael's Hospital; Faculty of Kinesiology and Physical Education (M.G.H.), Department of Medical Biophysics (S.J.G.), Faculty of Medicine (Neurosurgery) (T.A.S.), and Institute of Biomaterials and Biomedical Engineering (T.A.S.), University of Toronto; and Physical Sciences Platform (S.J.G.), Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Michael G Hutchison
- From the Keenan Research Centre for Biomedical Science (N.W.C., M.G.H., T.A.S.) and Neuroscience Research Program (N.W.C., T.A.S.), St. Michael's Hospital; Faculty of Kinesiology and Physical Education (M.G.H.), Department of Medical Biophysics (S.J.G.), Faculty of Medicine (Neurosurgery) (T.A.S.), and Institute of Biomaterials and Biomedical Engineering (T.A.S.), University of Toronto; and Physical Sciences Platform (S.J.G.), Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Simon J Graham
- From the Keenan Research Centre for Biomedical Science (N.W.C., M.G.H., T.A.S.) and Neuroscience Research Program (N.W.C., T.A.S.), St. Michael's Hospital; Faculty of Kinesiology and Physical Education (M.G.H.), Department of Medical Biophysics (S.J.G.), Faculty of Medicine (Neurosurgery) (T.A.S.), and Institute of Biomaterials and Biomedical Engineering (T.A.S.), University of Toronto; and Physical Sciences Platform (S.J.G.), Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Tom A Schweizer
- From the Keenan Research Centre for Biomedical Science (N.W.C., M.G.H., T.A.S.) and Neuroscience Research Program (N.W.C., T.A.S.), St. Michael's Hospital; Faculty of Kinesiology and Physical Education (M.G.H.), Department of Medical Biophysics (S.J.G.), Faculty of Medicine (Neurosurgery) (T.A.S.), and Institute of Biomaterials and Biomedical Engineering (T.A.S.), University of Toronto; and Physical Sciences Platform (S.J.G.), Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.
| |
Collapse
|
192
|
Iatrou A, Clark EM, Wang Y. Nuclear dynamics and stress responses in Alzheimer's disease. Mol Neurodegener 2021; 16:65. [PMID: 34535174 PMCID: PMC8447732 DOI: 10.1186/s13024-021-00489-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 09/01/2021] [Indexed: 02/06/2023] Open
Abstract
In response to extracellular and intracellular stressors, the nucleus and nuclear compartments undergo distinct molecular changes to maintain cell homeostasis. In the context of Alzheimer’s disease, misfolded proteins and various cellular stressors lead to profound structural and molecular changes at the nucleus. This review summarizes recent research on nuclear alterations in AD development, from the nuclear envelope changes to chromatin and epigenetic regulation and then to common nuclear stress responses. Finally, we provide our thoughts on the importance of understanding cell-type-specific changes and identifying upstream causal events in AD pathogenesis and highlight novel sequencing and gene perturbation technologies to address those challenges.
Collapse
Affiliation(s)
- Artemis Iatrou
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W. Harrison St., Chicago, IL, 60612, USA
| | - Eric M Clark
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W. Harrison St., Chicago, IL, 60612, USA
| | - Yanling Wang
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W. Harrison St., Chicago, IL, 60612, USA.
| |
Collapse
|
193
|
Jain N, Smirnovs M, Strojeva S, Murovska M, Skuja S. Chronic Alcoholism and HHV-6 Infection Synergistically Promote Neuroinflammatory Microglial Phenotypes in the Substantia Nigra of the Adult Human Brain. Biomedicines 2021; 9:biomedicines9091216. [PMID: 34572401 PMCID: PMC8472392 DOI: 10.3390/biomedicines9091216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 12/25/2022] Open
Abstract
Both chronic alcoholism and human herpesvirus-6 (HHV-6) infection have been identified as promoters of neuroinflammation and known to cause movement-related disorders. Substantia Nigra (SN), the dopaminergic neuron-rich region of the basal ganglia, is involved in regulating motor function and the reward system. Hence, we hypothesize the presence of possible synergism between alcoholism and HHV-6 infection in the SN region and report a comprehensive quantification and characterization of microglial functions and morphology in postmortem brain tissue from 44 healthy, age-matched alcoholics and chronic alcoholics. A decrease in the perivascular CD68+ microglia in alcoholics was noted in both the gray and white matter. Additionally, the CD68+/Iba1− microglial subpopulation was found to be the dominant type in the controls. Conversely, in alcoholics, dystrophic changes in microglia were seen with a significant increase in Iba1 expression and perivascular to diffuse migration. An increase in CD11b expression was noted in alcoholics, with the Iba1+/CD11b− subtype promoting inflammation. All the controls were found to be negative for HHV-6 whilst the alcoholics demonstrated HHV-6 positivity in both gray and white matter. Amongst HHV-6 positive alcoholics, all the above-mentioned changes were found to be heightened when compared with HHV-6 negative alcoholics, thereby highlighting the compounding relationship between alcoholism and HHV-6 infection that promotes microglia-mediated neuroinflammation.
