51
|
Maysinger D, Zhang I, Wu PY, Kagelmacher M, Luo HD, Kizhakkedathu JN, Dernedde J, Ballauff M, Haag R, Shobo A, Multhaup G, McKinney RA. Sulfated Hyperbranched and Linear Polyglycerols Modulate HMGB1 and Morphological Plasticity in Neural Cells. ACS Chem Neurosci 2023; 14:677-688. [PMID: 36717083 DOI: 10.1021/acschemneuro.2c00558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The objective of this study was to establish if polyglycerols with sulfate or sialic acid functional groups interact with high mobility group box 1 (HMGB1), and if so, which polyglycerol could prevent loss of morphological plasticity in excitatory neurons in the hippocampus. Considering that HMGB1 binds to heparan sulfate and that heparan sulfate has structural similarities with dendritic polyglycerol sulfates (dPGS), we performed the experiments to show if polyglycerols can mimic heparin functions by addressing the following questions: (1) do dendritic and linear polyglycerols interact with the alarmin molecule HMGB1? (2) Does dPGS interaction with HMGB1 influence the redox status of HMGB1? (3) Can dPGS prevent the loss of dendritic spines in organotypic cultures challenged with lipopolysaccharide (LPS)? LPS plays a critical role in infections with Gram-negative bacteria and is commonly used to test candidate therapeutic agents for inflammation and endotoxemia. Pathologically high LPS concentrations and other stressful stimuli cause HMGB1 release and post-translational modifications. We hypothesized that (i) electrostatic interactions of hyperbranched and linear polysulfated polyglycerols with HMGB1 will likely involve sites similar to those of heparan sulfate. (ii) dPGS can normalize HMGB1 compartmentalization in microglia exposed to LPS and prevent dendritic spine loss in the excitatory hippocampal neurons. We performed immunocytochemistry and biochemical analyses combined with confocal microscopy to determine cellular and extracellular locations of HMGB1 and morphological plasticity. Our results suggest that dPGS interacts with HMGB1 similarly to heparan sulfate. Hyperbranched dPGS and linear sulfated polymers prevent dendritic spine loss in hippocampal excitatory neurons. MS/MS analyses reveal that dPGS-HMGB1 interactions result in fully oxidized HMGB1 at critical cysteine residues (Cys23, Cys45, and Cys106). Triply oxidized HMGB1 leads to the loss of its pro-inflammatory action and could participate in dPGS-mediated spine loss prevention. LPG-Sia exposure to HMGB1 results in the oxidation of Cys23 and Cys106 but does not normalize spine density.
Collapse
Affiliation(s)
- Dusica Maysinger
- Department of Pharmacology and Therapeutics, McGill University, MontrealH3G 1Y6, Canada
| | - Issan Zhang
- Department of Pharmacology and Therapeutics, McGill University, MontrealH3G 1Y6, Canada
| | - Pei You Wu
- Department of Pharmacology and Therapeutics, McGill University, MontrealH3G 1Y6, Canada
| | - Marten Kagelmacher
- Institut für Chemie und Biochemie, Freie Universität Berlin, Berlin14195, Germany
| | - Haiming Daniel Luo
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, Life Science Institute, Department of Chemistry, School of Biomedical Engineering, University of British Columbia, VancouverV6T 1Z3, Canada
| | - Jayachandran N Kizhakkedathu
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, Life Science Institute, Department of Chemistry, School of Biomedical Engineering, University of British Columbia, VancouverV6T 1Z3, Canada
| | - Jens Dernedde
- Institute of Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Berlin13353, Germany
| | - Matthias Ballauff
- Institut für Chemie und Biochemie, Freie Universität Berlin, Berlin14195, Germany
| | - Rainer Haag
- Institut für Chemie und Biochemie, Freie Universität Berlin, Berlin14195, Germany
| | - Adeola Shobo
- Department of Pharmacology and Therapeutics, McGill University, MontrealH3G 1Y6, Canada
| | - Gerhard Multhaup
- Department of Pharmacology and Therapeutics, McGill University, MontrealH3G 1Y6, Canada
| | - R Anne McKinney
- Department of Pharmacology and Therapeutics, McGill University, MontrealH3G 1Y6, Canada
| |
Collapse
|
52
|
Hao L, Yang Y, Xu X, Guo X, Zhan Q. Modulatory effects of mesenchymal stem cells on microglia in ischemic stroke. Front Neurol 2023; 13:1073958. [PMID: 36742051 PMCID: PMC9889551 DOI: 10.3389/fneur.2022.1073958] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/28/2022] [Indexed: 01/20/2023] Open
Abstract
Ischemic stroke accounts for 70-80% of all stroke cases. Immunity plays an important role in the pathophysiology of ischemic stroke. Microglia are the first line of defense in the central nervous system. Microglial functions are largely dependent on their pro-inflammatory (M1-like) or anti-inflammatory (M2-like) phenotype. Modulating neuroinflammation via targeting microglia polarization toward anti-inflammatory phenotype might be a novel treatment for ischemic stroke. Mesenchymal stem cells (MSC) and MSC-derived extracellular vesicles (MSC-EVs) have been demonstrated to modulate microglia activation and phenotype polarization. In this review, we summarize the physiological characteristics and functions of microglia in the healthy brain, the activation and polarization of microglia in stroke brain, the effects of MSC/MSC-EVs on the activation of MSC in vitro and in vivo, and possible underlying mechanisms, providing evidence for a possible novel therapeutics for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Lei Hao
- Department of Neurology, The First Branch of The First Affiliated Hospital of Chongqing Medical University, Chongqing, China,Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China,Department of Neurology, The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Yongtao Yang
- Department of Neurology, The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Xiaoli Xu
- Department of Neurology, The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Xiuming Guo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China,*Correspondence: Xiuming Guo ✉
| | - Qunling Zhan
- Department of Neurology, The Fifth People's Hospital of Chongqing, Chongqing, China,Qunling Zhan ✉
| |
Collapse
|
53
|
Maurya SK, Gupta S, Mishra R. Transcriptional and epigenetic regulation of microglia in maintenance of brain homeostasis and neurodegeneration. Front Mol Neurosci 2023; 15:1072046. [PMID: 36698776 PMCID: PMC9870594 DOI: 10.3389/fnmol.2022.1072046] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/15/2022] [Indexed: 01/12/2023] Open
Abstract
The emerging role of microglia in brain homeostasis, neurodegeneration, and neurodevelopmental disorders has attracted considerable interest. In addition, recent developments in microglial functions and associated pathways have shed new light on their fundamental role in the immunological surveillance of the brain. Understanding the interconnections between microglia, neurons, and non-neuronal cells have opened up additional avenues for research in this evolving field. Furthermore, the study of microglia at the transcriptional and epigenetic levels has enhanced our knowledge of these native brain immune cells. Moreover, exploring various facets of microglia biology will facilitate the early detection, treatment, and management of neurological disorders. Consequently, the present review aimed to provide comprehensive insight on microglia biology and its influence on brain development, homeostasis, management of disease, and highlights microglia as potential therapeutic targets in neurodegenerative and neurodevelopmental diseases.
Collapse
Affiliation(s)
- Shashank Kumar Maurya
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India,*Correspondence: Shashank Kumar Maurya, ;
| | - Suchi Gupta
- Tech Cell Innovations Private Limited, Centre for Medical Innovation and Entrepreneurship (CMIE), All India Institute of Medical Sciences, New Delhi, India
| | - Rajnikant Mishra
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, India
| |
Collapse
|
54
|
Feng L, Lo H, You H, Wu W, Cheng X, Xin J, Ye Z, Chen X, Pan X. Loss of cannabinoid receptor 2 promotes α-Synuclein-induced microglial synaptic pruning in nucleus accumbens by modulating the pCREB-c-Fos signaling pathway and complement system. Exp Neurol 2023; 359:114230. [PMID: 36162511 DOI: 10.1016/j.expneurol.2022.114230] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/31/2022] [Accepted: 09/19/2022] [Indexed: 12/30/2022]
Abstract
The disruption of nucleus accumbens (NAc) function impacts mood and learning behavior in α-Synucleinopathy, in which microglial synaptic pruning plays a pivotal role in modulating the neuropathologic progression. Available literature documents that in microglia, the activation of cannabinoid receptor 2 (CB2R) decreases inflammation, but it remains obscured regarding the roles of CB2R in microglia-mediated synaptic pruning in the NAc during the neuropathological progression of α-Synucleinopathy. We adopted the fibrillar α-Synuclein (α-Syn) treatment to characterize the effect of genetic CB2R deletion on microglial function and the signaling pathway. CB2R knockout (CB2-/-) mice and wild-type (CB2+/+) mice were divided into the α-Syn or saline treatment groups. Biochemical and microscopy approaches, including immunofluorescence, real-time PCR, and western blotting, were employed to assess the changes in homeostasis of synaptic pruning in NAc under the α-Syn-induced microglia. Moreover, the underlying mechanisms of CB2R on α-Syn induced microglial activity was assessed in vitro. After the injection of α-Syn into the NAc, distinct microglial morphological changes and M1 phenotype transformation were observed between CB2-/- and CB2+/+ mice. Meanwhile, after the α-Syn treatment, CB2-/- mice showed an increased upregulation of CD68 protein and IL-1β mRNA but decreased brain-derived neurotrophic factor (BDNF) and TGF-β mRNA compared with CB2+/+ mice. Additionally, CB2-/- microglia after the treatment showed a highly enriched complement 3a receptor (C3aR) producing excessive pruning of cholinergic synapses but less engulfment of dopaminergic synapses. Mechanistically, the loss of CB2R function in the α-Syn stimulation triggered c-Fos activation in microglia, but not in neurons. Further inhibition of microglial CB2R functions under α-Syn stimulation activated the phosphorylated cAMP-response element-binding protein (pCREB)-c-Fos, which was closely related to the C3aR upregulation. Our results reveal a critical and mechanistic role of CB2R in altering the microglial function and its value in the homeostasis of synaptic circuits in the NAc under the α-Syn pathology.
Collapse
Affiliation(s)
- Linjuan Feng
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou 350001, China
| | - Hsuan Lo
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Hanlin You
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou 350001, China
| | - Wei Wu
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, 1 Xueyuan Road, Fuzhou 350001, China
| | - Xiaojuan Cheng
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou 350001, China
| | - Jiawei Xin
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China
| | - Zucheng Ye
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, 1 Xueyuan Road, Fuzhou 350001, China
| | - Xiaochun Chen
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou 350001, China
| | - Xiaodong Pan
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou 350001, China; Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, 1 Xueyuan Road, Fuzhou 350001, China.
| |
Collapse
|
55
|
Li H, Zhou B, Liao P, Liao D, Yang L, Wang J, Liu J, Jiang R, Chen L. Prolonged exposure of neonatal mice to sevoflurane leads to hyper-ramification in microglia, reduced contacts between microglia and synapses, and defects in adult behavior. Front Neurol 2023; 14:1142739. [PMID: 37025197 PMCID: PMC10072331 DOI: 10.3389/fneur.2023.1142739] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/24/2023] [Indexed: 04/08/2023] Open
Abstract
Background Prolonged exposure to general anesthetics during development is known to cause neurobehavioral abnormalities, but the cellular and molecular mechanisms involved are unclear. Microglia are the resident immune cells in the central nervous system and play essential roles in normal brain development. Materials and methods In the study, postnatal day 7 (P7) C57BL/6 mice were randomly assigned to two groups. In the sevoflurane (SEVO), mice were exposed to 2.5% sevoflurane for 4 h. In the control group, mice were exposed to carrier gas (30% O2/70% N2) for 4 h. Fixed brain slices from P14 to P21 mice were immunolabeled for ionized calcium-binding adapter molecule 1 (IBA-1) to visualize microglia. The morphological analysis of microglia in the somatosensory cortex was performed using ImageJ and Imaris software. Serial block face scanning electron microscopy (SBF-SEM) was performed to assess the ultrastructure of the microglia and the contacts between microglia and synapse in P14 and P21 mice. The confocal imaging of brain slices was performed to assess microglia surveillance in resting and activated states in P14 and P21 mice. Behavioral tests were used to assess the effect of microglia depletion and repopulation on neurobehavioral abnormalities caused by sevoflurane exposure. Results The prolonged exposure of neonatal mice to sevoflurane induced microglia hyper-ramification with an increase in total branch length, arborization area, and branch complexity 14 days after exposure. Prolonged neonatal sevoflurane exposure reduced contacts between microglia and synapses, without affecting the surveillance of microglia in the resting state or responding to laser-induced focal brain injury. These neonatal changes in microglia were associated with anxiety-like behaviors in adult mice. Furthermore, microglial depletion before sevoflurane exposure and subsequent repopulation in the neonatal brain mitigated anxiety-like behaviors caused by sevoflurane exposure. Conclusion Our experiments indicate that general anesthetics may harm the developing brain, and microglia may be an essential target of general anesthetic-related developmental neurotoxicity.
