1
|
Sadier NS, Hazimeh IA, Khazaal W, Al Sabouri AAK, Almutary AG, Alnuqaydan AM, Abou-Abbas L. Exploring the therapeutic potential of NLRP3 inhibitors in Parkinson's Disease: a systematic review of in-vivo studies. Inflammopharmacology 2025:10.1007/s10787-025-01733-x. [PMID: 40259110 DOI: 10.1007/s10787-025-01733-x] [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: 02/26/2024] [Accepted: 10/29/2024] [Indexed: 04/23/2025]
Abstract
BACKGROUND Parkinson's disease is a progressive neurodegenerative disorder characterized by motor symptoms such as tremors, rigidity, and bradykinesia. Although the exact etiology is unknown, the nod-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome-induced inflammation, plays a crucial role in the pathogenesis of Parkinson's disease. Many NLRP3 inhibitors are recognized for their role as potential therapeutic interventions for Parkinson's disease. METHODS A systematic literature search was performed in PubMed, Embase, and Science Direct databases for papers published during the 10 years prior to May 2023. All animal interventional studies assessing the effects of NLRP3 inhibitors on Parkinson's disease animal models were included. Primary outcomes included NLRP3 inflammasome inhibition, microglial activation reduction, oxidative stress, and anti-inflammatory marker reduction. The secondary outcomes included dopaminergic neuron loss alleviation and behavioral motor function attenuation. Quality assessment and narrative synthesis of the studies were performed. RESULTS Twenty-four studies out of 796 papers initially identified met the inclusion criteria. All the included studies, except one, found a reduction in NLRP3 inflammasome activation and anti-inflammatory markers in Parkinson's disease animal models after treatment with various NLRP3 inhibitors compared to control groups without inhibitors. Additionally, eighteen out of twenty-four inhibitors decreased microglial activation and behavioral deficits. Moreover, ten inhibitors attenuated oxidative stress, and twenty-two out of twenty-four alleviated dopaminergic neuron loss. The inhibitors utilized different mechanisms and pathways to exert their effects, including the NLRP3/Caspase-1 pathway, the NF-κB/NLRP3 pathway, inhibition of ROS and/or pyroptosis, as well as autophagy and mitophagy. CONCLUSION NLRP3 inhibitors represent a prospective therapy for Parkinson's disease, demonstrating efficacy in lowering neuroinflammation and protecting against dopaminergic loss. However, constraints, such as a male animal focus, apparent regional bias from China-centric studies, and diversity in induction models, entail the results presented herein require cautious interpretation. Further research, including preclinical and clinical studies, is required to thoroughly examine the safety, effectiveness, and generalizability of NLRP3 inhibitors in Parkinson's disease.
Collapse
Affiliation(s)
- Najwane Said Sadier
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Al Ain Road, PO Box 3838-111188, Abu Dhabi, UAE
- Department of Neurosciences, Neurosciences Research Center, Faculty of Medical Sciences, Lebanese University, 275 Old Saida Road, PO Box 6573/14, Beirut, Lebanon
| | - Inaam Ali Hazimeh
- Department of Neurosciences, Neurosciences Research Center, Faculty of Medical Sciences, Lebanese University, 275 Old Saida Road, PO Box 6573/14, Beirut, Lebanon
| | - Walaa Khazaal
- Department of Neurosciences, Neurosciences Research Center, Faculty of Medical Sciences, Lebanese University, 275 Old Saida Road, PO Box 6573/14, Beirut, Lebanon
| | - Amani Al Khayat Al Sabouri
- Department of Neurosciences, Neurosciences Research Center, Faculty of Medical Sciences, Lebanese University, 275 Old Saida Road, PO Box 6573/14, Beirut, Lebanon
| | - Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Al Ain Road, PO Box 3838-111188, Abu Dhabi, UAE
| | - Abdullah M Alnuqaydan
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Linda Abou-Abbas
- Department of Neurosciences, Neurosciences Research Center, Faculty of Medical Sciences, Lebanese University, 275 Old Saida Road, PO Box 6573/14, Beirut, Lebanon.
- INSPECT-LB (Institut National de Santé Publique, d'Épidémiologie Clinique Et de Toxicologie-Liban), Beirut, Lebanon.
