1
|
Basheer N, Muhammadi MK, Freites CL, Avila M, Momand MUD, Hryntsova N, Smolek T, Katina S, Zilka N. TLR4-mediated chronic neuroinflammation has no effect on tangle pathology in a tauopathy mouse model. Front Aging Neurosci 2024; 16:1468602. [PMID: 39503044 PMCID: PMC11536299 DOI: 10.3389/fnagi.2024.1468602] [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: 07/22/2024] [Accepted: 08/26/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Alzheimer's disease (AD) is marked by the accumulation of fibrillary aggregates composed of pathological tau protein. Although neuroinflammation is frequently observed in conjunction with tau pathology, current preclinical evidence does not sufficiently establish a direct causal role in tau tangle formation. This study aimed to evaluate whether chronic Toll-like receptor 4 (TLR4) stimulation, induced by a high dose of lipopolysaccharide (LPS, 5 mg/kg), exacerbates neurofibrillary tangle (NFT) pathology in a transgenic mouse model of tauopathy that expresses human truncated 151-391/3R tau, an early feature of sporadic AD. Methods We utilized a transgenic mouse model of tauopathy subjected to chronic TLR4 stimulation via weekly intraperitoneal injections of LPS over nine consecutive weeks. Neurofibrillary tangle formation, microglial activation, and tau hyperphosphorylation in the brainstem and hippocampus were assessed through immunohistochemistry, immunofluorescence, and detailed morphometric analysis of microglia. Results Chronic LPS treatment led to a significant increase in the number of Iba-1+ microglia in the LPS-treated group compared to the sham group (p < 0.0001). Notably, there was a 1.5- to 1.7-fold increase in microglia per tangle-bearing neuron in the LPS-treated group. These microglia exhibited a reactive yet exhausted phenotype, characterized by a significant reduction in cell area (p < 0.0001) without significant changes in other morphometric parameters, such as perimeter, circumference, solidity, aspect ratio, or arborization degree. Despite extensive microglial activation, there was no observed reduction in tau hyperphosphorylation or a decrease in tangle formation in the brainstem, where pathology predominantly develops in this model. Discussion These findings suggest that chronic TLR4 stimulation in tau-transgenic mice results in significant microglial activation but does not influence tau tangle formation. This underscores the complexity of the relationship between neuroinflammation and tau pathology, indicating that additional mechanisms may be required for neuroinflammation to directly contribute to tau tangle formation.
Collapse
Affiliation(s)
- Neha Basheer
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | - Carlos Leandro Freites
- Institute of Histology and Embryology of Mendoza (IHEM), National University of Cuyo, National Scientific and Technical Research Council (CONICET), Mendoza, Argentina
| | - Martin Avila
- Institute of Histology and Embryology of Mendoza (IHEM), National University of Cuyo, National Scientific and Technical Research Council (CONICET), Mendoza, Argentina
| | - Miraj Ud Din Momand
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Natalia Hryntsova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Tomas Smolek
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Stanislav Katina
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Institute of Mathematics and Statistics, Faculty of Science, Masaryk University, Brno, Czechia
| | - Norbert Zilka
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
2
|
Wang L, Wang Q, Wang X, Yang C, Wang X, Liu H, Wang H. Intermittent fasting alleviates postoperative cognitive dysfunction by reducing neuroinflammation in aged mice. Brain Res Bull 2024; 216:111034. [PMID: 39053649 DOI: 10.1016/j.brainresbull.2024.111034] [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: 04/27/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Elderly individuals undergoing surgical procedures are often confronted with the peril of experiencing postoperative cognitive dysfunction (POCD). Prior research has demonstrated the exacerbating effect of sevoflurane anesthesia on neuroinflammation, which can further deteriorate the condition of POCD in elderly patients. Intermittent fasting (IF) restricts food consumption to a specific time window and has been demonstrated to ameliorate cognitive dysfunction induced by neuropathic inflammation. We subjected 18-month-old male mice to 16 hours of fasting and 8 hours of unrestricted eating over a 24-hour period for 0, 1, 2, and 4 weeks, followed by abdominal exploration under sevoflurane anesthesia. In this study, we aim to explore the potential impact of IF on postoperative cognitive function in aged mice undergoing sevoflurane surgery through the preoperative implementation of IF measures. The findings indicate two weeks of IF leads to a significant enhancement of learning and memory capabilities in mice following surgery. The cognitive performance, as determined by the novel object recognition and Morris water maze tests, as well as the synaptic plasticity, as measured by in vivo electrophysiological recordings, has demonstrated marked improvements. Furthermore, the administration of IF markedly enhances the expression of synaptic-associated proteins in hippocampal neurons, concomitant with a decreasing expression of pro-inflammatory factors and a reduced density of microglial cells within the hippocampal brain region. To summarize, the results of this study indicate that IF may mitigate inflammation in the hippocampal area of the brain. Furthermore, IF appears to provide a safeguard against cognitive impairment and synaptic plasticity impairment brought on by sevoflurane anesthesia.
Collapse
Affiliation(s)
- Lei Wang
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China; Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, China
| | - Qiang Wang
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Xiaoqing Wang
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Chenyi Yang
- Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China; Nankai University 300071, Tianjin, China; Nankai University Affinity the Third Central Hospital, Tianjin 300170, China
| | - Xinyi Wang
- Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China; Nankai University 300071, Tianjin, China; Nankai University Affinity the Third Central Hospital, Tianjin 300170, China
| | - Huan Liu
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Haiyun Wang
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China; Nankai University 300071, Tianjin, China; Nankai University Affinity the Third Central Hospital, Tianjin 300170, China.
| |
Collapse
|
3
|
Frazier HN, Braun DJ, Bailey CS, Coleman MJ, Davis VA, Dundon SR, McLouth CJ, Muzyk HC, Powell DK, Rogers CB, Roy SM, Van Eldik LJ. A small molecule p38α MAPK inhibitor, MW150, attenuates behavioral deficits and neuronal dysfunction in a mouse model of mixed amyloid and vascular pathologies. Brain Behav Immun Health 2024; 40:100826. [PMID: 39161874 PMCID: PMC11331815 DOI: 10.1016/j.bbih.2024.100826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 07/20/2024] [Indexed: 08/21/2024] Open
Abstract
Background Inhibition of p38 alpha mitogen activated protein kinase (p38α) has shown great promise as a treatment for Alzheimer's disease (AD) in preclinical tests. However, previous preclinical studies were performed in "pure" models of AD pathology. A vast majority of AD patients have comorbid dementia-contributing pathologies, particularly some form of vascular damage. The present study therefore aimed to test the potential of p38α inhibition to address dysfunction in the context of comorbid amyloid and vascular pathologies. Methods An amyloid overexpressing mouse strain (5xFAD) was placed on an 8-week long diet to induce the hyperhomocysteinemia (HHcy) model of small vessel disease. Mice were treated with the brain-penetrant small molecule p38α inhibitor MW150 for the duration of the HHcy diet, and subsequently underwent behavioral, neuroimaging, electrophysiological, or biochemical/immunohistochemical analyses. Results MW150 successfully reduced behavioral impairment in the Morris Water Maze, corresponding with attenuation of synaptic loss, reduction in tau phosphorylation, and a partial normalization of electrophysiological parameters. No effect of MW150 was observed on the amyloid, vascular, or neuroinflammatory endpoints measured. Conclusions This study provides proof-of-principle that the inhibition of p38α is able to provide benefit even in the context of mixed pathological contributions to cognitive impairment. Interestingly, the benefit was mediated primarily via rescue of neuronal function without any direct effects on the primary pathologies. These data suggest a potential use for p38 inhibitors in the preservation of cognition across contexts, and in particular AD, either alone or as an adjunct to other AD therapies (i.e. anti-amyloid approaches). Future studies to delineate the precise neuronal pathways implicated in the benefit may help define other specific comorbid conditions amenable to this type of approach or suggest future refinement in pharmacological targeting.
Collapse
Affiliation(s)
- Hilaree N. Frazier
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - David J. Braun
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY, 40536, USA
| | - Caleb S. Bailey
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - Meggie J. Coleman
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - Verda A. Davis
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - Stephen R. Dundon
- Magnetic Resonance Imaging & Spectroscopy Center, University of Kentucky, Lexington, KY, 40536, USA
| | | | - Hana C. Muzyk
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - David K. Powell
- Department of Neuroscience, University of Kentucky, Lexington, KY, 40536, USA
- Magnetic Resonance Imaging & Spectroscopy Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Colin B. Rogers
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - Saktimayee M. Roy
- Department of Pharmacology, Northwestern University, Chicago, IL, 60611, USA
| | - Linda J. Van Eldik
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY, 40536, USA
| |
Collapse
|
4
|
Yang K, Wang X, Pan H, Wang X, Hu Y, Yao Y, Zhao X, Sun T. The roles of AIM2 in neurodegenerative diseases: insights and therapeutic implications. Front Immunol 2024; 15:1441385. [PMID: 39076969 PMCID: PMC11284019 DOI: 10.3389/fimmu.2024.1441385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/01/2024] [Indexed: 07/31/2024] Open
Abstract
AIM2, a cytosolic innate immune receptor, has the capability to recognize double-stranded DNA (dsDNA). This paper delineates the structural features of AIM2 and its mechanisms of activation, emphasizing its capacity to detect cytosolic DNA and initiate inflammasome assembly. Additionally, we explore the diverse functions of AIM2 in different cells. Insights into AIM2-mediated neuroinflammation provide a foundation for investigating novel therapeutic strategies targeting AIM2 signaling pathways. Furthermore, we present a comprehensive review of the roles of AIM2 in neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD). Finally, we discuss its therapeutic implications. In conclusion, a profound understanding of AIM2 in neurodegenerative diseases may facilitate the development of effective interventions to mitigate neuronal damage and slow disease progression.
Collapse
Affiliation(s)
- Kai Yang
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Xi Wang
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Hanyu Pan
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Xinqing Wang
- Hunan Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Yunhan Hu
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Yihe Yao
- Institute of WUT-AMU, Wuhan University of Technology, Wuhan, China
| | - Xinyue Zhao
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, China
| |
Collapse
|
5
|
Karnik SJ, Margetts TJ, Wang HS, Movila A, Oblak AL, Fehrenbacher JC, Kacena MA, Plotkin LI. Mind the Gap: Unraveling the Intricate Dance Between Alzheimer's Disease and Related Dementias and Bone Health. Curr Osteoporos Rep 2024; 22:165-176. [PMID: 38285083 PMCID: PMC10912190 DOI: 10.1007/s11914-023-00847-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 01/30/2024]
Abstract
PURPOSE OF REVIEW This review examines the linked pathophysiology of Alzheimer's disease/related dementia (AD/ADRD) and bone disorders like osteoporosis. The emphasis is on "inflammaging"-a low-level inflammation common to both, and its implications in an aging population. RECENT FINDINGS Aging intensifies both ADRD and bone deterioration. Notably, ADRD patients have a heightened fracture risk, impacting morbidity and mortality, though it is uncertain if fractures worsen ADRD. Therapeutically, agents targeting inflammation pathways, especially Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) and TNF-α, appear beneficial for both conditions. Additionally, treatments like Sirtuin 1 (SIRT-1), known for anti-inflammatory and neuroprotective properties, are gaining attention. The interconnectedness of AD/ADRD and bone health necessitates a unified treatment approach. By addressing shared mechanisms, we can potentially transform therapeutic strategies, enriching our understanding and refining care in our aging society. This review article is part of a series of multiple manuscripts designed to determine the utility of using artificial intelligence for writing scientific reviews.
Collapse
Affiliation(s)
- Sonali J Karnik
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Tyler J Margetts
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Hannah S Wang
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Alexandru Movila
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Adrian L Oblak
- Department of Radiology & Imaging Sciences, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jill C Fehrenbacher
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA.
| | - Lilian I Plotkin
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
6
|
Litvinenko IV, Naumov KM, Lobzin VY, Emelin AY, Dynin PS, Kolmakova KA, Nikishin VO. [Traumatic brain injury as risk factor of Alzheimer's disease and possibilities of pathogenetic therapy]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:45-54. [PMID: 38261283 DOI: 10.17116/jnevro202412401145] [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] [Indexed: 01/24/2024]
Abstract
The article examines the potential role of brain mechanical damage as a trigger for the development of neurodegenerative changes. Attention is paid to dysfunction of the neurovascular unit, and disruption of the functional and compensatory capabilities of blood flow. The importance of microhemorrhages that occur in the acute period of injury and the formation of first focal and then diffuse neuroinflammation is emphasized. The importance of mitochondrial dysfunction was separately determined as a significant factor in increasing the risk of developing Alzheimer's disease (AD) in patients after traumatic brain injury (TBI). In TBI, there is a decrease in the expression of tight junction (TC) proteins of endothelial cells, such as occludin, claudin, JP, which leads to increased permeability of the blood-brain barrier. TBI, provoking endothelial dysfunction, contributes to the development of metabolic disorders of β-amyloid and tau protein, which in turn leads to worsening vascular damage, resulting in a vicious circle that can ultimately lead to the development of AD and dementia. Age-related changes in cerebral arteries, which impair perivascular transport of interstitial fluid, are currently considered as an important part of the «amyloid cascade», especially against the background of genetically mediated disorders of glial membranes associated with defective aquaporin-4 (encoded by the APOE4). Studies in animal models of TBI have revealed an increase in tau protein immunoreactivity and its phosphorylation, which correlates with the severity of injury. A comprehensive analysis of research results shows that the cascade of reactions triggered by TBI includes all the main elements of the pathogenesis of AD: disorders of energy metabolism, microcirculation and clearance of cerebral metabolic products. This leads to a disruption in the metabolism of amyloid protein and its accumulation in brain tissue with the subsequent development of tauopathy. Cerebrolysin, by modulating the permeability of the blood-brain barrier, blocks the development of neuroinflammation, reduces the accumulation of pathological forms of proteins and may be slow down the progression of neurodegeneration.
