1
|
Pramanik S, Devi M H, Chakrabarty S, Paylar B, Pradhan A, Thaker M, Ayyadhury S, Manavalan A, Olsson PE, Pramanik G, Heese K. Microglia signaling in health and disease - Implications in sex-specific brain development and plasticity. Neurosci Biobehav Rev 2024; 165:105834. [PMID: 39084583 DOI: 10.1016/j.neubiorev.2024.105834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/21/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
Microglia, the intrinsic neuroimmune cells residing in the central nervous system (CNS), exert a pivotal influence on brain development, homeostasis, and functionality, encompassing critical roles during both aging and pathological states. Recent advancements in comprehending brain plasticity and functions have spotlighted conspicuous variances between male and female brains, notably in neurogenesis, neuronal myelination, axon fasciculation, and synaptogenesis. Nevertheless, the precise impact of microglia on sex-specific brain cell plasticity, sculpting diverse neural network architectures and circuits, remains largely unexplored. This article seeks to unravel the present understanding of microglial involvement in brain development, plasticity, and function, with a specific emphasis on microglial signaling in brain sex polymorphism. Commencing with an overview of microglia in the CNS and their associated signaling cascades, we subsequently probe recent revelations regarding molecular signaling by microglia in sex-dependent brain developmental plasticity, functions, and diseases. Notably, C-X3-C motif chemokine receptor 1 (CX3CR1), triggering receptors expressed on myeloid cells 2 (TREM2), calcium (Ca2+), and apolipoprotein E (APOE) emerge as molecular candidates significantly contributing to sex-dependent brain development and plasticity. In conclusion, we address burgeoning inquiries surrounding microglia's pivotal role in the functional diversity of developing and aging brains, contemplating their potential implications for gender-tailored therapeutic strategies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Subrata Pramanik
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| | - Harini Devi M
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Saswata Chakrabarty
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Berkay Paylar
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Ajay Pradhan
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Manisha Thaker
- Eurofins Lancaster Laboratories, Inc., 2425 New Holland Pike, Lancaster, PA 17601, USA
| | - Shamini Ayyadhury
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Arulmani Manavalan
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India
| | - Per-Erik Olsson
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Gopal Pramanik
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India.
| | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 133791, the Republic of Korea.
| |
Collapse
|
2
|
Islam R, Choudhary H, Rajan R, Vrionis F, Hanafy KA. An overview on microglial origin, distribution, and phenotype in Alzheimer's disease. J Cell Physiol 2024; 239:e30829. [PMID: 35822939 PMCID: PMC9837313 DOI: 10.1002/jcp.30829] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/12/2022] [Accepted: 07/04/2022] [Indexed: 01/17/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease that is responsible for about one-third of dementia cases worldwide. It is believed that AD is initiated with the deposition of Ab plaques in the brain. Genetic studies have shown that a high number of AD risk genes are expressed by microglia, the resident macrophages of brain. Common mode of action by microglia cells is neuroinflammation and phagocytosis. Moreover, it has been discovered that inflammatory marker levels are increased in AD patients. Recent studies advocate that neuroinflammation plays a major role in AD progression. Microglia have different activation profiles depending on the region of brain and stimuli. In different activation, profile microglia can generate either pro-inflammatory or anti-inflammatory responses. Microglia defend brain cells from pathogens and respond to injuries; also, microglia can lead to neuronal death along the way. In this review, we will bring the different roles played by microglia and microglia-related genes in the progression of AD.
Collapse
Affiliation(s)
- Rezwanul Islam
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL
| | - Hadi Choudhary
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL
| | - Robin Rajan
- Marcus Neuroscience Institute, Boca Raton Medical Center, Boca Raton, FL
| | - Frank Vrionis
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL
- Marcus Neuroscience Institute, Boca Raton Medical Center, Boca Raton, FL
| | - Khalid A. Hanafy
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL
- Marcus Neuroscience Institute, Boca Raton Medical Center, Boca Raton, FL
| |
Collapse
|
3
|
Qiu O, Zhao J, Shi Z, Li H, Wang S, Liao K, Tang M, Xie J, Huang X, Zhang W, Zhou L, Yang X, Zhou Z, Xu L, Huang R, Miao Y, Qiu Y, Lin Y. Asparagine endopeptidase deficiency mitigates radiation-induced brain injury by suppressing microglia-mediated neuronal senescence. iScience 2024; 27:109698. [PMID: 38655198 PMCID: PMC11035374 DOI: 10.1016/j.isci.2024.109698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 04/26/2024] Open
Abstract
Mounting evidence supports the role of neuroinflammation in radiation-induced brain injury (RIBI), a chronic disease characterized by delayed and progressive neurological impairment. Asparagine endopeptidase (AEP), also known as legumain (LGMN), participates in multiple malignancies and neurodegenerative diseases and may potentially be involved in RIBI. Here, we found AEP expression was substantially elevated in the cortex and hippocampus of wild-type (Lgmn+/+) mice following whole-brain irradiation. Lgmn knockout (Lgmn-/-) alleviated neurological impairment caused by whole-brain irradiation by suppressing neuronal senescence. Bulk RNA and metabolomic sequencing revealed AEP's involvement in the antigen processing and presentation pathway and neuroinflammation. This was further confirmed by co-culturing Lgmn+/+ primary neurons with the conditioned media derived from irradiated Lgmn+/+ or Lgmn-/- primary microglia. Furthermore, esomeprazole inhibited the enzymatic activity of AEP and RIBI. These findings identified AEP as a critical factor of neuroinflammation in RIBI, highlighting the prospect of targeting AEP as a therapeutic approach.
Collapse
Affiliation(s)
- Ouwen Qiu
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Jianyi Zhao
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Zhonggang Shi
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Huan Li
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Siyuan Wang
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Keman Liao
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Minchao Tang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Guangxi 530021, P.R. China
| | - Jieqiong Xie
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Guangxi 530007, P.R. China
| | - Xi Huang
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Guangxi 530021, P.R. China
| | - Wenrui Zhang
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Li Zhou
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Xi Yang
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Zhiyi Zhou
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Lei Xu
- Department of Radiation, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Renhua Huang
- Department of Radiation, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Yifeng Miao
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Yongming Qiu
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Yingying Lin
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| |
Collapse
|
4
|
Yeh TY, Chu WJ, Huang YS. GM1 ganglioside protects against LPS-induced neuroinflammatory and oxidative responses by inhibiting the activation of Akt, TAK1 and NADPH oxidase in MG6 microglial cells. Glycobiology 2024; 34:cwad087. [PMID: 37935390 DOI: 10.1093/glycob/cwad087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/13/2023] [Accepted: 10/14/2023] [Indexed: 11/09/2023] Open
Abstract
GM1 is a major brain ganglioside that exerts neurotrophic, neuroprotective and antineuroinflammatory effects. The aim of this study was to obtain insights into the antineuroinflammatory mechanisms of exogenous GM1 in lipopolysaccharide (LPS)-stimulated MG6 mouse transformed microglial cell line. First, we found that GM1 prevented the LPS-induced transformation of microglia into an amoeboid-like shape. GM1 treatment inhibited LPS-induced expression of inducible nitric oxide synthase, cyclooxygenase-2 (COX-2), and proinflammatory cytokines such as TNF-α, IL-1β and IL-6 in MG6 cells. In LPS-treated mice, GM1 also reduced striatal microglia activation and attenuated COX-2 expression. Subsequent mechanistic studies showed that GM1 suppressed LPS-induced nuclear translocation of nuclear factor κB (NF-κB) and activator protein-1 (AP-1), two critical transcription factors responsible for the production of proinflammatory mediators. GM1 exhibited antineuroinflammatory properties by suppressing Akt/NF-κB signaling and the activation of mitogen-activated protein kinases (MAPKs), including p38 MAPK, extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK). Furthermore, GM1 suppressed LPS-induced activation of transforming growth factor-β-activated kinase 1 (TAK1) and NADPH oxidase 2 (NOX2), upstream regulators of the IκBα/NF-κB and MAPK/AP-1 signaling pathways. GM1 also inhibited NOX-mediated reactive oxygen species (ROS) production and protected against LPS-induced MG6 cell death, suggesting an antioxidant role of GM1. In conclusion, GM1 exerts both antineuroinflammatory and antioxidative effects by inhibiting Akt, TAK1 and NOX2 activation.
Collapse
Affiliation(s)
- Ting-Yin Yeh
- Graduate Institute of Life Sciences, National Defense Medical Center, No. 161, Sec. 6, Minquan E. Rd., Neihu Dist, Taipei City 11490, Taiwan
| | - Wen-Jui Chu
- Department of Biology and Anatomy, National Defense Medical Center, No. 161, Sec. 6, Minquan E. Rd., Neihu Dist, Taipei City 11490, Taiwan
| | - Yuahn-Sieh Huang
- Graduate Institute of Life Sciences, National Defense Medical Center, No. 161, Sec. 6, Minquan E. Rd., Neihu Dist, Taipei City 11490, Taiwan
- Department of Biology and Anatomy, National Defense Medical Center, No. 161, Sec. 6, Minquan E. Rd., Neihu Dist, Taipei City 11490, Taiwan
| |
Collapse
|
5
|
Abbate C. The Adult Neurogenesis Theory of Alzheimer's Disease. J Alzheimers Dis 2023:JAD221279. [PMID: 37182879 DOI: 10.3233/jad-221279] [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: 05/16/2023]
Abstract
Alzheimer's disease starts in neural stem cells (NSCs) in the niches of adult neurogenesis. All primary factors responsible for pathological tau hyperphosphorylation are inherent to adult neurogenesis and migration. However, when amyloid pathology is present, it strongly amplifies tau pathogenesis. Indeed, the progressive accumulation of extracellular amyloid-β deposits in the brain triggers a state of chronic inflammation by microglia. Microglial activation has a significant pro-neurogenic effect that fosters the process of adult neurogenesis and supports neuronal migration. Unfortunately, this "reactive" pro-neurogenic activity ultimately perturbs homeostatic equilibrium in the niches of adult neurogenesis by amplifying tau pathogenesis in AD. This scenario involves NSCs in the subgranular zone of the hippocampal dentate gyrus in late-onset AD (LOAD) and NSCs in the ventricular-subventricular zone along the lateral ventricles in early-onset AD (EOAD), including familial AD (FAD). Neuroblasts carrying the initial seed of tau pathology travel throughout the brain via neuronal migration driven by complex signals and convey the disease from the niches of adult neurogenesis to near (LOAD) or distant (EOAD) brain regions. In these locations, or in close proximity, a focus of degeneration begins to develop. Then, tau pathology spreads from the initial foci to large neuronal networks along neural connections through neuron-to-neuron transmission.