| |
Collapse
|
2
|
Churchill NW, Hutchison MG, Graham SJ, Schweizer TA. Post-Concussion Brain Changes Relative to Pre-Injury White Matter and Cerebral Blood Flow: A Prospective Observational Study. Neurology 2025; 104:e213374. [PMID: 40073308 DOI: 10.1212/wnl.0000000000213374] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 12/06/2024] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Medical clearance for return to play (RTP) after sports-related concussion is based on clinical assessment. It is unknown whether brain physiology has entirely returned to preinjury baseline at the time of clearance. In this longitudinal study, we assessed whether concussed individuals show functional and structural MRI brain changes relative to preinjury levels that persist beyond medical clearance. Secondary objectives were to test whether postconcussion changes exceed uninjured brain variability and to correlate MRI findings with clinical recovery time. METHODS For this prospective observational study, healthy athletes without a history of psychiatric, neurologic, or sensory-motor conditions were recruited from a single university sport medicine clinic. Clinical and MRI data were collected at preseason baseline, and those who were later concussed were reassessed at 1-7 days after injury, RTP, 1-3 months after RTP, and 1 year after RTP. A demographically matched control cohort of uninjured athletes was also reassessed at their subsequent preseason baseline. Primary outcomes were postconcussion changes in MRI measures of cerebral blood flow (CBF), white matter mean diffusivity (MD), and fractional anisotropy (FA), evaluated using mixed models. Secondary outcomes were group differences in MRI change scores and correlations of change scores with days to RTP. RESULTS Of the 187 athletes enrolled in the study, 25 had concussion with follow-up imaging (20.3 ± 1.5 years, 56% male, 44% female) and were compared with 27 controls (19.7 ± 1.8 years, 44% male, 56% female). Concussed athletes showed statistically significant changes from baseline, including decreased frontoinsular CBF (mean and 95% CI -8.97 [-12.80, -5.01] mL/100 g/minute, z = -4.53), along with increased MD (1.94 × 10-5 [1.26, 2.69] × 10-5, z = 5.48) and reduced FA (-7.30 × 10-3 [-9.80, -5.05] × 10-3, z = -6.07) in the corona radiata and internal capsule. Effects persisted beyond RTP, although only CBF changes exceeded longitudinal variability in controls. For participants with longer recovery periods, significantly greater changes in medial temporal CBF were also seen (ρ = 0.64 [0.44, 0.81], z = 6.80). DISCUSSION This study provides direct evidence of persistent postconcussion changes in CBF and white matter at RTP and up to 1 year later. These results support incomplete recovery of brain physiology at medical clearance, with secondary analyses emphasizing the sensitivity of CBF to clinical recovery.
Collapse
Affiliation(s)
- Nathan W Churchill
- Brain Health and Wellness Research Program, St. Michael's Hospital, Unity Health Toronto, Ontario, Canada
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Unity Health Toronto, Ontario, Canada
- Department of Physics, Toronto Metropolitan University, Ontario, Canada
| | - Michael G Hutchison
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Unity Health Toronto, Ontario, Canada
- Faculty of Kinesiology and Physical Education, University of Toronto, Ontario, Canada
| | - Simon J Graham
- Department of Medical Biophysics, University of Toronto, Ontario, Canada
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; and
| | - Tom A Schweizer
- Brain Health and Wellness Research Program, St. Michael's Hospital, Unity Health Toronto, Ontario, Canada
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Unity Health Toronto, Ontario, Canada
- Faculty of Medicine (Neurosurgery), University of Toronto, Ontario, Canada
| |
Collapse
|
3
|
Zhan A, Zhong K, Zhang K. Novel subcellular regulatory mechanisms of protein homeostasis and its implications in amyotrophic lateral sclerosis. Biochem Biophys Res Commun 2025; 756:151582. [PMID: 40056503 DOI: 10.1016/j.bbrc.2025.151582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron degenerative disorder. Protein aggregates induce various forms of neuronal dysfunction and represent pathological hallmarks in ALS patients. Reducing protein aggregates could be a promising therapeutic strategy for ALS. While most studies have focused on cytoplasmic protein homeostasis, neurons adaptively reduce aggregates across subcellular compartments during stress through previously uncharacterized mechanisms. Here, we summarize novel compartment-specific proteostatic mechanisms: (1) the ERAD/RESET pathways, (2) HSPs-mediated nuclear sequestration, (3) mitochondrial aggregate import (MAGIC), (4) neurite-localized UPS/autophagosome and NMP, and (5) exopher-mediated extracellular disposal. These mechanisms collectively ensure cellular stress adaptation and provide novel therapeutic targets for ALS treatment.
Collapse
Affiliation(s)
- Aisheng Zhan
- Institute of Translational Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Keke Zhong
- Institute of Translational Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Kejing Zhang
- Institute of Translational Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China.