Collapse
Affiliation(s)
| | - K M Naumov
- Kirov Military Medical Academy, St. Petersburg, Russia
| | - V Yu Lobzin
- Kirov Military Medical Academy, St. Petersburg, Russia
- Mechnikov North-Western State Medical University, St. Petersburg, Russia
| | - A Yu Emelin
- Kirov Military Medical Academy, St. Petersburg, Russia
| | - P S Dynin
- Kirov Military Medical Academy, St. Petersburg, Russia
| | - K A Kolmakova
- Kirov Military Medical Academy, St. Petersburg, Russia
| | - V O Nikishin
- Kirov Military Medical Academy, St. Petersburg, Russia
| |
Collapse
|
7
|
Sun S, Li J, Wang S, Li J, Ren J, Bao Z, Sun L, Ma X, Zheng F, Ma S, Sun L, Wang M, Yu Y, Ma M, Wang Q, Chen Z, Ma H, Wang X, Wu Z, Zhang H, Yan K, Yang Y, Zhang Y, Zhang S, Lei J, Teng ZQ, Liu CM, Bai G, Wang YJ, Li J, Wang X, Zhao G, Jiang T, Belmonte JCI, Qu J, Zhang W, Liu GH. CHIT1-positive microglia drive motor neuron ageing in the primate spinal cord. Nature 2023; 624:611-620. [PMID: 37907096 DOI: 10.1038/s41586-023-06783-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 10/25/2023] [Indexed: 11/02/2023]
Abstract
Ageing is a critical factor in spinal-cord-associated disorders1, yet the ageing-specific mechanisms underlying this relationship remain poorly understood. Here, to address this knowledge gap, we combined single-nucleus RNA-sequencing analysis with behavioural and neurophysiological analysis in non-human primates (NHPs). We identified motor neuron senescence and neuroinflammation with microglial hyperactivation as intertwined hallmarks of spinal cord ageing. As an underlying mechanism, we identified a neurotoxic microglial state demarcated by elevated expression of CHIT1 (a secreted mammalian chitinase) specific to the aged spinal cords in NHP and human biopsies. In the aged spinal cord, CHIT1-positive microglia preferentially localize around motor neurons, and they have the ability to trigger senescence, partly by activating SMAD signalling. We further validated the driving role of secreted CHIT1 on MN senescence using multimodal experiments both in vivo, using the NHP spinal cord as a model, and in vitro, using a sophisticated system modelling the human motor-neuron-microenvironment interplay. Moreover, we demonstrated that ascorbic acid, a geroprotective compound, counteracted the pro-senescent effect of CHIT1 and mitigated motor neuron senescence in aged monkeys. Our findings provide the single-cell resolution cellular and molecular landscape of the aged primate spinal cord and identify a new biomarker and intervention target for spinal cord degeneration.
Collapse
Affiliation(s)
- Shuhui Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jiaming Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China
- Aging Biomarker Consortium, Beijing, China
- The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Jingyi Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Aging Biomarker Consortium, Beijing, China
| | - Jie Ren
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Aging Biomarker Consortium, Beijing, China
- Key Laboratory of RNA Science and Engineering, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Zhaoshi Bao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- The Chinese Glioma Genome Atlas, Beijing, China
| | - Le Sun
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xibo Ma
- MAIS, State Key Laboratory of Multimodal Artificial Intelligence Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
- College of Medicine and Biomedical Information Engineering, Northeastern University, Shenyang, China
| | - Fangshuo Zheng
- The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Aging Biomarker Consortium, Beijing, China
| | - Liang Sun
- Aging Biomarker Consortium, Beijing, China
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Min Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Science and Technology of China, Hefei, China
| | - Yan Yu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Miyang Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qiaoran Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhiyuan Chen
- MAIS, State Key Laboratory of Multimodal Artificial Intelligence Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - He Ma
- MAIS, State Key Laboratory of Multimodal Artificial Intelligence Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- College of Medicine and Biomedical Information Engineering, Northeastern University, Shenyang, China
| | - Xuebao Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zeming Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Hui Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kaowen Yan
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yuanhan Yang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yixin Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Sheng Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jinghui Lei
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhao-Qian Teng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chang-Mei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ge Bai
- The MOE Frontier Research Center of Brain & Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China
| | - Yan-Jiang Wang
- Aging Biomarker Consortium, Beijing, China
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Jian Li
- Aging Biomarker Consortium, Beijing, China
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Xiaoqun Wang
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, New Cornerstone Science Laboratory, Beijing Normal University, Beijing, China
| | - Guoguang Zhao
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, China
- Clinical Research Center for Epilepsy Capital Medical University, Beijing, China
- Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Tao Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- The Chinese Glioma Genome Atlas, Beijing, China
- Beijing Neurosurgical Institute, Beijing, China
| | | | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| | - Weiqi Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| |
Collapse
|
8
|
Bachstetter AD, Lutshumba J, Winford E, Abner EL, Martin BJ, Harp JP, Van Eldik LJ, Schmitt FA, Wilcock DM, Stowe AM, Jicha GA, Nikolajczyk BS. A blunted T H17 cytokine signature in women with mild cognitive impairment: insights from inflammatory profiling of a community-based cohort of older adults. Brain Commun 2023; 5:fcad259. [PMID: 37901041 PMCID: PMC10612408 DOI: 10.1093/braincomms/fcad259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 06/23/2023] [Accepted: 10/06/2023] [Indexed: 10/31/2023] Open
Abstract
People with dementia have an increase in brain inflammation, caused in part by innate and adaptive immune cells. However, it remains unknown whether dementia-associated diseases alter neuro-immune reflex arcs to impact the systemic immune system. We examined peripheral immune cells from a community-based cohort of older adults to test if systemic inflammatory cytokine signatures associated with early stages of cognitive impairment. Human peripheral blood mononuclear cells were cultured with monocyte or T-cell-targeted stimuli, and multiplex assays quantitated cytokines in the conditioned media. Following T-cell-targeted stimulation, cells from women with cognitive impairment produced lower amounts of TH17 cytokines compared with cells from cognitively healthy women, while myeloid-targeted stimuli elicited similar amounts of cytokines from cells of both groups. This TH17 signature correlated with the proportion of circulating CD4+ and CD8+ T cells and plasma glial fibrillary acidic protein and neurofilament light concentrations. These results suggest that decreases in TH17 cytokines could be an early systemic change in women at risk for developing dementia. Amelioration of TH17s cytokines in early cognitive impairment could, in part, explain the compromised ability of older adults to respond to vaccines or defend against infection.
Collapse
Affiliation(s)
- Adam D Bachstetter
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Jenny Lutshumba
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Edric Winford
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA
| | - Erin L Abner
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
- Department of Epidemiology, University of Kentucky, Lexington, KY 40536, USA
| | - Barbra J Martin
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Jordan P Harp
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
- Department of Neurology, University of Kentucky, Lexington, KY 40536, USA
| | - Linda J Van Eldik
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Frederick A Schmitt
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
- Department of Neurology, University of Kentucky, Lexington, KY 40536, USA
- Department of Behavioral Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Donna M Wilcock
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Ann M Stowe
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA
- Department of Neurology, University of Kentucky, Lexington, KY 40536, USA
| | - Gregory A Jicha
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
- Department of Neurology, University of Kentucky, Lexington, KY 40536, USA
| | - Barbara S Nikolajczyk
- Department of Pharmacology and Nutritional Science, and Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
9
|
Moon HR, Yun JM. Neuroprotective effects of hesperetin on H 2O 2-induced damage in neuroblastoma SH-SY5Y cells. Nutr Res Pract 2023; 17:899-916. [PMID: 37780221 PMCID: PMC10522820 DOI: 10.4162/nrp.2023.17.5.899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/12/2023] [Accepted: 07/06/2023] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND/OBJECTIVES Oxidative stress is a fundamental neurodegenerative disease trigger that damages and decimates nerve cells. Neurodegenerative diseases are chronic central nervous system disorders that progress and result from neuronal degradation and loss. Recent studies have extensively focused on neurodegenerative disease treatment and prevention using dietary compounds. Heseperetin is an aglycone hesperidin form with various physiological activities, such as anti-inflammation, antioxidant, and antitumor. However, few studies have considered hesperetin's neuroprotective effects and mechanisms; thus, our study investigated this in hydrogen peroxide (H2O2)-treated SH-SY5Y cells. MATERIALS/METHODS SH-SY5Y cells were treated with H2O2 (400 µM) in hesperetin absence or presence (10-40 µM) for 24 h. Three-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide assays detected cell viability, and 4',6-diamidino-2-phenylindole staining allowed us to observe nuclear morphology changes such as chromatin condensation and apoptotic nuclei. Reactive oxygen species (ROS) detection assays measured intracellular ROS production; Griess reaction assays assessed nitric oxide (NO) production. Western blotting and quantitative polymerase chain reactions quantified corresponding mRNA and proteins. RESULTS Subsequent experiments utilized various non-toxic hesperetin concentrations, establishing that hesperetin notably decreased intracellular ROS and NO production in H2O2-treated SH-SY5Y cells (P < 0.05). Furthermore, hesperetin inhibited H2O2-induced inflammation-related gene expression, including interluekin-6, tumor necrosis factor-α, and nuclear factor kappa B (NF-κB) p65 activation. In addition, hesperetin inhibited NF-κB translocation into H2O2-treated SH-SY5Y cell nuclei and suppressed mitogen-activated protein kinase protein expression, an essential apoptotic cell death regulator. Various apoptosis hallmarks, including shrinkage and nuclear condensation in H2O2-treated cells, were suppressed dose-dependently. Additionally, hesperetin treatment down-regulated Bax/Bcl-2 expression ratios and activated AMP-activated protein kinase-mammalian target of rapamycin autophagy pathways. CONCLUSION These results substantiate that hesperetin activates autophagy and inhibits apoptosis and inflammation. Hesperetin is a potentially potent dietary agent that reduces neurodegenerative disease onset, progression, and prevention.
Collapse
Affiliation(s)
- Ha-Rin Moon
- Department of Food and Nutrition, Chonnam National University, Gwangju 61186, Korea
| | - Jung-Mi Yun
- Department of Food and Nutrition, Chonnam National University, Gwangju 61186, Korea
| |
Collapse
|
10
|
Shahidehpour RK, Nelson AS, Sanders LG, Embry CR, Nelson PT, Bachstetter AD. The localization of molecularly distinct microglia populations to Alzheimer's disease pathologies using QUIVER. Acta Neuropathol Commun 2023; 11:45. [PMID: 36934255 PMCID: PMC10024857 DOI: 10.1186/s40478-023-01541-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/03/2023] [Indexed: 03/20/2023] Open
Abstract
New histological techniques are needed to examine protein distribution in human tissues, which can reveal cell shape and disease pathology connections. Spatial proteomics has changed the study of tumor microenvironments by identifying spatial relationships of immunomodulatory cells and proteins and contributing to the discovery of new cancer immunotherapy biomarkers. However, the fast-expanding toolkit of spatial proteomic approaches has yet to be systematically applied to investigate pathological alterations in the aging human brain in health and disease states. Moreover, post-mortem human brain tissue presents distinct technical problems due to fixation procedures and autofluorescence, which limit fluorescence methodologies. This study sought to develop a multiplex immunohistochemistry approach (visualizing the immunostain with brightfield microscopy). Quantitative multiplex Immunohistochemistry with Visual colorimetric staining to Enhance Regional protein localization (QUIVER) was developed to address these technical challenges. Using QUIVER, a ten-channel pseudo-fluorescent image was generated using chromogen removal and digital microscopy to identify unique molecular microglia phenotypes. Next, the study asked if the tissue environment, specifically the amyloid plaques and neurofibrillary tangles characteristic of Alzheimer's disease, has any bearing on microglia's cellular and molecular phenotypes. QUIVER allowed the visualization of five molecular microglia/macrophage phenotypes using digital pathology tools. The recognizable reactive and homeostatic microglia/macrophage phenotypes demonstrated spatial polarization towards and away from amyloid plaques, respectively. Yet, microglia morphology appearance did not always correspond to molecular phenotype. This research not only sheds light on the biology of microglia but also offers QUIVER, a new tool for examining pathological alterations in the brains of the elderly.
Collapse
Affiliation(s)
- Ryan K Shahidehpour
- Spinal Cord and Brain Injury Research Center, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY, 40536, USA
| | - Abraham S Nelson
- Spinal Cord and Brain Injury Research Center, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA
| | - Lydia G Sanders
- Spinal Cord and Brain Injury Research Center, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA
| | - Chloe R Embry
- Spinal Cord and Brain Injury Research Center, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA
| | - Peter T Nelson
- Department of Neuroscience, University of Kentucky, Lexington, KY, 40536, USA
- Sanders-Brown Center On Aging, University of Kentucky, Lexington, KY, 40536, USA
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Adam D Bachstetter
- Spinal Cord and Brain Injury Research Center, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA.