Collapse
Affiliation(s)
- Hong Li
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Bin Zhou
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Ping Liao
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Daqing Liao
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Linghui Yang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Wang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Ruotian Jiang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Ruotian Jiang,
| | - Lingmin Chen
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Lingmin Chen,
| |
Collapse
|
56
|
Qin J, Ma Z, Chen X, Shu S. Microglia activation in central nervous system disorders: A review of recent mechanistic investigations and development efforts. Front Neurol 2023; 14:1103416. [PMID: 36959826 PMCID: PMC10027711 DOI: 10.3389/fneur.2023.1103416] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 02/13/2023] [Indexed: 03/09/2023] Open
Abstract
Microglia are the principal resident immune cells in the central nervous system (CNS) and play important roles in the development of CNS disorders. In recent years, there have been significant developments in our understanding of microglia, and we now have greater insight into the temporal and spatial patterns of microglia activation in a variety of CNS disorders, as well as the interactions between microglia and neurons. A variety of signaling pathways have been implicated. However, to date, all published clinical trials have failed to demonstrate efficacy over placebo. This review summarizes the results of recent important studies and attempts to provide a mechanistic view of microglia activation, inflammation, tissue repair, and CNS disorders.
Collapse
|
57
|
Wang X, Liu X, Chen L, Zhang X. The inflammatory injury in the striatal microglia-dopaminergic-neuron crosstalk involved in Tourette syndrome development. Front Immunol 2023; 14:1178113. [PMID: 37187752 PMCID: PMC10175669 DOI: 10.3389/fimmu.2023.1178113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Background Tourette syndrome (TS) is associated with immunological dysfunction. The DA system is closely related to TS development, or behavioral stereotypes. Previous evidence suggested that hyper-M1-polarized microglia may exist in the brains of TS individuals. However, the role of microglia in TS and their interaction with dopaminergic neurons is unclear. In this study, we applied iminodipropionitrile (IDPN) to establish a TS model and focused on the inflammatory injury in the striatal microglia-dopaminergic-neuron crosstalk. Methods Male Sprague-Dawley rats were intraperitoneally injected with IDPN for seven consecutive days. Stereotypic behavior was observed to verify the TS model. Striatal microglia activation was evaluated based on different markers and expressions of inflammatory factors. The striatal dopaminergic neurons were purified and co-cultured with different microglia groups, and dopamine-associated markers were assessed. Results First, there was pathological damage to striatal dopaminergic neurons in TS rats, as indicated by decreased expression of TH, DAT, and PITX3. Next, the TS group showed a trend of increased Iba-1 positive cells and elevated levels of inflammatory factors TNF-α and IL-6, as well as an enhanced M1-polarization marker (iNOS) and an attenuated M2-polarization marker (Arg-1). Finally, in the co-culture experiment, IL-4-treated microglia could upregulate the expression of TH, DAT, and PITX3 in striatal dopaminergic neurons vs LPS-treated microglia. Similarly, the TS group (microglia from TS rats) caused a decreased expression of TH, DAT, and PITX3 compared with the Sham group (microglia from control rats) in the dopaminergic neurons. Conclusion In the striatum of TS rats, microglia activation is M1 hyperpolarized, which transmits inflammatory injury to striatal dopaminergic neurons and disrupts normal dopamine signaling.
Collapse
Affiliation(s)
- Xueming Wang
- Plastic Surgery Department, Fujian Children’s Hospital, Fuzhou, China
- Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiumei Liu
- Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Developmental and Behavior Pediatrics Department, Fujian Children’s Hospital, Fuzhou, China
- *Correspondence: Xiumei Liu,
| | - Liangliang Chen
- Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Developmental and Behavior Pediatrics Department, Fujian Children’s Hospital, Fuzhou, China
| | - Xiaoling Zhang
- Child Healthcare Department, Fuzhou Maternal and Child Health Hospital, Fuzhou, China
| |
Collapse
|
58
|
Xi L, Ji X, Ji W, Yang Y, Zhang Y, Long H. Jing-an oral liquid alleviates Tourette syndrome via the NMDAR/MAPK/CREB pathway in vivo and in vitro. PHARMACEUTICAL BIOLOGY 2022; 60:1790-1800. [PMID: 36102587 PMCID: PMC9487928 DOI: 10.1080/13880209.2022.2116056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/01/2022] [Accepted: 08/13/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT Jing-an oral liquid (JA) is a Chinese herbal formula used in the treatment of Tourette syndrome (TS); however, its mechanism is unclear. OBJECTIVE To investigate the effects of JA on amino acid neurotransmitters and microglia activation in vivo and in vitro. MATERIALS AND METHODS Sixty male Sprague-Dawley rats were divided into a control group and 5 TS groups. TS was induced in rats with intraperitoneal injection of 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (1 mg/kg) and in BV2 cells with lipopolysaccharide. Control and model rats were administered saline, whereas treatment groups were administered JA (5.18, 10.36, or 20.72 g/kg) or tiapride (a benzamide, 23.5 mg/kg) by gavage once daily for 21 days. Stereotypic behaviour was tested. The levels of N-methyl-d-aspartate receptor (NMDAR)/mitogen-activated protein kinase/cAMP response element-binding protein (CREB)-related proteins in striatum and BV2 cells were measured via western blots. CD11b and IBa1 levels were also measured. Ultra-high-performance liquid-chromatography was used to determine γ-aminobutyric acid (GABA), glutamic acid (Glu), and aspartic acid (ASP) levels. RESULTS JA markedly alleviated the stereotype behaviour (25.92 ± 0.35 to 13.78 ± 0.47) in rats. It also increased NMDAR1 (0.48 ± 0.09 to 0.67 ± 0.08; 0.54 ± 0.07 to 1.19 ± 0.18) expression and down-regulated the expression of p-ERK, p-JNK, p-P38, and p-CREB in BV2 cells and rat striatum. Additionally, Glu, ASP, GABA, CD11b, and IBa1 levels were significantly decreased by JA. DISCUSSION AND CONCLUSIONS JA suppressed microglia activation and regulated the levels of amino acid neurotransmitters, indicating that it could be a promising therapeutic agent for TS.
Collapse
Affiliation(s)
- Leying Xi
- Department of Pediatrics, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- Department of Pediatric, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xixi Ji
- Department of Pediatrics, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- Department of Pediatric, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenxiu Ji
- Department of Pediatrics, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- Department of Pediatric, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yue’e Yang
- Department of Pediatrics, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Yajie Zhang
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- Clinical Biobank of Nanjing Hospital of Chinese Medicine, Nanjing Hospital of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongyan Long
- Department of Pediatrics, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- Department of Pediatric, Nanjing University of Chinese Medicine, Nanjing, China
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- Clinical Biobank of Nanjing Hospital of Chinese Medicine, Nanjing Hospital of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
59
|
Mahony C, O'Ryan C. A molecular framework for autistic experiences: Mitochondrial allostatic load as a mediator between autism and psychopathology. Front Psychiatry 2022; 13:985713. [PMID: 36506457 PMCID: PMC9732262 DOI: 10.3389/fpsyt.2022.985713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 11/07/2022] [Indexed: 11/27/2022] Open
Abstract
Molecular autism research is evolving toward a biopsychosocial framework that is more informed by autistic experiences. In this context, research aims are moving away from correcting external autistic behaviors and toward alleviating internal distress. Autism Spectrum Conditions (ASCs) are associated with high rates of depression, suicidality and other comorbid psychopathologies, but this relationship is poorly understood. Here, we integrate emerging characterizations of internal autistic experiences within a molecular framework to yield insight into the prevalence of psychopathology in ASC. We demonstrate that descriptions of social camouflaging and autistic burnout resonate closely with the accepted definitions for early life stress (ELS) and chronic adolescent stress (CAS). We propose that social camouflaging could be considered a distinct form of CAS that contributes to allostatic overload, culminating in a pathophysiological state that is experienced as autistic burnout. Autistic burnout is thought to contribute to psychopathology via psychological and physiological mechanisms, but these remain largely unexplored by molecular researchers. Building on converging fields in molecular neuroscience, we discuss the substantial evidence implicating mitochondrial dysfunction in ASC to propose a novel role for mitochondrial allostatic load in the relationship between autism and psychopathology. An interplay between mitochondrial, neuroimmune and neuroendocrine signaling is increasingly implicated in stress-related psychopathologies, and these molecular players are also associated with neurodevelopmental, neurophysiological and neurochemical aspects of ASC. Together, this suggests an increased exposure and underlying molecular susceptibility to ELS that increases the risk of psychopathology in ASC. This article describes an integrative framework shaped by autistic experiences that highlights novel avenues for molecular research into mechanisms that directly affect the quality of life and wellbeing of autistic individuals. Moreover, this framework emphasizes the need for increased access to diagnoses, accommodations, and resources to improve mental health outcomes in autism.
Collapse
Affiliation(s)
| | - Colleen O'Ryan
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
60
|
Immunosenescence and Aging: Neuroinflammation Is a Prominent Feature of Alzheimer's Disease and Is a Likely Contributor to Neurodegenerative Disease Pathogenesis. J Pers Med 2022; 12:jpm12111817. [PMID: 36579548 PMCID: PMC9698256 DOI: 10.3390/jpm12111817] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/25/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Alzheimer's disease (AD) is a chronic multifactorial and complex neuro-degenerative disorder characterized by memory impairment and the loss of cognitive ability, which is a problem affecting the elderly. The pathological intracellular accumulation of abnormally phosphorylated Tau proteins, forming neurofibrillary tangles, and extracellular amyloid-beta (Aβ) deposition, forming senile plaques, as well as neural disconnection, neural death and synaptic dysfunction in the brain, are hallmark pathologies that characterize AD. The prevalence of the disease continues to increase globally due to the increase in longevity, quality of life, and medical treatment for chronic diseases that decreases the mortality and enhance the survival of elderly. Medical awareness and the accurate diagnosis of the disease also contribute to the high prevalence observed globally. Unfortunately, no definitive treatment exists that can be used to modify the course of AD, and no available treatment is capable of mitigating the cognitive decline or reversing the pathology of the disease as of yet. A plethora of hypotheses, ranging from the cholinergic theory and dominant Aβ cascade hypothesis to the abnormally excessive phosphorylated Tau protein hypothesis, have been reported. Various explanations for the pathogenesis of AD, such as the abnormal excitation of the glutamate system and mitochondrial dysfunction, have also been suggested. Despite the continuous efforts to deliver significant benefits and an effective treatment for this distressing, globally attested aging illness, multipronged approaches and strategies for ameliorating the disease course based on knowledge of the underpinnings of the pathogenesis of AD are urgently needed. Immunosenescence is an immune deficit process that appears with age (inflammaging process) and encompasses the remodeling of the lymphoid organs, leading to alterations in the immune function and neuroinflammation during advanced aging, which is closely linked to the outgrowth of infections, autoimmune diseases, and malignant cancers. It is well known that long-standing inflammation negatively influences the brain over the course of a lifetime due to the senescence of the immune system. Herein, we aim to trace the role of the immune system in the pathogenesis of AD. Thus, we explore alternative avenues, such as neuroimmune involvement in the pathogenesis of AD. We determine the initial triggers of neuroinflammation, which is an early episode in the pre-symptomatic stages of AD and contributes to the advancement of the disease, and the underlying key mechanisms of brain damage that might aid in the development of therapeutic strategies that can be used to combat this devastating disease. In addition, we aim to outline the ways in which different aspects of the immune system, both in the brain and peripherally, behave and thus to contribute to AD.