| |
Collapse
|
4
|
Luo W, Xu C, Li L, Ji Y, Wang Y, Li Y, Ye Y. Perfluoropentane-based oxygen-loaded nanodroplets reduce microglial activation through metabolic reprogramming. Neural Regen Res 2025; 20:1178-1191. [PMID: 38989955 PMCID: PMC11438333 DOI: 10.4103/nrr.nrr-d-23-01299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/05/2024] [Indexed: 07/12/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202504000-00032/figure1/v/2024-07-06T104127Z/r/image-tiff Microglia, the primary immune cells within the brain, have gained recognition as a promising therapeutic target for managing neurodegenerative diseases within the central nervous system, including Parkinson's disease. Nanoscale perfluorocarbon droplets have been reported to not only possess a high oxygen-carrying capacity, but also exhibit remarkable anti-inflammatory properties. However, the role of perfluoropentane in microglia-mediated central inflammatory reactions remains poorly understood. In this study, we developed perfluoropentane-based oxygen-loaded nanodroplets (PFP-OLNDs) and found that pretreatment with these droplets suppressed the lipopolysaccharide-induced activation of M1-type microglia in vitro and in vivo, and suppressed microglial activation in a mouse model of Parkinson's disease. Microglial suppression led to a reduction in the inflammatory response, oxidative stress, and cell migration capacity in vitro. Consequently, the neurotoxic effects were mitigated, which alleviated neuronal degeneration. Additionally, ultrahigh-performance liquid chromatography-tandem mass spectrometry showed that the anti-inflammatory effects of PFP-OLNDs mainly resulted from the modulation of microglial metabolic reprogramming. We further showed that PFP-OLNDs regulated microglial metabolic reprogramming through the AKT-mTOR-HIF-1α pathway. Collectively, our findings suggest that the novel PFP-OLNDs constructed in this study alleviate microglia-mediated central inflammatory reactions through metabolic reprogramming.
Collapse
Affiliation(s)
- Wanxian Luo
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Chuanhui Xu
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Linxi Li
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yunxiang Ji
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yezhong Wang
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yingjia Li
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yongyi Ye
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
5
|
You Z, Jain S, Shen S, Mao J, Martyn JJ. Pathophysiology and management of burn injury-induced pain. BURNS OPEN 2025; 10:100396. [PMID: 40255244 PMCID: PMC12007888 DOI: 10.1016/j.burnso.2025.100396] [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] [Indexed: 04/22/2025] Open
Abstract
This review examines the pathophysiology and therapeutic management of burn injury-induced pain (BIP). Burn injury, occurring globally in about 11 million people, often induces the most intense pain, but its management remains suboptimal. The pain often persists even after complete wound healing and hospital discharge causing both long-term disability and neurological dysfunction. The fact that BIP persists well beyond the initial hospitalization is not well recognized and should be underscored as the pain involves even non-burned areas. The pathophysiology of the latter problem is poorly understood and needs further study. Opioids, the mainstay for moderate to severe pain relief after major burn injury, with time, have poor analgesic and serious side effects. Accurate assessment pain of BIP and its biology at different stages of treatment helps to provide effective treatments of the different etiological factors that cause BIP and their sequelae. Based on clinical and pre-clinical studies, we discuss the current knowledge on the underlying cellular and molecular mechanisms in the initiation and persistence of BIP during the acute phase and later phases of injury. Opioid receptor-mediated signaling changes per se and immune microglia responses in concert exaggerate nociceptive behavior. Both burn injury and opioids upregulate spinal NMDA receptor expression and microglia changes, which further exaggerate pain. BIP has inflammatory and neuropathic components. Pharmacological and non-pharmacological approaches currently available for management of BIP is discussed. Areas that need further study include the role of other central and peripheral factors in the exaggeration of pain well beyond wound healing. Novel non-opioid methods to rectify BIP is important to develop in view of the potential for opioid use disorder. The role of microbiome in chronic pain syndromes is an unexplored territory and its relevance to BIP needs further examination. Pruritus or itch, though very common and important in the pharmacotherapy of burns, the discussion of this topic is brief. Extensive review of this topic is beyond the scope of this review in view of the vast body of knowledge and varying and multiple treatment options.