Collapse
Affiliation(s)
- Carlo Abbate
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| |
Collapse
|
6
|
Liang Z, Lou Y, Hao Y, Li H, Feng J, Liu S. The Relationship of Astrocytes and Microglia with Different Stages of Ischemic Stroke. Curr Neuropharmacol 2023; 21:2465-2480. [PMID: 37464832 PMCID: PMC10616922 DOI: 10.2174/1570159x21666230718104634] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/31/2023] [Accepted: 02/04/2023] [Indexed: 07/20/2023] Open
Abstract
Ischemic stroke is the predominant cause of severe morbidity and mortality worldwide. Post-stroke neuroinflammation has recently received increasing attention with the aim of providing a new effective treatment strategy for ischemic stroke. Microglia and astrocytes are major components of the innate immune system of the central nervous system. They can be involved in all phases of ischemic stroke, from the early stage, contributing to the first wave of neuronal cell death, to the late stage involving phagocytosis and repair. In the early stage of ischemic stroke, a vicious cycle exists between the activation of microglia and astrocytes (through astrocytic connexin 43 hemichannels), aggravating neuroinflammatory injury post-stroke. However, in the late stage of ischemic stroke, repeatedly activated microglia can induce the formation of glial scars by triggering reactive astrogliosis in the peri-infarct regions, which may limit the movement of activated microglia in reverse and restrict the diffusion of inflammation to healthy brain tissues, alleviating the neuroinflammatory injury poststroke. In this review, we elucidated the various roles of astrocytes and microglia and summarized their relationship with neuroinflammation. We also examined how astrocytes and microglia influence each other at different stages of ischemic stroke. Several potential therapeutic approaches targeting astrocytes and microglia in ischemic stroke have been reviewed. Understanding the details of astrocytemicroglia interaction processes will contribute to a better understanding of the mechanisms underlying ischemic stroke, contributing to the identification of new therapeutic interventions.
Collapse
Affiliation(s)
- Zhen Liang
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Yingyue Lou
- Department of Rehabilitation, The Second Hospital of Jilin University, Changchun, China
| | - Yulei Hao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Hui Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Songyan Liu
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
7
|
Ma X, Zhao Y, Yang T, Gong N, Chen X, Liu G, Xiao J. Integration of network pharmacology and molecular docking to explore the molecular mechanism of Cordycepin in the treatment of Alzheimer's disease. Front Aging Neurosci 2022; 14:1058780. [PMID: 36620771 PMCID: PMC9817107 DOI: 10.3389/fnagi.2022.1058780] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/21/2022] [Indexed: 12/25/2022] Open
Abstract
Background Cordycepin is a nucleoside adenosine analog and an active ingredient isolated from the liquid fermentation of Cordyceps. This study sought to explore the mechanism underlying the therapeutic effect of Cordycepin against Alzheimer's disease using network pharmacology and molecular docking technology. Methods TCMSP, SYMMAP, CTD, Super-pred, SEA, GeneCards, DisGeNET database, and STRING platform were used to screen and construct the target and protein interaction network of Cordycepin for Alzheimer's disease. The results of Gene Ontology annotation and KEGG pathway enrichment analysis were obtained based on the DAVID database. The Omicshare database was also applied in GO and KEGG pathway enrichment analysis of the key targets. The protein-protein interaction network was constructed using the STRING database, and the potential effective targets for AD were screened based on the degree values. The correlation between the potential targets of Cordycepin in the treatment of AD and APP, MAPT, and PSEN2 was analyzed using (GEPIA) databases. We obtained potential targets related to aging using the Aging Altas database. Molecular docking analysis was performed by AutoDock Vina and Pymol software. Finally, we validated the significant therapeutic targets in the Gene Expression Omnibus (GEO) database. Results A total of 74 potential targets of Cordycepin for treating Alzheimer's disease were identified. The potential targets of Cordycepin for the treatment of AD mainly focused on Lipid and atherosclerosis (hsa05417), Platinum drug resistance (hsa01524), Apoptosis (hsa04210), and Pathways in cancer (hsa05200). Our findings suggest that the therapeutic effect of Cordycepin on AD is primarily associated with these biological processes. We obtained 12 potential therapeutic targets for AD using the degree value in Cytoscape. Interestingly, AKT1, MAPK8, BCL2L1, FOXO3, and CTNNB1 were not only significantly associated with pathogenic genes (APP, MAPT, and PSEN2) but also with longevity in Alzheimer's Disease. Thus we speculated that the five target genes were potential core targets mediating the therapeutic effect of Cordycepin against AD. Moreover, molecular docking results analysis showed good binding affinity between Cordycepin and the five core targets. Overall, MAPK8, FOXO3 and CTNNB1 may have significant clinical and treatment implications. Conclusion Network pharmacology demonstrated that Cordycepin exerts a therapeutic effect against Alzheimer's disease via multiple targets and signaling pathways and has huge prospects for application in treating neurodegenerative diseases.
Collapse
|
8
|
Miao BB, Gao D, Hao JP, Li YL, Li L, Wang JB, Xiao XH, Yang CC, Zhang L. Tetrahydroxy stilbene glucoside alters neurogenesis and neuroinflammation to ameliorate radiation-associated cognitive disability via AMPK/Tet2. Int Immunopharmacol 2022; 110:108928. [DOI: 10.1016/j.intimp.2022.108928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/23/2022] [Accepted: 06/05/2022] [Indexed: 11/26/2022]
|
9
|
Microglia in Alzheimer’s Disease: A Favorable Cellular Target to Ameliorate Alzheimer’s Pathogenesis. Mediators Inflamm 2022; 2022:6052932. [PMID: 35693110 PMCID: PMC9184163 DOI: 10.1155/2022/6052932] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 05/09/2022] [Indexed: 11/18/2022] Open
Abstract
Microglial cells serve as molecular sensors of the brain that play a role in physiological and pathological conditions. Under normal physiology, microglia are primarily responsible for regulating central nervous system homeostasis through the phagocytic clearance of redundant protein aggregates, apoptotic cells, damaged neurons, and synapses. Furthermore, microglial cells can promote and mitigate amyloid β phagocytosis and tau phosphorylation. Dysregulation of the microglial programming alters cellular morphology, molecular signaling, and secretory inflammatory molecules that contribute to various neurodegenerative disorders especially Alzheimer’s disease (AD). Furthermore, microglia are considered primary sources of inflammatory molecules and can induce or regulate a broad spectrum of cellular responses. Interestingly, in AD, microglia play a double-edged role in disease progression; for instance, the detrimental microglial effects increase in AD while microglial beneficiary mechanisms are jeopardized. Depending on the disease stages, microglial cells are expressed differently, which may open new avenues for AD therapy. However, the disease-related role of microglial cells and their receptors in the AD brain remain unclear. Therefore, this review represents the role of microglial cells and their involvement in AD pathogenesis.
Collapse
|
10
|
Huang Y, Cai Q, Liu H, Wang Y, Ma W. Remifentanil inhibits the inflammatory response of BV2 microglia and protects PC12 cells from damage caused by microglia activation. Bioengineered 2022; 13:13944-13955. [PMID: 35726401 PMCID: PMC9275917 DOI: 10.1080/21655979.2022.2080421] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Microglia acts as a critical player in neuroinflammation and neuronal injury. Remifentanil (Rem) has been reported to exert anti-inflammatory activity in several types of diseases. However, the role of Rem in microglia-mediated neuroinflammation is unclear. The present study was designed to investigate the effects of Rem against lipopolysaccharide (LPS)-activated BV2 microglial and PC12 cell induced by activated BV2 microglia. Cell proliferative ability was assessed with cell counting kit-8 assay and cellular morphology was observed. ELISA assay was used to measure the expressions of PGE2 and inflammatory factors. The contents of p-NF-KB p65, p-IKKα/β, and COX2 were evaluated with the aid of western blot. The levels of NO and iNOS were assessed with Griess assay, qRT-PCR, and western blot. In addition, Tunel assay and western blot were performed to assess cell apoptosis. The data revealed that Rem alleviated BV2 microglial morphological injury induced by LPS. Furthermore, Rem suppressed inflammatory releases, iNOS, NO and PGE2 stimulated by LPS in activated BV2 cells. Moreover, Rem suppressed PC12 cell injury, the generations of inflammatory factors and cell apoptosis triggered by inflammatory mediators secreted from activated BV2 cells. These results suggest that Rem exhibited anti-neuroinflammatory activity in protecting PC12 cells against injury derived from LPS-stimulated BV2 microglia.
Collapse
Affiliation(s)
- Yankui Huang
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Qingxiang Cai
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Huihui Liu
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Yong Wang
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Wuhua Ma
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| |
Collapse
|
11
|
Feng YQ, Xu ZZ, Wang YT, Xiong Y, Xie W, He YY, Chen L, Liu GY, Li X, Liu J, Wu Q. Targeting C–C Chemokine Receptor 5: Key to Opening the Neurorehabilitation Window After Ischemic Stroke. Front Cell Neurosci 2022; 16:876342. [PMID: 35573839 PMCID: PMC9095921 DOI: 10.3389/fncel.2022.876342] [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: 02/16/2022] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
Stroke is the world’s second major cause of adult death and disability, resulting in the destruction of brain tissue and long-term neurological impairment; induction of neuronal plasticity can promote recovery after stroke. C–C chemokine receptor 5 (CCR5) can direct leukocyte migration and localization and is a co-receptor that can mediate human immunodeficiency virus (HIV) entry into cells. Its role in HIV infection and immune response has been extensively studied. Furthermore, CCR5 is widely expressed in the central nervous system (CNS), is engaged in various physiological activities such as brain development, neuronal differentiation, communication, survival, and learning and memory capabilities, and is also involved in the development of numerous neurological diseases. CCR5 is differentially upregulated in neurons after stroke, and the inhibition of CCR5 in specific regions of the brain promotes motor and cognitive recovery. The mechanism by which CCR5 acts as a therapeutic target to promote neurorehabilitation after stroke has rarely been systematically reported yet. Thus, this review aims to discuss the function of CCR5 in the CNS and the mechanism of its effect on post-stroke recovery by regulating neuroplasticity and the inflammatory response to provide an effective basis for clinical rehabilitation after stroke.
Collapse
|
12
|
Treatment of Radiation-Induced Brain Necrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:4793517. [PMID: 34976300 PMCID: PMC8720020 DOI: 10.1155/2021/4793517] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 11/25/2021] [Accepted: 12/08/2021] [Indexed: 02/07/2023]
Abstract
Radiation-induced brain necrosis (RBN) is a serious complication of intracranial as well as skull base tumors after radiotherapy. In the past, due to the lack of effective treatment, radiation brain necrosis was considered to be progressive and irreversible. With better understanding in histopathology and neuroimaging, the occurrence and development of RBN have been gradually clarified, and new treatment methods are constantly emerging. In recent years, some scholars have tried to treat RBN with bevacizumab, nerve growth factor, and gangliosides and have achieved similar results. Some cases of brain necrosis can be repairable and reversible. We aimed to summarize the incidence, pathogenesis, and treatment of RBN.
Collapse
|
13
|
Lu X, Lu F, Yu J, Xue X, Jiang H, Jiang L, Yang Y. Gramine promotes functional recovery after spinal cord injury via ameliorating microglia activation. J Cell Mol Med 2021; 25:7980-7992. [PMID: 34382745 PMCID: PMC8358888 DOI: 10.1111/jcmm.16728] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 05/14/2021] [Accepted: 05/31/2021] [Indexed: 11/29/2022] Open
Abstract
In recent years, a large number of studies have reported that neuroinflammation aggravates the occurrence of secondary injury after spinal cord injury. Gramine (GM), a natural indole alkaloid, possesses various pharmacological properties; however, the anti-inflammation property remains unclear. In our study, Gramine was investigated in vitro and in vivo to explore the neuroprotection effects. In vitro experiment, our results suggest that Gramine treatment can inhibit release of pro-inflammatory mediators. Moreover, Gramine prevented apoptosis of PC12 cells which was caused by activated HAPI microglia, and the inflammatory secretion ability of microglia was inhibited by Gramine through NF-κB pathway. The in vivo experiment is that 80 mg/kg Gramine was injected orthotopically to rats after spinal cord injury (SCI). Behavioural and histological analyses demonstrated that Gramine treatment may alleviate microglia activation and then boost recovery of motor function after SCI. Overall, our research has demonstrated that Gramine exerts suppressed microglia activation and promotes motor functional recovery after SCI through NF-κB pathway, which may put forward the prospect of clinical treatment of inflammation-related central nervous diseases.