- Department of Neuroscience, University of Kentucky, Lexington, KY, 40536, USA.
- Sanders-Brown Center On Aging, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
11
|
Penke B, Szűcs M, Bogár F. New Pathways Identify Novel Drug Targets for the Prevention and Treatment of Alzheimer's Disease. Int J Mol Sci 2023; 24:5383. [PMID: 36982456 PMCID: PMC10049476 DOI: 10.3390/ijms24065383] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Alzheimer's disease (AD) is an incurable, progressive neurodegenerative disorder. AD is a complex and multifactorial disease that is responsible for 60-80% of dementia cases. Aging, genetic factors, and epigenetic changes are the main risk factors for AD. Two aggregation-prone proteins play a decisive role in AD pathogenesis: β-amyloid (Aβ) and hyperphosphorylated tau (pTau). Both of them form deposits and diffusible toxic aggregates in the brain. These proteins are the biomarkers of AD. Different hypotheses have tried to explain AD pathogenesis and served as platforms for AD drug research. Experiments demonstrated that both Aβ and pTau might start neurodegenerative processes and are necessary for cognitive decline. The two pathologies act in synergy. Inhibition of the formation of toxic Aβ and pTau aggregates has been an old drug target. Recently, successful Aβ clearance by monoclonal antibodies has raised new hopes for AD treatments if the disease is detected at early stages. More recently, novel targets, e.g., improvements in amyloid clearance from the brain, application of small heat shock proteins (Hsps), modulation of chronic neuroinflammation by different receptor ligands, modulation of microglial phagocytosis, and increase in myelination have been revealed in AD research.
Collapse
Affiliation(s)
- Botond Penke
- Department of Medical Chemistry, University of Szeged, Dóm Square 8, H-6720 Szeged, Hungary
| | - Mária Szűcs
- Department of Medical Chemistry, University of Szeged, Dóm Square 8, H-6720 Szeged, Hungary
| | - Ferenc Bogár
- ELKH-SZTE Biomimetic Systems Research Group, Eötvös Loránd Research Network (ELKH), Dóm Square 8, H-6720 Szeged, Hungary
| |
Collapse
|
12
|
Chen X, Jiang S, Wang R, Bao X, Li Y. Neural Stem Cells in the Treatment of Alzheimer's Disease: Current Status, Challenges, and Future Prospects. J Alzheimers Dis 2023; 94:S173-S186. [PMID: 36336934 PMCID: PMC10473082 DOI: 10.3233/jad-220721] [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] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
Alzheimer's disease (AD), a progressive dementia, is one of the world's most dangerous and debilitating diseases. Clinical trial results of amyloid-β (Aβ) and tau regulators based on the pretext of straightforward amyloid and tau immunotherapy were disappointing. There are currently no effective strategies for slowing the progression of AD. Further understanding of the mechanisms underlying AD and the development of novel therapeutic options are critical. Neurogenesis is impaired in AD, which contributes to memory deficits. Transplanted neural stem cells (NSCs) can regenerate degraded cholinergic neurons, and new neurons derived from NSCs can form synaptic connections with neighboring neurons. In theory, employing NSCs to replace and restore damaged cholinergic neurons and brain connections may offer new treatment options for AD. However there remain barriers to surmount before NSC-based therapy can be used clinically. The objective of this article is to describe recent advances in the treatment of AD models and clinical trials involving NSCs. In addition, we discuss the challenges and prospects associated with cell transplant therapy for AD.
Collapse
Affiliation(s)
- Xiaokun Chen
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Shenzhong Jiang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xinjie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yongning Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
13
|
Bi W, Lei T, Cai S, Zhang X, Yang Y, Xiao Z, Wang L, Du H. Potential of astrocytes in targeting therapy for Alzheimer’s disease. Int Immunopharmacol 2022; 113:109368. [DOI: 10.1016/j.intimp.2022.109368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/06/2022] [Accepted: 10/15/2022] [Indexed: 11/05/2022]
|
14
|
Zhou C, Li T, Dong Q, Liang H, Xu L. SARM suppresses glioma progression in GL261 glioma cells and regulates microglial polarization. Cell Biol Int 2022; 46:1927-1936. [PMID: 35971755 DOI: 10.1002/cbin.11881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 07/14/2022] [Indexed: 11/09/2022]
Abstract
Microglia is the major cellular component of glioma mass that promotes glioma growth, invasion, and chemoresistance by releasing inflammatory factors. Sterile alpha and HEAT/Armadillo motif (SARM), a member of the Toll-interleukin-1 receptor (TIR) domain-containing adaptor family, is primarily expressed in the central nervous system. However, the role of SARM in glioma is still undefined. In the present work, we examined the function of SARM in microglial polarization and glioma progression. Our results showed that forced the expression of SARM in GL261 glioma cells inhibited tumor growth, and reduced interleukin (IL)-6 secretion in conditioned media. Silencing of SARM in microglia cells inhibited IL-4-induced M2 polarization, enhanced lipopolysaccharide -induced M1 microglial polarization. Furthermore, overexpression of SARM increased the migration of microglia cells upon TGFβ stimulation. These data suggested that SARM is involved in neuro-inflammation and microglia activation. In summary, this study provides novel insight into the mechanisms of microglial polarization.
Collapse
Affiliation(s)
- Chun Zhou
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Tianzun Li
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Qian Dong
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Hong Liang
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Lunshan Xu
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
15
|
Peng LS, Xu Y, Wang QS. YY1 PROMOTES MICROGLIA M2 POLARIZATION THROUGH THE MIR-130A-3P/TREM-2 AXIS TO ALLEVIATE SEPSIS-ASSOCIATED ENCEPHALOPATHY. Shock 2022; 58:128-136. [PMID: 35234205 DOI: 10.1097/shk.0000000000001914] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Purpose: Sepsis-associated encephalopathy (SAE) induces cognitive dysfunction via mechanisms that commonly involve neuroinflammation. Yin Yang 1 (YY1) is an important transcription factor that acts as a key role in sepsis and neuroepithelium development. However, the function of YY1 in SAE remains unclear. Our study aimed to probe the intrinsic and concrete molecular mechanism of YY1 in SAE. Methods: SAE cell model and SAE animal model were constructed by lipopolysaccharide (LPS) treatment and cecal ligation and puncture surgery, respectively. Behavioral tests were performed to analyze the cognitive function. The polarization state of mouse microglia (BV-2 cells) was assessed by flow cytometry assay. The mRNA and protein expressions were assessed by qRT-PCR and western blot. Finally, the binding relationships between YY1, miR-130a-3p, andTREM-2were verified by dual luciferase reporter gene assay and/or ChIP assay. Results: Here our results described that YY1 and TREM-2 were downregulated and miR-130a-3p was upregulated in SAE. YY1 overexpression could promote M2 polarization of microglia, and alleviate neuroinflammation and behavioral deficits in vitro and in vivo. YY1 could inhibit miR-130a-3p promoter activity. As expected, miR-130a-3p overexpression abolished the effects of YY1 overexpression on LPS-treated BV-2 cells. Besides, TREM-2 was identified as the target of miR-130a-3p. TREM-2 silencing could reverse the effects of miR-130a-3p inhibition on LPS-treated BV-2 cells. Conclusion: Taken together, YY1 promoted microglia M2 polarization via upregulating TREM-2 by interacting with miR-130a-3p promoter, suggesting YY1 overexpression might be a novel therapeutic strategy of SAE.
Collapse
Affiliation(s)
- Liang-Shan Peng
- The First Affiliated Hospital, Department of Critical Care Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | | | | |
Collapse
|
16
|
Reis PA, Castro-Faria-Neto HC. Systemic Response to Infection Induces Long-Term Cognitive Decline: Neuroinflammation and Oxidative Stress as Therapeutical Targets. Front Neurosci 2022; 15:742158. [PMID: 35250433 PMCID: PMC8895724 DOI: 10.3389/fnins.2021.742158] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 12/31/2021] [Indexed: 12/29/2022] Open
Abstract
In response to pathogens or damage signs, the immune system is activated in order to eliminate the noxious stimuli. The inflammatory response to infectious diseases induces systemic events, including cytokine storm phenomenon, vascular dysfunction, and coagulopathy, that can lead to multiple-organ dysfunction. The central nervous system (CNS) is one of the major organs affected, and symptoms such as sickness behavior (depression and fever, among others), or even delirium, can be observed due to activation of endothelial and glial cells, leading to neuroinflammation. Several reports have been shown that, due to CNS alterations caused by neuroinflammation, some sequels can be developed in special cognitive decline. There is still no any treatment to avoid cognitive impairment, especially those developed due to systemic infectious diseases, but preclinical and clinical trials have pointed out controlling neuroinflammatory events to avoid the development of this sequel. In this minireview, we point to the possible mechanisms that triggers long-term cognitive decline, proposing the acute neuroinflammatory events as a potential therapeutical target to treat this sequel that has been associated to several infectious diseases, such as malaria, sepsis, and, more recently, the new SARS-Cov2 infection.
Collapse
Affiliation(s)
- Patricia Alves Reis
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
- Biochemistry Department, Roberto Alcântara Gomes Biology Institute, Rio de Janeiro State University, Rio de Janeiro, Brazil
- *Correspondence: Patricia Alves Reis,
| | | |
Collapse
|
17
|
Cyr B, Hadad R, Keane RW, de Rivero Vaccari JP. The Role of Non-canonical and Canonical Inflammasomes in Inflammaging. Front Mol Neurosci 2022; 15:774014. [PMID: 35221912 PMCID: PMC8864077 DOI: 10.3389/fnmol.2022.774014] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/17/2022] [Indexed: 01/11/2023] Open
Abstract
Neurodegenerative diseases currently affect millions of people worldwide and continues to increase in the expanding elderly population. Neurodegenerative diseases usually involve cognitive decline and are among the top causes of death. Thus, there is a critical need for the development of treatments and preventive strategies for neurodegenerative diseases. One of the risk factors of neurodegeneration is inflammaging, a low level of chronic inflammation due to old age. We have previously shown that the inflammasome contributes to inflammaging in the central nervous system (CNS). The inflammasome is a multiprotein complex of the innate immune response consisting of a sensor protein, apoptosis speck-like protein containing a CARD (ASC), and caspase-1. Our lab has developed a humanized monoclonal antibody against ASC (anti-ASC). Here, we analyzed cortical lysates from young (3 months old), aged (18 months old), and aged anti-ASC treated mice for the expression of canonical and non-canonical inflammasome proteins. We show that the protein levels of NLRP1, ASC, caspase-1, and caspase-8 were elevated in the cortex of aged mice, and that anti-ASC decreased the expression of these proteins, consistent with lower levels of the pro-inflammatory cytokine interleukin (IL)-1β. Additionally, we show that these proteins form a novel NLRP1-caspase-8 non-canonical inflammasome comprised of NLRP1, caspase-8 and ASC. Moreover, these inflammasome proteins were present in neurons in young and aged mice. Together, these results indicate that a novel NLRP1-caspase-8 non-canonical inflammasome is present in the cortex of mice and that anti-ASC is a potential therapeutic to decrease inflammasome-mediated inflammaging in the CNS.
Collapse
Affiliation(s)
- Brianna Cyr
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Roey Hadad
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Robert W. Keane
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
- Center for Cognitive Neuroscience and Aging University of Miami Miller School of Medicine, Miami, FL, United States
- *Correspondence: Juan Pablo de Rivero Vaccari,
| |
Collapse
|
18
|
In Vitro Methodologies to Study the Role of Advanced Glycation End Products (AGEs) in Neurodegeneration. Nutrients 2022; 14:nu14020363. [PMID: 35057544 PMCID: PMC8777776 DOI: 10.3390/nu14020363] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 02/07/2023] Open
Abstract
Advanced glycation end products (AGEs) can be present in food or be endogenously produced in biological systems. Their formation has been associated with chronic neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and amyotrophic lateral sclerosis. The implication of AGEs in neurodegeneration is related to their ability to bind to AGE-specific receptors and the ability of their precursors to induce the so-called “dicarbonyl stress”, resulting in cross-linking and protein damage. However, the mode of action underlying their role in neurodegeneration remains unclear. While some research has been carried out in observational clinical studies, further in vitro studies may help elucidate these underlying modes of action. This review presents and discusses in vitro methodologies used in research on the potential role of AGEs in neuroinflammation and neurodegeneration. The overview reveals the main concepts linking AGEs to neurodegeneration, the current findings, and the available and advisable in vitro models to study their role. Moreover, the major questions regarding the role of AGEs in neurodegenerative diseases and the challenges and discrepancies in the research field are discussed.