Collapse
Affiliation(s)
- Zerong You
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, USA
- Shriners Hospital for Children, Boston, MA, USA
| | - Shubhika Jain
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, USA
| | - Shiqian Shen
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, USA
| | - Jianren Mao
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, USA
| | - J.A. Jeevendra Martyn
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, USA
- Shriners Hospital for Children, Boston, MA, USA
| |
Collapse
|
6
|
Chen Y, Zhu Y, Tan Z, Zhang X, Hu J, Zhu R, Xie M, Wang J, Chen L, Guo Z. Jiajian Shuyu pills effectively ameliorate cognitive impairment via regulating the inflammation of microglia in an Alzheimer's disease mouse model. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119508. [PMID: 39971019 DOI: 10.1016/j.jep.2025.119508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by progressive cognitive decline and behavioral impairments in the elderly. Microglia, the resident immune cells of the central nervous system, play a crucial role in modulating the pathological processes associated with AD. Jiajian Shuyu Pills (JJSYP) are frequently employed in the treatment of AD, purportedly by enhancing the physiological functions of human tissues and organs to modulate the immune response. Nevertheless, the underlying mechanisms by which JJSYP exert their therapeutic effects in the context of AD remain inadequately elucidated. AIM OF THE STUDY This study aimed to assess the effects of JJSYP on cognitive enhancement and the alleviation of neuroinflammation in the treatment of AD, as well as to explore the underlying mechanisms using mouse models. MATERIALS AND METHODS The components of JJSYP in serum were analyzed using HPLC-Q/TOF-MS. APP/PS1 transgenic mice served as AD models in this investigation. Cognitive function in the AD mice was assessed through the Mirror Water Maze Test and the Novel Object Recognition Test. The quantification of apoptotic hippocampal cells was conducted using Nissl staining and TUNEL staining. Immunofluorescence (IF) and Western blot (WB) analyses were employed to examine microglial activation and the expression of relevant proteins. Transcriptomic sequencing analysis and network pharmacology were administrated to explore the potential mechanisms of JJSYP in AD treatment. Inflammatory cytokine levels in the brain were measured using RT-PCR. RESULTS A total of 74 absorbed prototype components from JJSYP were identified. JJSYP effectively improved cognitive function and neuroapoptosis in AD model mice by modulating the activation of microglia. The JJSYP intervention alleviated neuroinflammation by suppressing microglial activation and reducing the accumulation of amyloid β-protein. Through transcriptome sequencing and WB verification, 34 differentially expressed genes (DEGs) were identified, including ACKR3, NR1H3 and Adra1a. Following treatment with a high dose of JJSYP, both ACKR3 and NR1H3 showed a significant decrease compared to the model group. Conversely, ADRA1A expression was reduced in model group compared to the control group, but increased following high dose JJSYP treatment. Research involving RNA sequencing and network pharmacology indicated that JJSYP altered the activation of CXCL12/ACKR3 signaling pathways in the hippocampus. CONCLUSIONS JJSYP exhibits potential anti-Alzheimer's Disease effects and warrants further investigation and development as a prosper treatment for AD.
Collapse
Affiliation(s)
- Yan Chen
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, No. 11 Lingjiaohu Road, Wuhan, Hubei, 430015, China.
| | - Yan Zhu
- Wuhan No.1 Hospital, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, Hubei, 430015, China
| | - Zihu Tan
- Department of Geriatrics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
| | - Xueyi Zhang
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, No. 11 Lingjiaohu Road, Wuhan, Hubei, 430015, China
| | - Jiafeng Hu
- Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Ruichi Zhu
- Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Jing Wang
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, No. 11 Lingjiaohu Road, Wuhan, Hubei, 430015, China
| | - Lizhu Chen
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, No. 11 Lingjiaohu Road, Wuhan, Hubei, 430015, China
| | - Zhenli Guo
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, No. 11 Lingjiaohu Road, Wuhan, Hubei, 430015, China.
| |
Collapse
|
7
|
Cheng WY, Lee XZ, Lai MSL, Ho YS, Chang RCC. PKR modulates sterile systemic inflammation-triggered neuroinflammation and brain glucose metabolism disturbances. Front Immunol 2025; 16:1469737. [PMID: 40070845 PMCID: PMC11893411 DOI: 10.3389/fimmu.2025.1469737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 02/06/2025] [Indexed: 03/14/2025] Open
Abstract
Sterile systemic inflammation may contribute to neuroinflammation and accelerate the progression of neurodegenerative diseases. The double-stranded RNA-dependent protein kinase (PKR) is a key signaling molecule that regulates immune responses by regulating macrophage activation, various inflammatory pathways, and inflammasome formation. This study aims to study the role of PKR in regulating sterile systemic inflammation-triggered neuroinflammation and cognitive dysfunctions. Here, the laparotomy mouse model was used to study neuroimmune responses triggered by sterile systemic inflammation. Our study revealed that genetic deletion of PKR in mice potently attenuated the laparotomy-induced peripheral and neural inflammation and cognitive deficits. Furthermore, intracerebroventricular injection of rAAV-DIO-PKR-K296R to inhibit PKR in cholinergic neurons of ChAT-IRES-Cre-eGFP mice rescued the laparotomy-induced changes in key metabolites of brain glucose metabolism, particularly the changes in phosphoenolpyruvate and succinate levels, and cognitive impairment in short-term and spatial working memory. Our results demonstrated the critical role of PKR in regulating neuroinflammation, brain glucose metabolism and cognitive dysfunctions in a peripheral inflammation model. PKR could be a novel pharmacological target for treating systemic inflammation-induced neuroinflammation and cognitive dysfunctions.