Collapse
Affiliation(s)
- Xiaolang Lu
- Department of OrthopedicsThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- Zhejiang Provincial Key Laboratory of OrthopedicsWenzhouChina
| | - Fengfeng Lu
- Department of OrthopedicsThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- Zhejiang Provincial Key Laboratory of OrthopedicsWenzhouChina
| | - Jiachen Yu
- Department of OrthopedicsThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- Zhejiang Provincial Key Laboratory of OrthopedicsWenzhouChina
| | - Xinghe Xue
- Department of OrthopedicsThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- Zhejiang Provincial Key Laboratory of OrthopedicsWenzhouChina
| | - Hongyi Jiang
- Department of OrthopedicsThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- Zhejiang Provincial Key Laboratory of OrthopedicsWenzhouChina
| | - Liting Jiang
- Department of OrthopedicsThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- Zhejiang Provincial Key Laboratory of OrthopedicsWenzhouChina
| | - Yang Yang
- Department of OrthopedicsThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- Zhejiang Provincial Key Laboratory of OrthopedicsWenzhouChina
| |
Collapse
|
14
|
Edler MK, Mhatre-Winters I, Richardson JR. Microglia in Aging and Alzheimer's Disease: A Comparative Species Review. Cells 2021; 10:1138. [PMID: 34066847 PMCID: PMC8150617 DOI: 10.3390/cells10051138] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia are the primary immune cells of the central nervous system that help nourish and support neurons, clear debris, and respond to foreign stimuli. Greatly impacted by their environment, microglia go through rapid changes in cell shape, gene expression, and functional behavior during states of infection, trauma, and neurodegeneration. Aging also has a profound effect on microglia, leading to chronic inflammation and an increase in the brain's susceptibility to neurodegenerative processes that occur in Alzheimer's disease. Despite the scientific community's growing knowledge in the field of neuroinflammation, the overall success rate of drug treatment for age-related and neurodegenerative diseases remains incredibly low. Potential reasons for the lack of translation from animal models to the clinic include the use of a single species model, an assumption of similarity in humans, and ignoring contradictory data or information from other species. To aid in the selection of validated and predictive animal models and to bridge the translational gap, this review evaluates similarities and differences among species in microglial activation and density, morphology and phenotype, cytokine expression, phagocytosis, and production of oxidative species in aging and Alzheimer's disease.
Collapse
Affiliation(s)
- Melissa K. Edler
- Department of Anthropology, School of Biomedical Sciences, Brain Health Research Institute, Kent State University, Kent, OH 44240, USA;
| | - Isha Mhatre-Winters
- School of Biomedical Sciences, College of Arts and Sciences, Kent State University, Kent, OH 44240, USA;
- Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Jason R. Richardson
- Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
15
|
Celorrio M, Abellanas MA, Rhodes J, Goodwin V, Moritz J, Vadivelu S, Wang L, Rodgers R, Xiao S, Anabayan I, Payne C, Perry AM, Baldridge MT, Aymerich MS, Steed A, Friess SH. Gut microbial dysbiosis after traumatic brain injury modulates the immune response and impairs neurogenesis. Acta Neuropathol Commun 2021; 9:40. [PMID: 33691793 PMCID: PMC7944629 DOI: 10.1186/s40478-021-01137-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 02/26/2021] [Indexed: 12/14/2022] Open
Abstract
The influence of the gut microbiota on traumatic brain injury (TBI) is presently unknown. This knowledge gap is of paramount clinical significance as TBI patients are highly susceptible to alterations in the gut microbiota by antibiotic exposure. Antibiotic-induced gut microbial dysbiosis established prior to TBI significantly worsened neuronal loss and reduced microglia activation in the injured hippocampus with concomitant changes in fear memory response. Importantly, antibiotic exposure for 1 week after TBI reduced cortical infiltration of Ly6Chigh monocytes, increased microglial pro-inflammatory markers, and decreased T lymphocyte infiltration, which persisted through 1 month post-injury. Moreover, microbial dysbiosis was associated with reduced neurogenesis in the dentate gyrus 1 week after TBI. By 3 months after injury (11 weeks after discontinuation of the antibiotics), we observed increased microglial proliferation, increased hippocampal neuronal loss, and modulation of fear memory response. These data demonstrate that antibiotic-induced gut microbial dysbiosis after TBI impacts neuroinflammation, neurogenesis, and fear memory and implicate gut microbial modulation as a potential therapeutic intervention for TBI.
Collapse
Affiliation(s)
- Marta Celorrio
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Miguel A Abellanas
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
- Departamento de Bioquímica Y Genética, Facultad de Ciencias, Universidad de Navarra, Pamplona, Spain
- CIMA, Programa de Neurociencias, Universidad de Navarra, Pamplona, Spain
| | - James Rhodes
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Victoria Goodwin
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Jennie Moritz
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Sangeetha Vadivelu
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Leran Wang
- Division of Infectious Diseases, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Rachel Rodgers
- Division of Infectious Diseases, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Sophia Xiao
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Ilakkia Anabayan
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Camryn Payne
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Alexandra M Perry
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Megan T Baldridge
- Division of Infectious Diseases, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Maria S Aymerich
- Departamento de Bioquímica Y Genética, Facultad de Ciencias, Universidad de Navarra, Pamplona, Spain
- CIMA, Programa de Neurociencias, Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Ashley Steed
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Stuart H Friess
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA.
| |
Collapse
|
16
|
Maksoud MJE, Tellios V, Xiang YY, Lu WY. Nitric oxide displays a biphasic effect on calcium dynamics in microglia. Nitric Oxide 2021; 108:28-39. [PMID: 33418057 DOI: 10.1016/j.niox.2021.01.001] [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: 07/21/2020] [Revised: 11/11/2020] [Accepted: 01/01/2021] [Indexed: 01/13/2023]
Abstract
Calcium is a critical secondary messenger in microglia. In response to inflammation, microglia mobilize intracellular calcium and increase the expression of inducible nitric oxide synthase (iNOS), which produces nitric oxide (NO). This study set to explore whether NO regulates intracellular calcium dynamics through transient receptor potential (TRP) channels in primary wildtype (WT) and iNOS knockout (iNOS-/-) microglia, and the BV2 microglial cell line using calcium imaging and voltage-clamp recordings. Our results demonstrated that application of the NO-donor SNAP induced a biphasic calcium response in naïve murine microglia. Specifically, phase I was characterized by a rapid decline in calcium influx that was attenuated by pretreatment of the store operated calcium channel (SOCC) inhibitor 2APB, while phase II presented as a slow calcium influx that was abolished by pretreatment with the TRP vanilloid type 2 (TRPV2) channel inhibitor tranilast. Importantly, in the presence of a protein kinase G (PKG) inhibitor, the SNAP-mediated calcium decline in phase I persisted while the calcium influx in phase II was abolished. Application of thapsigargin to activate SOCCs caused a calcium influx through a nonselective cation conductance in BV2 microglia, which was abruptly attenuated by SNAP. Importantly, iNOS-/- microglia displayed a significantly larger calcium influx though SOCCs while expressing less stromal interaction molecule 1, Orai1, and TRP canonical type 1 and 3 mRNA, when compared to WT microglia. Together, these results demonstrate that NO signaling restricts calcium influx through SOCCs independent of PKG signaling and increases calcium influx through TRPV2 channels in a PKG-dependent mechanism in microglia.
Collapse
Affiliation(s)
- Matthew J E Maksoud
- Graduate Program of Neuroscience, The University of Western Ontario, Canada; Robarts Research Institute, The University of Western Ontario, Canada.
| | - Vasiliki Tellios
- Graduate Program of Neuroscience, The University of Western Ontario, Canada; Robarts Research Institute, The University of Western Ontario, Canada.
| | - Yun-Yan Xiang
- Robarts Research Institute, The University of Western Ontario, Canada.
| | - Wei-Yang Lu
- Graduate Program of Neuroscience, The University of Western Ontario, Canada; Robarts Research Institute, The University of Western Ontario, Canada; Department of Physiology and Pharmacology, University of Western Ontario, Canada.
| |
Collapse
|
17
|
Gagne C, Piot A, Brake WG. Depression, Estrogens, and Neuroinflammation: A Preclinical Review of Ketamine Treatment for Mood Disorders in Women. Front Psychiatry 2021; 12:797577. [PMID: 35115970 PMCID: PMC8804176 DOI: 10.3389/fpsyt.2021.797577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/24/2021] [Indexed: 12/11/2022] Open
Abstract
Ketamine has been shown to acutely and rapidly ameliorate depression symptoms and suicidality. Given that women suffer from major depression at twice the rate of men, it is important to understand how ketamine works in the female brain. This review explores three themes. First, it examines our current understanding of the etiology of depression in women. Second, it examines preclinical research on ketamine's antidepressant effects at a neurobiological level as well as how ovarian hormones present a unique challenge in interpreting these findings. Lastly, the neuroinflammatory hypothesis of depression is highlighted to help better understand how ovarian hormones might interact with ketamine in the female brain.
Collapse
Affiliation(s)
- Collin Gagne
- Department of Psychology, Centre for Studies in Behavioural Neurobiology Concordia University, Montreal, QC, Canada
| | - Alexandre Piot
- Department of Psychology, Centre for Studies in Behavioural Neurobiology Concordia University, Montreal, QC, Canada
| | - Wayne G Brake
- Department of Psychology, Centre for Studies in Behavioural Neurobiology Concordia University, Montreal, QC, Canada
| |
Collapse
|
18
|
Liu G, Fiock KL, Levites Y, Golde TE, Hefti MM, Lee G. Fyn depletion ameliorates tau P301L-induced neuropathology. Acta Neuropathol Commun 2020; 8:108. [PMID: 32665013 PMCID: PMC7362472 DOI: 10.1186/s40478-020-00979-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/24/2020] [Indexed: 02/06/2023] Open
Abstract
The Src family non-receptor tyrosine kinase Fyn has been implicated in neurodegeneration of Alzheimer's disease through interaction with amyloid β (Aβ). However, the role of Fyn in the pathogenesis of primary tauopathies such as FTDP-17, where Aβ plaques are absent, is poorly understood. In the current study, we used AAV2/8 vectors to deliver tauP301L to the brains of WT and Fyn KO mice, generating somatic transgenic tauopathy models with the presence or absence of Fyn. Although both genotypes developed tau pathology, Fyn KO developed fewer neurofibrillary tangles on Bielschowsky and Thioflavin S stained sections and showed lower levels of phosphorylated tau. In addition, tauP301L-induced behavior abnormalities and depletion of synaptic proteins were not observed in the Fyn KO model. Our work provides evidence for Fyn being a critical protein in the disease pathogenesis of FTDP-17.