Collapse
|
19
|
Abd Al Haleem EN, Ahmed HI, El-Naga RN. Lycopene and Chrysin through Mitigation of Neuroinflammation and Oxidative Stress Exerted Antidepressant Effects in Clonidine-Induced Depression-like Behavior in Rats. J Diet Suppl 2021; 20:391-410. [PMID: 34633271 DOI: 10.1080/19390211.2021.1988797] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Depression is a severely debilitating psychiatric disorder that influences more than 15% of the population worldwide. It has been demonstrated that it is associated with a high risk of developing other diseases such as cardiovascular diseases, diabetes, stroke, epilepsy, and cancer. The current study examines the possibility of chrysin and lycopene having an antidepressant effect in a rat model of depression induced by clonidine, as well as the mechanisms underlying this effect, including the role of neuroinflammation and oxidative stress. Rats were allotted into seven groups. The rats in group 1 served as a control. Group 2 received lycopene only. Group 3 was provided chrysin only. Group 4 was administered clonidine and served as the model. Group 5 was offered lycopene and clonidine. Group 6 was administered chrysin and clonidine. Group 7 was given FLX and clonidine and represented the standard. The experiment lasted two weeks, during which behavioral, biochemical, histopathological, and immunohistochemical measurements were performed. Lycopene and chrysin were used to correct the concentrations of noradrenaline and serotonin hippocampal tissue concentrations. These findings were also improved by immunohistochemical analysis of GFAP, VEGF, caspase3, and histopathological examinations, in which pretreatment of rats with lycopene and chrysin reversed all clonidine-induced alterations. The current research demonstrates that lycopene and chrysin have an auspicious antidepressant effect against clonidine that provoked behavioral hopelessness in rats. Manipulating oxidative stress, inflammation, and apoptosis may partially represent the corrective mechanism for the neuroprotective actions against the depressive effect of clonidine.
Collapse
Affiliation(s)
- Ekram Nemr Abd Al Haleem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Hebatalla I Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Reem N El-Naga
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
20
|
Chidambaram SB, Essa MM, Rathipriya AG, Bishir M, Ray B, Mahalakshmi AM, Tousif AH, Sakharkar MK, Kashyap RS, Friedland RP, Monaghan TM. Gut dysbiosis, defective autophagy and altered immune responses in neurodegenerative diseases: Tales of a vicious cycle. Pharmacol Ther 2021; 231:107988. [PMID: 34536490 DOI: 10.1016/j.pharmthera.2021.107988] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/16/2021] [Accepted: 08/25/2021] [Indexed: 02/08/2023]
Abstract
The human microbiota comprises trillions of symbiotic microorganisms and is involved in regulating gastrointestinal (GI), immune, nervous system and metabolic homeostasis. Recent observations suggest a bidirectional communication between the gut microbiota and the brain via immune, circulatory and neural pathways, termed the Gut-Brain Axis (GBA). Alterations in gut microbiota composition, such as seen with an increased number of pathobionts and a decreased number of symbionts, termed gut dysbiosis or microbial intestinal dysbiosis, plays a prominent role in the pathogenesis of central nervous system (CNS)-related disorders. Clinical reports confirm that GI symptoms often precede neurological symptoms several years before the development of neurodegenerative diseases (NDDs). Pathologically, gut dysbiosis disrupts the integrity of the intestinal barrier leading to ingress of pathobionts and toxic metabolites into the systemic circulation causing GBA dysregulation. Subsequently, chronic neuroinflammation via dysregulated immune activation triggers the accumulation of neurotoxic misfolded proteins in and around CNS cells resulting in neuronal death. Emerging evidence links gut dysbiosis to the aggravation and/or spread of proteinopathies from the peripheral nervous system to the CNS and defective autophagy-mediated proteinopathies. This review summarizes the current understanding of the role of gut microbiota in NDDs, and highlights a vicious cycle of gut dysbiosis, immune-mediated chronic neuroinflammation, impaired autophagy and proteinopathies, which contributes to the development of neurodegeneration in Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis, amyotrophic lateral sclerosis and frontotemporal lobar degeneration. We also discuss novel therapeutic strategies targeting the modulation of gut dysbiosis through prebiotics, probiotics, synbiotics or dietary interventions, and faecal microbial transplantation (FMT) in the management of NDDs.
Collapse
Affiliation(s)
- Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India; Centre for Experimental Pharmacology and Toxicology (CPT), JSS Academy of Higher Education & Research, Mysuru 570015, KA, India.
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat 123, Oman; Ageing and Dementia Research Group, Sultan Qaboos University, Muscat 123, Oman; Biomedical Sciences Department, University of Pacific, Sacramento, CA, USA.
| | - A G Rathipriya
- Food and Brain Research Foundation, Chennai 600 094, Tamil Nadu, India
| | - Muhammed Bishir
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India
| | - Bipul Ray
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India; Centre for Experimental Pharmacology and Toxicology (CPT), JSS Academy of Higher Education & Research, Mysuru 570015, KA, India
| | - Arehally M Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India
| | - A H Tousif
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India; Centre for Experimental Pharmacology and Toxicology (CPT), JSS Academy of Higher Education & Research, Mysuru 570015, KA, India
| | - Meena K Sakharkar
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
| | - Rajpal Singh Kashyap
- Research Centre, Dr G. M. Taori Central India Institute of Medical Sciences (CIIMS), Nagpur, Maharashtra, India
| | - Robert P Friedland
- Department of Neurology, University of Louisville, Louisville, KY 40292, USA
| | - Tanya M Monaghan
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham NG7 2UH, UK; Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK.
| |
Collapse
|
21
|
Patel AG, Nehete PN, Krivoshik SR, Pei X, Cho EL, Nehete BP, Ramani MD, Shao Y, Williams LE, Wisniewski T, Scholtzova H. Innate immunity stimulation via CpG oligodeoxynucleotides ameliorates Alzheimer's disease pathology in aged squirrel monkeys. Brain 2021; 144:2146-2165. [PMID: 34128045 PMCID: PMC8502485 DOI: 10.1093/brain/awab129] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/09/2020] [Accepted: 12/17/2020] [Indexed: 11/15/2022] Open
Abstract
Alzheimer's disease is the most common cause of dementia and the only illness among the top 10 causes of death for which there is no disease-modifying therapy. The failure rate of clinical trials is very high, in part due to the premature translation of successful results in transgenic mouse models to patients. Extensive evidence suggests that dysregulation of innate immunity and microglia/macrophages plays a key role in Alzheimer's disease pathogenesis. Activated resident microglia and peripheral macrophages can display protective or detrimental phenotypes depending on the stimulus and environment. Toll-like receptors (TLRs) are a family of innate immune regulators known to play an important role in governing the phenotypic status of microglia. We have shown in multiple transgenic Alzheimer's disease mouse models that harnessing innate immunity via TLR9 agonist CpG oligodeoxynucleotides (ODNs) modulates age-related defects associated with immune cells and safely reduces amyloid plaques, oligomeric amyloid-β, tau pathology, and cerebral amyloid angiopathy (CAA) while promoting cognitive benefits. In the current study we have used a non-human primate model of sporadic Alzheimer's disease pathology that develops extensive CAA-elderly squirrel monkeys. The major complications in current immunotherapeutic trials for Alzheimer's disease are amyloid-related imaging abnormalities, which are linked to the presence and extent of CAA; hence, the prominence of CAA in elderly squirrel monkeys makes them a valuable model for studying the safety of the CpG ODN-based concept of immunomodulation. We demonstrate that long-term use of Class B CpG ODN 2006 induces a favourable degree of innate immunity stimulation without producing excessive or sustained inflammation, resulting in efficient amelioration of both CAA and tau Alzheimer's disease-related pathologies in association with behavioural improvements and in the absence of microhaemorrhages in aged elderly squirrel monkeys. CpG ODN 2006 has been well established in numerous human trials for a variety of diseases. The present evidence together with our earlier, extensive preclinical research, validates the beneficial therapeutic outcomes and safety of this innovative immunomodulatory approach, increasing the likelihood of CpG ODN therapeutic efficacy in future clinical trials.
Collapse
Affiliation(s)
- Akash G Patel
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Pramod N Nehete
- Department of Comparative Medicine, the University of Texas MD Anderson Cancer Center, Bastrop, TX 78602, USA
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Sara R Krivoshik
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Xuewei Pei
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Elizabeth L Cho
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Bharti P Nehete
- Department of Comparative Medicine, the University of Texas MD Anderson Cancer Center, Bastrop, TX 78602, USA
| | - Margish D Ramani
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Yongzhao Shao
- Division of Biostatistics, Departments of Population Health and Environmental Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Lawrence E Williams
- Department of Comparative Medicine, the University of Texas MD Anderson Cancer Center, Bastrop, TX 78602, USA
| | - Thomas Wisniewski
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
- Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA
| | - Henrieta Scholtzova
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
- Department of Comparative Medicine, the University of Texas MD Anderson Cancer Center, Bastrop, TX 78602, USA
| |
Collapse
|
22
|
Oberman K, Hovens I, de Haan J, Falcao-Salles J, van Leeuwen B, Schoemaker R. Acute pre-operative ibuprofen improves cognition in a rat model for postoperative cognitive dysfunction. J Neuroinflammation 2021; 18:156. [PMID: 34238316 PMCID: PMC8265047 DOI: 10.1186/s12974-021-02206-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/23/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Inflammation is considered a key factor in the development of postoperative cognitive dysfunction (POCD). Therefore, we hypothesized that pre-operative anti-inflammatory treatment with ibuprofen would inhibit POCD in our rat-model. METHODS Male Wistar rats of 3 or 23 months old received a single injection of ibuprofen (15 mg/kg i.p.) or were control handled before abdominal surgery. Timed blood and fecal samples were collected for analyses of inflammation markers and gut microbiome changes. Behavioral testing was performed from 9 to 14 days after surgery, in the open field, novel object- and novel location-recognition tests and Morris water maze. Neuroinflammation and neurogenesis were assessed by immune histochemistry after sacrifice on postoperative day 14. RESULTS Ibuprofen improved short-term spatial memory in the novel location recognition test, and increased hippocampal neurogenesis. However, these effects were associated with increased hippocampal microglia activity. Whereas plasma cytokine levels (IL1-β, IL6, IL10, and TNFα) were not significantly affected, VEGF levels increased and IFABP levels decreased after ibuprofen. Long-term memory in the Morris water maze was not significantly improved by ibuprofen. The gut microbiome was neither significantly affected by surgery nor by ibuprofen treatment. In general, effects in aged rats appeared similar to those in young rats, though less pronounced. CONCLUSION A single injection of ibuprofen before surgery improved hippocampus-associated short-term memory after surgery and increased neurogenesis. However, this favorable outcome seemed not attributable to inhibition of (neuro)inflammation. Potential contributions of intestinal and blood-brain barrier integrity need further investigation. Although less pronounced compared to young rats, effects in aged rats indicate that even elderly individuals could benefit from ibuprofen treatment.
Collapse
Affiliation(s)
- Klaske Oberman
- Department of Neurobiology, GELIFES, University of Groningen, Nijenborgh 7, 9747, AG, Groningen, The Netherlands
| | - Iris Hovens
- Department of Neurobiology, GELIFES, University of Groningen, Nijenborgh 7, 9747, AG, Groningen, The Netherlands
| | - Jacco de Haan
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - Joana Falcao-Salles
- Department of Microbial Ecology, GELIFES, University of Groningen, Groningen, The Netherlands
| | - Barbara van Leeuwen
- Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Regien Schoemaker
- Department of Neurobiology, GELIFES, University of Groningen, Nijenborgh 7, 9747, AG, Groningen, The Netherlands.
| |
Collapse
|
23
|
Beneficial Effects of Metformin on the Central Nervous System, with a Focus on Epilepsy and Lafora Disease. Int J Mol Sci 2021; 22:ijms22105351. [PMID: 34069559 PMCID: PMC8160983 DOI: 10.3390/ijms22105351] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022] Open
Abstract
Metformin is a drug in the family of biguanide compounds that is widely used in the treatment of type 2 diabetes (T2D). Interestingly, the therapeutic potential of metformin expands its prescribed use as an anti-diabetic drug. In this sense, it has been described that metformin administration has beneficial effects on different neurological conditions. In this work, we review the beneficial effects of this drug as a neuroprotective agent in different neurological diseases, with a special focus on epileptic disorders and Lafora disease, a particular type of progressive myoclonus epilepsy. In addition, we review the different proposed mechanisms of action of metformin to understand its function at the neurological level.
Collapse
|
24
|
Duggan MR, Parikh V. Microglia and modifiable life factors: Potential contributions to cognitive resilience in aging. Behav Brain Res 2021; 405:113207. [PMID: 33640394 PMCID: PMC8005490 DOI: 10.1016/j.bbr.2021.113207] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/27/2021] [Accepted: 02/20/2021] [Indexed: 02/08/2023]
Abstract
Given the increasing prevalence of age-related cognitive decline, it is relevant to consider the factors and mechanisms that might facilitate an individual's resiliency to such deficits. Growing evidence suggests a preeminent role of microglia, the prime mediator of innate immunity within the central nervous system. Human and animal investigations suggest aberrant microglial functioning and neuroinflammation are not only characteristic of the aged brain, but also might contribute to age-related dementia and Alzheimer's Disease. Conversely, accumulating data suggest that modifiable lifestyle factors (MLFs), such as healthy diet, exercise and cognitive engagement, can reliably afford cognitive benefits by potentially suppressing inflammation in the aging brain. The present review highlights recent advances in our understanding of the role for microglia in maintaining brain homeostasis and cognitive functioning in aging. Moreover, we propose an integrated, mechanistic model that postulates an individual's resiliency to cognitive decline afforded by MLFs might be mediated by the mitigation of aberrant microglia activation in aging, and subsequent suppression of neuroinflammation.