Collapse
Affiliation(s)
- Wai-Yin Cheng
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Xin-Zin Lee
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Michael Siu-Lun Lai
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yuen-Shan Ho
- School of Nursing, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Raymond Chuen-Chung Chang
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
8
|
Mielcarska MB, Rouse BT. Viruses and the Brain-A Relationship Prone to Trouble. Viruses 2025; 17:203. [PMID: 40006958 PMCID: PMC11860391 DOI: 10.3390/v17020203] [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: 01/06/2025] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Neurological disorders, some of which are associated with viral infections, are growing due to the aging and expanding population. Despite strong defenses of the central nervous system, some viruses have evolved ways to breach them, which often result in dire consequences. In this review, we recount the various ways by which different viruses can enter the CNS, and we describe the consequences of such invasions. Consequences may manifest as acute disease, such as encephalitis, meningitis, or result in long-term effects, such as neuromuscular dysfunction, as occurs in poliomyelitis. We discuss evidence for viral involvement in the causation of well-known chronic neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, as well as vascular dementia in the elderly. We also describe the approaches currently available to control a few of the neural viral infections. These include antivirals that are effective against human immunodeficiency virus and herpes simplex virus, as well as vaccines valuable for controlling rabies virus, poliomyelitis virus, and some flavivirus infections. There is an urgent need to better understand, at a molecular level, how viruses contribute to acute and, especially, chronic neurological diseases and to develop more precise and effective vaccines and therapies.
Collapse
Affiliation(s)
- Matylda Barbara Mielcarska
- Department of Preclinical Sciences, Institute of Veterinary Sciences, Warsaw University of Life Sciences–SGGW, Jana Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Barry T. Rouse
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
9
|
Fu Q, Qiu R, Liang J, Wu S, Huang D, Qin Y, Li Q, Shi X, Xiong X, Jiang Z, Chen Y, Cheng Y. Sugemule-7 alleviates oxidative stress, neuroinflammation, and cell death, promoting synaptic plasticity recovery in mice with postpartum depression. Sci Rep 2025; 15:1426. [PMID: 39789071 PMCID: PMC11718020 DOI: 10.1038/s41598-025-85276-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/01/2025] [Indexed: 01/12/2025] Open
Abstract
Postpartum depression (PPD) profoundly impacts the mental and physical health of women globally and is an incurable psychological disorder. Traditional pharmacological treatments often have strong side effects and may adversely affect infant health through breastfeeding, underscoring the critical need for natural and gentle treatment strategies. Sugemule-7, a traditional Chinese medicine comprising multiple natural plant ingredients, represents a potentially safer and more effective alternative. To investigate its preventive effects on PPD, we established an animal model and administered the drug Sugemule-7. Our study demonstrated that varying doses of Sugemule-7 effectively alleviated depressive and anxiety-like behaviors in PPD mice, as assessed through a battery of tests, including the open field test, tail suspension test, sucrose preference test, forced swim test, novelty-suppressed feeding test, and elevated plus maze test. Furthermore, Sugemule-7 significantly improved oxidative stress levels in the serum, prefrontal cortex, and hippocampus of PPD-induced mice while also suppressing inflammatory responses and abnormal neuronal death in these brain regions. Transcriptomic sequencing of hippocampal and prefrontal cortex tissues supported our findings, revealing that differential gene expression is primarily involved in regulating synaptic plasticity. Overall, our study confirms the efficacy of Sugemule-7 in treating PPD at different concentrations, potentially alleviating depressive behaviors by enhancing synaptic plasticity, mitigating oxidative stress, reducing inflammation, and protecting neurons.
Collapse
Affiliation(s)
- Qiang Fu
- Center on Translational Neuroscience, Institute of National Security, Minzu University of China, Beijing, China
- School of Ethnology and Sociology, Minzu University of China, Beijing, China
| | - Rui Qiu
- Center on Translational Neuroscience, Institute of National Security, Minzu University of China, Beijing, China
- School of Ethnology and Sociology, Minzu University of China, Beijing, China
| | - Jiaquan Liang
- Center on Translational Neuroscience, Institute of National Security, Minzu University of China, Beijing, China
| | - Shuai Wu
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Dezhi Huang
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Yuxiang Qin
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Qiaosheng Li
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Xiaojie Shi
- School of Pharmacy, Minzu University of China, Beijing, China
| | - Xiyue Xiong
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Zhongyong Jiang
- Department of Medical Laboratory, Affiliated Cancer Hospital of Chengdu Medical College, Chengdu Seventh People's Hospital, Chengdu, Sichuan, China
| | - Yuewen Chen
- Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Chinese Academy of Sciences, Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, Guangdong, China.
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, 518057, Guangdong, China.
- Xili Shenzhen University Town, No.1068 Xueyuan Avenue, Nanshan District, Shenzhen, 518055, China.
| | - Yong Cheng
- Center on Translational Neuroscience, Institute of National Security, Minzu University of China, Beijing, China.
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China.