Collapse
|
61
|
Preeti K, Sood A, Fernandes V. Metabolic Regulation of Glia and Their Neuroinflammatory Role in Alzheimer's Disease. Cell Mol Neurobiol 2022; 42:2527-2551. [PMID: 34515874 PMCID: PMC11421648 DOI: 10.1007/s10571-021-01147-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 09/02/2021] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is an aging-related neurodegenerative disorder. It is characterized clinically by progressive memory loss and impaired cognitive function. Its progression occurs from neuronal synapse loss to amyloid pathology and Tau deposit which eventually leads to the compromised neuronal function. Neurons in central nervous tissue work in a composite and intricate network with the glia and vascular cells. Microglia and astrocytes are becoming the prime focus due to their involvement in various aspects of neurophysiology, such as trophic support to neurons, synaptic modulation, and brain surveillance. AD is also often considered as the sequela of prolonged metabolic dyshomeostasis. The neuron and glia have different metabolic profiles as cytosolic glycolysis and mitochondrial-dependent oxidative phosphorylation (OXPHOS), especially under dyshomeostasis or with aging pertaining to their unique genetic built-up. Various efforts are being put in to decipher the role of mitochondrial dynamics regarding their trafficking, fission/fusion imbalance, and mitophagy spanning over both neurons and glia to improve aging-related brain health. The mitochondrial dysfunction may lead to activation in various signaling mechanisms causing metabolic reprogramming in glia cells, further accelerating AD-related pathogenic events. The glycolytic-dominant astrocytes switch to the neurotoxic phenotype, i.e., disease-associated astrocyte under metabolic stress. The microglia also transform from resting to reactive phenotype, i.e., disease-associated microglia. It may also exist in otherwise a misconception an M1, glycolytic, or M2, an OXPHOS-dependent phenotype. Further, glial transformation plays a vital role in regulating hallmarks of AD pathologies like synapse maintenance, amyloid, and Tau clearance. In this updated review, we have tried to emphasize the metabolic regulation of glial reactivity, mitochondrial quality control mechanisms, and their neuroinflammatory response in Alzheimer's progression.
Collapse
Affiliation(s)
- Kumari Preeti
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India.
| | - Anika Sood
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Valencia Fernandes
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| |
Collapse
|
62
|
Yang G, Fan X, Mazhar M, Guo W, Zou Y, Dechsupa N, Wang L. Neuroinflammation of microglia polarization in intracerebral hemorrhage and its potential targets for intervention. Front Mol Neurosci 2022; 15:1013706. [PMID: 36304999 PMCID: PMC9592761 DOI: 10.3389/fnmol.2022.1013706] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS) and play a key role in neurological diseases, including intracerebral hemorrhage (ICH). Microglia are activated to acquire either pro-inflammatory or anti-inflammatory phenotypes. After the onset of ICH, pro-inflammatory mediators produced by microglia at the early stages serve as a crucial character in neuroinflammation. Conversely, switching the microglial shift to an anti-inflammatory phenotype could alleviate inflammatory response and incite recovery. This review will elucidate the dynamic profiles of microglia phenotypes and their available shift following ICH. This study can facilitate an understanding of the self-regulatory functions of the immune system involving the shift of microglia phenotypes in ICH. Moreover, suggestions for future preclinical and clinical research and potential intervention strategies are discussed.
Collapse
Affiliation(s)
- Guoqiang Yang
- Research Center for Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Acupuncture and Rehabilitation Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Xuehui Fan
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Maryam Mazhar
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Wubin Guo
- Department of General Surgery, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Yuanxia Zou
- Research Center for Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Nathupakorn Dechsupa
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- *Correspondence: Li Wang Nathupakorn Dechsupa
| | - Li Wang
- Research Center for Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
- *Correspondence: Li Wang Nathupakorn Dechsupa
| |
Collapse
|
63
|
Promoted CD4 + T cell-derived IFN-γ/IL-10 by photobiomodulation therapy modulates neurogenesis to ameliorate cognitive deficits in APP/PS1 and 3xTg-AD mice. J Neuroinflammation 2022; 19:253. [PMID: 36217178 PMCID: PMC9549637 DOI: 10.1186/s12974-022-02617-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 10/03/2022] [Indexed: 11/16/2022] Open
Abstract
Background The immune system has been implicated in synaptic plasticity, inflammation, and the progression of Alzheimer's disease (AD). However, there were few studies on improving the niche microenvironment of neural stem cells (NSCs) in the brain of AD to promote adult hippocampal neurogenesis (AHN) by regulating the function of non-parenchymal immune cells. Methods The lymph nodes of amyloid precursor protein/presenilin 1 (APP/PS1) and 3xTg (APP/PS1/tau) mouse models of AD were treated with photobiomodulation therapy (PBMT) for 10 J/cm2 per day for 1 month (10 min for each day), T lymphocytes isolated from these two AD models were treated with PBMT for 2 J/cm2 (5 min for each time). The NSCs isolated from hippocampus of these two AD models at E14, and the cells were co-cultivated with PBMT-treated T lymphocyte conditioned medium for NSCs differentiation. Results Our results showed that PBMT treatment could promote AHN and reverse cognitive deficits in AD mouse model. The expression of interferon-γ (IFN-γ) and interleukin-10 (IL-10) was upregulated in the brain of these two AD models after PBMT treated, which was induced by the activation of Janus kinase 2 (JAK2)-mediated signal transducer and activator of transcription 4 (STAT4)/STAT5 signaling pathway in CD4+ T cells. In addition, elevated CD4+ T cell levels and upregulated transforming growth factor-β1 (TGFβ1)/insulin-like growth factors-1 (IGF-1)/brain-derived neurotrophic factor (BDNF) protein expression levels were also detected in the brain. More importantly, co-cultivated the PBMT-treated T lymphocyte conditioned medium with NSCs derived from these two AD models was shown to promote NSCs differentiation, which was reflected in the upregulation of both neuronal class-III β-tubulin (Tuj1) and postsynaptic density protein 95 (PSD95), but the effects of PBMT was blocked by reactive oxygen species (ROS) scavenger or JAK2 inhibitor. Conclusion Our research suggests that PBMT exerts a beneficial neurogenesis modulatory effect through activating the JAK2/STAT4/STAT5 signaling pathway to promote the expression of IFN-γ/IL-10 in non-parenchymal CD4+ T cells, induction of improvement of brain microenvironmental conditions and alleviation of cognitive deficits in APP/PS1 and 3xTg-AD mouse models. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02617-5.
Collapse
|
64
|
Kumar P, Mishra J, Kumar N. Mechanistic Role of Jak3 in Obesity-Associated Cognitive Impairments. Nutrients 2022; 14:nu14183715. [PMID: 36145091 PMCID: PMC9505565 DOI: 10.3390/nu14183715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Background and Aims: A compromise in intestinal mucosal functions is associated with several chronic inflammatory diseases. Previously, we reported that obese humans have a reduced expression of intestinal Janus kinase-3 (Jak3), a non-receptor tyrosine kinase, and a deficiency of Jak3 in mice led to predisposition to obesity-associated metabolic syndrome. Since meta-analyses show cognitive impairment as co-morbidity of obesity, the present study demonstrates the mechanistic role of Jak3 in obesity associated cognitive impairment. Our data show that high-fat diet (HFD) suppresses Jak3 expression both in intestinal mucosa and in the brain of wild-type mice. Methodology: Recapitulating these conditions using global (Jak3-KO) and intestinal epithelial cell-specific conditional (IEC-Jak3-KO) mice and using cognitive testing, western analysis, flow cytometry, immunofluorescence microscopy and 16s rRNA sequencing, we demonstrate that HFD-induced Jak3 deficiency is responsible for cognitive impairments in mice, and these are, in part, specifically due to intestinal epithelial deficiency of Jak3. Results: We reveal that Jak3 deficiency leads to gut dysbiosis, compromised TREM-2-functions-mediated activation of microglial cells, increased TLR-4 expression and HIF1-α-mediated inflammation in the brain. Together, these lead to compromised microglial-functions-mediated increased deposition of β-amyloid (Aβ) and hyperphosphorylated Tau (pTau), which are responsible for cognitive impairments. Collectively, these data illustrate how the drivers of obesity promote cognitive impairment and demonstrate the underlying mechanism where HFD-mediated impact on IEC-Jak3 deficiency is responsible for Jak3 deficiency in the brain, reduced microglial TREM2 expression, microglial activation and compromised clearance of Aβ and pTau as the mechanism during obesity-associated cognitive impairments. Conclusion: Thus, we not only demonstrate the mechanism of obesity-associated cognitive impairments but also characterize the tissue-specific role of Jak3 in such conditions through mucosal tolerance, gut–brain axis and regulation of microglial functions.
Collapse
|
65
|
Gao C, Shen X, Tan Y, Chen S. Pathogenesis, therapeutic strategies and biomarker development based on "omics" analysis related to microglia in Alzheimer's disease. J Neuroinflammation 2022; 19:215. [PMID: 36058959 PMCID: PMC9441025 DOI: 10.1186/s12974-022-02580-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/27/2022] [Indexed: 11/10/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease and the most common cause of dementia. Among various pathophysiological aspects, microglia are considered to play important roles in the pathogenesis of AD. Genome wide association studies (GWAS) showed that the majority of AD risk genes are highly or exclusively expressed in microglia, underscoring the critical roles of microglia in AD pathogenesis. Recently, omics technologies have greatly advanced our knowledge of microglia biology in AD. Omics approaches, including genomics, epigenomics, transcriptomics, proteomics, and metabolomics/lipidomics, present remarkable opportunities to delineate the underlying mechanisms, discover novel diagnostic biomarkers, monitor disease progression, and shape therapeutic strategies for diseases. In this review, we summarized research based on microglial "omics" analysis in AD, especially the recent research advances in the identification of AD-associated microglial subsets. This review reinforces the important role of microglia in AD and advances our understanding of the mechanism of microglia in AD pathogenesis. Moreover, we proposed the value of microglia-based omics in the development of therapeutic strategies and biomarkers for AD.
Collapse
Affiliation(s)
- Chao Gao
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin Shen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuyan Tan
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, Shanghai, 201210, China.
| |
Collapse
|
66
|
Barragán-Álvarez CP, Flores-Fernandez JM, Hernández-Pérez OR, Ávila-Gónzalez D, Díaz NF, Padilla-Camberos E, Dublan-García O, Gómez-Oliván LM, Diaz-Martinez NE. Recent advances in the use of CRISPR/Cas for understanding the early development of molecular gaps in glial cells. Front Cell Dev Biol 2022; 10:947769. [PMID: 36120556 PMCID: PMC9479146 DOI: 10.3389/fcell.2022.947769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/01/2022] [Indexed: 12/03/2022] Open
Abstract
Glial cells are non-neuronal elements of the nervous system (NS) and play a central role in its development, maturation, and homeostasis. Glial cell interest has increased, leading to the discovery of novel study fields. The CRISPR/Cas system has been widely employed for NS understanding. Its use to study glial cells gives crucial information about their mechanisms and role in the central nervous system (CNS) and neurodegenerative disorders. Furthermore, the increasingly accelerated discovery of genes associated with the multiple implications of glial cells could be studied and complemented with the novel screening methods of high-content and single-cell screens at the genome-scale as Perturb-Seq, CRISP-seq, and CROPseq. Besides, the emerging methods, GESTALT, and LINNAEUS, employed to generate large-scale cell lineage maps have yielded invaluable information about processes involved in neurogenesis. These advances offer new therapeutic approaches to finding critical unanswered questions about glial cells and their fundamental role in the nervous system. Furthermore, they help to better understanding the significance of glial cells and their role in developmental biology.
Collapse
Affiliation(s)
- Carla Patricia Barragán-Álvarez
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño Del Estado de Jalisco, Guadalajara, Mexico
| | - José Miguel Flores-Fernandez
- Departamento de Investigación e Innovación, Universidad Tecnológica de Oriental, Oriental, Mexico
- Department of Biochemistry & Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
| | | | - Daniela Ávila-Gónzalez
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño Del Estado de Jalisco, Guadalajara, Mexico
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología, México City, Mexico
| | - Nestor Fabian Díaz
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología, México City, Mexico
| | - Eduardo Padilla-Camberos
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño Del Estado de Jalisco, Guadalajara, Mexico
| | - Octavio Dublan-García
- Laboratorio de Alimentos y Toxicología Ambiental, Facultad de Química, Universidad Autónoma Del Estado de México, Toluca, México
| | - Leobardo Manuel Gómez-Oliván
- Laboratorio de Alimentos y Toxicología Ambiental, Facultad de Química, Universidad Autónoma Del Estado de México, Toluca, México
| | - Nestor Emmanuel Diaz-Martinez
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño Del Estado de Jalisco, Guadalajara, Mexico
| |
Collapse
|
67
|
Ma'arif B, Muslikh FA, Amalia D, Mahardiani A, Muchlasi LA, Riwanti P, Taek MM, Laswati H, Agil M. Metabolite Profiling of the Environmental-Controlled Growth of Marsilea crenata Presl. and Its In Vitro and In Silico Antineuroinflammatory Properties. BORNEO JOURNAL OF PHARMACY 2022. [DOI: 10.33084/bjop.v5i3.3262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
This study was aimed to evaluate the metabolite contents and antineuroinflammatory potential of Marsilea crenata Presl. grown under a controlled environmental condition. The antineuroinflammatory test has been carried out in vitro using ethanolic extract of M. crenata leaves on HMC3 microglia cells. An in silico approach was applied to predict the active compounds of the extract. The HMC3 microglia cells were induced with IFNγ to create prolonged inflammatory conditions and then treated with 96% ethanolic extract of the M. crenata leaves of 62.5, 125, and 250 μg/mL. The expression of MHC II was analyzed using the ICC method with the CLSM instrument. Metabolites of the extract were profiled using UPLC-QToF-MS/MS instrument and MassLynx 4.1 software. In silico evaluation was conducted with molecular docking on 3OLS protein using PyRx 0.8 software, and physicochemical properties of the compounds were analyzed using SwissADME webtool. The ethanolic extract of M. crenata leaves could reduce the MHC II expression in HMC3 microglia cells in all concentrations with the values 97.458, 139.574, and 82.128 AU. The result of metabolite profiling found 79 compounds in the extract. In silico evaluation showed that 19 compounds gave agonist interaction toward 3OLS, and three met all parameters of physicochemical analysis. The ethanolic extract of the environmental-controlled growth of M. crenata leaves antineuroinflammatory activity on HMC3 microglia cells. The extract was predicted to contain some phytoestrogen compounds which act as 3OLS agonists.