Collapse
Affiliation(s)
- Guanghao Liu
- Interdisciplinary Program in Neuroscience, University of Iowa Carver College of Medicine, Iowa City, IA USA
| | - Kimberly L. Fiock
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA USA
| | - Yona Levites
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL USA
| | - Todd E. Golde
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL USA
| | - Marco M. Hefti
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA USA
| | - Gloria Lee
- Interdisciplinary Program in Neuroscience, University of Iowa Carver College of Medicine, Iowa City, IA USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 500 Newton Road, ML B191, Iowa City, IA 52242 USA
| |
Collapse
|
19
|
Shi YX, Chen WS. Monascin ameliorate inflammation in the lipopolysaccharide-induced BV-2 microglial cells via suppressing the NF-κB/p65 pathway. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:461-468. [PMID: 32489561 PMCID: PMC7239424 DOI: 10.22038/ijbms.2020.41045.9702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Objectives The pathophysiology of neurodegenerative diseases is complicated, in which inflammatory reactions play a vital role. Microglia cells activation, an essential process of neuroinflammation, can produce neurotoxic molecules and neurotrophic factors, which aggravate inflammation and neuronal injury. Monascin, a major component of red yeast rice, is an azaphilonoid pigment with potential anti-inflammatory effects; however, the effects in central nervous system have not been evaluated. Our goal in this project was to explore the therapeutic effect and the underlying mechanism of Monascin, which may be via anti-inflammatory action. Materials and Methods We used lipopolysaccharide to induce BV-2 microglial cells in order to form an inflammation model in vitro. The anti-inflammatory effects of Monascin were measured by enzyme-linked immunosorbent assay (ELISA), real time-polymerase chain reaction (RT-PCR), Western Blot and Immunofluorescent staining. Results Our data indicated that inflammatory cytokines including interleukin-1β (IL-1β), IL-6, tumor necrosis factor-alpha (TNF-α) and nitric oxide were suppressed by Monascin treatment. Furthermore, the related pro-inflammatory genes were inhibited consistent with the results of ELISA assay. Western blotting results showed that the phosphorylation of nuclear factor kappa B (NF-κB/p65) was reduced by Monascin treatment may be through suppressing the activation of IκB. Furthermore, immunofluorescence staining showed that the translocation of NF-κB/p65 to the cellular nuclear was blockaded after Monascin treatment. Conclusion Taken together, Monascin exerts anti-inflammatory effect and suppressed microglia activation, which suggested its potential therapeutic effect for inflammation-related diseases.
Collapse
Affiliation(s)
- Yong-Xiang Shi
- Department of Orthopedics Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - Wei-Shan Chen
- Department of Orthopedics Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, People's Republic of China
| |
Collapse
|
20
|
Niziolek GM, Boudreau RM, Baker J, Friend LA, Makley AT, Edwards MJ, Gulbins E, Goodman MD. Acid Sphingomyelinase Inhibition Mitigates Histopathological and Behavioral Changes in a Murine Model of Traumatic Brain Injury. J Neurotrauma 2020; 37:1902-1909. [PMID: 32138594 DOI: 10.1089/neu.2019.6436] [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] [Indexed: 01/05/2023] Open
Abstract
Traumatic brain injury (TBI) can lead to the development of chronic traumatic encephalopathy as a result of neuronal phosphorylated tau (p-tau) protein aggregation and neuroinflammation. Acid sphingomyelinase (Asm) may also contribute to post-TBI neurodegenerative disorders. We hypothesized that Asm inhibition would ameliorate p-tau aggregation, neuroinflammation, and behavioral changes after TBI in a murine model. TBI was generated using a weight-drop method. Asm inhibition in wild-type mice was achieved with a single injection of amitriptyline 1 h after TBI. Genetic Asm ablation was achieved using Asm-deficient mice (Asm-/-). Thirty days after TBI, mice underwent behavioral testing with the forced swim test for symptoms of depression or were euthanized for neurohistological analysis. Neuroinflammation was quantified using the microglial markers, ionized calcium-binding adaptor molecule 1 and transmembrane protein 119. Compared to sham mice, TBI mice demonstrated increased hippocampal p-tau. Mice that received amitriptyline after TBI demonstrated decreased p-tau compared to mice that received a saline control. Further, post-TBI Asm-/- mice demonstrated lower levels of p-tau compared to wild-type mice. Though a decrease in neuroinflammation was observed at 1 month post-TBI, no change was demonstrated with mice treated with amitriptyline. Similarly, TBI mice were more likely to show depression compared to mice that received amitriptyline after TBI. Utilizing a weight-drop method to induce moderate TBI, we have shown that genetic deficiency or pharmacological inhibition of Asm prevented hippocampal p-tau aggregation 1 month after injury as well as decreased symptoms of depression. These findings highlight an opportunity to potentially reduce the long-term consequences of TBI.
Collapse
Affiliation(s)
- Grace M Niziolek
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| | - Ryan M Boudreau
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jennifer Baker
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| | - Lou Ann Friend
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| | - Amy T Makley
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| | - Michael J Edwards
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| | - Erich Gulbins
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA.,Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | - Michael D Goodman
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
21
|
Shenqi Fuzheng Injection Ameliorates Radiation-induced Brain Injury. Curr Med Sci 2019; 39:965-971. [DOI: 10.1007/s11596-019-2129-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 10/28/2019] [Indexed: 10/25/2022]
|
22
|
Takeshita Y, Watanabe K, Kakeda S, Hamamura T, Sugimoto K, Masaki H, Ueda I, Igata N, Ohguri T, Korogi Y. Early volume reduction of the hippocampus after whole-brain radiation therapy: an automated brain structure segmentation study. Jpn J Radiol 2019; 38:118-125. [PMID: 31664663 DOI: 10.1007/s11604-019-00895-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 10/16/2019] [Indexed: 11/25/2022]
Abstract
PURPOSE To assess atrophy differences among brain regions and time-dependent changes after whole-brain radiation therapy (WBRT). MATERIALS AND METHODS Twenty patients with lung cancer who underwent both WBRT and chemotherapy (WBRT group) and 18 patients with lung cancer who underwent only chemotherapy (control group) were recruited. Three-dimensional T1WI were analyzed to calculate volume reduction ratio after WBRT in various brain structures. The volume reduction ratio of the hippocampus was compared among following 3 periods: 0-3, 4-7, and 8-11 months after WBRT. RESULTS The volume reduction ratio of the hippocampus was significantly higher in the WBRT group than in the control group (p < 0.05). In WBRT group, the volume reduction ratio of the hippocampus was significantly higher than that of the cortex and white matter (p < 0.05). There were significant differences in the volume reduction ratio between of 0-3 months and that of 4-7 months (p = 0.02) and between 4-7 months and that of 8-11 months (p = 0.01). CONCLUSION The hippocampus is more vulnerable to the radiation compared with other brain regions and may become atrophic even in the early stage after WBRT.
Collapse
Affiliation(s)
- Yohei Takeshita
- Department of Radiology, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Keita Watanabe
- Department of Radiology, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan.
| | - Shingo Kakeda
- Department of Radiology, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Toshihiko Hamamura
- Department of Radiology, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Koichiro Sugimoto
- Department of Radiology, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Hiromi Masaki
- Department of Radiology, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Issei Ueda
- Department of Radiology, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Natsuki Igata
- Department of Radiology, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Takayuki Ohguri
- Department of Radiology, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Yukunori Korogi
- Department of Radiology, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| |
Collapse
|
23
|
Anti-inflammatory treatment with β-asarone improves impairments in social interaction and cognition in MK-801 treated mice. Brain Res Bull 2019; 150:150-159. [DOI: 10.1016/j.brainresbull.2019.05.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 05/09/2019] [Accepted: 05/21/2019] [Indexed: 01/10/2023]
|
24
|
Asari MA, Zulkaflee MH, Sirajudeen K, Mohd Yusof NA, Mohd Sairazi NS. Tualang honey and DHA-rich fish oil reduce the production of pro-inflammatory cytokines in the rat brain following exposure to chronic stress. J Taibah Univ Med Sci 2019; 14:317-323. [PMID: 31488962 PMCID: PMC6717079 DOI: 10.1016/j.jtumed.2019.06.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/16/2019] [Accepted: 06/21/2019] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVES The aim of the present study was to investigate the effects of Tualang honey (TH), DHA-rich fish oil, and their combination on the concentrations of selected pro-inflammatory cytokines in rat brains following exposure to chronic stress. METHODS Fifty male Sprague-Dawley rats were divided into (i) control, (ii) stress-exposed, (iii) stress-exposed and treated with TH (1 g/kg body weight twice daily via oral gavage), (iv) stress-exposed and treated with DHA-rich fish oil (450 mg/kg body weight twice daily via oral gavage), and (v) stress-exposed and treated with a combination of TH and DHA-rich fish oil. The chronic stress regimen consisted of a combination of restraint stress and a swim stress test for 28 days. The concentrations of selected pro-inflammatory cytokines in brain homogenates (TNF-α, IL6, and IFN-γ) were measured by ELISA. RESULTS The concentrations of TNF-α, IL6, and IFN-γ in brain homogenates from the DHA, TH, and TH + DHA-treated groups were significantly lower compared to the control and stress-only-exposed groups (p < 0.05), but no difference was observed between treatment groups. CONCLUSION Consumption of DHA-rich fish oil and TH can be effective in lowering pro-inflammatory cytokine levels in the brains of rats under chronic stress conditions. However, consuming these agents together does not provide additional benefits compared to taking them separately.
Collapse
Affiliation(s)
- Mohd A. Asari
- Department of Anatomy, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia
| | - Mohd H. Zulkaflee
- Department of Anatomy, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia
| | - K.N.S. Sirajudeen
- Department of Chemical Pathology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia
| | - Nurul A. Mohd Yusof
- Department of Anatomy, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia
| | - Nur S. Mohd Sairazi
- Department of Chemical Pathology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia
| |
Collapse
|
25
|
Involvement of the microglial NLRP3 inflammasome in the anti-inflammatory effect of the antidepressant clomipramine. J Affect Disord 2019; 254:15-25. [PMID: 31082627 DOI: 10.1016/j.jad.2019.05.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/25/2019] [Accepted: 05/04/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Depression has recently been referred to as a neuroimmune disease because it is characterized by inflammatory changes in the cerebral cortex and hippocampus. Studies have demonstrated that microglial activation plays a crucial role in releasing inflammatory cytokines in the central nervous system (CNS), thereby contributing to depression, the mechanism underlying which remains unclear. METHODS First, we examined microglial activation and inflammatory changes in C57BL/6 male mice injected with lipopolysaccharide (LPS; 1 mg/kg), which leads to depressive behaviors in mice that were attenuated by the antidepressant clomipramine. Second, we utilized a BV2 cell line and primary microglial cultures to determine the inflammatory response in vitro, and the effects of clomipramine exerted on the inflammatory response using real-time polymerase chain reaction and ELISA. Third, we utilized NLRP3 (NOD-like receptor protein 3) knock-out (KO) mice to prove that NLRP3 is involved in the effects of clomipramine. RESULTS The results showed that LPS injection induced depressive-like behaviors in mice, as assessed using several behavioral tests including body weight, and forced swimming and tail suspension tests. The LPS-induced expression of interleukin-1beta (IL-1β), IL-6, and tumor necrosis factor alpha could be downregulated by clomipramine pre-treatment both in vivo and in vitro. The inhibitory effect of clomipramine on the LPS-induced increase in cytokines was found at both the protein and gene levels. Clomipramine significantly reduced the LPS-induced increase in NLRP3 gene expression in BV2 cells. Furthermore, we utilized NLRP3 KO mice to explore whether NLPP3 was involved in this process and found that clomipramine treatment inhibits the LPS-induced increased expression of IL-1β. CONCLUSION These results imply that clomipramine could attenuate depressive behaviors and neuroinflammation induced by LPS via partial regulation of NLRP3.