Collapse
Affiliation(s)
- Michael R Duggan
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, United States
| | - Vinay Parikh
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, United States.
| |
Collapse
|
25
|
Maurya SK, Bhattacharya N, Mishra S, Bhattacharya A, Banerjee P, Senapati S, Mishra R. Microglia Specific Drug Targeting Using Natural Products for the Regulation of Redox Imbalance in Neurodegeneration. Front Pharmacol 2021; 12:654489. [PMID: 33927630 PMCID: PMC8076853 DOI: 10.3389/fphar.2021.654489] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/08/2021] [Indexed: 12/14/2022] Open
Abstract
Microglia, a type of innate immune cell of the brain, regulates neurogenesis, immunological surveillance, redox imbalance, cognitive and behavioral changes under normal and pathological conditions like Alzheimer's, Parkinson's, Multiple sclerosis and traumatic brain injury. Microglia produces a wide variety of cytokines to maintain homeostasis. It also participates in synaptic pruning and regulation of neurons overproduction by phagocytosis of neural precursor cells. The phenotypes of microglia are regulated by the local microenvironment of neurons and astrocytes via interaction with both soluble and membrane-bound mediators. In case of neuron degeneration as observed in acute or chronic neurodegenerative diseases, microglia gets released from the inhibitory effect of neurons and astrocytes, showing activated phenotype either of its dual function. Microglia shows neuroprotective effect by secreting growths factors to heal neurons and clears cell debris through phagocytosis in case of a moderate stimulus. But the same microglia starts releasing pro-inflammatory cytokines like TNF-α, IFN-γ, reactive oxygen species (ROS), and nitric oxide (NO), increasing neuroinflammation and redox imbalance in the brain under chronic signals. Therefore, pharmacological targeting of microglia would be a promising strategy in the regulation of neuroinflammation, redox imbalance and oxidative stress in neurodegenerative diseases. Some studies present potentials of natural products like curcumin, resveratrol, cannabidiol, ginsenosides, flavonoids and sulforaphane to suppress activation of microglia. These natural products have also been proposed as effective therapeutics to regulate the progression of neurodegenerative diseases. The present review article intends to explain the molecular mechanisms and functions of microglia and molecular dynamics of microglia specific genes and proteins like Iba1 and Tmem119 in neurodegeneration. The possible interventions by curcumin, resveratrol, cannabidiol, ginsenosides, flavonoids and sulforaphane on microglia specific protein Iba1 suggest possibility of natural products mediated regulation of microglia phenotypes and its functions to control redox imbalance and neuroinflammation in management of Alzheimer's, Parkinson's and Multiple Sclerosis for microglia-mediated therapeutics.
Collapse
Affiliation(s)
| | - Neetu Bhattacharya
- Department of Zoology, Dyal Singh College, University of Delhi, Delhi, India
| | - Suman Mishra
- Department of Molecular Medicine and Biotechnology, SGPGI, Lucknow, India
| | - Amit Bhattacharya
- Department of Zoology, Ramjas College, University of Delhi, Delhi, India
| | - Pratibha Banerjee
- Immunogenomics Laboratory, Department of Human Genetics & Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Sabyasachi Senapati
- Immunogenomics Laboratory, Department of Human Genetics & Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Rajnikant Mishra
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, India
| |
Collapse
|
26
|
Hsueh SC, Luo W, Tweedie D, Kim DS, Kim YK, Hwang I, Gil JE, Han BS, Chiang YH, Selman W, Hoffer BJ, Greig NH. N-Adamantyl Phthalimidine: A New Thalidomide-like Drug That Lacks Cereblon Binding and Mitigates Neuronal and Synaptic Loss, Neuroinflammation, and Behavioral Deficits in Traumatic Brain Injury and LPS Challenge. ACS Pharmacol Transl Sci 2021; 4:980-1000. [PMID: 33860215 DOI: 10.1021/acsptsci.1c00042] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Indexed: 02/07/2023]
Abstract
Neuroinflammation contributes to delayed secondary cell death following traumatic brain injury (TBI), has the potential to chronically exacerbate the initial insult, and represents a therapeutic target that has largely failed to translate into human efficacy. Thalidomide-like drugs have effectively mitigated neuroinflammation across cellular and animal models of TBI and neurodegeneration but are complicated by adverse actions in humans. We hence developed N-adamantyl phthalimidine (NAP) as a new thalidomide-like drug to mitigate inflammation without binding to cereblon, a key target associated with the antiproliferative, antiangiogenic, and teratogenic actions seen in this drug class. We utilized a phenotypic drug discovery approach that employed multiple cellular and animal models and ultimately examined immunohistochemical, biochemical, and behavioral measures following controlled cortical impact (CCI) TBI in mice. NAP mitigated LPS-induced inflammation across cellular and rodent models and reduced oligomeric α-synuclein and amyloid-β mediated inflammation. Following CCI TBI, NAP mitigated neuronal and synaptic loss, neuroinflammation, and behavioral deficits, and is unencumbered by cereblon binding, a key protein underpinning the teratogenic and adverse actions of thalidomide-like drugs in humans. In summary, NAP represents a new class of thalidomide-like drugs with anti-inflammatory actions for promising efficacy in the treatment of TBI and potentially longer-term neurodegenerative disorders.
Collapse
Affiliation(s)
- Shih Chang Hsueh
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, Maryland 21224, United States
| | - Weiming Luo
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, Maryland 21224, United States
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, Maryland 21224, United States
| | - Dong Seok Kim
- AevisBio, Inc., Gaithersburg Maryland 20878, United States.,Aevis Bio, Inc., Daejeon 34141, Republic of Korea
| | - Yu Kyung Kim
- Aevis Bio, Inc., Daejeon 34141, Republic of Korea
| | - Inho Hwang
- Aevis Bio, Inc., Daejeon 34141, Republic of Korea
| | - Jung-Eun Gil
- Aevis Bio, Inc., Daejeon 34141, Republic of Korea
| | - Baek-Soo Han
- Research Center for Biodefence, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - Yung-Hsiao Chiang
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 110, Taiwan.,Neuroscience Research Center, Taipei Medical University, Taipei 110, Taiwan.,Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 110, Taiwan
| | - Warren Selman
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Barry J Hoffer
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, Maryland 21224, United States
| |
Collapse
|
27
|
Potter H, Woodcock JH, Boyd TD, Coughlan CM, O'Shaughnessy JR, Borges MT, Thaker AA, Raj BA, Adamszuk K, Scott D, Adame V, Anton P, Chial HJ, Gray H, Daniels J, Stocker ME, Sillau SH. Safety and efficacy of sargramostim (GM-CSF) in the treatment of Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12158. [PMID: 33778150 PMCID: PMC7988877 DOI: 10.1002/trc2.12158] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 02/05/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Inflammatory markers have long been observed in the brain, cerebrospinal fluid (CSF), and plasma of Alzheimer's disease (AD) patients, suggesting that inflammation contributes to AD and might be a therapeutic target. However, non-steroidal anti-inflammatory drug trials in AD and mild cognitive impairment (MCI) failed to show benefit. Our previous work seeking to understand why people with the inflammatory disease rheumatoid arthritis are protected from AD found that short-term treatment of transgenic AD mice with the pro-inflammatory cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) led to an increase in activated microglia, a 50% reduction in amyloid load, an increase in synaptic area, and improvement in spatial memory to normal. These results called into question the consensus view that inflammation is solely detrimental in AD. Here, we tested our hypothesis that modulation of the innate immune system might similarly be used to treat AD in humans by investigating the ability of GM-CSF/sargramostim to safely ameliorate AD symptoms/pathology. METHODS A randomized, double-blind, placebo-controlled trial was conducted in mild-to-moderate AD participants (NCT01409915). Treatments (20 participants/group) occurred 5 days/week for 3 weeks plus two follow-up (FU) visits (FU1 at 45 days and FU2 at 90 days) with neurological, neuropsychological, blood biomarker, and imaging assessments. RESULTS Sargramostim treatment expectedly changed innate immune system markers, with no drug-related serious adverse events or amyloid-related imaging abnormalities. At end of treatment (EOT), the Mini-Mental State Examination score of the sargramostim group increased compared to baseline (P = .0074) and compared to placebo (P = .0370); the treatment effect persisted at FU1 (P = .0272). Plasma markers of amyloid beta (Aβ40 [decreased in AD]) increased 10% (P = .0105); plasma markers of neurodegeneration (total tau and UCH-L1) decreased 24% (P = .0174) and 42% (P = .0019), respectively, after sargramostim treatment compared to placebo. DISCUSSION The innate immune system is a viable target for therapeutic intervention in AD. An extended treatment trial testing the long-term safety and efficacy of GM-CSF/sargramostim in AD is warranted.
Collapse
Affiliation(s)
- Huntington Potter
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Jonathan H. Woodcock
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
| | - Timothy D. Boyd
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Christina M. Coughlan
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - John R. O'Shaughnessy
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
| | - Manuel T. Borges
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- Department of RadiologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Ashesh A. Thaker
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- Department of RadiologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | | | | | | | - Vanesa Adame
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Paige Anton
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Heidi J. Chial
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Helen Gray
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
| | - Joseph Daniels
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
| | - Michelle E. Stocker
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
| | - Stefan H. Sillau
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
| |
Collapse
|
28
|
Zheng Y, Zhang J, Zhao Y, Zhang Y, Zhang X, Guan J, Liu Y, Fu J. Curcumin protects against cognitive impairments in a rat model of chronic cerebral hypoperfusion combined with diabetes mellitus by suppressing neuroinflammation, apoptosis, and pyroptosis. Int Immunopharmacol 2021; 93:107422. [PMID: 33548579 DOI: 10.1016/j.intimp.2021.107422] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/12/2021] [Accepted: 01/19/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) is regarded as a high-risk factor for cognitive decline in vascular dementia (VaD). We have previously shown that diabetes mellitus (DM) synergistically promotes CCH-induced cognitive dysfunction via exacerbating neuroinflammation. Furthermore, curcumin has been shown to exhibit anti-inflammatory and neuroprotective activities. However, the effects of curcumin on CCH-induced cognitive impairments in DM have remained unknown. METHODS Rats were fed with a high-fat diet (HFD) and injected with low-dose streptozotocin (STZ), followed by bilateral common carotid artery occlusion (BCCAO), to model DM and CCH in vivo. After BCCAO, curcumin (50 mg/kg) was administered intraperitoneally every two days for eight weeks to evaluate its therapeutic effects. Additionally, mouse BV2 microglial cells were exposed to hypoxia and high glucose to model CCH and DM pathologies in vitro. RESULTS Curcumin treatment significantly improved DM/CCH-induced cognitive deficits and attenuated neuronal cell death. Molecular analysis revealed that curcumin exerted protective effects via suppressing neuroinflammation induced by microglial activation, regulating the triggering receptor expressed on myeloid cells 2 (TREM2)/toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) pathway, alleviating apoptosis, and reducing nod-like receptor protein 3 (NLRP3)-dependent pyroptosis. CONCLUSIONS Taken together, our findings suggest that curcumin represents a promising therapy for DM/CCH-induced cognitive impairments.
Collapse
Affiliation(s)
- Yaling Zheng
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Jiawei Zhang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Yao Zhao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Yaxuan Zhang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Xiaojie Zhang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Jian Guan
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yu Liu
- Department of Medicine, Shanghai Eighth People's Hospital, Shanghai 200235, China
| | - Jianliang Fu
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China; Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
29
|
Li Y, Shen PP, Wang B. Induced pluripotent stem cell technology for spinal cord injury: a promising alternative therapy. Neural Regen Res 2021; 16:1500-1509. [PMID: 33433463 PMCID: PMC8323703 DOI: 10.4103/1673-5374.303013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury has long been a prominent challenge in the trauma repair process. Spinal cord injury is a research hotspot by virtue of its difficulty to treat and its escalating morbidity. Furthermore, spinal cord injury has a long period of disease progression and leads to complications that exert a lot of mental and economic pressure on patients. There are currently a large number of therapeutic strategies for treating spinal cord injury, which range from pharmacological and surgical methods to cell therapy and rehabilitation training. All of these strategies have positive effects in the course of spinal cord injury treatment. This review mainly discusses the problems regarding stem cell therapy for spinal cord injury, including the characteristics and action modes of all relevant cell types. Induced pluripotent stem cells, which represent a special kind of stem cell population, have gained impetus in cell therapy development because of a range of advantages. Induced pluripotent stem cells can be developed into the precursor cells of each neural cell type at the site of spinal cord injury, and have great potential for application in spinal cord injury therapy.