Collapse
|
68
|
Rahman-Enyart A, Yaggie RE, Bollinger JL, Arvanitis C, Winter DR, Schaeffer AJ, Klumpp DJ. Acyloxyacyl hydrolase regulates microglia-mediated pelvic pain. PLoS One 2022; 17:e0269140. [PMID: 35980963 PMCID: PMC9387837 DOI: 10.1371/journal.pone.0269140] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 05/15/2022] [Indexed: 12/04/2022] Open
Abstract
Chronic pelvic pain conditions such as interstitial cystitis/bladder pain syndrome (IC/BPS) remain clinical and mechanistic enigmas. Microglia are resident immune cells of the central nervous system (CNS) that respond to changes in the gut microbiome, and studies have linked microglial activation to acute and chronic pain in a variety of models, including pelvic pain. We have previously reported that mice deficient for the lipase acyloxyacyl hydrolase (AOAH) develop pelvic allodynia and exhibit symptoms, comorbidities, and gut dysbiosis mimicking IC/BPS. Here, we assessed the role of AOAH in microglial activation and pelvic pain. RNAseq analyses using the ARCHS4 database and confocal microscopy revealed that AOAH is highly expressed in wild type microglia but at low levels in astrocytes, suggesting a functional role for AOAH in microglia. Pharmacologic ablation of CNS microglia with PLX5622 resulted in decreased pelvic allodynia in AOAH-deficient mice and resurgence of pelvic pain upon drug washout. Skeletal analyses revealed that AOAH-deficient mice have an activated microglia morphology in the medial prefrontal cortex and paraventricular nucleus, brain regions associated with pain modulation. Because microglia express Toll-like receptors and respond to microbial components, we also examine the potential role of dysbiosis in microglial activation. Consistent with our hypothesis of microglia activation by leakage of gut microbes, we observed increased serum endotoxins in AOAH-deficient mice and increased activation of cultured BV2 microglial cells by stool of AOAH-deficient mice. Together, these findings demonstrate a role for AOAH in microglial modulation of pelvic pain and thus identify a novel therapeutic target for IC/BPS.
Collapse
Affiliation(s)
- Afrida Rahman-Enyart
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
- Currently Proteintech Group Incorporated, Rosemont, Illinois, United States of America
| | - Ryan E. Yaggie
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Justin L. Bollinger
- Department of Pharmacology & Systems Physiology, College of Medicine University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Constadina Arvanitis
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
- Center for Advanced Microscopy & Nikon Imaging Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Deborah R. Winter
- Division of Rheumatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Anthony J. Schaeffer
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - David J. Klumpp
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
69
|
Yousefizadeh A, Piccioni G, Saidi A, Triaca V, Mango D, Nisticò R. Pharmacological targeting of microglia dynamics in Alzheimer's disease: Preclinical and clinical evidence. Pharmacol Res 2022; 184:106404. [PMID: 35988869 DOI: 10.1016/j.phrs.2022.106404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 10/15/2022]
Abstract
Numerous clinical trials of anti-amyloid agents for Alzheimer's disease (AD) were so far unsuccessful thereby challenging the validity of the amyloid hypothesis. This lack of progress has encouraged researchers to investigate alternative mechanisms in non-neuronal cells, among which microglia represent nowadays an attractive target. Microglia play a key role in the developing brain and contribute to synaptic remodeling in the mature brain. On the other hand, the intimate relationship between microglia and synapses led to the so-called synaptic stripping hypothesis, a process in which microglia selectively remove synapses from injured neurons. Synaptic stripping, along with the induction of a microglia-mediated chronic neuroinflammatory environment, promote the progressive synaptic degeneration in AD. Therefore, targeting microglia may pave the way for a new disease modifying approach. This review provides an overview of the pathophysiological roles of the microglia cells in AD and describes putative targets for pharmacological intervention. It also provides evidence for microglia-targeted strategies in preclinical AD studies and in early clinical trials.
Collapse
Affiliation(s)
- Atrin Yousefizadeh
- School of Pharmacy, Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Gaia Piccioni
- Department of Physiology and Pharmacology "V.Erspamer", Sapienza University of Rome, Rome, Italy; Laboratory Pharmacology of Synaptic Plasticity, European Brain Research (EBRI) Institute, Rome, Italy
| | - Amira Saidi
- Department of Physiology and Pharmacology "V.Erspamer", Sapienza University of Rome, Rome, Italy; Laboratory Pharmacology of Synaptic Plasticity, European Brain Research (EBRI) Institute, Rome, Italy
| | - Viviana Triaca
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Rome, Italy
| | - Dalila Mango
- School of Pharmacy, Department of Biology, University of Rome "Tor Vergata", Rome, Italy; Laboratory Pharmacology of Synaptic Plasticity, European Brain Research (EBRI) Institute, Rome, Italy
| | - Robert Nisticò
- School of Pharmacy, Department of Biology, University of Rome "Tor Vergata", Rome, Italy; Laboratory Pharmacology of Synaptic Plasticity, European Brain Research (EBRI) Institute, Rome, Italy.
| |
Collapse
|
70
|
Woo A, Botta A, Shi SSW, Paus T, Pausova Z. Obesity-Related Neuroinflammation: Magnetic Resonance and Microscopy Imaging of the Brain. Int J Mol Sci 2022; 23:8790. [PMID: 35955925 PMCID: PMC9368789 DOI: 10.3390/ijms23158790] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 12/01/2022] Open
Abstract
Obesity is a major risk factor of Alzheimer's disease and related dementias. The principal feature of dementia is a loss of neurons and brain atrophy. The mechanistic links between obesity and the neurodegenerative processes of dementias are not fully understood, but recent research suggests that obesity-related systemic inflammation and subsequent neuroinflammation may be involved. Adipose tissues release multiple proinflammatory molecules (fatty acids and cytokines) that impact blood and vessel cells, inducing low-grade systemic inflammation that can transition to tissues, including the brain. Inflammation in the brain-neuroinflammation-is one of key elements of the pathobiology of neurodegenerative disorders; it is characterized by the activation of microglia, the resident immune cells in the brain, and by the structural and functional changes of other cells forming the brain parenchyma, including neurons. Such cellular changes have been shown in animal models with direct methods, such as confocal microscopy. In humans, cellular changes are less tangible, as only indirect methods such as magnetic resonance (MR) imaging are usually used. In these studies, obesity and low-grade systemic inflammation have been associated with lower volumes of the cerebral gray matter, cortex, and hippocampus, as well as altered tissue MR properties (suggesting microstructural variations in cellular and molecular composition). How these structural variations in the human brain observed using MR imaging relate to the cellular variations in the animal brain seen with microscopy is not well understood. This review describes the current understanding of neuroinflammation in the context of obesity-induced systemic inflammation, and it highlights need for the bridge between animal microscopy and human MR imaging studies.
Collapse
Affiliation(s)
- Anita Woo
- The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Amy Botta
- The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Sammy S. W. Shi
- The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Tomas Paus
- Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montreal, QC H3T 1C5, Canada
- Departments of Psychiatry of Neuroscience, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
- Departments of Psychology and Psychiatry, University of Toronto, Toronto, ON M5S 1A1, Canada
- ECOGENE-21, Chicoutimi, QC G7H 7K9, Canada
| | - Zdenka Pausova
- The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON M5S 1A1, Canada
- ECOGENE-21, Chicoutimi, QC G7H 7K9, Canada
| |
Collapse
|
71
|
Badrah A, Al-Tuwairqi S. Modeling the dynamics of innate immune response to Parkinson disease with therapeutic approach. Phys Biol 2022; 19. [PMID: 35901788 DOI: 10.1088/1478-3975/ac8516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/28/2022] [Indexed: 11/12/2022]
Abstract
This paper aims to mathematically model the dynamics of Parkinson's disease with therapeutic strategies. The constructed model consists of five state variables: healthy neurons, infected neurons, extracellular $\alpha$-syn, active microglia, and resting microglia. The qualitative analysis of the model produced an unstable free equilibrium point and a stable endemic equilibrium point. Moreover, these results are validated by numerical experiments with different initial values. Two therapeutic interventions, reduction of extracellular $\alpha$-syn and reduction of inflammation induced by activated microglia in the central nervous system, are investigated. It is observed that the latter has no apparent effect in delaying the deterioration of neurons. However, treatment to reduce extracellular $\alpha$-syn preserves neurons and delays the onset of Parkinson's disease, whether alone or in combination with another treatment.
Collapse
Affiliation(s)
- Asma Badrah
- Mathematics, King Abdulaziz University, Jeddah, Saudi Arabia, Jeddah, 21589, SAUDI ARABIA
| | - Salma Al-Tuwairqi
- Mathematics, King Abdulaziz University, Jeddah, Saudi Arabia, Jeddah, 21589, SAUDI ARABIA
| |
Collapse
|
72
|
Solomon S, Sampathkumar NK, Carre I, Mondal M, Chennell G, Vernon AC, Ruepp MD, Mitchell JC. Heterozygous expression of the Alzheimer's disease-protective PLCγ2 P522R variant enhances Aβ clearance while preserving synapses. Cell Mol Life Sci 2022; 79:453. [PMID: 35895133 PMCID: PMC9329165 DOI: 10.1007/s00018-022-04473-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/29/2022] [Accepted: 07/05/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND A rare coding variant, P522R, in the phospholipase C gamma 2 (PLCG2) gene has been identified as protective against late-onset Alzheimer's disease (AD), but the mechanism is unknown. PLCG2 is exclusively expressed in microglia within the central nervous system, and altered microglial function has been implicated in the progression of AD. METHODS Healthy control hiPSCs were CRISPR edited to generate cells heterozygous and homozygous for the PLCG2P522R variant. Microglia derived from these hiPSC's were used to investigate the impact of PLCγ2P522R on disease relevant processes, specifically microglial capacity to take up amyloid beta (Aβ) and synapses. Targeted qPCR assessment was conducted to explore expression changes in core AD linked and microglial genes, and mitochondrial function was assessed using an Agilent Seahorse assay. RESULTS Heterozygous expression of the P522R variant resulted in increased microglial clearance of Aβ, while preserving synapses. This was associated with the upregulation of a number of genes, including the anti-inflammatory cytokine Il-10, and the synapse-linked CX3CR1, as well as alterations in mitochondrial function, and increased cellular motility. The protective capacity of PLCγ2P522R appeared crucially dependent on (gene) 'dose', as cells homozygous for the variant showed reduced synapse preservation, and a differential gene expression profile relative to heterozygous cells. CONCLUSION These findings suggest that PLCγ2P522R may result in increased surveillance by microglia, and prime them towards an anti-inflammatory state, with an increased capacity to respond to increasing energy demands, but highlights the delicate balance of this system, with increasing PLCγ2P522R 'dose' resulting in reduced beneficial impacts.