Collapse
Affiliation(s)
- Nityanand Jain
- Joint Laboratory of Electron Microscopy, Institute of Anatomy and Anthropology, Rīga Stradiņš University, LV-1010 Riga, Latvia;
- Correspondence: (N.J.); (S.S.); Tel.: +371-673-204-21 (N.J. & S.S.)
| | - Marks Smirnovs
- Joint Laboratory of Electron Microscopy, Institute of Anatomy and Anthropology, Rīga Stradiņš University, LV-1010 Riga, Latvia;
| | - Samanta Strojeva
- Institute of Microbiology and Virology, Rīga Stradiņš University, LV-1067 Riga, Latvia; (S.S.); (M.M.)
| | - Modra Murovska
- Institute of Microbiology and Virology, Rīga Stradiņš University, LV-1067 Riga, Latvia; (S.S.); (M.M.)
| | - Sandra Skuja
- Joint Laboratory of Electron Microscopy, Institute of Anatomy and Anthropology, Rīga Stradiņš University, LV-1010 Riga, Latvia;
- Correspondence: (N.J.); (S.S.); Tel.: +371-673-204-21 (N.J. & S.S.)
| |
Collapse
|
194
|
Energy homeostasis deregulation is attenuated by TUDCA treatment in streptozotocin-induced Alzheimer's disease mice model. Sci Rep 2021; 11:18114. [PMID: 34518585 PMCID: PMC8437965 DOI: 10.1038/s41598-021-97624-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/27/2021] [Indexed: 12/04/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia. While cognitive deficits remain the major manifestation of AD, metabolic and non-cognitive abnormalities, such as alterations in food intake, body weight and energy balance are also present, both in AD patients and animal models. In this sense, the tauroursodeoxycholic acid (TUDCA) has shown beneficial effects both in reducing the central and cognitive markers of AD, as well as in attenuating the metabolic disorders associated with it. We previously demonstrated that TUDCA improves glucose homeostasis and decreases the main AD neuromarkers in the streptozotocin-induced AD mouse model (Stz). Besides that, TUDCA-treated Stz mice showed lower body weight and adiposity. Here, we investigated the actions of TUDCA involved in the regulation of body weight and adiposity in Stz mice, since the effects of TUDCA in hypothalamic appetite control and energy homeostasis have not yet been explored in an AD mice model. The TUDCA-treated mice (Stz + TUDCA) displayed lower food intake, higher energy expenditure (EE) and respiratory quotient. In addition, we observed in the hypothalamus of the Stz + TUDCA mice reduced fluorescence and gene expression of inflammatory markers, as well as normalization of the orexigenic neuropeptides AgRP and NPY expression. Moreover, leptin-induced p-JAK2 and p-STAT3 signaling in the hypothalamus of Stz + TUDCA mice was improved, accompanied by reduced acute food intake after leptin stimulation. Taken together, we demonstrate that TUDCA treatment restores energy metabolism in Stz mice, a phenomenon that is associated with reduced food intake, increased EE and improved hypothalamic leptin signaling. These findings suggest treatment with TUDCA as a promising therapeutic intervention for the control of energy homeostasis in AD individuals.
Collapse
|
195
|
Takata K, Ginhoux F, Shimohama S. Roles of microglia in Alzheimer's disease and impact of new findings on microglial heterogeneity as a target for therapeutic intervention. Biochem Pharmacol 2021; 192:114754. [PMID: 34480881 DOI: 10.1016/j.bcp.2021.114754] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 12/25/2022]
Abstract
Microglia are specialized macrophages that reside within the central nervous system and play key roles in brain immunity, development and homeostasis. Recent studies also revealed functions of microglia in neuroprotection and neuroinflammation, leading to the discovery that microglia are involved in several brain pathologies including Alzheimer's disease (AD). However, the beneficial and detrimental actions of this intriguing cell population can be challenging to dissect: the advent of single-cell and single-nucleus transcriptomic technologies has revolutionized our understanding of the heterogeneity of multiple cell types and is now being applied to the study of microglia in health and disease. Here, we review recent findings on microglial biology, focusing on insights from single cell transcriptomic studies and the heterogeneity that they reveal, and consider the impact of these findings on our understanding of AD. We also discuss how microglia might represent a next-generation therapeutic target for treatment of AD and other neuroinflammatory conditions.