- , 27th South Zhongguancun Avenue, Beijing, 100081, China.
| |
Collapse
|
10
|
Ghaith WZ, Wadie W, El-Yamany MF. Crosstalk between SIRT1/Nrf2 signaling and NLRP3 inflammasome/pyroptosis as a mechanistic approach for the neuroprotective effect of linagliptin in Parkinson's disease. Int Immunopharmacol 2025; 145:113716. [PMID: 39642562 DOI: 10.1016/j.intimp.2024.113716] [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: 08/31/2024] [Revised: 11/24/2024] [Accepted: 11/24/2024] [Indexed: 12/09/2024]
Abstract
In recent years, special attention has been paid to highlighting the antiparkinsonian effect of linagliptin. However, the mechanism of its action has not yet been well investigated. The present study aimed to verify the neuroprotective effect of linagliptin in the rotenone model of Parkinson's disease (PD) and further explore its potential molecular mechanisms. Rats were intoxicated with rotenone (2 mg/kg/day; sc) and treated with linagliptin (10 mg/kg/day; po) for 14 consecutive days. The present finding showed that linagliptin ameliorated the histopathological changes of rotenone on substantia nigra and striata. Linagliptin decreased α-synuclein immunoreactivity along with an increase in tyrosine hydroxylase immunoreactivity and striatal dopamine content. This was reflected in the marked improvement of the behavior and motor deficits in rotenone-intoxicated rats. On the molecular level, linagliptin upregulated sirtuin 1 (SIRT1)/ nuclear factor erythroid 2-related factor 2 (Nrf2) signaling, reduced ionized calcium-binding adaptor molecule 1 (Iba1) protein expression, restored glutathione (GSH) content, and elevated heme oxygenase-1 (HO-1) level in rats with rotenone intoxication. Moreover, linagliptin inhibited NOD-like receptor protein 3 (NLRP3)/caspase-1/interleukin-1β (IL-1β) cascade with subsequent reduction in gasdermin D (GSDMD) expression. Therefore, the present study reveals the ability of linagliptin, through the activation of SIRT1/Nrf2 signaling, to suppress NLRP3 inflammasome-mediated pyroptosis and protect against rotenone-induced parkinsonism.
Collapse
Affiliation(s)
| | - Walaa Wadie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Egypt
| | - Mohammed F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Egypt.
| |
Collapse
|
11
|
Kim M, Kang S, Oh S. The Anti-Inflammatory Activities of Benzylideneacetophenone Derivatives in LPS Stimulated BV2 Microglia Cells and Mice. Biomol Ther (Seoul) 2025; 33:106-116. [PMID: 39390761 PMCID: PMC11704402 DOI: 10.4062/biomolther.2024.049] [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: 03/22/2024] [Revised: 05/28/2024] [Accepted: 06/09/2024] [Indexed: 10/12/2024] Open
Abstract
A previously reported study highlighted the neuroprotective potential of the novel benzylideneacetophenone derivative, JC3, in mice. In pursuit of compounds with even more robust neuroprotective and anti-inflammatory properties compared to JC3, we synthesized substituted 1,3-diphenyl-2-propen-1-ones based on chalcones. Molecular modeling studies aimed at discerning the chemical structural features conducive to heightened biological activity revealed that JCII-8,10,11 exhibited the widest HOMOLUMO gap within this category, indicating facile electron and radical transfer between HOMO and LUMO in model assessments. From the pool of synthesized compounds, JCII-8,10,11 were selected for the present investigation. The biological assays involving JCII-8,10,11 demonstrated their concentration-dependent suppression of iNOS and COX-2 protein levels, alongside various cytokine mRNA expressions in LPS-induced murine microglial BV2 cells. Furthermore, western blot analyses were conducted to investigate the MAPK pathways and NF-κB/p65 nuclear translocation. These evaluations conclusively confirmed the inflammatory inhibition effects in both in vitro and in vivo inflammation models. These findings establish JCII-8,10,11 as potent anti-inflammatory agents, hindering inflammatory mediators and impeding NF-κB/p65 nuclear translocation via JNK and ERK MAPK phosphorylation in BV2 cells. The study positions them as potential therapeutics for inflammation-related conditions. Additionally, JCII-11 exhibited greater activity compared to other tested JCII compounds.