Collapse
Affiliation(s)
- Shiden Solomon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- UK Dementia Research Institute, King’s College London, London, UK
| | - Nirmal Kumar Sampathkumar
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- UK Dementia Research Institute, King’s College London, London, UK
- Present Address: Alzheimer’s Research UK Oxford Drug Discovery Institute, Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Ivo Carre
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- UK Dementia Research Institute, King’s College London, London, UK
| | - Mrityunjoy Mondal
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- UK Dementia Research Institute, King’s College London, London, UK
| | - George Chennell
- Wohl Cellular Imaging Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Anthony C. Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, UK
| | - Marc-David Ruepp
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- UK Dementia Research Institute, King’s College London, London, UK
| | - Jacqueline C Mitchell
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| |
Collapse
|
73
|
Seitkazina A, Kim KH, Fagan E, Sung Y, Kim YK, Lim S. The Fate of Tau Aggregates Between Clearance and Transmission. Front Aging Neurosci 2022; 14:932541. [PMID: 35923541 PMCID: PMC9339952 DOI: 10.3389/fnagi.2022.932541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/22/2022] [Indexed: 11/30/2022] Open
Abstract
Neuronal accumulation of mis-folded tau is the pathological hallmark of multiple neurodegenerative disorders, including Alzheimer’s disease. Distinct from amyloid plaques, which appear simultaneously throughout the brain, tau pathology develops first in a specific brain region and then propagates to neuroanatomically connected brain regions, exacerbating the disease. Due to the implication in disease progression, prevention of tau transmission is recognized as an important therapeutic strategy that can halt disease progression in the brain. Recently, accumulating studies have demonstrated diverse cellular mechanisms associated with cell-to-cell transmission of tau. Once transmitted, mis-folded tau species act as a prion-like seed for native tau aggregation in the recipient neuron. In this review, we summarize the diverse cellular mechanisms associated with the secretion and uptake of tau, and highlight tau-trafficking receptors, which mediate tau clearance or cell-to-cell tau transmission.
Collapse
Affiliation(s)
- Assel Seitkazina
- Convergence Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, Korea Institute of Science and Technology (KIST) School, University of Science and Technology (UST), Seoul, South Korea
| | - Kyu Hyeon Kim
- Convergence Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, Korea Institute of Science and Technology (KIST) School, University of Science and Technology (UST), Seoul, South Korea
| | - Erin Fagan
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, United States
| | - Yoonsik Sung
- Convergence Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, Korea Institute of Science and Technology (KIST) School, University of Science and Technology (UST), Seoul, South Korea
| | - Yun Kyung Kim
- Convergence Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, Korea Institute of Science and Technology (KIST) School, University of Science and Technology (UST), Seoul, South Korea
- *Correspondence: Yun Kyung Kim,
| | - Sungsu Lim
- Convergence Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Sungsu Lim,
| |
Collapse
|
74
|
de Araújo FM, Cuenca-Bermejo L, Fernández-Villalba E, Costa SL, Silva VDA, Herrero MT. Role of Microgliosis and NLRP3 Inflammasome in Parkinson's Disease Pathogenesis and Therapy. Cell Mol Neurobiol 2022; 42:1283-1300. [PMID: 33387119 PMCID: PMC11421755 DOI: 10.1007/s10571-020-01027-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder marked primarily by motor symptoms such as rigidity, bradykinesia, postural instability and resting tremor associated with dopaminergic neuronal loss in the Substantia Nigra pars compacta (SNpc) and deficit of dopamine in the basal ganglia. These motor symptoms can be preceded by pre-motor symptoms whose recognition can be useful to apply different strategies to evaluate risk, early diagnosis and prevention of PD progression. Although clinical characteristics of PD are well defined, its pathogenesis is still not completely known, what makes discoveries of therapies capable of curing patients difficult to be reached. Several theories about the cause of idiopathic PD have been investigated and among them, the key role of inflammation, microglia and the inflammasome in the pathogenesis of PD has been considered. In this review, we describe the role and relation of both the inflammasome and microglial activation with the pathogenesis, symptoms, progression and the possibilities for new therapeutic strategies in PD.
Collapse
Affiliation(s)
- Fillipe M de Araújo
- Clinical and Experimental Neuroscience (NiCE), Institute for Bio-Health Research of Murcia (IMIB), Institute for Aging Research (IUIE), School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, Spain
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, 40110-100, Brazil
| | - Lorena Cuenca-Bermejo
- Clinical and Experimental Neuroscience (NiCE), Institute for Bio-Health Research of Murcia (IMIB), Institute for Aging Research (IUIE), School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, Spain
| | - Emiliano Fernández-Villalba
- Clinical and Experimental Neuroscience (NiCE), Institute for Bio-Health Research of Murcia (IMIB), Institute for Aging Research (IUIE), School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, Spain
| | - Silvia L Costa
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, 40110-100, Brazil.
| | - Victor Diogenes A Silva
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, 40110-100, Brazil.
| | - Maria Trinidad Herrero
- Clinical and Experimental Neuroscience (NiCE), Institute for Bio-Health Research of Murcia (IMIB), Institute for Aging Research (IUIE), School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, Spain.
| |
Collapse
|
75
|
Roles of Fatty Acids in Microglial Polarization: Evidence from In Vitro and In Vivo Studies on Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms23137300. [PMID: 35806302 PMCID: PMC9266841 DOI: 10.3390/ijms23137300] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
Microglial polarization to the M1 phenotype (classically activated) or the M2 phenotype (alternatively activated) is critical in determining the fate of immune responses in neurodegenerative diseases (NDs). M1 macrophages contribute to neurotoxicity, neuronal and synaptic damage, and oxidative stress and are the first line of defense, and M2 macrophages elicit an anti-inflammatory response to regulate neuroinflammation, clear cell debris, and promote neuroregeneration. Various studies have focused on the ability of natural compounds to promote microglial polarization from the M1 phenotype to the M2 phenotype in several diseases, including NDs. However, studies on the roles of fatty acids in microglial polarization and their implications in NDs are a rare find. Most of the studies support the role of polyunsaturated fatty acids (PUFAs) in microglial polarization using cell and animal models. Thus, we aimed to collect data and provide a narrative account of microglial types, markers, and studies pertaining to fatty acids, particularly PUFAs, on microglial polarization and their neuroprotective effects. The involvement of only PUFAs in the chosen topic necessitates more in-depth research into the role of unexplored fatty acids in microglial polarization and their mechanistic implications. The review also highlights limitations and future challenges.
Collapse
|
76
|
Ascorbic acid along with ciprofloxacin regulates S. aureus induced microglial inflammatory responses and oxidative stress through TLR-2 and glucocorticoid receptor modulation. Inflammopharmacology 2022; 30:1303-1322. [PMID: 35704229 DOI: 10.1007/s10787-022-01012-z] [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: 10/21/2021] [Accepted: 05/13/2022] [Indexed: 11/05/2022]
Abstract
Microglial inflammatory responses play a central role in the pathogenesis of S. aureus induced brain infections. Upon activation, microglia produces free radicals (ROS/RNS) and disrupts the cellular antioxidant defense to combat invading microorganisms. Despite conventional antibiotic or steroid therapy, microglial over-activation could not be controlled. So, an attempt had been taken by using a natural antioxidant ascorbic acid along with ciprofloxacin to regulate microglial over-activation by involving TLR-2 and glucocorticoid receptor (GR) in an in-vitro cell culture-based study. Combinatorial treatment during TLR-2 neutralization effectively reduced the bacterial burden at 60 min compared to the GR blocking condition (p < 0.05). Moreover, the infection-induced H2O2, O2.-, and NO release in microglial cell culture was diminished possibly by enhancing SOD and catalase activities in the same condition (p < 0.05). The arginase activity was markedly increased after TLR-2 blocking in the combinatorial group compared to single treatments (p < 0.05). Experimental results indicated that combinatorial treatment may act through up-regulating GR expression by augmenting endogenous corticosterone levels. However, better bacterial clearance could further suppress the TLR-2 mediated pro-inflammatory NF-κB signaling. From Western blot analysis, it was concluded that ciprofloxacin-ascorbic acid combination in presence of anti-TLR-2 antibody exhibited 81.25% inhibition of TLR-2 expression while the inhibition for GR was 3.57% with respect to the infected group. Therefore, during TLR-2 blockade ascorbic acid combination might be responsible for the restoration of redox balance in microglia via modulating TLR-2/GR interaction. The combination treatment could play a major role in the neuroendocrine-immune regulation of S. aureus induced microglial activation.
Collapse
|
77
|
Lima MN, Barbosa-Silva MC, Maron-Gutierrez T. Microglial Priming in Infections and Its Risk to Neurodegenerative Diseases. Front Cell Neurosci 2022; 16:878987. [PMID: 35783096 PMCID: PMC9240317 DOI: 10.3389/fncel.2022.878987] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/26/2022] [Indexed: 11/29/2022] Open
Abstract
Infectious diseases of different etiologies have been associated with acute and long-term neurological consequences. The primary cause of these consequences appears to be an inflammatory process characterized primarily by a pro-inflammatory microglial state. Microglial cells, the local effectors' cells of innate immunity, once faced by a stimulus, alter their morphology, and become a primary source of inflammatory cytokines that increase the inflammatory process of the brain. This inflammatory scenario exerts a critical role in the pathogenesis of neurodegenerative diseases. In recent years, several studies have shown the involvement of the microglial inflammatory response caused by infections in the development of neurodegenerative diseases. This has been associated with a transitory microglial state subsequent to an inflammatory response, known as microglial priming, in which these cells are more responsive to stimuli. Thus, systemic inflammation and infections induce a transitory state in microglia that may lead to changes in their state and function, making priming them for subsequent immune challenges. However, considering that microglia are long-lived cells and are repeatedly exposed to infections during a lifetime, microglial priming may not be beneficial. In this review, we discuss the relationship between infections and neurodegenerative diseases and how this may rely on microglial priming.
Collapse
Affiliation(s)
- Maiara N. Lima
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, Brazil
| | - Maria C. Barbosa-Silva
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, Brazil
| | - Tatiana Maron-Gutierrez
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation, Rio de Janeiro, Brazil
| |
Collapse
|
78
|
Rahmani B, Ghashghayi E, Zendehdel M, Baghbanzadeh A, Khodadadi M. Molecular mechanisms highlighting the potential role of COVID-19 in the development of neurodegenerative diseases. Physiol Int 2022; 109:135-162. [DOI: 10.1556/2060.2022.00019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/17/2022] [Accepted: 04/11/2022] [Indexed: 01/08/2023]
Abstract
Abstract
Coronavirus disease 2019 (COVID-19) is a contagious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In addition to the pulmonary manifestations, COVID-19 patients may present a wide range of neurological disorders as extrapulmonary presentations. In this view, several studies have recently documented the worsening of neurological symptoms within COVID-19 morbidity in patients previously diagnosed with neurodegenerative diseases (NDs). Moreover, several cases have also been reported in which the patients presented parkinsonian features after initial COVID-19 symptoms. These data raise a major concern about the possibility of communication between SARS-CoV-2 infection and the initiation and/or worsening of NDs. In this review, we have collected compelling evidence suggesting SARS-CoV-2, as an environmental factor, may be capable of developing NDs. In this respect, the possible links between SARS-CoV-2 infection and molecular pathways related to most NDs and the pathophysiological mechanisms of the NDs such as Alzheimer's disease, vascular dementia, frontotemporal dementia, Parkinson's disease, and amyotrophic lateral sclerosis will be explained.
Collapse
Affiliation(s)
- Behrouz Rahmani
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, 14155-6453 Tehran, Iran
| | - Elham Ghashghayi
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, 14155-6453 Tehran, Iran
| | - Morteza Zendehdel
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, 14155-6453 Tehran, Iran
| | - Ali Baghbanzadeh
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, 14155-6453 Tehran, Iran
| | - Mina Khodadadi
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, 14155-6453 Tehran, Iran
| |
Collapse
|
79
|
Tabaa MME, Aboalazm HM, Shaalan M, Khedr NF. Silymarin constrains diacetyl-prompted oxidative stress and neuroinflammation in rats: involvements of Dyn/GDNF and MAPK signaling pathway. Inflammopharmacology 2022; 30:961-980. [PMID: 35366745 PMCID: PMC9135832 DOI: 10.1007/s10787-022-00961-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/25/2022] [Indexed: 11/26/2022]
Abstract
Neuroinflammation, a major component of many CNS disorders, has been suggested to be associated with diacetyl (DA) exposure. DA is commonly used as a food flavoring additive and condiment. Lately, silymarin (Sily) has shown protective and therapeutic effects on neuronal inflammation. The study aimed to explore the role of Sily in protecting and/or treating DA-induced neuroinflammation. Neuroinflammation was induced in rats by administering DA (25 mg/kg) orally. Results revealed that Sily (50 mg/kg) obviously maintained cognitive and behavioral functions, alleviated brain antioxidant status, and inhibited microglial activation. Sily enhanced IL-10, GDNF and Dyn levels, reduced IFN-γ, TNFα, and IL-1β levels, and down-regulated the MAPK pathway. Immunohistochemical investigation of EGFR and GFAP declared that Sily could conserve neurons from inflammatory damage. However, with continuing DA exposure during Sily treatment, oxidative stress and neuroinflammation were less mitigated. These findings point to a novel mechanism involving the Dyn/GDNF and MAPK pathway through which Sily might prevent and treat DA-induced neuroinflammation.