Collapse
Affiliation(s)
- Yu Li
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, School of Life Science, Nanjing University, Nanjing, Jiangsu Province, China
| | - Ping-Ping Shen
- State Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive Cancer Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, School of Life Science, Nanjing University, Nanjing, Jiangsu Province, China
| | - Bin Wang
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| |
Collapse
|
30
|
Inestrosa NC, Tapia-Rojas C, Cerpa W, Cisternas P, Zolezzi JM. WNT Signaling Is a Key Player in Alzheimer's Disease. Handb Exp Pharmacol 2021; 269:357-382. [PMID: 34486097 DOI: 10.1007/164_2021_532] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The cellular processes regulated by WNT signaling have been mainly studied during embryonic development and cancer. In the last two decades, the role of WNT in the adult central nervous system has been the focus of interest in our laboratory. In this chapter, we will be summarized β-catenin-dependent and -independent WNT pathways, then we will be revised WNT signaling function at the pre- and post-synaptic level. Concerning Alzheimer's disease (AD) initially, we found that WNT/β-catenin signaling activation exerts a neuroprotective mechanism against the amyloid β (Αβ) peptide toxicity. Later, we found that WNT/β-catenin participates in Tau phosphorylation and in learning and memory. In the last years, we demonstrated that WNT/β-catenin signaling is instrumental in the amyloid precursor protein (APP) processing and that WNT/β-catenin dysfunction results in Aβ production and aggregation. We highlight the importance of WNT/β-catenin signaling dysfunction in the onset of AD and propose that the loss of WNT/β-catenin signaling is a triggering factor of AD. The WNT pathway is therefore positioned as a therapeutic target for AD and could be a valid concept for improving AD therapy. We think that metabolism and inflammation will be relevant when defining future research in the context of WNT signaling and the neurodegeneration associated with AD.
Collapse
Affiliation(s)
- Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile.
| | - Cheril Tapia-Rojas
- Centro de Biología Celular y Biomedicina (CEBICEM), Laboratory of Neurobiology of Aging, Facultad de Medicina y Ciencia, Universidad de San Sebastián, Sede Los Leones, Santiago, Chile
| | - Waldo Cerpa
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
| | - Pedro Cisternas
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Instituto de Ciencias de la Salud, Universidad de O´Higgins, Rancagua, Chile
| | - Juan M Zolezzi
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
31
|
Zhong J, Qiu X, Yu Q, Chen H, Yan C. A novel polysaccharide from Acorus tatarinowii protects against LPS-induced neuroinflammation and neurotoxicity by inhibiting TLR4-mediated MyD88/NF-κB and PI3K/Akt signaling pathways. Int J Biol Macromol 2020; 163:464-475. [PMID: 32621930 DOI: 10.1016/j.ijbiomac.2020.06.266] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 06/02/2020] [Accepted: 06/28/2020] [Indexed: 12/26/2022]
Abstract
Our previous study has indicated that a crude polysaccharide derived from Acorus tatarinowii, AT50, remarkably improves learning and memory in scopolamine-induced amnesic mice and prevents the release of inflammatory mediators. To further explore the bioactive constituents of AT50, a novel polysaccharide (ATP50-3) was purified, and its anti-neuroinflammatory effects and underlying mechanisms were investigated. ATP50-3 significantly reduced abnormal elevation of inflammatory mediators in lipopolysaccharide (LPS)-induced proinflammatory BV2 cells in vitro and inhibited the activation of nuclear factor kappa B (NF-κB). Moreover, ATP50-3 down-regulated LPS-induced protein levels of Toll-like receptor 4 (TLR4), myeloid differentiation primary response protein (MyD88), p-PI3K (phosphoinositide 3-kinase), and p-Akt (protein kinase B). Further experiments demonstrated that TAK242 (a TLR4 inhibitor) and LY294002 (a PI3K inhibitor) remarkably augmented ATP50-3's down-regulation on LPS-induced proinflammatory mediators. Importantly, ATP50-3 provided neuroprotection against neuroinflammation-induced neurotoxicity in primary cortical and hippocampal neurons by mitigating overproduction of reactive oxygen species and damage to the mitochondrial membrane potential (MMP). Taken together, our findings suggest that ATP50-3 exerts anti-neuroinflammatory and neuroprotective effects through modulation of TLR4-mediated MyD88/NF-κB and PI3K/Akt signaling pathways.
Collapse
Affiliation(s)
- Jing Zhong
- Center for Clinical Precision Medication, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510006, China; School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xian Qiu
- Center for Clinical Precision Medication, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qian Yu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Haiyun Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Chunyan Yan
- Center for Clinical Precision Medication, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510006, China; School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
32
|
Pasqualini C, Kozaki T, Bruschi M, Nguyen THH, Minard-Colin V, Castel D, Grill J, Ginhoux F. Modeling the Interaction between the Microenvironment and Tumor Cells in Brain Tumors. Neuron 2020; 108:1025-1044. [PMID: 33065047 DOI: 10.1016/j.neuron.2020.09.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/26/2020] [Accepted: 09/09/2020] [Indexed: 12/18/2022]
Abstract
Despite considerable recent advances in understanding and treating many other cancers, malignant brain tumors remain associated with low survival or severe long-term sequelae. Limited progress, including development of immunotherapies, relates in part to difficulties in accurately reproducing brain microenvironment with current preclinical models. The cellular interactions among resident microglia, recruited tumor-associated macrophages, stromal cells, glial cells, neurons, and cancer cells and how they affect tumor growth or behavior are emerging, yet many questions remain. The role of the blood-brain barrier, extracellular matrix components, and heterogeneity among tumor types and within different regions of a single tumor further complicate the matter. Here, we focus on brain microenvironment features impacted by tumor biology. We also discuss limits of current preclinical models and how complementary models, such as humanized animals and organoids, will allow deeper mechanistic insights on cancer biology, allowing for more efficient testing of therapeutic strategies, including immunotherapy, for brain cancers.
Collapse
Affiliation(s)
- Claudia Pasqualini
- Children and Adolescent Oncology Department, Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - Tatsuya Kozaki
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Marco Bruschi
- Genomics & Oncogenesis of Pediatric Brain Tumors, INSERM U981, Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - Thi Hai Hoa Nguyen
- Genomics & Oncogenesis of Pediatric Brain Tumors, INSERM U981, Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - Véronique Minard-Colin
- Children and Adolescent Oncology Department, Gustave Roussy, Paris-Saclay University, Villejuif, France; INSERM U1015, Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - David Castel
- Genomics & Oncogenesis of Pediatric Brain Tumors, INSERM U981, Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - Jacques Grill
- Children and Adolescent Oncology Department, Gustave Roussy, Paris-Saclay University, Villejuif, France; Genomics & Oncogenesis of Pediatric Brain Tumors, INSERM U981, Gustave Roussy, Paris-Saclay University, Villejuif, France.
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore; Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.
| |
Collapse
|
33
|
Zhang RY, Tu JB, Ran RT, Zhang WX, Tan Q, Tang P, Kuang T, Cheng SQ, Chen CZ, Jiang XJ, Chen C, Han TL, Zhang T, Cao XQ, Peng B, Zhang H, Xia YY. Using the Metabolome to Understand the Mechanisms Linking Chronic Arsenic Exposure to Microglia Activation, and Learning and Memory Impairment. Neurotox Res 2020; 39:720-739. [PMID: 32955723 DOI: 10.1007/s12640-020-00286-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/28/2020] [Accepted: 09/07/2020] [Indexed: 12/19/2022]
Abstract
The activation of microglia is a hallmark of neuroinflammation and contributes to various neurodegenerative diseases. Chronic inorganic arsenic exposure is associated with impaired cognitive ability and increased risk of neurodegeneration. The present study aimed to investigate whether chronic inorganic arsenic-induced learning and memory impairment was associated with microglial activation, and how organic (DMAV 600 μM, MMAV 0.1 μM) and inorganic arsenic (NaAsO2 0.6 μM) affect the microglia. Male C57BL/6J mice were divided into two groups: a control group and a group exposed to arsenic in their drinking water (50 mg/L NaAsO2 for 24 weeks). The Morris water maze was performed to analyze neuro-behavior and transmission electron microscopy was used to assess alterations in cellular ultra-structures. Hematoxylin-eosin and Nissl staining were used to observe pathological changes in the cerebral cortex and hippocampus. Flow cytometry was used to reveal the polarization of the arsenic-treated microglia phenotype and GC-MS was used to assess metabolomic differences in the in vitro microglia BV-2 cell line model derived from mice. The results showed learning and memory impairments and activation of microglia in the cerebral cortex and dentate gyrus (DG) zone of the hippocampus, in mice chronically exposed to arsenic. Flow cytometry demonstrated that BV-2 cells were activated with the treatment of different arsenic species. The GC-MS data showed three important metabolites to be at different levels according to the different arsenic species used to treat the microglia. These included tyrosine, arachidonic acid, and citric acid. Metabolite pathway analysis showed that a metabolic pathways associated with tyrosine metabolism, the dopaminergic synapse, Parkinson's disease, and the citrate cycle were differentially affected when comparing exposure to organic arsenic and inorganic arsenic. Organic arsenic MMAV was predominantly pro-inflammatory, and inorganic arsenic exposure contributed to energy metabolism disruptions in BV-2 microglia. Our findings provide novel insight into understanding the neurotoxicity mechanisms of chronic arsenic exposure and reveal the changes of the metabolome in response to exposure to different arsenic species in the microglia.
Collapse
Affiliation(s)
- Rui-Yuan Zhang
- Department of Occupational and Environmental Hygiene, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People's Republic of China
| | - Jie-Bai Tu
- Department of Occupational and Environmental Hygiene, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People's Republic of China
| | - Rui-Tu Ran
- Department of Urinary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Wen-Xuan Zhang
- Department of Occupational and Environmental Hygiene, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People's Republic of China
| | - Qiang Tan
- Department of Occupational and Environmental Hygiene, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People's Republic of China
| | - Ping Tang
- Department of Occupational and Environmental Hygiene, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People's Republic of China
| | - Tao Kuang
- Department of Occupational and Environmental Hygiene, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People's Republic of China
| | - Shu-Qun Cheng
- Department of Occupational and Environmental Hygiene, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People's Republic of China
| | - Cheng-Zhi Chen
- Department of Occupational and Environmental Hygiene, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People's Republic of China
| | - Xue-Jun Jiang
- Department of Occupational and Environmental Hygiene, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People's Republic of China
| | - Chang Chen
- Institute of Life Sciences, Chongqing Medical University, Chongqing, People's Republic of China
| | - Ting-Li Han
- Institute of Life Sciences, Chongqing Medical University, Chongqing, People's Republic of China.,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing Medical University, Chongqing, People's Republic of China
| | - Ting Zhang
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing Medical University, Chongqing, People's Republic of China
| | - Xian-Qing Cao
- Department of Occupational and Environmental Hygiene, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People's Republic of China
| | - Bin Peng
- Department of Statistics, School of Public Health and Management, Chongqing Medical University, Chongqing, People's Republic of China
| | - Hua Zhang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, People's Republic of China.,Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yin-Yin Xia
- Department of Occupational and Environmental Hygiene, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
34
|
Neuroinflammation and progressive myoclonus epilepsies: from basic science to therapeutic opportunities. Expert Rev Mol Med 2020; 22:e4. [PMID: 32938505 PMCID: PMC7520540 DOI: 10.1017/erm.2020.5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Progressive myoclonus epilepsies (PMEs) are a group of genetic neurological disorders characterised by the occurrence of epileptic seizures, myoclonus and progressive neurological deterioration including cerebellar involvement and dementia. The primary cause of PMEs is variable and alterations in the corresponding mutated genes determine the progression and severity of the disease. In most cases, they lead to the death of the patient after a period of prolonged disability. PMEs also share poor information on the pathophysiological bases and the lack of a specific treatment. Recent reports suggest that neuroinflammation is a common trait under all these conditions. Here, we review similarities and differences in neuroinflammatory response in several PMEs and discuss the window of opportunity of using anti-inflammatory drugs in the treatment of several of these conditions.
Collapse
|
35
|
Mori T, Koyama N, Yokoo T, Segawa T, Maeda M, Sawmiller D, Tan J, Town T. Gallic acid is a dual α/β-secretase modulator that reverses cognitive impairment and remediates pathology in Alzheimer mice. J Biol Chem 2020; 295:16251-16266. [PMID: 32913125 DOI: 10.1074/jbc.ra119.012330] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 09/02/2020] [Indexed: 01/22/2023] Open
Abstract
Several plant-derived compounds have demonstrated efficacy in pre-clinical Alzheimer's disease (AD) rodent models. Each of these compounds share a gallic acid (GA) moiety, and initial assays on this isolated molecule indicated that it might be responsible for the therapeutic benefits observed. To test this hypothesis in a more physiologically relevant setting, we investigated the effect of GA in the mutant human amyloid β-protein precursor/presenilin 1 (APP/PS1) transgenic AD mouse model. Beginning at 12 months, we orally administered GA (20 mg/kg) or vehicle once daily for 6 months to APP/PS1 mice that have accelerated Alzheimer-like pathology. At 18 months of age, GA therapy reversed impaired learning and memory as compared with vehicle, and did not alter behavior in nontransgenic littermates. GA-treated APP/PS1 mice had mitigated cerebral amyloidosis, including brain parenchymal and cerebral vascular β-amyloid deposits, and decreased cerebral amyloid β-proteins. Beneficial effects co-occurred with reduced amyloidogenic and elevated nonamyloidogenic APP processing. Furthermore, brain inflammation, gliosis, and oxidative stress were alleviated. We show that GA simultaneously elevates α- and reduces β-secretase activity, inhibits neuroinflammation, and stabilizes brain oxidative stress in a pre-clinical mouse model of AD. We further demonstrate that GA increases abundance of a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10, Adam10) proprotein convertase furin and activates ADAM10, directly inhibits β-site APP cleaving enzyme 1 (BACE1, Bace1) activity but does not alter Adam10 or Bace1 transcription. Thus, our data reveal novel post-translational mechanisms for GA. We suggest further examination of GA supplementation in humans will shed light on the exciting therapeutic potential of this molecule.