Collapse
Affiliation(s)
- Mijin Kim
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Seungmin Kang
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Seikwan Oh
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| |
Collapse
|
12
|
Askarizadeh A, Vahdat-Lasemi F, Karav S, Kesharwani P, Sahebkar A. Lipid nanoparticle-based delivery of small interfering RNAs: New possibilities in the treatment of diverse diseases. Eur Polym J 2025; 223:113624. [DOI: 10.1016/j.eurpolymj.2024.113624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
13
|
Paoletti I, Coccurello R. Irisin: A Multifaceted Hormone Bridging Exercise and Disease Pathophysiology. Int J Mol Sci 2024; 25:13480. [PMID: 39769243 PMCID: PMC11676223 DOI: 10.3390/ijms252413480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/03/2025] Open
Abstract
The fibronectin domain-containing protein 5 (FNDC5), or irisin, is an adipo-myokine hormone produced during exercise, which shows therapeutic potential for conditions like metabolic disorders, osteoporosis, sarcopenia, obesity, type 2 diabetes, and neurodegenerative diseases, including Alzheimer's disease (AD). This review explores its potential across various pathophysiological processes that are often considered independent. Elevated in healthy states but reduced in diseases, irisin improves muscle-adipose communication, insulin sensitivity, and metabolic balance by enhancing mitochondrial function and reducing oxidative stress. It promotes osteogenesis and mitigates bone loss in osteoporosis and sarcopenia. Irisin exhibits anti-inflammatory effects by inhibiting NF-κB signaling and countering insulin resistance. In the brain, it reduces amyloid-β toxicity, inflammation, and oxidative stress, enhancing brain-derived neurotrophic factor (BDNF) signaling, which improves cognition and synaptic health in AD models. It also regulates dopamine pathways, potentially alleviating neuropsychiatric symptoms like depression and apathy. By linking physical activity to systemic health, irisin emphasizes its role in the muscle-bone-brain axis. Its multifaceted benefits highlight its potential as a therapeutic target for AD and related disorders, with applications in prevention, in treatment, and as a complement to exercise strategies.
Collapse
Affiliation(s)
- Ilaria Paoletti
- IRCSS Santa Lucia Foundation, European Center for Brain Research, 00143 Rome, Italy;
| | - Roberto Coccurello
- IRCSS Santa Lucia Foundation, European Center for Brain Research, 00143 Rome, Italy;
- Institute for Complex Systems (ISC), National Research Council (C.N.R.), 00185 Rome, Italy
| |
Collapse
|
14
|
Roy P, Guo Y, Muzik O, Woodcock EA, Jiang H. Fully automated radiosynthesis of [ 18F]FCPPC for imaging microglia with PET. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2024; 14:351-356. [PMID: 39840376 PMCID: PMC11744358 DOI: 10.62347/qfgp5253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/09/2024] [Indexed: 01/23/2025]
Abstract
Colony-stimulating factor 1 receptor (CSF1R) is almost exclusively expressed on microglia in the human brain and thus, has promise as a biomarker for imaging microglia density as a proxy for neuroinflammation. [11C]CPPC is a radiotracer with selective affinity to CSF1R, and has been evaluated for in-human microglia PET imaging. The flourine-18 labeled CPPC derivative, 5-cyano-N-(4-(4-(2-[18F]fluoroethyl)piperazin-1-yl)-2-(piperidin-1-yl)phenyl)furan-2-carboxamide ([18F]FCPPC), was previously synthesized, however, with a low radiochemical yield using manual radiosynthesis. In this work, we report a fully automated radiosynthesis of [18F]FCPPC on a Synthra RNplus research module. In a total synthesis time of 50 min, [18F]FCPPC was obtained in decay corrected radiochemical yields of 26.8 ± 0.1% (n = 3) with >99% radiochemical purities. Quality control testing showed that [18F]FCPPC met all release criteria. In sum, we report the first fully automated radiosynthesis of [18F]FCPPC, a promising radiopharmaceutical for imaging microglia in humans.
Collapse
Affiliation(s)
- Pritam Roy
- Cyclotron and Radiochemistry Core, Karmanos Cancer InstituteDetroit, MI, USA
- PET Center, Karmanos Cancer InstituteDetroit, MI, USA
- Department of Oncology, Wayne State UniversityDetroit, MI, USA
| | - Yan Guo
- Cyclotron and Radiochemistry Core, Karmanos Cancer InstituteDetroit, MI, USA
- PET Center, Karmanos Cancer InstituteDetroit, MI, USA
- Department of Oncology, Wayne State UniversityDetroit, MI, USA
| | - Otto Muzik
- PET Center, Karmanos Cancer InstituteDetroit, MI, USA
- Department of Pediatrics and Neurology, Wayne State UniversityDetroit, MI, USA
| | - Eric A Woodcock
- Department of Psychiatry and Behavioral Neurosciences and Pharmacology, Wayne State University School of MedicineDetroit, MI, USA
| | - Huailei Jiang
- Cyclotron and Radiochemistry Core, Karmanos Cancer InstituteDetroit, MI, USA
- PET Center, Karmanos Cancer InstituteDetroit, MI, USA
- Department of Oncology, Wayne State UniversityDetroit, MI, USA
| |
Collapse
|
15
|
Yang Z, Chen J, Zhang C, Peng H. Pathological mechanisms of glial cell activation and neurodegenerative and neuropsychiatric disorders caused by Toxoplasma gondii infection. Front Microbiol 2024; 15:1512233. [PMID: 39723133 PMCID: PMC11668811 DOI: 10.3389/fmicb.2024.1512233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Toxoplasma gondii is an intracellular opportunistic parasite that exists in a latent form within the human central nervous system (CNS), even in immune-competent hosts. During acute infection, T. gondii traverses the blood-brain barrier (BBB). In the subsequent chronic infection phase, the infiltration of immune cells into the brain, driven by T. gondii infection and the formation of parasitic cysts, leads to persistent activation and proliferation of astrocytes and microglia. This process results in neuronal damages that are fatal in some cases. Through inducing systemic immune responses, T. gondii infection can dramatically alter the behavior of rodents and increase the risk of various neuropsychiatric disorders in humans. In this review, we explore some recent research progress on the major events involved in BBB disruption, glial cell activation and neuronal damage following T. gondii infection in hosts. It further discusses potential pathological mechanisms and the feasible treatment approaches for the neurodegenerative and neuropsychiatric disorders caused by T. gondii infection to extend our understanding for pathogenesis and preventive control of toxoplasmosis in humans.