Collapse
Affiliation(s)
- Manar Mohammed El Tabaa
- Pharmacology & Environmental Toxicology, Environmental Studies & Research Institute (ESRI), University of Sadat City, Minofia Governorate, Sadat city, Egypt
| | - Hamdi M. Aboalazm
- Biochemistry, Environmental Studies & Research Institute (ESRI), University of Sadat City, Sadat City, Egypt
| | - Mohamed Shaalan
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | | |
Collapse
|
80
|
Polarization of Microglia and Its Therapeutic Potential in Sepsis. Int J Mol Sci 2022; 23:ijms23094925. [PMID: 35563317 PMCID: PMC9101892 DOI: 10.3390/ijms23094925] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 02/04/2023] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection, leaving the inflammation process without a proper resolution, leading to tissue damage and possibly sequelae. The central nervous system (CNS) is one of the first regions affected by the peripheral inflammation caused by sepsis, exposing the neurons to an environment of oxidative stress, triggering neuronal dysfunction and apoptosis. Sepsis-associated encephalopathy (SAE) is the most frequent sepsis-associated organ dysfunction, with symptoms such as deliriums, seizures, and coma, linked to increased mortality, morbidity, and cognitive disability. However, the current therapy does not avoid those patients’ symptoms, evidencing the search for a more optimal approach. Herein we focus on microglia as a prominent therapeutic target due to its multiple functions maintaining CNS homeostasis and its polarizing capabilities, stimulating and resolving neuroinflammation depending on the stimuli. Microglia polarization is a target of multiple studies involving nerve cell preservation in diseases caused or aggravated by neuroinflammation, but in sepsis, its therapeutic potential is overlooked. We highlight the peroxisome proliferator-activated receptor gamma (PPARγ) neuroprotective properties, its role in microglia polarization and inflammation resolution, and the interaction with nuclear factor-κB (NF-κB) and mitogen-activated kinases (MAPK), making PPARγ a molecular target for sepsis-related studies to come.
Collapse
|
81
|
Choe K, Park HY, Ikram M, Lee HJ, Park TJ, Ullah R, Kim MO. Systematic Review of the Common Pathophysiological Mechanisms in COVID-19 and Neurodegeneration: The Role of Bioactive Compounds and Natural Antioxidants. Cells 2022; 11:cells11081298. [PMID: 35455977 PMCID: PMC9031507 DOI: 10.3390/cells11081298] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/08/2022] [Accepted: 04/09/2022] [Indexed: 02/06/2023] Open
Abstract
The novel coronavirus (2019-nCoVCOVID-19) belongs to the Beta coronavirus family, which contains MERS-CoV (Middle East respiratory syndrome coronavirus) and SARS-CoV (severe acute respiratory syndrome coronavirus). SARS-CoV-2 activates the innate immune system, thereby activating the inflammatory mechanism, causing the release of inflammatory cytokines. Moreover, it has been suggested that COVID-19 may penetrate the central nervous system, and release inflammatory cytokines in the brains, inducing neuroinflammation and neurodegeneration. Several links connect COVID-19 with Alzheimer’s disease (AD), such as elevated oxidative stress, uncontrolled release of the inflammatory cytokines, and mitochondrial apoptosis. There are severe concerns that excessive immune cell activation in COVID-19 may aggravate the neurodegeneration and amyloid-beta pathology of AD. Here, we have collected the evidence, showing the links between the two diseases. The focus has been made to collect the information on the activation of the inflammation, its contributors, and shared therapeutic targets. Furthermore, we have given future perspectives, research gaps, and overlapping pathological bases of the two diseases. Lastly, we have given the short touch to the drugs that have equally shown rescuing effects against both diseases. Although there is limited information available regarding the exact links between COVID-19 and neuroinflammation, we have insight into the pathological contributors of the diseases. Based on the shared pathological features and therapeutic targets, we hypothesize that the activation of the immune system may induce neurological disorders by triggering oxidative stress and neuroinflammation.
Collapse
Affiliation(s)
- Kyonghwan Choe
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (K.C.); (M.I.); (H.J.L.); (R.U.)
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Hyun Young Park
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands;
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht Medical Center, 6229 ER Maastricht, The Netherlands
| | - Muhammad Ikram
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (K.C.); (M.I.); (H.J.L.); (R.U.)
| | - Hyeon Jin Lee
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (K.C.); (M.I.); (H.J.L.); (R.U.)
| | - Tae Ju Park
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences (MVLS), University of Glasgow, Glasgow G12 0ZD, UK;
| | - Rahat Ullah
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (K.C.); (M.I.); (H.J.L.); (R.U.)
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (K.C.); (M.I.); (H.J.L.); (R.U.)
- Alz-Dementia Korea Co., Jinju 52828, Korea
- Correspondence: ; Tel.: +82-55-772-1345; Fax: +82-55-772-2656
| |
Collapse
|
82
|
Farooq RK, Alamoudi W, Alhibshi A, Rehman S, Sharma AR, Abdulla FA. Varied Composition and Underlying Mechanisms of Gut Microbiome in Neuroinflammation. Microorganisms 2022; 10:705. [PMID: 35456757 PMCID: PMC9032006 DOI: 10.3390/microorganisms10040705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/21/2022] [Accepted: 03/17/2022] [Indexed: 11/16/2022] Open
Abstract
The human gut microbiome has been implicated in a host of bodily functions and their regulation, including brain development and cognition. Neuroinflammation is a relatively newer piece of the puzzle and is implicated in the pathogenesis of many neurological disorders. The microbiome of the gut may alter the inflammatory signaling inside the brain through the secretion of short-chain fatty acids, controlling the availability of amino acid tryptophan and altering vagal activation. Studies in Korea and elsewhere highlight a strong link between microbiome dynamics and neurocognitive states, including personality. For these reasons, re-establishing microbial flora of the gut looks critical for keeping neuroinflammation from putting the whole system aflame through probiotics and allotransplantation of the fecal microbiome. However, the numerosity of the microbiome remains a challenge. For this purpose, it is suggested that wherever possible, a fecal microbial auto-transplant may prove more effective. This review summarizes the current knowledge about the role of the microbiome in neuroinflammation and the various mechanism involved in this process. As an example, we have also discussed the autism spectrum disorder and the implication of neuroinflammation and microbiome in its pathogenesis.
Collapse
Affiliation(s)
- Rai Khalid Farooq
- Department of Neuroscience Research, Institute of Research and Medical Consultations, Imam Abdul Rahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (W.A.); (A.A.); (F.A.A.)
| | - Widyan Alamoudi
- Department of Neuroscience Research, Institute of Research and Medical Consultations, Imam Abdul Rahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (W.A.); (A.A.); (F.A.A.)
| | - Amani Alhibshi
- Department of Neuroscience Research, Institute of Research and Medical Consultations, Imam Abdul Rahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (W.A.); (A.A.); (F.A.A.)
| | - Suriya Rehman
- Department of Epidemic Diseases Research, Institute of Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si 24252, Gangwon-do, Korea;
| | - Fuad A. Abdulla
- Department of Neuroscience Research, Institute of Research and Medical Consultations, Imam Abdul Rahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (W.A.); (A.A.); (F.A.A.)
- Department of Physical Therapy, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, P.O. Box 2435, Dammam 31441, Saudi Arabia
| |
Collapse
|
83
|
Alfarhan M, Liu F, Shan S, Pichavaram P, Somanath PR, Narayanan SP. Pharmacological Inhibition of Spermine Oxidase Suppresses Excitotoxicity Induced Neuroinflammation in Mouse Retina. Int J Mol Sci 2022; 23:2133. [PMID: 35216248 PMCID: PMC8875684 DOI: 10.3390/ijms23042133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 02/07/2023] Open
Abstract
Polyamine oxidation plays a major role in neurodegenerative diseases. Previous studies from our laboratory demonstrated that spermine oxidase (SMOX, a member of the polyamine oxidase family) inhibition using MDL 72527 reduced neurodegeneration in models of retinal excitotoxicity and diabetic retinopathy. However, the mechanisms behind the neuroprotection offered by SMOX inhibition are not completely studied. Utilizing the experimental model of retinal excitotoxicity, the present study determined the impact of SMOX blockade in retinal neuroinflammation. Our results demonstrated upregulation in the number of cells positive for Iba-1 (ionized calcium-binding adaptor molecule 1), CD (Cluster Differentiation) 68, and CD16/32 in excitotoxicity-induced retinas, while MDL 72527 treatment reduced these changes, along with increases in the number of cells positive for Arginase1 and CD206. When retinal excitotoxicity upregulated several pro-inflammatory genes, MDL 72527 treatment reduced many of them and increased anti-inflammatory genes. Furthermore, SMOX inhibition upregulated antioxidant signaling (indicated by elevated Nrf2 and HO-1 levels) and reduced protein-conjugated acrolein in excitotoxic retinas. In vitro studies using C8-B4 cells showed changes in cellular morphology and increased reactive oxygen species formation in response to acrolein (a product of SMOX activity) treatment. Overall, our findings indicate that the inhibition SMOX pathway reduced neuroinflammation and upregulated antioxidant signaling in the retina.
Collapse
Affiliation(s)
- Moaddey Alfarhan
- Clinical and Experimental Therapeutics Program, Department of Clinical and Administrative Pharmacy, University of Georgia, Augusta, GA 30912, USA; (M.A.); (F.L.); (S.S.); (P.R.S.)
- Research Division, Charlie Norwood VA Medical Center, Augusta, GA 30901, USA
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA;
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Fang Liu
- Clinical and Experimental Therapeutics Program, Department of Clinical and Administrative Pharmacy, University of Georgia, Augusta, GA 30912, USA; (M.A.); (F.L.); (S.S.); (P.R.S.)
- Research Division, Charlie Norwood VA Medical Center, Augusta, GA 30901, USA
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA;
| | - Shengshuai Shan
- Clinical and Experimental Therapeutics Program, Department of Clinical and Administrative Pharmacy, University of Georgia, Augusta, GA 30912, USA; (M.A.); (F.L.); (S.S.); (P.R.S.)
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA;
| | | | - Payaningal R. Somanath
- Clinical and Experimental Therapeutics Program, Department of Clinical and Administrative Pharmacy, University of Georgia, Augusta, GA 30912, USA; (M.A.); (F.L.); (S.S.); (P.R.S.)
- Research Division, Charlie Norwood VA Medical Center, Augusta, GA 30901, USA
| | - S. Priya Narayanan
- Clinical and Experimental Therapeutics Program, Department of Clinical and Administrative Pharmacy, University of Georgia, Augusta, GA 30912, USA; (M.A.); (F.L.); (S.S.); (P.R.S.)
- Research Division, Charlie Norwood VA Medical Center, Augusta, GA 30901, USA
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA;
| |
Collapse
|
84
|
Sanchez K, Darling JS, Kakkar R, Wu SL, Zentay A, Lowry CA, Fonken LK. Mycobacterium vaccae immunization in rats ameliorates features of age-associated microglia activation in the amygdala and hippocampus. Sci Rep 2022; 12:2165. [PMID: 35140249 PMCID: PMC8828872 DOI: 10.1038/s41598-022-05275-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 01/05/2022] [Indexed: 12/14/2022] Open
Abstract
Aging and reduced exposure to environmental microbes can both potentiate neuroinflammatory responses. Prior studies indicate that immunization with the immunoregulatory and anti-inflammatory bacterium, Mycobacterium vaccae (M. vaccae), in aged rats limits neuroimmune activation and cognitive impairments. However, the mechanisms by which M. vaccae immunization ameliorates age-associated neuroinflammatory “priming” and whether microglia are a primary target remain unclear. Here, we investigated whether M. vaccae immunization protects against microglia morphological changes in response to aging. Adult (3 mos) and aged (24 mos) Fisher 344 × Brown Norway rats were immunized with either M. vaccae or vehicle once every week for 3 weeks. Aging led to elevated Iba1 immunoreactivity, microglial density, and deramification of microglia processes in the hippocampus and amygdala but not other brain regions. Additionally, aged rats exhibited larger microglial somas in the dorsal hippocampus, suggestive of a more activated phenotype. Notably, M. vaccae treatment ameliorated indicators of microglia activation in both the amygdala and hippocampus. While changes in morphology appeared to be region-specific, gene markers indicative of microglia activation were upregulated by age and lowered in response to M. vaccae in all brain regions evaluated. Taken together, these data suggest that peripheral immunization with M. vaccae quells markers of age-associated microglia activation.