Collapse
Affiliation(s)
- Kazuyuki Takata
- Division of Integrated Pharmaceutical Sciences, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan.
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore; Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Translational Immunology Institute, SingHealth/Duke-NUS, Academic Medical Centre, The Academia, Singapore 169856, Singapore
| | - Shun Shimohama
- Department of Neurology, Sapporo Medical University, School of Medicine, Sapporo 060-8543, Japan
| |
Collapse
|
196
|
Huang X, Ying J, Yang D, Fang P, Wang X, Zhou B, Zhang L, Fang Y, Yu W, Liu X, Zhen Q, Hua F. The Mechanisms of Sevoflurane-Induced Neuroinflammation. Front Aging Neurosci 2021; 13:717745. [PMID: 34421578 PMCID: PMC8375153 DOI: 10.3389/fnagi.2021.717745] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/13/2021] [Indexed: 12/17/2022] Open
Abstract
Sevoflurane is one of the most commonly used inhaled anesthetics due to its low blood gas coefficient, fast onset, low airway irritation, and aromatic smell. However, recent studies have reported that sevoflurane exposure may have deleterious effects on cognitive function. Although neuroinflammation was most widely mentioned among the established mechanisms of sevoflurane-induced cognitive dysfunction, its upstream mechanisms have yet to be illustrated. Thus, we reviewed the relevant literature and discussed the most mentioned mechanisms, including the modulation of the microglial function, blood–brain barrier (BBB) breakdown, changes in gut microbiota, and ease of cholinergic neurotransmission to help us understand the properties of sevoflurane, providing us new perspectives for the prevention of sevoflurane-induced cognitive impairment.
Collapse
Affiliation(s)
- Xiangfei Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Jun Ying
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Danying Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Pu Fang
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xifeng Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bin Zhou
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Lieliang Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Yang Fang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Wen Yu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Xing Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Qingcui Zhen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| |
Collapse
|
197
|
El Mahmoudi N, Rastoldo G, Marouane E, Péricat D, Watabe I, Tonetto A, Hautefort C, Chabbert C, Sargolini F, Tighilet B. Breaking a dogma: acute anti-inflammatory treatment alters both post-lesional functional recovery and endogenous adaptive plasticity mechanisms in a rodent model of acute peripheral vestibulopathy. J Neuroinflammation 2021; 18:183. [PMID: 34419105 PMCID: PMC8380392 DOI: 10.1186/s12974-021-02222-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 07/19/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Due to their anti-inflammatory action, corticosteroids are the reference treatment for brain injuries and many inflammatory diseases. However, the benefits of acute corticotherapy are now being questioned, particularly in the case of acute peripheral vestibulopathies (APV), characterized by a vestibular syndrome composed of sustained spinning vertigo, spontaneous ocular nystagmus and oscillopsia, perceptual-cognitive, posturo-locomotor, and vegetative disorders. We assessed the effectiveness of acute corticotherapy, and the functional role of acute inflammation observed after sudden unilateral vestibular loss. METHODS We used the rodent model of unilateral vestibular neurectomy, mimicking the syndrome observed in patients with APV. We treated the animals during the acute phase of the vestibular syndrome, either with placebo or methylprednisolone, an anti-inflammatory corticosteroid. At the cellular level, impacts of methylprednisolone on endogenous plasticity mechanisms were assessed through analysis of cell proliferation and survival, glial reactions, neuron's membrane excitability, and stress marker. At the behavioral level, vestibular and posturo-locomotor functions' recovery were assessed with appropriate qualitative and quantitative evaluations. RESULTS We observed that acute treatment with methylprednisolone significantly decreases glial reactions, cell proliferation and survival. In addition, stress and excitability markers were significantly impacted by the treatment. Besides, vestibular syndrome's intensity was enhanced, and vestibular compensation delayed under acute methylprednisolone treatment. CONCLUSIONS We show here, for the first time, that acute anti-inflammatory treatment alters the expression of the adaptive plasticity mechanisms in the deafferented vestibular nuclei and generates enhanced and prolonged vestibular and postural deficits. These results strongly suggest a beneficial role for acute endogenous neuroinflammation in vestibular compensation. They open the way to a change in dogma for the treatment and therapeutic management of vestibular patients.