Collapse
|
26
|
Lauretti E, Praticò D. Novel Key Players in the Development of Tau Neuropathology: Focus on the 5-Lipoxygenase. J Alzheimers Dis 2019; 64:S481-S489. [PMID: 29758943 DOI: 10.3233/jad-179931] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Tauopathies belong to a large group of neurodegenerative diseases characterized by progressive accumulation of hyperphosphorylated tau. Tau is a microtubule binding protein which is necessary for their assembly and stability. However, tau affinity for microtubules mainly depends on its phosphorylation status, which is the result of a delicate balance between kinases and phosphatases activity. Any significant changes in this equilibrium can promote tau fibrillation, aggregation, neuronal dysfunction, and ultimately neuronal loss. Despite intensive research, the molecular mechanism(s) leading to tau hyperphosphorylation are still unknown and there is no cure for these diseases. Development of an effective strategy that successfully prevents tau excessive phosphorylation and/or tau aggregation may offer a real therapeutic opportunity for these less investigated neurodegenerative conditions. Beside tau, chronic brain inflammation is a common feature of all tauopathies and 5-lipoxygenase, an inflammatory enzyme, is upregulated in brain regions affected by tau pathology. Recently, in vitro studies and preclinical investigations with animal models of tauopathy have implicated 5-lipoxygenase in the regulation of tau phosphorylation through activation of the cyclin-dependent kinase 5 pathway, supporting the novel hypothesis that this protein is a promising therapeutic target for the treatment of tauopathies. In this article, we will discuss the contribution of the 5-lipoxygenase signaling pathway in the development of tau neuropathology, and the promising potential that drugs targeting this enzyme activation hold as a novel disease-modifying therapeutic approach for tauopathies.
Collapse
Affiliation(s)
- Elisabetta Lauretti
- Alzheimer's Center at Temple, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Domenico Praticò
- Alzheimer's Center at Temple, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
27
|
Carlomagno Y, Chung DEC, Yue M, Kurti A, Avendano NM, Castanedes-Casey M, Hinkle KM, Jansen-West K, Daughrity LM, Tong J, Phillips V, Rademakers R, DeTure M, Fryer JD, Dickson DW, Petrucelli L, Cook C. Enhanced phosphorylation of T153 in soluble tau is a defining biochemical feature of the A152T tau risk variant. Acta Neuropathol Commun 2019; 7:10. [PMID: 30674342 PMCID: PMC6345061 DOI: 10.1186/s40478-019-0661-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 01/16/2019] [Indexed: 12/30/2022] Open
Abstract
Pathogenic mutations in the tau gene (microtubule associated protein tau, MAPT) are linked to the onset of tauopathy, but the A152T variant is unique in acting as a risk factor for a range of disorders including Alzheimer’s disease (AD), progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), and dementia with Lewy bodies (DLB). In order to provide insight into the mechanism by which A152T modulates disease risk, we developed a novel mouse model utilizing somatic brain transgenesis with adeno-associated virus (AAV) to drive tau expression in vivo, and validated the model by confirming the distinct biochemical features of A152T tau in postmortem brain tissue from human carriers. Specifically, TauA152T-AAV mice exhibited increased tau phosphorylation that unlike animals expressing the pathogenic P301L mutation remained localized to the soluble fraction. To investigate the possibility that the A152T variant might alter the phosphorylation state of tau on T152 or the neighboring T153 residue, we generated a novel antibody that revealed significant accumulation of soluble tau species that were hyperphosphorylated on T153 (pT153) in TauA152T-AAV mice, which were absent the soluble fraction of TauP301L-AAV mice. Providing new insight into the role of A152T in modifying risk of tauopathy, as well as validating the TauA152T-AAV model, we demonstrate that the presence of soluble pT153-positive tau species in human postmortem brain tissue differentiates A152T carriers from noncarriers, independent of disease classification. These results implicate both phosphorylation of T153 and an altered solubility profile in the mechanism by which A152T modulates disease risk.
Collapse
|
28
|
Yan L, Yang J, Yu M, Lu Y, Huang L, Wang J, Lu X, Jin C, Wu S, Cai Y. Lanthanum chloride induces neuron damage by activating the nuclear factor-kappa B signaling pathway in activated microglia. Metallomics 2019; 11:1277-1287. [DOI: 10.1039/c9mt00108e] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Lanthanum is a rare earth element which can have adverse effects on the central nervous system (CNS).
Collapse
|
29
|
Edler MK, Sherwood CC, Meindl RS, Munger E, Hopkins WD, Ely JJ, Erwin JM, Perl DP, Mufson EJ, Hof PR, Raghanti MA. Microglia changes associated to Alzheimer's disease pathology in aged chimpanzees. J Comp Neurol 2018; 526:2921-2936. [PMID: 30069930 PMCID: PMC6283685 DOI: 10.1002/cne.24484] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 05/11/2018] [Accepted: 05/16/2018] [Indexed: 01/01/2023]
Abstract
In Alzheimer's disease (AD), the brain's primary immune cells, microglia, become activated and are found in close apposition to amyloid beta (Aβ) protein plaques and neurofibrillary tangles (NFT). The present study evaluated microglia density and morphology in a large group of aged chimpanzees (n = 20, ages 37-62 years) with varying degrees of AD-like pathology. Using immunohistochemical and stereological techniques, we quantified the density of activated microglia and morphological variants (ramified, intermediate, and amoeboid) in postmortem chimpanzee brain samples from prefrontal cortex, middle temporal gyrus, and hippocampus, areas that show a high degree of AD pathology in humans. Microglia measurements were compared to pathological markers of AD in these cases. Activated microglia were consistently present across brain areas. In the hippocampus, CA3 displayed a higher density than CA1. Aβ42 plaque volume was positively correlated with higher microglial activation and with an intermediate morphology in the hippocampus. Aβ42-positive vessel volume was associated with increased hippocampal microglial activation. Activated microglia density and morphology were not associated with age, sex, pretangle density, NFT density, or tau neuritic cluster density. Aged chimpanzees displayed comparable patterns of activated microglia phenotypes as well as an association of increased microglial activation and morphological changes with Aβ deposition similar to AD patients. In contrast to human AD brains, activated microglia density was not significantly correlated with tau lesions. This evidence suggests that the chimpanzee brain may be relatively preserved during normal aging processes but not entirely protected from neurodegeneration as previously assumed.
Collapse
Affiliation(s)
- Melissa K. Edler
- School of Biomedical Sciences, Kent State University, Kent, OH 44242
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
| | - Chet C. Sherwood
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC 20052
| | | | - Emily Munger
- School of Biomedical Sciences, Kent State University, Kent, OH 44242
| | - William D. Hopkins
- Division of Developmental and Cognitive Neuroscience, Yerkes National Primate Research Center, Atlanta, GA 30322
- Neuroscience Institute, Georgia State University, Atlanta, GA 30302
| | | | - Joseph M. Erwin
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC 20052
| | - Daniel P. Perl
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Elliott J. Mufson
- Departments of Neurobiology and Neurology, Barrow Neurological Institute, Phoenix, AZ 85013
| | - Patrick R. Hof
- Fishberg Department of Neuroscience, Ronald M. Loeb Center for Alzheimer’s Disease, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- New York Consortium for Evolutionary Primatology, New York, NY 10468
| | - Mary Ann Raghanti
- School of Biomedical Sciences, Kent State University, Kent, OH 44242
- Department of Anthropology, Kent State University, Kent, OH 44242
| |
Collapse
|
30
|
Zakharov S, Hlusicka J, Nurieva O, Kotikova K, Lischkova L, Kacer P, Kacerova T, Urban P, Vaneckova M, Seidl Z, Diblik P, Kuthan P, Heissigerova J, Lesovsky J, Rulisek J, Vojtova L, Hubacek JA, Navratil T. Neuroinflammation markers and methyl alcohol induced toxic brain damage. Toxicol Lett 2018; 298:60-69. [DOI: 10.1016/j.toxlet.2018.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 04/22/2018] [Accepted: 05/03/2018] [Indexed: 12/29/2022]
|
31
|
Abstract
OBJECTIVES To address the estimated rates of incidence, potential underlying etiologies, and cognitive domains affected from diagnosis and treatment. To describe potential cognitive function interventions. DATA SOURCES PubMed. CONCLUSION Adults with gliomas report that the most distressing, persistent, and greatest negative impact on their lives relates to the cognitive impairment they experience. However, there are several potential interventions that may prevent cognitive decline during treatment or maintain cognitive function long term. IMPLICATIONS FOR NURSING PRACTICE Awareness of cognitive sequela that adults with gliomas face can lead to early identification, full neurocognitive profiling, and implementation of evidence-based interventions for those experiencing cognitive impairments following cancer treatment.
Collapse
|
32
|
Nurieva O, Diblik P, Kuthan P, Sklenka P, Meliska M, Bydzovsky J, Heissigerova J, Urban P, Kotikova K, Navratil T, Komarc M, Seidl Z, Vaneckova M, Pelclova D, Zakharov S. Progressive Chronic Retinal Axonal Loss Following Acute Methanol-induced Optic Neuropathy: Four-Year Prospective Cohort Study. Am J Ophthalmol 2018; 191:100-115. [PMID: 29709459 DOI: 10.1016/j.ajo.2018.04.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 03/03/2018] [Accepted: 04/19/2018] [Indexed: 12/24/2022]
Abstract
PURPOSE To study the dynamics and clinical determinants of chronic retinal nerve fiber layer thickness (RNFL) loss after methanol-induced optic neuropathy. DESIGN Prospective cohort study. METHODS All patients underwent complete ophthalmic evaluation including spectral-domain optical coherence tomography 3 times during 4 years of observation: 4.9 (±0.6), 25.0 (±0.6), and 49.9 (±0.5) months after discharge. PARTICIPANTS Eighty-four eyes of 42 survivors of methanol poisoning, mean age (standard deviation) of 45.7 (±4.4) years; and 82 eyes of 41 controls, mean age 44.0 (±4.2) years. MAIN OUTCOME MEASURES Global and temporal RNFL loss. RESULTS Abnormal RNFL thickness was registered in 13 of 42 (31%) survivors of methanol poisoning and chronic axonal loss in 10 of 42 (24%) patients. Significant decrease of global/temporal RNFL thickness during the observation period was found in the study population compared to the controls (P < .001). The risk estimate of chronic global RNFL loss for arterial blood pH < 7.3 at admission was 11.65 (95% confidence interval 1.91-71.12) after adjusting for age and sex. The patients with chronic axonal degeneration demonstrated progressive visual loss in 7 of 10 cases. The patients with abnormal RNFL thickness had magnetic resonance signs of brain damage in 10 of 13 vs 8 of 29 cases with normal RNFL thickness (P = .003). Signs of brain hemorrhages were present in 7 of 13 patients with abnormal RNFL thickness vs 5 of 29 cases with normal RNFL thickness (P = .015). CONCLUSIONS Methanol-induced optic neuropathy may lead to chronic retinal axonal loss during the following years. Arterial blood pH on admission is the strongest predictor of chronic RNFL thickness decrease. Chronic retinal neurodegeneration is associated with the progressive loss of visual functions and necrotic brain lesions.