Collapse
Affiliation(s)
- Kevin Sanchez
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, 107 W Dean Keeton St 3.510C, Austin, TX, 78712, USA
| | - Jeffrey S Darling
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, 107 W Dean Keeton St 3.510C, Austin, TX, 78712, USA
| | - Reha Kakkar
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, 107 W Dean Keeton St 3.510C, Austin, TX, 78712, USA
| | - Sienna L Wu
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, 107 W Dean Keeton St 3.510C, Austin, TX, 78712, USA
| | - Andrew Zentay
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, 107 W Dean Keeton St 3.510C, Austin, TX, 78712, USA
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Laura K Fonken
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, 107 W Dean Keeton St 3.510C, Austin, TX, 78712, USA.
| |
Collapse
|
85
|
Merging pruning and neuroevolution: towards robust and efficient controllers for modular soft robots. KNOWL ENG REV 2022. [DOI: 10.1017/s0269888921000151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Abstract
Artificial neural networks (ANNs) can be employed as controllers for robotic agents. Their structure is often complex, with many neurons and connections, especially when the robots have many sensors and actuators distributed across their bodies and/or when high expressive power is desirable. Pruning (removing neurons or connections) reduces the complexity of the ANN, thus increasing its energy efficiency, and has been reported to improve the generalization capability, in some cases. In addition, it is well-known that pruning in biological neural networks plays a fundamental role in the development of brains and their ability to learn. In this study, we consider the evolutionary optimization of neural controllers for the case study of Voxel-based soft robots, a kind of modular, bio-inspired soft robots, applying pruning during fitness evaluation. For a locomotion task, and for centralized as well as distributed controllers, we experimentally characterize the effect of different forms of pruning on after-pruning effectiveness, life-long effectiveness, adaptability to new terrains, and behavior. We find that incorporating some forms of pruning in neuroevolution leads to almost equally effective controllers as those evolved without pruning, with the benefit of higher robustness to pruning. We also observe occasional improvements in generalization ability.
Collapse
|
86
|
Ayub M, Jin HK, Bae JS. Sphingosine kinase-dependent regulation of pro-resolving lipid mediators in Alzheimer's disease. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159126. [DOI: 10.1016/j.bbalip.2022.159126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/03/2022] [Indexed: 12/14/2022]
|
87
|
Acid-Sensing Ion Channels in Glial Cells. MEMBRANES 2022; 12:membranes12020119. [PMID: 35207041 PMCID: PMC8878633 DOI: 10.3390/membranes12020119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/29/2021] [Accepted: 01/17/2022] [Indexed: 12/13/2022]
Abstract
Acid-sensing ion channels (ASICs) are proton-gated cation channels and key mediators of responses to neuronal injury. ASICs exhibit unique patterns of distribution in the brain, with high expression in neurons and low expression in glial cells. While there has been a lot of focus on ASIC in neurons, less is known about the roles of ASICs in glial cells. ASIC1a is expressed in astrocytes and might contribute to synaptic transmission and long-term potentiation. In oligodendrocytes, constitutive activation of ASIC1a participates in demyelinating diseases. ASIC1a, ASIC2a, and ASIC3, found in microglial cells, could mediate the inflammatory response. Under pathological conditions, ASIC dysregulation in glial cells can contribute to disease states. For example, activation of astrocytic ASIC1a may worsen neurodegeneration and glioma staging, activation of microglial ASIC1a and ASIC2a may perpetuate ischemia and inflammation, while oligodendrocytic ASIC1a might be involved in multiple sclerosis. This review concentrates on the unique ASIC components in each of the glial cells and integrates these glial-specific ASICs with their physiological and pathological conditions. Such knowledge provides promising evidence for targeting of ASICs in individual glial cells as a therapeutic strategy for a diverse range of conditions.
Collapse
|
88
|
Rohm TV, Meier DT, Olefsky JM, Donath MY. Inflammation in obesity, diabetes, and related disorders. Immunity 2022; 55:31-55. [PMID: 35021057 PMCID: PMC8773457 DOI: 10.1016/j.immuni.2021.12.013] [Citation(s) in RCA: 858] [Impact Index Per Article: 286.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/13/2021] [Accepted: 12/17/2021] [Indexed: 01/13/2023]
Abstract
Obesity leads to chronic, systemic inflammation and can lead to insulin resistance (IR), β-cell dysfunction, and ultimately type 2 diabetes (T2D). This chronic inflammatory state contributes to long-term complications of diabetes, including non-alcoholic fatty liver disease (NAFLD), retinopathy, cardiovascular disease, and nephropathy, and may underlie the association of type 2 diabetes with other conditions such as Alzheimer's disease, polycystic ovarian syndrome, gout, and rheumatoid arthritis. Here, we review the current understanding of the mechanisms underlying inflammation in obesity, T2D, and related disorders. We discuss how chronic tissue inflammation results in IR, impaired insulin secretion, glucose intolerance, and T2D and review the effect of inflammation on diabetic complications and on the relationship between T2D and other pathologies. In this context, we discuss current therapeutic options for the treatment of metabolic disease, advances in the clinic and the potential of immune-modulatory approaches.
Collapse
Affiliation(s)
- Theresa V. Rohm
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniel T. Meier
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, CH-4031 Basel, Switzerland.,Department of Biomedicine (DBM), University of Basel, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Jerrold M. Olefsky
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Marc Y. Donath
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, CH-4031 Basel, Switzerland.,Department of Biomedicine (DBM), University of Basel, University Hospital Basel, CH-4031 Basel, Switzerland.,Correspondence:
| |
Collapse
|
89
|
Jonas LA, Jain T, Li YM. Functional insight into LOAD-associated microglial response genes. Open Biol 2022; 12:210280. [PMID: 35078351 PMCID: PMC8790339 DOI: 10.1098/rsob.210280] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/01/2021] [Indexed: 01/07/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by the presence of amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs), neuronal and synaptic loss and inflammation of the central nervous system (CNS). The majority of AD research has been dedicated to the understanding of two major AD hallmarks (i.e. Aβ and NFTs); however, recent genome-wide association studies (GWAS) data indicate neuroinflammation as having a critical role in late-onset AD (LOAD) development, thus unveiling a novel avenue for AD therapeutics. Recent evidence has provided much support to the innate immune system's involvement with AD progression; however, much remains to be uncovered regarding the role of glial cells, specifically microglia, in AD. Moreover, numerous variants in immune and/or microglia-related genes have been identified in whole-genome sequencing and GWAS analyses, including such genes as TREM2, CD33, APOE, API1, MS4A, ABCA7, BIN1, CLU, CR1, INPP5D, PICALM and PLCG2. In this review, we aim to provide an insight into the function of the major LOAD-associated microglia response genes.
Collapse
Affiliation(s)
- Lauren A. Jonas
- Weill Cornell, Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10065, USA
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tanya Jain
- Weill Cornell, Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10065, USA
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yue-Ming Li
- Weill Cornell, Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10065, USA
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
90
|
Haidar MA, Ibeh S, Shakkour Z, Reslan MA, Nwaiwu J, Moqidem YA, Sader G, Nickles RG, Babale I, Jaffa AA, Salama M, Shaito A, Kobeissy F. Crosstalk between Microglia and Neurons in Neurotrauma: An Overview of the Underlying Mechanisms. Curr Neuropharmacol 2022; 20:2050-2065. [PMID: 34856905 PMCID: PMC9886840 DOI: 10.2174/1570159x19666211202123322] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 11/22/2022] Open
Abstract
Microglia are the resident immune cells of the brain and play a crucial role in housekeeping and maintaining homeostasis of the brain microenvironment. Upon injury or disease, microglial cells become activated, at least partly, via signals initiated by injured neurons. Activated microglia, thereby, contribute to both neuroprotection and neuroinflammation. However, sustained microglial activation initiates a chronic neuroinflammatory response which can disturb neuronal health and disrupt communications between neurons and microglia. Thus, microglia-neuron crosstalk is critical in a healthy brain as well as during states of injury or disease. As most studies focus on how neurons and microglia act in isolation during neurotrauma, there is a need to understand the interplay between these cells in brain pathophysiology. This review highlights how neurons and microglia reciprocally communicate under physiological conditions and during brain injury and disease. Furthermore, the modes of microglia-neuron communication are exposed, focusing on cell-contact dependent signaling and communication by the secretion of soluble factors like cytokines and growth factors. In addition, it has been discussed that how microglia-neuron interactions could exert either beneficial neurotrophic effects or pathologic proinflammatory responses. We further explore how aberrations in microglia-neuron crosstalk may be involved in central nervous system (CNS) anomalies, namely traumatic brain injury (TBI), neurodegeneration, and ischemic stroke. A clear understanding of how the microglia-neuron crosstalk contributes to the pathogenesis of brain pathologies may offer novel therapeutic avenues of brain trauma treatment.
Collapse
Affiliation(s)
- Muhammad Ali Haidar
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Stanley Ibeh
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Zaynab Shakkour
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mohammad Amine Reslan
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Judith Nwaiwu
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Yomna Adel Moqidem
- Biotechnology Program, School of Science and Engineering, The American University in Cairo, Cairo, Egypt
| | - Georgio Sader
- Faculty of Medicine, University of Balamand, Balamand, Lebanon
| | - Rachel G. Nickles
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Ismail Babale
- Department of Biomedical Engineering, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Aneese A. Jaffa
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Beirut, Lebanon
| | - Mohamed Salama
- Institute of Global Health and Human Ecology (I-GHHE), The American University in Cairo, New Cairo 11835, Egypt
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
| | - Abdullah Shaito
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Biomedical Engineering, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
91
|
Chen K, Stieger KC, Kozai TD. Challenges and opportunities of advanced gliomodulation technologies for excitation-inhibition balance of brain networks. Curr Opin Biotechnol 2021; 72:112-120. [PMID: 34773740 PMCID: PMC8671375 DOI: 10.1016/j.copbio.2021.10.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 10/02/2021] [Accepted: 10/16/2021] [Indexed: 12/13/2022]
Abstract
Recent neuroscience studies have highlighted the critical role of glial cells in information processing. This has increased the demand for technologies that selectively modulate glial cells that regulate the excitation-inhibition balance of neural network function. Engineered technologies that modulate glial activity may be necessary for precise tuning of neural network activity in higher-order brain function. This perspective summarizes how glial cells regulate excitation and inhibition of neural circuits, highlights available technologies for glial modulation, and discusses current challenges and potential opportunities for glial engineering technologies.
Collapse
Affiliation(s)
- Keying Chen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kevin C Stieger
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takashi Dy Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA.
| |
Collapse
|
92
|
Rapaka D, Bitra VR, Challa SR, Adiukwu PC. Potentiation of microglial endocannabinoid signaling alleviates neuroinflammation in Alzheimer's disease. Neuropeptides 2021; 90:102196. [PMID: 34508923 DOI: 10.1016/j.npep.2021.102196] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) isaprogressive neurodegenerative disorder characterized by chronic inflammation due to the presence of neurotoxic Aβ and tau proteins. Increased microglial activation and inflated immune response are the other factors to be considered in AD pathology. Microglial cells own biochemical machinery that synthesizes and release endocannabinoids. The exploitation of therapeutic actions of endocannabinoid system has newly emerged in the field of Alzheimer's disease. The activation of cannabinoid receptors/ cannabinoid system modulates inflammatory responses. This review assesses the association between the microglial endocannabinoid system and neuroinflammation in AD. The data supporting the anti-inflammatory role of pharmacological agents modulating cannabinoid system are also reviewed.
Collapse
Affiliation(s)
- Deepthi Rapaka
- A.U. College of Pharmaceutical Sciences, Andhra University, Visakhapatnam 530003, India.
| | | | - Siva Reddy Challa
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL 61614, USA
| | - Paul C Adiukwu
- School of Pharmacy, University of Botswana, P/Bag-0022, Gaborone, Botswana
| |
Collapse
|
93
|
Doremus-Fitzwater TL, Deak T. Adolescent neuroimmune function and its interaction with alcohol. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 161:167-208. [PMID: 34801169 DOI: 10.1016/bs.irn.2021.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Adolescence is an evolutionarily conserved developmental period associated with behavioral change, including increased risk-taking and alcohol use. Experimentation with alcohol typically begins in adolescence and transitions to binge-like patterns of consumption. Alcohol exposure during adolescence can alter normative changes in brain structure and function. Understanding mechanisms by which ethanol impacts neurodevelopmental processes is important for preventing and ameliorating the deleterious consequences of adolescent alcohol abuse. This review focuses on the neuroimmune system as a key contributor to ethanol-induced changes in adolescent brain and behavior. After brief review of neuroimmune system development, acute and chronic effects of ethanol on adolescent neuroimmune functioning are addressed. Comparisons between stress/immunological challenges and ethanol on adolescent neuroimmunity are reviewed, as cross-sensitization is relevant during adolescence. The mechanisms by which ethanol alters neuroimmune functioning are then discussed, as they may portend development of neuropathological consequences and thus increase vulnerability to subsequent challenges and potentiate addictive behaviors.