Collapse
Affiliation(s)
- Nada El Mahmoudi
- Aix-Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
- Centre Saint-Charles, Aix-Marseille Université CNRS, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
| | - Guillaume Rastoldo
- Aix-Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
- Centre Saint-Charles, Aix-Marseille Université CNRS, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
| | - Emna Marouane
- Aix-Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
- Centre Saint-Charles, Aix-Marseille Université CNRS, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
| | - David Péricat
- Institut de Pharmacologie Et de Biologie Structurale, Université de Toulouse Paul Sabatier-CNRS, Toulouse, France
| | - Isabelle Watabe
- Aix-Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
- Centre Saint-Charles, Aix-Marseille Université CNRS, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
| | - Alain Tonetto
- Centrale Marseille, FSCM (FR 1739), PRATIM, Aix Marseille Université-CNRS, 13397, Marseille, France
| | - Charlotte Hautefort
- Department of Head and Neck Surgery, Lariboisière University Hospital, Paris, France
| | - Christian Chabbert
- Aix-Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
- Centre Saint-Charles, Aix-Marseille Université CNRS, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
- GDR Physiopathologie Vestibulaire-Unité GDR2074 CNRS, Marseille, France
| | - Francesca Sargolini
- Aix-Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
| | - Brahim Tighilet
- Aix-Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France.
- GDR Physiopathologie Vestibulaire-Unité GDR2074 CNRS, Marseille, France.
| |
Collapse
|
198
|
Apostol CR, Tanguturi P, Szabò LZ, Varela D, Gilmartin T, Streicher JM, Polt R. Synthesis and In Vitro Characterization of Glycopeptide Drug Candidates Related to PACAP 1-23. Molecules 2021; 26:4932. [PMID: 34443519 PMCID: PMC8401035 DOI: 10.3390/molecules26164932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 01/05/2023] Open
Abstract
The search for efficacious treatment of neurodegenerative and progressive neuroinflammatory diseases continues, as current therapies are unable to halt or reverse disease progression. PACAP represents one potential therapeutic that provides neuroprotection effects on neurons, and also modulates inflammatory responses and circulation within the brain. However, PACAP is a relatively long peptide hormone that is not trivial to synthesize. Based on previous observations that the shortened isoform PACAP1-23 is capable of inducing neuroprotection in vitro, we were inspired to synthesize shortened glycopeptide analogues of PACAP1-23. Herein, we report the synthesis and in vitro characterization of glycosylated PACAP1-23 analogues that interact strongly with the PAC1 and VPAC1 receptors, while showing reduced activity at the VPAC2 receptor.
Collapse
Affiliation(s)
- Christopher R. Apostol
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, 1306 E. University Blvd, Tucson, AZ 85721, USA; (C.R.A.); (L.Z.S.)
| | - Parthasaradhireddy Tanguturi
- Department of Pharmacology, College of Medicine, The University of Arizona, 1501 N. Campbell Ave, Tucson, AZ 85724, USA; (P.T.); (J.M.S.)
| | - Lajos Z. Szabò
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, 1306 E. University Blvd, Tucson, AZ 85721, USA; (C.R.A.); (L.Z.S.)
| | - Daniel Varela
- Facultat de Quìmica Tarragona, Universitat Rovera I Virgili, 43007 Barcelona, Spain; (D.V.); (T.G.)
| | - Thiago Gilmartin
- Facultat de Quìmica Tarragona, Universitat Rovera I Virgili, 43007 Barcelona, Spain; (D.V.); (T.G.)
| | - John M. Streicher
- Department of Pharmacology, College of Medicine, The University of Arizona, 1501 N. Campbell Ave, Tucson, AZ 85724, USA; (P.T.); (J.M.S.)
| | - Robin Polt
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, 1306 E. University Blvd, Tucson, AZ 85721, USA; (C.R.A.); (L.Z.S.)