Collapse
Affiliation(s)
- Olga Nurieva
- Toxicological Information Centre, Department of Occupational Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Pavel Diblik
- Clinic of Ophthalmology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Pavel Kuthan
- Clinic of Ophthalmology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Petr Sklenka
- Clinic of Ophthalmology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Martin Meliska
- Clinic of Ophthalmology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jan Bydzovsky
- Clinic of Ophthalmology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jarmila Heissigerova
- Clinic of Ophthalmology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Pavel Urban
- Toxicological Information Centre, Department of Occupational Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Katerina Kotikova
- Toxicological Information Centre, Department of Occupational Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Tomas Navratil
- Department of Biomimetic Electrochemistry, J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czech Republic; Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Martin Komarc
- Department of Methodology, Faculty of Physical Education and Sport, Charles University, Prague, Czech Republic
| | - Zdenek Seidl
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Manuela Vaneckova
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Daniela Pelclova
- Toxicological Information Centre, Department of Occupational Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Sergey Zakharov
- Toxicological Information Centre, Department of Occupational Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic.
| |
Collapse
|
33
|
Al-Amin MM, Choudhury MFR, Chowdhury AS, Chowdhury TR, Jain P, Kazi M, Alkholief M, Alshehri SM, Reza HM. Pretreatment With Risperidone Ameliorates Systemic LPS-Induced Oxidative Stress in the Cortex and Hippocampus. Front Neurosci 2018; 12:384. [PMID: 29937710 PMCID: PMC6002684 DOI: 10.3389/fnins.2018.00384] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 05/22/2018] [Indexed: 11/30/2022] Open
Abstract
Risperidone (RIS), an atypical antipsychotic has been found to show anti-inflammatory effect against lipopolysaccharide (LPS)-induced inflammation. In vitro study has revealed that RIS inhibits the LPS-induced reactive oxygen species (ROS) formation. We investigated the antioxidant effects of RIS on LPS-induced oxidative stress markers in Swiss albino mice. Ten weeks old male Swiss albino mice (30 ± 2 g) were pretreated with either distilled water (control) or RIS (3 mg/kg) for 7 days. On day 8, animals were challenged with a single dose of LPS (0.8 mg/kg) while control animals received distilled water only. The animals were sacrificed after 24 h of LPS administration and tissue samples were collected. RIS administration significantly (p < 0.05) reduced the LPS-induced elevated levels of lipid peroxidation product malondialdehyde (MDA), advanced protein oxidation products, and nitric oxide (NO) in the cortex. Catalase (CAT) and superoxide dismutase (SOD) levels were also diminished while the level of glutathione (GSH) was enhanced. Hippocampus data showed that RIS significantly (p < 0.05) reduced the LPS-induced increased levels of MDA and NO, and SOD activity. Our results suggest that LPS-induced neuronal oxidative damage can be alleviated by the pretreatment with RIS and the effect is shown presumably by scavenging of the ROS by risperidone as an antioxidant.
Collapse
Affiliation(s)
- Md Mamun Al-Amin
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | | | - Al Saad Chowdhury
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | | | - Preeti Jain
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Mohsin Kazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Musaed Alkholief
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sultan M Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hasan Mahmud Reza
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| |
Collapse
|
34
|
You P, Fu S, Yu K, Xia Y, Wu H, Yang Y, Ma C, Liu D, Chen X, Wang J, Ye X, Liu Y. Scutellarin suppresses neuroinflammation via the inhibition of the AKT/NF-κB and p38/JNK pathway in LPS-induced BV-2 microglial cells. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:743-751. [PMID: 29680917 DOI: 10.1007/s00210-018-1503-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 04/09/2018] [Indexed: 02/08/2023]
Abstract
In vitro and in vivo studies indicate that scutellarin (SCU) exerts anti-inflammatory effects in the central nervous system (CNS) and inhibits microglia activation. This study investigated the anti-neuroinflammation molecular mechanisms exerted by scutellarin in LPS-induced BV-2 cells. The results showed that production of TNF-α, IL-1β, IL-6, and NO and TNF-α, IL-1β, IL-6, and iNOS mRNA were inhibited by scutellarin, which was independent of cytotoxicity as assessed by a CCK8 assay. Western blot analysis indicated that NF-κB-p65 phosphorylation was suppressed by scutellarin via inhibition of IκB degradation and IKKβ activation, which coincided with blockage of nuclear translocation of NF-κB as shown by immunofluorescent staining. Consistent with the inhibition of NF-κB, scutellarin inhibited the phosphorylation of p38, JNK, and AKT without affecting phosphorylation of ERK1/2 or PI3K in LPS-induced BV-2 cells. Overall, the present study suggests that scutellarin inhibits the production of pro-inflammatory mediators via inhibition of the IKK-dependent NF-κB and p38/JNK signaling pathway, which inhibits microglia activation and exerts anti-inflammation, indicating its potential therapeutic effect for neurodegenerative and cerebrovascular diseases.
Collapse
Affiliation(s)
- Pengtao You
- Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, 1 HuangJia Hu Road West, Hongshan District, Wuhan, Hubei, 430065, People's Republic of China.
| | - San Fu
- Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, 1 HuangJia Hu Road West, Hongshan District, Wuhan, Hubei, 430065, People's Republic of China
| | - Kun Yu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, People's Republic of China
| | - Yu Xia
- Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, 1 HuangJia Hu Road West, Hongshan District, Wuhan, Hubei, 430065, People's Republic of China
| | - Hezhen Wu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, People's Republic of China
| | - Yanfang Yang
- Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, 1 HuangJia Hu Road West, Hongshan District, Wuhan, Hubei, 430065, People's Republic of China
| | - Chaozhi Ma
- Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, 1 HuangJia Hu Road West, Hongshan District, Wuhan, Hubei, 430065, People's Republic of China
| | - Dan Liu
- Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, 1 HuangJia Hu Road West, Hongshan District, Wuhan, Hubei, 430065, People's Republic of China
| | - Xin Chen
- Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, 1 HuangJia Hu Road West, Hongshan District, Wuhan, Hubei, 430065, People's Republic of China
| | - Jun Wang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, People's Republic of China
| | - Xiaochuan Ye
- Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, 1 HuangJia Hu Road West, Hongshan District, Wuhan, Hubei, 430065, People's Republic of China
| | - Yanwen Liu
- Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, 1 HuangJia Hu Road West, Hongshan District, Wuhan, Hubei, 430065, People's Republic of China
| |
Collapse
|
35
|
Kim DH, Li H, Han YE, Jeong JH, Lee HJ, Ryu JH. Modulation of Inducible Nitric Oxide Synthase Expression in LPS-Stimulated BV-2 Microglia by Prenylated Chalcones from Cullen corylifolium (L.) Medik. through Inhibition of I-κBα Degradation. Molecules 2018; 23:molecules23010109. [PMID: 29300354 PMCID: PMC6017879 DOI: 10.3390/molecules23010109] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/22/2017] [Accepted: 12/28/2017] [Indexed: 12/13/2022] Open
Abstract
The overproduction of nitric oxide (NO) and prostaglandin E₂ (PGE₂) by microglia may cause neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease. From the activity-guided purification of Cullen corylifolium (L.) Medik. (syn. Psoralea corylifolia L.), three prenylated chalcones were identified: isobavachalcone (1), bavachromene (2), and kanzonol B (3). These prenylated chalcones showed concentration-dependent inhibitory effects on NO and PGE₂ production in lipopolysaccharide (LPS)-activated microglia. Western blotting and RT-PCR analysis demonstrated that these prenylchalcones reduced the expression of protein and mRNA of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in LPS-activated microglia. Furthermore, three prenylated chalcones blocked the inhibitory-κBα (I-κBα) degradation and down-regulated nuclear factor κB (NF-κB) level of nucleus in LPS-stimulated BV-2 microglia. Therefore, these prenylated chalcones from Psoralea corylifolia may be beneficial for the treatment of neuro-inflammatory diseases by modulating iNOS and COX-2 expressions in activated microglial cells.
Collapse
Affiliation(s)
- Do Hee Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul 04310, Korea.
| | - Hua Li
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul 04310, Korea.
| | - Yeong Eun Han
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul 04310, Korea.
| | - Ji Hye Jeong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul 04310, Korea.
| | - Hwa Jin Lee
- Department of Natural Medicine Resources, Semyung University, Jecheon 27136, Korea.
| | - Jae-Ha Ryu
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul 04310, Korea.
| |
Collapse
|
36
|
Novak P, Cente M, Kosikova N, Augustin T, Kvetnansky R, Novak M, Filipcik P. Stress-Induced Alterations of Immune Profile in Animals Suffering by Tau Protein-Driven Neurodegeneration. Cell Mol Neurobiol 2018; 38:243-259. [PMID: 28405903 DOI: 10.1007/s10571-017-0491-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 03/29/2017] [Indexed: 01/01/2023]
Abstract
Alzheimer's disease (AD) is a multifactorial disorder; neurofibrillary pathology composed of tau protein is found side by side with amyloid-β deposits and extensive neuroinflammation. The immune system of the brain is considered as one of the factors that could influence the speed of the progression of AD neuropathology as a potential mediator of the damage induced by AD protein deposits. Alzheimer's disease pathology can be impacted by psychological stress; however, signalling pathways in background are not well known. We have explored possible avenues of how stress could influence the brain's immune system in a rat model of AD. Animals were subjected either to a single or multiple instances of immobilization stress. The analysis of a panel of immunity-related genes was used to evaluate the impact of stress on the immune response in the brain. We have identified 19 stress-responsive genes that are involved in neuroinflammation accompanying tau pathology: Nos2, Ptgs2, IL-8rb, C5, Mmp9, Cx3cr1, CD40lg, Adrb2, IL-6, IL-6r, IL-1r2, Ccl2, Ccl3, Ccl4, Ccl12, TNF-α, IL-1α, IL-1β, IL-10. Most of them are deregulated under the stress conditions also in control animals; however, the magnitude of the response to either acute or chronic stress differs. This can lead to serious influence, most probably to acceleration of neurodegenerative phenotype in diseased animals. Several of the genes (IL-1β, Casp1, Cx3cr1 and C5) are deregulated solely in tauopathic animals. The stress-induced changes in the inflammatory picture of the brain highlight the fact that the brain's immune response is highly responsive to environmental stimuli. The pattern of changes is indicative of an attempt to protect the brain in the short term, while being potentially detrimental to the response against a long-term pathological process such as neurofibrillary degeneration.
Collapse
Affiliation(s)
- Petr Novak
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Martin Cente
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Nina Kosikova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Tomas Augustin
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Richard Kvetnansky
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Michal Novak
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Peter Filipcik
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia.
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia.
| |
Collapse
|
37
|
Attiq A, Jalil J, Husain K. Annonaceae: Breaking the Wall of Inflammation. Front Pharmacol 2017; 8:752. [PMID: 29104539 PMCID: PMC5654839 DOI: 10.3389/fphar.2017.00752] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/03/2017] [Indexed: 12/12/2022] Open
Abstract
Inventories of tropical forests have listed Annonaceae as one of the most diverse plant families. For centuries, it is employed in traditional medicines to cure various pathological conditions including snakebite, analgesic, astringent, diarrhea, dysentery, arthritis pain, rheumatism, neuralgia, and weight loss etc. Phytochemical analysis of Annonaceae family have reported the occurrence of alkaloids, flavonoids, triterpenes, diterpenes and diterpene flavone glycosides, sterols, lignans, and annonaceous acetogenin characteristically affiliated with Annonaceae sp. Numerous past studies have underlined the pleotropic pharmacological activities of the crude extracts and isolated compounds from Annonaceae species. This review is an effort to abridge the ethnobotany, morphology, phytochemistry, toxicity, and particularly focusing on the anti-inflammatory activity of the Annonaceae species.