Collapse
Affiliation(s)
- Takashi Mori
- Department of Biomedical Sciences, Saitama Medical Center and University, Kawagoe, Saitama, Japan; Department of Pathology, Saitama Medical Center and University, Kawagoe, Saitama, Japan.
| | - Naoki Koyama
- Department of Biomedical Sciences, Saitama Medical Center and University, Kawagoe, Saitama, Japan
| | - Tomotaka Yokoo
- The Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, Japan
| | - Tatsuya Segawa
- The Immuno-Biological Laboratories Co., Ltd., Fujioka, Gunma, Japan
| | - Masahiro Maeda
- The Immuno-Biological Laboratories Co., Ltd., Fujioka, Gunma, Japan
| | - Darrell Sawmiller
- The Department of Neurosurgery and Brain Repair, Center for Aging and Brain Repair, Morsoni College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Jun Tan
- The Department of Psychiatry and Behavioral Neurosciences, Morsoni College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Terrence Town
- The Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
36
|
Kimono D, Bose D, Seth RK, Mondal A, Saha P, Janulewicz P, Sullivan K, Lasley S, Horner R, Klimas N, Chatterjee S. Host Akkermansia muciniphila Abundance Correlates With Gulf War Illness Symptom Persistence via NLRP3-Mediated Neuroinflammation and Decreased Brain-Derived Neurotrophic Factor. Neurosci Insights 2020; 15:2633105520942480. [PMID: 32832901 PMCID: PMC7440889 DOI: 10.1177/2633105520942480] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 06/25/2020] [Indexed: 12/16/2022] Open
Abstract
Neurological disorders are commonly reported among veterans who returned from the Gulf war. Veterans who suffer from Gulf War illness (GWI) complain of continued symptom persistence that includes neurological disorders, muscle weakness, headaches, and memory loss, that developed during or shortly after the war. Our recent research showed that chemical exposure associated microbial dysbiosis accompanied by a leaky gut connected the pathologies in the intestine, liver, and brain. However, the mechanisms that caused the symptoms to persist even 30 years after the war remained elusive to investigators. In this study, we used a rodent model of GWI to investigate the persistence of microbiome alterations, resultant chronic inflammation, and its effect on neurotrophic and synaptic plasticity marker BDNF. The results showed that exposure to GW chemicals (the pesticide permethrin and prophylactic drug pyridostigmine bromide) resulted in persistent pathology characterized by the low relative abundance of the probiotic bacteria Akkermansia muciniphila in the gut, which correlated with high circulatory HMGB1 levels, blood-brain barrier dysfunction, neuroinflammation and lowered neurotrophin BDNF levels. Mechanistically, we used mice lacking the NLRP3 gene to investigate this inflammasome's role in observed pathology. These mice had significantly decreased inflammation and a subsequent increase in BDNF in the frontal cortex. This suggests that a persistently low species abundance of Akkermansia muciniphila and associated chronic inflammation due to inflammasome activation might be playing a significant role in contributing to chronic neurological problems in GWI. A therapeutic approach with various small molecules that can target both the restoration of a healthy microbiome and decreasing inflammasome activation might have better outcomes in treating GWI symptom persistence.
Collapse
Affiliation(s)
- Diana Kimono
- Environmental Health and Disease Laboratory, NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, USA
| | - Dipro Bose
- Environmental Health and Disease Laboratory, NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, USA
| | - Ratanesh K Seth
- Environmental Health and Disease Laboratory, NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, USA
| | - Ayan Mondal
- Environmental Health and Disease Laboratory, NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, USA
| | - Punnag Saha
- Environmental Health and Disease Laboratory, NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, USA
| | - Patricia Janulewicz
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Kimberly Sullivan
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Stephen Lasley
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA
| | - Ronnie Horner
- Department of Health Services Policy and Management, University of South Carolina, Columbia, SC, USA
| | - Nancy Klimas
- Department of Clinical Immunology, College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
37
|
Silva MC, Haggarty SJ. Human pluripotent stem cell-derived models and drug screening in CNS precision medicine. Ann N Y Acad Sci 2020; 1471:18-56. [PMID: 30875083 PMCID: PMC8193821 DOI: 10.1111/nyas.14012] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/02/2019] [Accepted: 01/07/2019] [Indexed: 12/12/2022]
Abstract
Development of effective therapeutics for neurological disorders has historically been challenging partly because of lack of accurate model systems in which to investigate disease etiology and test new therapeutics at the preclinical stage. Human stem cells, particularly patient-derived induced pluripotent stem cells (iPSCs) upon differentiation, have the ability to recapitulate aspects of disease pathophysiology and are increasingly recognized as robust scalable systems for drug discovery. We review advances in deriving cellular models of human central nervous system (CNS) disorders using iPSCs along with strategies for investigating disease-relevant phenotypes, translatable biomarkers, and therapeutic targets. Given their potential to identify novel therapeutic targets and leads, we focus on phenotype-based, small-molecule screens employing human stem cell-derived models. Integrated efforts to assemble patient iPSC-derived cell models with deeply annotated clinicopathological data, along with molecular and drug-response signatures, may aid in the stratification of patients, diagnostics, and clinical trial success, shifting translational science and precision medicine approaches. A number of remaining challenges, including the optimization of cost-effective, large-scale culture of iPSC-derived cell types, incorporation of aging into neuronal models, as well as robustness and automation of phenotypic assays to support quantitative drug efficacy, toxicity, and metabolism testing workflows, are covered. Continued advancement of the field is expected to help fully humanize the process of CNS drug discovery.
Collapse
Affiliation(s)
- M. Catarina Silva
- Chemical Neurobiology Laboratory, Departments of Neurology and Psychiatry, Massachusetts General Hospital, Center for Genomic Medicine, Harvard Medical School, Boston MA, USA
| | - Stephen J. Haggarty
- Chemical Neurobiology Laboratory, Departments of Neurology and Psychiatry, Massachusetts General Hospital, Center for Genomic Medicine, Harvard Medical School, Boston MA, USA
| |
Collapse
|
38
|
Kaur H, Nagamoto-Combs K, Golovko S, Golovko MY, Klug MG, Combs CK. Probiotics ameliorate intestinal pathophysiology in a mouse model of Alzheimer's disease. Neurobiol Aging 2020; 92:114-134. [PMID: 32417748 DOI: 10.1016/j.neurobiolaging.2020.04.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 02/06/2023]
Abstract
Evidence suggests that changes in intestinal microbiota may affect the central nervous system. However, it is unclear whether alteration of intestinal microbiota affects progression of Alzheimer's disease (AD). To understand this, wild-type control (C57BL/6) mice were compared with the AppNL-G-F model of disease. We used probiotic supplementation to manipulate the gut microbiota. Fecal samples were collected for microbiota profiling. To study brain and intestinal inflammation, biochemical and histological analyses were performed. Altered metabolic pathways were examined by quantifying eicosanoid and bile acid profiles in the brain and serum using ultraperformance liquid chromatography-tandem mass spectrometry. We observed that brain pathology was associated with intestinal dysbiosis and increased intestinal inflammation and leakiness in AppNL-G-F mice. Probiotic supplementation significantly decreased intestinal inflammation and gut permeability with minimal effect on amyloid-β, cytokine, or gliosis levels in the brain. Concentrations of several bile acids and prostaglandins were altered in the serum and brain because of AD or probiotic supplementation. Our study characterizes intestinal dysfunction in an AD mouse model and the potential of probiotic intervention to ameliorate this condition.
Collapse
Affiliation(s)
- Harpreet Kaur
- Department of Biomedical Sciences, University of North Dakota, School of Medicine & Health Sciences, Grand Forks, ND, USA
| | - Kumi Nagamoto-Combs
- Department of Pathology, University of North Dakota, School of Medicine & Health Sciences, Grand Forks, ND, USA
| | - Svetlana Golovko
- Department of Biomedical Sciences, University of North Dakota, School of Medicine & Health Sciences, Grand Forks, ND, USA
| | - Mikhail Y Golovko
- Department of Biomedical Sciences, University of North Dakota, School of Medicine & Health Sciences, Grand Forks, ND, USA
| | - Marilyn G Klug
- Department of Population Health, University of North Dakota, School of Medicine & Health Sciences, Grand Forks, ND, USA
| | - Colin Kelly Combs
- Department of Biomedical Sciences, University of North Dakota, School of Medicine & Health Sciences, Grand Forks, ND, USA.
| |
Collapse
|
39
|
Oxidative Stress, a Crossroad Between Rare Diseases and Neurodegeneration. Antioxidants (Basel) 2020; 9:antiox9040313. [PMID: 32326494 PMCID: PMC7222183 DOI: 10.3390/antiox9040313] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/06/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress is an imbalance between production and accumulation of oxygen reactive species and/or reactive nitrogen species in cells and tissues, and the capacity of detoxifying these products, using enzymatic and non-enzymatic components, such as glutathione. Oxidative stress plays roles in several pathological processes in the nervous system, such as neurotoxicity, neuroinflammation, ischemic stroke, and neurodegeneration. The concepts of oxidative stress and rare diseases were formulated in the eighties, and since then, the link between them has not stopped growing. The present review aims to expand knowledge in the pathological processes associated with oxidative stress underlying some groups of rare diseases: Friedreich’s ataxia, diseases with neurodegeneration with brain iron accumulation, Charcot-Marie-Tooth as an example of rare neuromuscular disorders, inherited retinal dystrophies, progressive myoclonus epilepsies, and pediatric drug-resistant epilepsies. Despite the discrimination between cause and effect may not be easy on many occasions, all these conditions are Mendelian rare diseases that share oxidative stress as a common factor, and this may represent a potential target for therapies.
Collapse
|
40
|
Devanney NA, Stewart AN, Gensel JC. Microglia and macrophage metabolism in CNS injury and disease: The role of immunometabolism in neurodegeneration and neurotrauma. Exp Neurol 2020; 329:113310. [PMID: 32289316 DOI: 10.1016/j.expneurol.2020.113310] [Citation(s) in RCA: 169] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 03/25/2020] [Accepted: 04/10/2020] [Indexed: 12/12/2022]
Abstract
Innate immune responses, particularly activation of macrophages and microglia, are increasingly implicated in CNS disorders. It is now appreciated that the heterogeneity of functions adopted by these cells dictates neuropathophysiology. Research efforts to characterize the range of pro-inflammatory and anti-inflammatory phenotypes and functions adopted by microglia and macrophages are fueled by the potential for inflammatory cells to both exacerbate neurodegeneration and promote repair/disease resolution. The stimulation-based, M1/M2 classification system has emerged over the last decade as a common language to discuss macrophage and microglia heterogeneity across different fields. However, discontinuities between phenotypic markers and function create potential hurdles for the utility of the M1/M2 system in the development of effective immunomodulatory therapeutics for neuroinflammation. A framework to approach macrophage and microglia heterogeneity from a function-based phenotypic approach comes from rapidly emerging evidence that metabolic processes regulate immune cell activation. This concept of immunometabolism, however, is only beginning to unfold in the study of neurodegeneration and has yet to receive much focus in the context of neurotrauma. In this review, we first discuss the current views of macrophage and microglia heterogeneity and limitations of the M1/M2 classification system for neuropathological studies. We then review and discuss the current literature supporting metabolism as a regulator of microglia function in vitro. Lastly, we evaluate the evidence that metabolism regulates microglia and macrophage phenotype in vivo in models of Alzheimer's disease (AD), stroke, traumatic brain injury (TBI) and spinal cord injury (SCI).