| |
Collapse
|
199
|
Lin D, Chen J, Duan K, Perrone-Bizzozero N, Sui J, Calhoun V, Liu J. Network modules linking expression and methylation in prefrontal cortex of schizophrenia. Epigenetics 2021; 16:876-893. [PMID: 33079616 PMCID: PMC8331039 DOI: 10.1080/15592294.2020.1827718] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/13/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023] Open
Abstract
Tremendous work has demonstrated the critical roles of genetics, epigenetics as well as their interplay in brain transcriptional regulations in the pathology of schizophrenia (SZ). There is great success currently in the dissection of the genetic components underlying risk-conferring transcriptomic networks. However, the study of regulating effect of epigenetics in the etiopathogenesis of SZ still faces many challenges. In this work, we investigated DNA methylation and gene expression from the dorsolateral prefrontal cortex (DLPFC) region of schizophrenia patients and healthy controls using weighted correlation network approach. We identified and replicated two expression and two methylation modules significantly associated with SZ. Among them, one pair of expression and methylation modules were significantly overlapped in the module genes which were significantly enriched in astrocyte-associated functional pathways, and specifically expressed in astrocytes. Another two linked expression-methylation module pairs were involved ageing process with module genes mostly related to oligodendrocyte development and myelination, and specifically expressed in oligodendrocytes. Further examination of underlying quantitative trait loci (QTLs) showed significant enrichment in genetic risk of most psychiatric disorders for expression QTLs but not for methylation QTLs. These results support the coherence between methylation and gene expression at the network level, and suggest a combinatorial effect of genetics and epigenetics in regulating gene expression networks specific to glia cells in relation to SZ and ageing process.
Collapse
Affiliation(s)
- Dongdong Lin
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS): {Georgia State University, Georgia Institute of Technology, and Emory University}, Atlanta, USA
| | - Jiayu Chen
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS): {Georgia State University, Georgia Institute of Technology, and Emory University}, Atlanta, USA
| | - Kuaikuai Duan
- Department of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, USA
| | - Nora Perrone-Bizzozero
- Department of Neurosciences, University of New Mexico, Albuquerque, NM, USA
- Department of Psychiatry, University of New Mexico, Albuquerque, NM, USA
| | - Jing Sui
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS): {Georgia State University, Georgia Institute of Technology, and Emory University}, Atlanta, USA
| | - Vince Calhoun
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS): {Georgia State University, Georgia Institute of Technology, and Emory University}, Atlanta, USA
- Department of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, USA
- Department of Psychology, Georgia State University, Atlanta, USA
- Department of Computer Science, Georgia State University, Atlanta, USA
| | - Jingyu Liu
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS): {Georgia State University, Georgia Institute of Technology, and Emory University}, Atlanta, USA
- Department of Computer Science, Georgia State University, Atlanta, USA
| |
Collapse
|
200
|
Yazar T, Olgun Yazar H, Cihan M. Evaluation of serum galectin-3 levels at Alzheimer patients by stages: a preliminary report. Acta Neurol Belg 2021; 121:949-954. [PMID: 32852752 DOI: 10.1007/s13760-020-01477-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/17/2020] [Indexed: 02/11/2023]
Abstract
BACKGROUND AND AIMS Neuroinflammation has a critic role in the pathophysiology of neurological diseases. The activation of microglia is the main actor in this process. The aim of this study to collect data on the role of microglial activation in the etiology, and the possible continuum at the stage of disease through the evaluation of serum galectin-3 levels in patients with Alzheimer's disease (AD). METHODS This was a prospective and cross-sectional study conducted on patients who were diagnosed as having AD using the criteria of the National Institute of Neurological and Communicative Disorders and Stroke-Alzheimer's Disease and Related Disorders Association (NINCDS-ADRDA) and stages determined with the scales of Clinical Dementia Rating (CDR) and Mini-Mental State Examination (MMSE) with healthy controls. RESULTS In our study, we studied 118 people, 57 with AD and 61 healthy people as a control group. In the AD patient group, serum galectin-3 levels were higher compared with the control group (p = 0.003). There were no significant differences in either group in other collected parameters (p > 0.05). It was observed that in all patients with AD, parallel to the stage of the disease, serum galectin-3 levels, patience's age, and duration of disease were statically and significantly increased (p < 0.05). CONCLUSION In conclusion, serum galactin-3 levels may be associated with AD and maybe a potential biomarker for the identification of disease in the early stages. In future years, serum galectin-3 levels may become an important biomarker and therapeutic agent for chronic neurodegenerative diseases such as AD.
Collapse
|