Collapse
Affiliation(s)
- T L Doremus-Fitzwater
- Department of Psychology, Ithaca College, Ithaca, NY, United States; Developmental Exposure Alcohol Research Center (DEARC), Binghamton, NY, United States.
| | - T Deak
- Developmental Exposure Alcohol Research Center (DEARC), Binghamton, NY, United States; Binghamton University-SUNY, Binghamton, NY, United States
| |
Collapse
|
94
|
Kikel-Coury NL, Brandt JP, Correia IA, O’Dea MR, DeSantis DF, Sterling F, Vaughan K, Ozcebe G, Zorlutuna P, Smith CJ. Identification of astroglia-like cardiac nexus glia that are critical regulators of cardiac development and function. PLoS Biol 2021; 19:e3001444. [PMID: 34793438 PMCID: PMC8601506 DOI: 10.1371/journal.pbio.3001444] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 10/18/2021] [Indexed: 01/09/2023] Open
Abstract
Glial cells are essential for functionality of the nervous system. Growing evidence underscores the importance of astrocytes; however, analogous astroglia in peripheral organs are poorly understood. Using confocal time-lapse imaging, fate mapping, and mutant genesis in a zebrafish model, we identify a neural crest-derived glial cell, termed nexus glia, which utilizes Meteorin signaling via Jak/Stat3 to drive differentiation and regulate heart rate and rhythm. Nexus glia are labeled with gfap, glast, and glutamine synthetase, markers that typically denote astroglia cells. Further, analysis of single-cell sequencing datasets of human and murine hearts across ages reveals astrocyte-like cells, which we confirm through a multispecies approach. We show that cardiac nexus glia at the outflow tract are critical regulators of both the sympathetic and parasympathetic system. These data establish the crucial role of glia on cardiac homeostasis and provide a description of nexus glia in the PNS.
Collapse
Affiliation(s)
- Nina L. Kikel-Coury
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Jacob P. Brandt
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Isabel A. Correia
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Michael R. O’Dea
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Dana F. DeSantis
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Felicity Sterling
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Kevin Vaughan
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Gulberk Ozcebe
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Pinar Zorlutuna
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Cody J. Smith
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, United States of America
| |
Collapse
|
95
|
Andoh NE, Gyan BA. The Potential Roles of Glial Cells in the Neuropathogenesis of Cerebral Malaria. Front Cell Infect Microbiol 2021; 11:741370. [PMID: 34692564 PMCID: PMC8529055 DOI: 10.3389/fcimb.2021.741370] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/17/2021] [Indexed: 01/02/2023] Open
Abstract
Cerebral malaria (CM) is a severe neurological complication of malaria caused by the Plasmodium falciparum parasite. It is one of the leading causes of death in children under 5 years of age in Sub-Saharan Africa. CM is associated with blood-brain barrier disruption and long-term neurological sequelae in survivors of CM. Despite the vast amount of research on cerebral malaria, the cause of neurological sequelae observed in CM patients is poorly understood. In this article, the potential roles of glial cells, astrocytes, and microglia, in cerebral malaria pathogenesis are reviewed. The possible mechanisms by which glial cells contribute to neurological damage in CM patients are also examined.
Collapse
Affiliation(s)
- Nana Efua Andoh
- Noguchi Memorial Institute for Medical Research, Department of Parasitology, University of Ghana, Accra, Ghana
| | - Ben Adu Gyan
- Noguchi Memorial Institute for Medical Research, Department of Immunology, University of Ghana, Accra, Ghana
| |
Collapse
|
96
|
The Need for New Biomarkers to Assist with Stroke Prevention and Prediction of Post-Stroke Therapy Based on Plasma-Derived Extracellular Vesicles. Biomedicines 2021; 9:biomedicines9091226. [PMID: 34572411 PMCID: PMC8466486 DOI: 10.3390/biomedicines9091226] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/07/2021] [Accepted: 09/13/2021] [Indexed: 12/24/2022] Open
Abstract
The risk of having a stroke event doubles each decade after the age of 55. Therefore, it is of great interest to develop neurorestorative therapies of stroke which occurs mostly in elderly people. However, to date, patients at risk for these sequels of stroke are not duly diagnosed and treated due to the lack of reliable biomarkers. Extracellular vesicles (EVs) are lipid bilayer-delimited particles that are shed by the brain cells and are able to cross the blood–brain barrier and enter the blood stream; thus, they may be used to interrogate molecular and cellular events in the brain damaged area. In this review, we summarize the major molecular and cellular responses of astroglia and neurons to cerebral ischemia and assess their impact on post-stroke recovery and rehabilitation. In particular, we ask if EVs secreted by brain cells are responses to cerebral ischemia, and they may shed new light on the interplay between exosomes-mediated interactions between brain cells and the question of how to exploit it in order to predict the individual course of the disease and to introduce specific preventive or therapeutic strategies. Given these findings, we are left with two options: either to (i) transplant neuronal precursors into the damaged cortical area or (ii) to covert abundantly present proliferating astrocytes in the perilesional area into neurons by using recently developed genetic technologies. However, given the complexity of molecular and cellular responses to cerebral ischemia and our limited capabilities to restore brain structure and function, we are left with only one realistic aim: to invest more in prevention.
Collapse
|
97
|
Cornell J, Salinas S, Huang HY, Zhou M. Microglia regulation of synaptic plasticity and learning and memory. Neural Regen Res 2021; 17:705-716. [PMID: 34472455 PMCID: PMC8530121 DOI: 10.4103/1673-5374.322423] [Citation(s) in RCA: 225] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Microglia are the resident macrophages of the central nervous system. Microglia possess varied morphologies and functions. Under normal physiological conditions, microglia mainly exist in a resting state and constantly monitor their microenvironment and survey neuronal and synaptic activity. Through the C1q, C3 and CR3 “Eat Me” and CD47 and SIRPα “Don’t Eat Me” complement pathways, as well as other pathways such as CX3CR1 signaling, resting microglia regulate synaptic pruning, a process crucial for the promotion of synapse formation and the regulation of neuronal activity and synaptic plasticity. By mediating synaptic pruning, resting microglia play an important role in the regulation of experience-dependent plasticity in the barrel cortex and visual cortex after whisker removal or monocular deprivation, and also in the regulation of learning and memory, including the modulation of memory strength, forgetfulness, and memory quality. As a response to brain injury, infection or neuroinflammation, microglia become activated and increase in number. Activated microglia change to an amoeboid shape, migrate to sites of inflammation and secrete proteins such as cytokines, chemokines and reactive oxygen species. These molecules released by microglia can lead to synaptic plasticity and learning and memory deficits associated with aging, Alzheimer’s disease, traumatic brain injury, HIV-associated neurocognitive disorder, and other neurological or mental disorders such as autism, depression and post-traumatic stress disorder. With a focus mainly on recently published literature, here we reviewed the studies investigating the role of resting microglia in synaptic plasticity and learning and memory, as well as how activated microglia modulate disease-related plasticity and learning and memory deficits. By summarizing the function of microglia in these processes, we aim to provide an overview of microglia regulation of synaptic plasticity and learning and memory, and to discuss the possibility of microglia manipulation as a therapeutic to ameliorate cognitive deficits associated with aging, Alzheimer’s disease, traumatic brain injury, HIV-associated neurocognitive disorder, and mental disorders.
Collapse
Affiliation(s)
- Jessica Cornell
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Shelbi Salinas
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Hou-Yuan Huang
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Miou Zhou
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
98
|
Dziabis JE, Bilbo SD. Microglia and Sensitive Periods in Brain Development. Curr Top Behav Neurosci 2021; 53:55-78. [PMID: 34463934 DOI: 10.1007/7854_2021_242] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
From embryonic neuronal migration to adolescent circuit refinement, the immune system plays an essential role throughout central nervous system (CNS) development. Immune signaling molecules serve as a common language between the immune system and CNS, allowing them to work together to modulate brain function both in health and disease. As the resident CNS macrophage, microglia comprise the majority of immune cells in the brain. Much like their peripheral counterparts, microglia survey their environment for pathology, clean up debris, and propagate inflammatory responses when necessary. Beyond this, recent studies have highlighted that microglia perform a number of complex tasks during neural development, from directing neuronal and axonal positioning to pruning synapses, receptors, and even whole cells. In this chapter, we discuss this literature within the framework that immune activation during discrete windows of neural development can profoundly impact brain function long-term, and thus the risk of neurodevelopmental and neuropsychiatric disorders. In this chapter, we review three sensitive developmental periods - embryonic wiring, early postnatal synaptic pruning, and adolescent circuit refinement - in order to highlight the diversity of functions that microglia perform in building a brain. In reviewing this literature, it becomes obvious that timing matters, perhaps more so than the nature of the immune activation itself; largely conserved patterns of microglial response to diverse insults result in different functional impacts depending on the stage of brain maturation at the time of the challenge.
Collapse
Affiliation(s)
- Julia E Dziabis
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA.,Department of Neurobiology, Duke University, Durham, NC, USA
| | - Staci D Bilbo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA. .,Department of Neurobiology, Duke University, Durham, NC, USA.
| |
Collapse
|
99
|
Piec PA, Pons V, Rivest S. Triggering Innate Immune Receptors as New Therapies in Alzheimer's Disease and Multiple Sclerosis. Cells 2021; 10:cells10082164. [PMID: 34440933 PMCID: PMC8393987 DOI: 10.3390/cells10082164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis and Alzheimer's disease are two complex neurodegenerative diseases involving the immune system. So far, available treatments provide at best mild improvements to patients' conditions. For decades now, a new set of molecules have been used to modulate and regulate the innate immunity in these pathologies. Most studies have been carried out in rodents and some of them have reported tremendous beneficial effects on the disease course. The modulation of innate immune cells is of great interest since it provides new hope for patients. In this review, we will briefly overview the therapeutic potential of some molecules and receptors in multiple sclerosis and Alzheimer's disease and how they could be used to exploit new therapeutic avenues.
Collapse
|
100
|
Alam M, Yadav RK, Minj E, Tiwari A, Mehan S. Exploring Molecular Approaches in Amyotrophic Lateral Sclerosis: Drug Targets from Clinical and Pre-Clinical Findings. Curr Mol Pharmacol 2021; 14:263-280. [PMID: 32342825 DOI: 10.2174/1566524020666200427214356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/24/2019] [Accepted: 12/26/2019] [Indexed: 11/22/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disease (MND) characterized by the death of upper and lower motor neurons (corticospinal tract) in the motor cortex, basal ganglia, brain stem, and spinal cord. The patient experiences the sign and symptoms between 55 to 75 years of age, which include impaired motor movement, difficulty in speaking and swallowing, grip loss, muscle atrophy, spasticity, and sometimes associated with memory and cognitive impairments. Median survival is 3 to 5 years after diagnosis and 5 to 10% of the patients live for more than 10 years. The limited intervention of pharmacologically active compounds, that are used clinically, is majorly associated with the narrow therapeutic index. Pre-clinically established experimental models, where neurotoxin methyl mercury mimics the ALS like behavioural and neurochemical alterations in rodents associated with neuronal mitochondrial dysfunctions and downregulation of adenyl cyclase mediated cAMP/CREB, is the main pathological hallmark for the progression of ALS in central as well in the peripheral nervous system. Despite the considerable investigation into neuroprotection, it still constrains treatment choices to strong care and organization of ALS complications. Therefore, this current review specially targeted the investigation of clinical and pre-clinical features available for ALS to understand the pathogenic mechanisms and to explore the pharmacological interventions associated with the up-regulation of intracellular adenyl cyclase/cAMP/ CREB and activation of mitochondrial-ETC coenzyme-Q10 as a future drug target in the amelioration of ALS mediated motor neuronal dysfunctions.
Collapse
Affiliation(s)
- Mamtaj Alam
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Rajeshwar K Yadav
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Elizabeth Minj
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Aarti Tiwari
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Sidharth Mehan
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| |
Collapse
|