Collapse
Affiliation(s)
- Nicholas A Devanney
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America
| | - Andrew N Stewart
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America; Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America
| | - John C Gensel
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America; Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America.
| |
Collapse
|
41
|
Kempuraj D, Ahmed ME, Selvakumar GP, Thangavel R, Dhaliwal AS, Dubova I, Mentor S, Premkumar K, Saeed D, Zahoor H, Raikwar SP, Zaheer S, Iyer SS, Zaheer A. Brain Injury-Mediated Neuroinflammatory Response and Alzheimer's Disease. Neuroscientist 2020; 26:134-155. [PMID: 31092147 PMCID: PMC7274851 DOI: 10.1177/1073858419848293] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Traumatic brain injury (TBI) is a major health problem in the United States, which affects about 1.7 million people each year. Glial cells, T-cells, and mast cells perform specific protective functions in different regions of the brain for the recovery of cognitive and motor functions after central nervous system (CNS) injuries including TBI. Chronic neuroinflammatory responses resulting in neuronal death and the accompanying stress following brain injury predisposes or accelerates the onset and progression of Alzheimer's disease (AD) in high-risk individuals. About 5.7 million Americans are currently living with AD. Immediately following brain injury, mast cells respond by releasing prestored and preactivated mediators and recruit immune cells to the CNS. Blood-brain barrier (BBB), tight junction and adherens junction proteins, neurovascular and gliovascular microstructural rearrangements, and dysfunction associated with increased trafficking of inflammatory mediators and inflammatory cells from the periphery across the BBB leads to increase in the chronic neuroinflammatory reactions following brain injury. In this review, we advance the hypothesis that neuroinflammatory responses resulting from mast cell activation along with the accompanying risk factors such as age, gender, food habits, emotional status, stress, allergic tendency, chronic inflammatory diseases, and certain drugs can accelerate brain injury-associated neuroinflammation, neurodegeneration, and AD pathogenesis.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs’, Columbia, MO 65201, USA
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Mohammad Ejaz Ahmed
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs’, Columbia, MO 65201, USA
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Govindhasamy Pushpavathi Selvakumar
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs’, Columbia, MO 65201, USA
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs’, Columbia, MO 65201, USA
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Arshdeep S. Dhaliwal
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Iuliia Dubova
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs’, Columbia, MO 65201, USA
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Shireen Mentor
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Keerthivaas Premkumar
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Daniyal Saeed
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Haris Zahoor
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Sudhanshu P. Raikwar
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs’, Columbia, MO 65201, USA
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Smita Zaheer
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Shankar S. Iyer
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs’, Columbia, MO 65201, USA
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Asgar Zaheer
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs’, Columbia, MO 65201, USA
- Department of Neurology, and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
42
|
Guo Q, Xu S, Yang P, Wang P, Lu S, Sheng D, Qian K, Cao J, Lu W, Zhang Q. A dual-ligand fusion peptide improves the brain-neuron targeting of nanocarriers in Alzheimer's disease mice. J Control Release 2020; 320:347-362. [DOI: 10.1016/j.jconrel.2020.01.039] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 12/17/2022]
|
43
|
Anwar S, Rivest S. Alzheimer's disease: microglia targets and their modulation to promote amyloid phagocytosis and mitigate neuroinflammation. Expert Opin Ther Targets 2020; 24:331-344. [PMID: 32129117 DOI: 10.1080/14728222.2020.1738391] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Introduction: Despite the revolutionary progress in neurodegenerative disease research, there is no cure for Alzheimer's disease (AD). This is a chronic progressive neurodegenerative disease affecting aged people and is associated with chronic neuroinflammation and amyloid-beta (Aβ) deposition in the brain parenchyma. Microglia, the resident myeloid cells in the central nervous system, are critically involved in the pathogenesis of AD and have emerged as a potential therapeutic target for treating or preventing AD. The failure of microglia to keep up with persistent amyloid-beta development along with secretion of inflammatory cytokines is detrimental to neurons and favors Aβ accumulation.Areas covered: This review illuminates the latest research that is focused on molecules and their intracellular targets that promote microglial phagocytosis and /or its polarization to an anti-inflammatory state.Expert opinion: A robust inflammatory response of microglia is not necessary to improve their efficiency of Aβ clearance. The challenge is to master inflammatory/anti-inflammatory phenotypes depending on the stage of AD and to maintain efficient responses to remove Aβ. Therefore, promoting microglia phagocytosis without a persistent excessive inflammatory response could be a potential therapeutic strategy.
Collapse
Affiliation(s)
- Shehata Anwar
- Neuroscience Laboratory, CHU de Québec Research Center (CHUL), Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC, Canada.,Department of Pathology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Québec Research Center (CHUL), Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| |
Collapse
|
44
|
Fu Q, Li J, Qiu L, Ruan J, Mao M, Li S, Mao Q. Inhibiting NLRP3 inflammasome with MCC950 ameliorates perioperative neurocognitive disorders, suppressing neuroinflammation in the hippocampus in aged mice. Int Immunopharmacol 2020; 82:106317. [PMID: 32087497 DOI: 10.1016/j.intimp.2020.106317] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/09/2020] [Accepted: 02/13/2020] [Indexed: 01/16/2023]
Abstract
Perioperative neurocognitive disorders (PND) are characterized by deficits in cognitive functions in the elderly following anesthesia and surgery. Effective clinical interventions for preventing this disease are limited. Growing evidence demonstrates that activation of NOD-like receptor protein3 (NLRP3) inflammasome is involved in neurodegenerative diseases. We therefore hypothesized that activation of NLRP3 inflammasome is linked to neuroinflammation and the subsequent cognitive impairments that occurred in an animal model of PND. In this study, 18-month-old C57BL/6 mice were subjected to an exploratory laparotomy under isoflurane anesthesia to mimic clinical human abdominal surgery. For interventional studies, mice received NLRP3 specific inhibitor MCC950 (10 mg/kg) or the vehicle only intraperitoneally. Behavioral studies were performed at 6 and 7 d after surgery using open field and fear conditioning tests, respectively. Interleukin-1β (IL-1β), interleukin-18 (IL-18), tumor necrosis factor-α (TNF-α), ionized calcium-binding adaptor molecule-1 (IBA1) positive cells, glial fibrillary acidic protein (GFAP) positive cells, NLRP3, apoptosis-associated speck-like protein containing a CARD (ASC), and cleaved caspase-1 were measured at 3 days post-surgery. Brain-derived neurotrophic factor (BDNF) and postsynaptic density protein 95 (PSD95) were measured at 7 days post-surgery. Our data indicates that surgery-induced cognitive impairments were associated with significant increases in IL-1β, IL-18, TNF-α, NLRP3, ASC, cleaved caspase-1, IBA1-positive cells and GFAP-positive cells, and decreases in BDNF and PSD95 expression in the hippocampus. Notably, administration with MCC950 attenuated inflammatory changes and rescued surgery-induced cognitive impairments. Our study suggests that surgery induces neuroinflammation and cognitive deficits that are partly attributed to the activation of NLRP3 inflammasome in the hippocampus of aged mice.
Collapse
Affiliation(s)
- Qun Fu
- Department of Anesthesiology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Jing Li
- Fenghuang Community Health Service Center, Gulou District, Nanjing 210029, China
| | - Lili Qiu
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jiaping Ruan
- Department of Anesthesiology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Mingjie Mao
- Department of Anesthesiology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Shuming Li
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Qinghong Mao
- Department of Anesthesiology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China.
| |
Collapse
|
45
|
Zheng F, Zhou YT, Feng DD, Li PF, Tang T, Luo JK, Wang Y. Metabolomics analysis of the hippocampus in a rat model of traumatic brain injury during the acute phase. Brain Behav 2020; 10:e01520. [PMID: 31908160 PMCID: PMC7010586 DOI: 10.1002/brb3.1520] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 11/26/2019] [Accepted: 12/02/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) has increased in rank among traumatic injuries worldwide. Traumatic brain injury is a serious obstacle given that its complex pathology represents a long-term process. Recently, systems biology strategies such as metabolomics to investigate the multifactorial nature of TBI have facilitated attempts to find biomarkers and probe molecular pathways for its diagnosis and therapy. METHODS This study included a group of 20 rats with controlled cortical impact and a group of 20 sham rats. We utilized mNSS tests to investigate neurological metabolic impairments on day 1 and day 3. Furthermore, we applied metabolomics and bioinformatics to determine the metabolic perturbation caused by TBI during the acute period in the hippocampus tissue of controlled cortical impact (CCI) rats. Notably, TBI-protein-metabolite subnetworks identified from a database were assessed for associations between metabolites and TBI by the dysregulation of related enzymes and transporters. RESULTS Our results identified 7 and 8 biomarkers on day 1 and day 3, respectively. Additionally, related pathway disorders showed effects on arginine and proline metabolism as well as taurine and hypotaurine metabolism on day 3 in acute TBI. Furthermore, according to metabolite-protein database searches, 25 metabolite-protein pairs were established as causally associated with TBI. Further, bioinformation indicated that these TBI-associated proteins mainly take part in 5'-nucleotidase activity and carboxylic acid transmembrane transport. In addition, interweaved networks were constructed to show that the development of TBI might be affected by metabolite-related proteins and their protein pathways. CONCLUSION The overall results show that acute TBI is susceptible to metabolic disorders, and the joint metabolite-protein network analysis provides a favorable prediction of TBI pathogenesis mechanisms in the brain. The signatures in the hippocampus might be promising for the development of biomarkers and pathways relevant to acute TBI and could further guide testable predictions of the underlying mechanism of TBI.
Collapse
Affiliation(s)
- Fei Zheng
- College of Electrical and Information Engineering, Hunan University, Changsha, China
| | - Yan-Tao Zhou
- College of Electrical and Information Engineering, Hunan University, Changsha, China
| | - Dan-Dan Feng
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Peng-Fei Li
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Tao Tang
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Jie-Kun Luo
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yang Wang
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
46
|
Shimada A. Principles of neuroanatomical architecture supporting brain–immune cell–cell interactions. ACTA ACUST UNITED AC 2020. [DOI: 10.1111/cen3.12559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
47
|
Ciccocioppo F, Bologna G, Ercolino E, Pierdomenico L, Simeone P, Lanuti P, Pieragostino D, Del Boccio P, Marchisio M, Miscia S. Neurodegenerative diseases as proteinopathies-driven immune disorders. Neural Regen Res 2020; 15:850-856. [PMID: 31719246 PMCID: PMC6990794 DOI: 10.4103/1673-5374.268971] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
In the pathophysiology of neurodegenerative disorders, the role of misfolded protein deposition leading to neurodegeneration has been primarily discussed. In the last decade, however, it has been proposed a parallel involvement of innate immune activation, chronic inflammation and adaptive immunity in the neurodegeneration mechanisms triggered by proteinopathies. New insights in the neurodegenerative field strongly suggest a role for the immune system in the pathophysiology of neurodegenerative disorders. Therefore, the hypothesis underlining the modulation of the innate and the adaptive immune system in the events linked to brain deposition of misfolded proteins could open new perspectives in the setting of specific immunotherapeutic strategies for the treatment of neurodegenerative diseases. Therefore, we have reviewed the pathogenic hypothesis in neurodegenerative pathologies, underling the links between the deposition of misfolded protein mechanisms and the immune activation.
Collapse
Affiliation(s)
- Fausta Ciccocioppo
- Department of Medicine and Aging Science; Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Giuseppina Bologna
- Department of Medicine and Aging Science; Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Eva Ercolino
- Department of Medicine and Aging Science; Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Laura Pierdomenico
- Department of Medicine and Aging Science; Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Pasquale Simeone
- Department of Medicine and Aging Science; Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Paola Lanuti
- Department of Medicine and Aging Science; Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Damiana Pieragostino
- Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT); Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Piero Del Boccio
- Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT); Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Marco Marchisio
- Department of Medicine and Aging Science; Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Sebastiano Miscia
- Department of Medicine and Aging Science; Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
48
|
Nouri A, Hashemzadeh F, Soltani A, Saghaei E, Amini-Khoei H. Progesterone exerts antidepressant-like effect in a mouse model of maternal separation stress through mitigation of neuroinflammatory response and oxidative stress. PHARMACEUTICAL BIOLOGY 2020; 58:64-71. [PMID: 31873049 PMCID: PMC6968520 DOI: 10.1080/13880209.2019.1702704] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 11/01/2019] [Accepted: 12/04/2019] [Indexed: 06/10/2023]
Abstract
Context: Experiencing early-life adversity plays a key role in the development of mood disorders in adulthood. Experiencing adversities during early life period negatively affects brain development. Sex steroids such as progesterone affect the brain structure and functions and subsequently affects behaviour.Objective: We assess the antidepressant-like effect of progesterone in a mouse model of maternal separation (MS) stress, focussing on its anti-neuroinflammatory and antioxidative effects.Materials and methods: NMRI mice were treated with progesterone (10, 50, and 100 mg/kg, i.p., respectively) for 14 days. Valid behavioural tests including forced swimming test (FST), splash test and open field test (OFT) were used. Quantitative reverse transcription-PCR (qRT-PCR) was used for evaluation of genetic expression in the hippocampus. Antioxidant capacity was assessed by the FRAP method and the level of malondialdehide by TBA.Results: MS provoked depressive-like behaviour in mice. Treatment of MS mice with progesterone increased the grooming activity time in the splash test and decreased the immobility time in the FST. In addition, progesterone decreased the expression of inflammatory genes related to neuroinflammation (IL-1β, TNF-α, TLR4 and NLRP3) as well as increased the antioxidant capacity and decreased the lipid peroxidation (MDA) in the hippocampus.Discussion and Conclusion: Administration of progesterone significantly mitigated the negative effects of MS on behaviours relevant to depressive-like behaviour as well as attenuated neuro-immune response and oxidative stress in the hippocampus of MS mice. In this context, we conclude that progesterone, at least partially, via attenuation of oxidative stress and neuroinflammation, exerts antidepressant-like effects.
Collapse
Affiliation(s)
- Ali Nouri
- Medical plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Farzaneh Hashemzadeh
- Medical plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Amin Soltani
- Medical plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Elham Saghaei
- Medical plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hossein Amini-Khoei
- Medical plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
49
|
Peng S, Jia J, Sun J, Xie Q, Zhang X, Deng Y, Yi L. LXW7 attenuates inflammation via suppressing Akt/nuclear factor kappa B and mitogen-activated protein kinases signaling pathways in lipopolysaccharide-stimulated BV2 microglial cells. Int Immunopharmacol 2019; 77:105963. [DOI: 10.1016/j.intimp.2019.105963] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/07/2019] [Accepted: 10/03/2019] [Indexed: 02/06/2023]
|
50
|
Lindsay CB, Zolezzi JM, Rivera DS, Cisternas P, Bozinovic F, Inestrosa NC. Andrographolide Reduces Neuroinflammation and Oxidative Stress in Aged Octodon degus. Mol Neurobiol 2019; 57:1131-1145. [DOI: 10.1007/s12035-019-01784-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 09/12/2019] [Indexed: 12/12/2022]
|