Collapse
Affiliation(s)
- Ali Attiq
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Juriyati Jalil
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Khairana Husain
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
38
|
Intranasal telmisartan ameliorates brain pathology in five familial Alzheimer's disease mice. Brain Behav Immun 2017; 64:80-90. [PMID: 28385651 DOI: 10.1016/j.bbi.2017.04.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/09/2017] [Accepted: 04/01/2017] [Indexed: 11/23/2022] Open
Abstract
The renin-angiotensin system (RAS) is a major circulative system engaged in homeostasis modulation. Angiotensin II (Ang II) serves as its main effector hormone upon binding to its primary receptor, Ang II receptor type 1 (AT1R). It is well established that an intrinsic independent brain RAS exists. Abnormal AT1R activation both in the periphery and in the brain probably contributes to the development of Alzheimer's disease (AD) pathology that is characterized, among others, by brain inflammation. Moreover, treatment with drugs that block AT1R (AT1R blockers, ARBs) ameliorates most of the clinical risk factors leading to AD. Previously we showed that short period of intranasal treatment with telmisartan (a brain penetrating ARB) reduced brain inflammation and ameliorated amyloid burden (a component of Alzheimer's plaques) in AD transgenic mouse model. In the present study, we aimed to examine the long-term effect of intranasally administrated telmisartan on brain inflammation features including microglial activation, astrogliosis, neuronal loss and hippocampus-dependent cognition in five-familial AD mouse model (5XFAD). Five month of intranasal treatment with telmisartan significantly reduced amyloid burden in the cortex and hippocampus of 5XFAD mice as compared with the vehicle-treated 5XFAD group. Similar effects were also observed for CD11b staining, which is a marker for microglial accumulation. Telmisartan also significantly reduced astrogliosis and neuronal loss in the cortex of 5XFAD mice compared with the vehicle-treated group. Improved spatial acquisition of the 5XFAD mice following long-term intranasal administration of telmisartan was also observed. Taken together, our data suggest a significant role for AT1R blockage in mediating neuronal loss and cognitive behavior, possibly through regulation of amyloid burden and glial inflammation.
Collapse
|
39
|
Yang W, Sabi-Mouka EMB, Wang L, Shu C, Wang Y, Ding J, Ding L. Determination of tranilast in bio-samples by LC-MS/MS: Application to a pharmacokinetic and brain tissue distribution study in rats. J Pharm Biomed Anal 2017; 147:479-484. [PMID: 28774678 DOI: 10.1016/j.jpba.2017.06.047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 06/11/2017] [Accepted: 06/17/2017] [Indexed: 10/19/2022]
Abstract
As a potent drug used to improve the neurodegenerative conditions, there is few information about the brain tissue distribution of tranilast by now. In this study, a novel sensitive LC-MS/MS method has been developed and validated to determine tranilast in rat brain tissue samples. The calibration curve showed good linearity ranged from 2.140 to 428.0ng·mL-1. The method was fully validated and successfully applied in the brain tissue distribution study of tranilast in rats, which had never been reported in detail by now. Furthermore, a rapid LC-MS/MS method with a short run time of 3min was developed and validated for the determination of tranilast in rat plasma and the application to a pharmacokinetic study of tranilast in rats. After oral dosage of 10.5mg·kg-1 tranilast, the maximum plasma concentration (Cmax1) of tranilast was (18.59±5.40) μg·mL-1 at (0.667±0.408) h while the area under the curve (AUC0-24) was (54.87±14.13) μg·h·mL-1 with the elimination half-life of (2.93±0.41) h. The ratio calculated by dividing the concentration of tranilast in brain with the concentration of tranilast in the plasma, was (0.6042%±0.0572%), (0.7484%±0.0883%), (0.5914%±0.0416%) and (0.3830%±0.1632%) at 0.167, 0.5, 2 and 8h, respectively. The results showed that tranilast with fast absorption could penetrate the rat brain blood barrier after oral gavage. The obtained data also showed that tranilast could be quickly distributed and eliminated in brain tissue.
Collapse
Affiliation(s)
- Wen Yang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China
| | - Eboka Majolene B Sabi-Mouka
- Department of Pharmaceutical Analysis, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China
| | - Lei Wang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China
| | - Chang Shu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China
| | - Yan Wang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China; College of Pharmacy and Chemistry, Dali University, Wanhua Road, Dali 671000, China
| | - Juefang Ding
- Department of Pharmaceutical Analysis, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China; Nanjing Clinical Tech Laboratories Inc., 18 Zhilan Road, Jiangning District, Nanjing 211000, China
| | - Li Ding
- Department of Pharmaceutical Analysis, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China.
| |
Collapse
|
40
|
Lumniczky K, Szatmári T, Sáfrány G. Ionizing Radiation-Induced Immune and Inflammatory Reactions in the Brain. Front Immunol 2017; 8:517. [PMID: 28529513 PMCID: PMC5418235 DOI: 10.3389/fimmu.2017.00517] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/18/2017] [Indexed: 01/17/2023] Open
Abstract
Radiation-induced late brain injury consisting of vascular abnormalities, demyelination, white matter necrosis, and cognitive impairment has been described in patients subjected to cranial radiotherapy for brain tumors. Accumulating evidence suggests that various degrees of cognitive deficit can develop after much lower doses of ionizing radiation, as well. The pathophysiological mechanisms underlying these alterations are not elucidated so far. A permanent deficit in neurogenesis, chronic microvascular alterations, and blood–brain barrier dysfunctionality are considered among the main causative factors. Chronic neuroinflammation and altered immune reactions in the brain, which are inherent complications of brain irradiation, have also been directly implicated in the development of cognitive decline after radiation. This review aims to give a comprehensive overview on radiation-induced immune alterations and inflammatory reactions in the brain and summarizes how these processes can influence cognitive performance. The available data on the risk of low-dose radiation exposure in the development of cognitive impairment and the underlying mechanisms are also discussed.
Collapse
Affiliation(s)
- Katalin Lumniczky
- Division of Radiation Medicine, National Public Health Centre, National Research Directorate for Radiobiology and Radiohygiene, Budapest, Hungary
| | - Tünde Szatmári
- Division of Radiation Medicine, National Public Health Centre, National Research Directorate for Radiobiology and Radiohygiene, Budapest, Hungary
| | - Géza Sáfrány
- Division of Radiation Medicine, National Public Health Centre, National Research Directorate for Radiobiology and Radiohygiene, Budapest, Hungary
| |
Collapse
|
41
|
Burns TC, Awad AJ, Li MD, Grant GA. Radiation-induced brain injury: low-hanging fruit for neuroregeneration. Neurosurg Focus 2017; 40:E3. [PMID: 27132524 DOI: 10.3171/2016.2.focus161] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Brain radiation is a fundamental tool in neurooncology to improve local tumor control, but it leads to profound and progressive impairments in cognitive function. Increased attention to quality of life in neurooncology has accelerated efforts to understand and ameliorate radiation-induced cognitive sequelae. Such progress has coincided with a new understanding of the role of CNS progenitor cell populations in normal cognition and in their potential utility for the treatment of neurological diseases. The irradiated brain exhibits a host of biochemical and cellular derangements, including loss of endogenous neurogenesis, demyelination, and ablation of endogenous oligodendrocyte progenitor cells. These changes, in combination with a state of chronic neuroinflammation, underlie impairments in memory, attention, executive function, and acquisition of motor and language skills. Animal models of radiation-induced brain injury have demonstrated a robust capacity of both neural stem cells and oligodendrocyte progenitor cells to restore cognitive function after brain irradiation, likely through a combination of cell replacement and trophic effects. Oligodendrocyte progenitor cells exhibit a remarkable capacity to migrate, integrate, and functionally remyelinate damaged white matter tracts in a variety of preclinical models. The authors here critically address the opportunities and challenges in translating regenerative cell therapies from rodents to humans. Although valiant attempts to translate neuroprotective therapies in recent decades have almost uniformly failed, the authors make the case that harnessing human radiation-induced brain injury as a scientific tool represents a unique opportunity to both successfully translate a neuroregenerative therapy and to acquire tools to facilitate future restorative therapies for human traumatic and degenerative diseases of the central nervous system.
Collapse
Affiliation(s)
- Terry C Burns
- Department of Neurosurgery and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California
| | - Ahmed J Awad
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York;,Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine; and
| | - Matthew D Li
- Stanford University School of Medicine, Stanford, California
| | - Gerald A Grant
- Department of Neurosurgery and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California
| |
Collapse
|
42
|
Zakharov S, Kotikova K, Nurieva O, Hlusicka J, Kacer P, Urban P, Vaneckova M, Seidl Z, Diblik P, Kuthan P, Navratil T, Pelclova D. Leukotriene-mediated neuroinflammation, toxic brain damage, and neurodegeneration in acute methanol poisoning. Clin Toxicol (Phila) 2017; 55:249-259. [DOI: 10.1080/15563650.2017.1284332] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Sergey Zakharov
- Department of Occupational Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Toxicological Information Centre, General University Hospital, Prague, Czech Republic
| | - Katerina Kotikova
- Department of Occupational Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Olga Nurieva
- Department of Occupational Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jiri Hlusicka
- Department of Occupational Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Toxicological Information Centre, General University Hospital, Prague, Czech Republic
| | - Petr Kacer
- Institute of Chemical Technology, Prague, Czech Republic
| | - Pavel Urban
- Department of Occupational Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Centre for Industrial Hygiene and Occupational Medicine, National Institute of Public Health, Prague, Czech Republic
| | - Manuela Vaneckova
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Zdenek Seidl
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Pavel Diblik
- Department of Ophthalmology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Pavel Kuthan
- Department of Ophthalmology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Tomas Navratil
- Toxicological Information Centre, General University Hospital, Prague, Czech Republic
- Department of Biomimetic Electrochemistry, J. Heyrovský Institute of Physical Chemistry of the AS CR, v.v.i, Prague, Czech Republic
| | - Daniela Pelclova
- Department of Occupational Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Toxicological Information Centre, General University Hospital, Prague, Czech Republic
| |
Collapse
|
43
|
Abstract
OBJECTIVE Epilepsy is a chronic neurological disease characterised with seizures. The aetiology of the most generalised epilepsies cannot be explicitly determined and the seizures are pronounced to be genetically determined by disturbances of receptors in central nervous system. Besides, neurotransmitter distributions or other metabolic problems are supposed to involve in epileptogenesis. Lack of adequate data about pharmacological agents that have antiepileptogenic effects point to need of research on this field. Thus, in this review, inflammatory aspects of epileptogenesis has been focussed via considering several concepts like role of immune system, blood-brain barrier and antibody involvement in epileptogenesis. METHODS We conducted an evidence-based review of the literatures in order to evaluate the possible participation of inflammatory processes to epileptogenesis and also, promising agents which are effective to these processes. We searched PubMed database up to November 2015 with no date restrictions. RESULTS In the present review, 163 appropriate articles were included. Obtained data suggests that inflammatory processes participate to epileptogenesis in several ways like affecting fibroblast growth factor-2 and tropomyosin receptor kinase B signalling pathways, detrimental proinflammatory pathways [such as the interleukin-1 beta (IL-1β)-interleukin-1 receptor type 1 (IL-1R1) system], mammalian target of rapamycin pathway, microglial activities, release of glial inflammatory proteins (such as macrophage inflammatory protein, interleukin 6, C-C motif ligand 2 and IL-1β), adhesion molecules that are suggested to function in signalling pathways between neurons and microglia and also linkage between these molecules and proinflammatory cytokines. CONCLUSION The literature research indicated that inflammation is a part of epileptogenesis. For this reason, further studies are necessary for assessing agents that will be effective in clinical use for therapeutic treatment of epileptogenesis.
Collapse
|
44
|
Rodriguez-Callejas JD, Fuchs E, Perez-Cruz C. Evidence of Tau Hyperphosphorylation and Dystrophic Microglia in the Common Marmoset. Front Aging Neurosci 2016; 8:315. [PMID: 28066237 PMCID: PMC5177639 DOI: 10.3389/fnagi.2016.00315] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/08/2016] [Indexed: 01/22/2023] Open
Abstract
Common marmosets (Callithrix jacchus) have recently gained popularity in biomedical research as models of aging research. Basically, they confer advantages from other non-human primates due to their shorter lifespan with onset of appearance of aging at 8 years. Old marmosets present some markers linked to neurodegeneration in the brain such as amyloid beta (Aβ)1-42 and Aβ1-40. However, there are no studies exploring other cellular markers associated with neurodegenerative diseases in this non-human primate. Using immunohistochemistry, we analyzed brains of male adolescent, adult, old, and aged marmosets. We observed accumulation of Aβ1-40 and Aβ1-42 in the cortex of aged subjects. Tau hyperphosphorylation was already detected in the brain of adolescent animals and increased with aging in a more fibrillary form. Microglia activation was also observed in the aging process, while a dystrophic phenotype accumulates in aged subjects. Interestingly, dystrophic microglia contained hyperphosphorylated tau, but active microglia did not. These results support previous findings regarding microglia dysfunctionality in aging and neurodegenerative diseases as Alzheimer's disease. Further studies should explore the functional consequences of these findings to position this non-human primate as animal model of aging and neurodegeneration.
Collapse
Affiliation(s)
- Juan D Rodriguez-Callejas
- Laboratory of Neuroplasticity and Neurodegeneration, Department of Pharmacology, Center for Research and Advanced Studies (CINVESTAV) Mexico City, Mexico
| | - Eberhard Fuchs
- Clinical Neurobiology Laboratory, German Primate Center - Leibniz Institute for Primate Research, Göttingen Germany
| | - Claudia Perez-Cruz
- Laboratory of Neuroplasticity and Neurodegeneration, Department of Pharmacology, Center for Research and Advanced Studies (CINVESTAV) Mexico City, Mexico
| |
Collapse
|
45
|
Park E, Chun HS. Melatonin Attenuates Manganese and Lipopolysaccharide-Induced Inflammatory Activation of BV2 Microglia. Neurochem Res 2016; 42:656-666. [DOI: 10.1007/s11064-016-2122-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/26/2016] [Accepted: 11/21/2016] [Indexed: 01/28/2023]
|
46
|
Bolós M, Llorens-Martín M, Jurado-Arjona J, Hernández F, Rábano A, Avila J. Direct Evidence of Internalization of Tau by Microglia In Vitro and In Vivo. J Alzheimers Dis 2016; 50:77-87. [PMID: 26638867 DOI: 10.3233/jad-150704] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The microtubule-associated protein (MAP) tau plays a critical role in the pathogenesis of tauopathies. Excess tau can be released into the extracellular medium in a physiological or pathological manner to be internalized by surrounding neurons-a process that contributes to the spread of this protein throughout the brain. Such spreading may correlate with the progression of the abovementioned diseases. In addition to neurons, tau can be internalized into other cells. Here we demonstrate that microglia take up tau in vitro and in vivo. In this regard, microglia from primary cultures internalized soluble (human recombinant tau42) and insoluble (homogenates derived from human AD brain) tau in vitro. Furthermore, using stereotaxic injection of tau in mice in vivo, we show that murine microglia internalize human tau. In addition, we demonstrate, for the first time, that microglia colocalize with various forms of tau in postmortem brain tissue of patients with Alzheimer's disease and non-demented control subjects. Our data reveal a potential role of microglia in the internalization of tau that might be relevant for the design of strategies to enhance the clearance of extracellular tau in neurodegenerative diseases characterized by the accumulation of this protein.
Collapse
Affiliation(s)
- Marta Bolós
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa" CSIC-UAM, Madrid, Spain
| | - María Llorens-Martín
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa" CSIC-UAM, Madrid, Spain
| | - Jerónimo Jurado-Arjona
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa" CSIC-UAM, Madrid, Spain
| | - Félix Hernández
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa" CSIC-UAM, Madrid, Spain
| | - Alberto Rábano
- Neuropathology Department, CIEN Foundation, Madrid, Spain
| | - Jesús Avila
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa" CSIC-UAM, Madrid, Spain
| |
Collapse
|
47
|
Ahmed ME, Tucker D, Dong Y, Lu Y, Zhao N, Wang R, Zhang Q. Methylene Blue promotes cortical neurogenesis and ameliorates behavioral deficit after photothrombotic stroke in rats. Neuroscience 2016; 336:39-48. [PMID: 27590267 DOI: 10.1016/j.neuroscience.2016.08.036] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/06/2016] [Accepted: 08/22/2016] [Indexed: 01/02/2023]
Abstract
Ischemic stroke in rodents stimulates neurogenesis in the adult brain and the proliferation of newborn neurons that migrate into the penumbra zone. The present study investigated the effect of Methylene Blue (MB) on neurogenesis and functional recovery in a photothrombotic (PT) model of ischemic stroke in rats. PT stroke model was induced by photo-activation of Rose Bengal dye in cerebral blood flow by cold fiber light. Rats received intraperitoneal injection of either MB (0.5mg/kg/day) from day 1 to day 5 after stroke or an equal volume of saline solution as a control. Cell proliferative marker 5-bromodeoxyuridine (BrdU) was injected twice daily (50mg/kg) from day 2 to day 8 and animals were sacrificed on day 12 after PT induction. We report that MB significantly enhanced cell proliferation and neurogenesis, as evidenced by the increased co-localizations of BrdU/NeuN, BrdU/DCX, BrdU/MAP2 and BrdU/Ki67 in the peri-infarct zone compared with vehicle controls. MB thus effectively limited infarct volume and improved neurological deficits compared to PT control animals. The effects of MB were accompanied with an attenuated level of reactive gliosis and release of pro-inflammatory cytokines, as well as elevated levels of cytochrome c oxidase activity and ATP production in peri-infarct regions. Our study provides important information that MB has the ability to promote neurogenesis and enhance the newborn-neurons' survival in ischemic brain repair by inhibiting microenvironmental inflammation and increasing mitochondrial function.
Collapse
Affiliation(s)
- Mohammad Ejaz Ahmed
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA
| | - Donovan Tucker
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA
| | - Yan Dong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA
| | - Yujiao Lu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA
| | - Ningjun Zhao
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA
| | - Ruimin Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA
| | - Quanguang Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA
| |
Collapse
|
48
|
Daulatzai MA. Dysfunctional Sensory Modalities, Locus Coeruleus, and Basal Forebrain: Early Determinants that Promote Neuropathogenesis of Cognitive and Memory Decline and Alzheimer’s Disease. Neurotox Res 2016; 30:295-337. [DOI: 10.1007/s12640-016-9643-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 06/08/2016] [Accepted: 06/10/2016] [Indexed: 12/22/2022]
|
49
|
Abstract
BACKGROUND Previous literature indicates that flow-mediated dilation (FMD) is associated with impaired cognition among patients with stroke. The relationship between FMD and cognition in individuals without cerebrovascular disease has yet to be systematically reviewed. METHODS The literature was searched using MEDLINE. Exclusion criteria were as follows: focus on neurological disease (e.g., stroke), animal studies, no quantitative measure of endothelial function or cognition, newborn studies, articles with no original data, and articles that are irrelevant to the topic of interest. Neurocognitive tests were categorized in the following domains: executive function, memory (general, working, episodic/semantic, verbal, visual), global cognitive function, information processing speed, language, psychomotor speed, and visual-spatial ability. RESULTS The search yielded 700 articles, of which 10 articles, consisting of 2791 participants, met the criteria for inclusion. Most studies conclude that impaired FMD is associated with poorer neuropsychological functioning, particularly in executive functioning (effect sizes: r = 0.07-0.58) and working memory tasks (effect sizes: r = 0.19-0.39). No association was found between other subdomains of memory and FMD. Visual spatial tasks, information processing speed, language tasks, and global cognition were not associated with FMD overall; however fewer studies examined these domains. CONCLUSIONS Even in the absence of cerebrovascular disease, there are links between cognition, particularly executive tasks, and vascular function. Public health implications include the potential value of examining FMD as a predictor of cognitive decline, as well as the potential value of improving cognition through pharmacological and behavioral interventions that improve vascular function. Future studies incorporating neuroimaging measures of cerebral blood flow are warranted.
Collapse
|
50
|
Tong F, Zhang J, Liu L, Gao X, Cai Q, Wei C, Dong J, Hu Y, Wu G, Dong X. Corilagin Attenuates Radiation-Induced Brain Injury in Mice. Mol Neurobiol 2015; 53:6982-6996. [PMID: 26666668 DOI: 10.1007/s12035-015-9591-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 11/30/2015] [Indexed: 01/12/2023]
Abstract
Cranial irradiation-induced inflammation plays a critical role in the initiation and progression of radiation-induced brain injury (RIBI). Anti-inflammation treatment may provide therapeutic benefits. Corilagin (beta-1-O-galloyl-3, 6-(R)-hexahydroxydiphenoyl-D-glucose, C27H22O18) was a novel member of the tannin family with anti-inflammatory properties and is isolated from some medicinal plants, such as Phyllanthus amarus and Caesalpinia coriaria. In this study, the effect of Corilagin on RIBI was investigated and the underlying mechanisms were explored. Spatial learning and memory ability of mice were investigated by the Morris water maze test. Evans blue leakage and electron microscopy were used to assess the integrity of blood-brain barrier (BBB). The mRNA and protein expressions of inflammatory cytokines, TNF-α and IL-1β, were measured by using real-time PCR and Western blotting. The activation of microglial cells and expression of TNF-α were examined by immunofluorescence staining. Phosphorylated signal transducers and activators of transcription 3 (p-STAT3) and IκBα, and the translocation of p65 from cytoplasm to nucleus were detected by using Western blotting. Morris water maze test showed that Corilagin ameliorated the neurocognitive deficits in RIBI mice. Evans blue leakage and electron microscopy exhibited that Corilagin partially protected the BBB integrity from cranial irradiation-caused damage; immunofluorescence staining showed that Corilagin could inhibit microglial activation and TNF-α expression. Real-time PCR and Western blotting revealed that Corilagin downregulated the expression of TNF-α and IL-1β and inhibited the irradiation-induced activation of NF-κB pathways by upregulating p-STAT3 expression. In conclusion, Corilagin could attenuate RIBI through inhibiting microglial activation and the expressions of inflammatory cytokines. Corilagin might inhibit the activation of NF-κB pathway in a STAT3-associated manner, thereby downregulating the inflammatory cytokine expressions.
Collapse
Affiliation(s)
- Fan Tong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Jian Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Li Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Xican Gao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Qian Cai
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Chunhua Wei
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Jihua Dong
- Experimental Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
| | - Xiaorong Dong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
| |
Collapse
|