Collapse
Affiliation(s)
| | | | | | - Hongjuan Peng
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Diseases Research, School of Public Health, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
16
|
Kim H, Le B, Goshi N, Zhu K, Grodzki AC, Lein PJ, Zhao M, Seker E. Primary cortical cell tri-culture to study effects of amyloid-β on microglia function and neuroinflammatory response. J Alzheimers Dis 2024; 102:730-741. [PMID: 39501607 PMCID: PMC11758989 DOI: 10.1177/13872877241291142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2024]
Abstract
BACKGROUND Microglia play a critical role in neurodegenerative disorders, such as Alzheimer's disease, where alterations in microglial function may result in pathogenic amyloid-β (Aβ) accumulation, chronic neuroinflammation, and deleterious effects on neuronal function. However, studying these complex factors in vivo, where numerous confounding processes exist, is challenging, and until recently, in vitro models have not allowed sustained culture of critical cell types in the same culture. OBJECTIVE We employed a rat primary tri-culture (neurons, astrocytes, and microglia) model and compared it to co-culture (neurons and astrocytes) and mono-culture (microglia) to study microglial function (i.e., motility and Aβ clearance) and proteomic response to exogenous Aβ. METHODS The cultures were exposed to fluorescently-labeled Aβ (FITC-Aβ) particles for varying durations. Epifluorescence microscopy images were analyzed to quantify the number of FITC-Aβ particles and assess cytomorphological features. Cytokine profiles from conditioned media were obtained. Live-cell imaging was employed to extract microglia motility parameters. RESULTS FITC-Aβ particles were more effectively cleared in the tri-culture compared to the co-culture. This was attributed to microglia engulfing FITC-Aβ particles, as confirmed via epifluorescence and confocal microscopy. FITC-Aβ treatment significantly increased microglia size, but had no significant effect on neuronal surface coverage or astrocyte size. Upon FITC-Aβ treatment, there was a significant increase in proinflammatory cytokines in tri-culture, but not in co-culture. Aβ treatment altered microglia motility evident as a swarming-like motion. CONCLUSIONS The results suggest that neuron-astrocyte-microglia interactions influence microglia function and highlight the utility of the tri-culture model for studies of neuroinflammation, neurodegeneration, and cell-cell communication.
Collapse
Affiliation(s)
- Hyehyun Kim
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, USA
| | - Bryan Le
- Department of Ophthalmology and Vision Science, University of California-Davis, Davis, CA, USA
| | - Noah Goshi
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, USA
| | - Kan Zhu
- Department of Ophthalmology and Vision Science, University of California-Davis, Davis, CA, USA
| | - Ana Cristina Grodzki
- Department of Molecular Biosciences, University of California-Davis, Davis, CA, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, University of California-Davis, Davis, CA, USA
| | - Min Zhao
- Department of Ophthalmology and Vision Science, University of California-Davis, Davis, CA, USA
| | - Erkin Seker
- Department of Electrical and Computer Engineering, University of California-Davis, Davis, CA, USA
| |
Collapse
|
17
|
Sian-Hulsmann J, Riederer P. Virus-induced brain pathology and the neuroinflammation-inflammation continuum: the neurochemists view. J Neural Transm (Vienna) 2024; 131:1429-1453. [PMID: 38261034 PMCID: PMC11608394 DOI: 10.1007/s00702-023-02723-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/18/2023] [Indexed: 01/24/2024]
Abstract
Fascinatingly, an abundance of recent studies has subscribed to the importance of cytotoxic immune mechanisms that appear to increase the risk/trigger for many progressive neurodegenerative disorders, including Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis, and multiple sclerosis. Events associated with the neuroinflammatory cascades, such as ageing, immunologic dysfunction, and eventually disruption of the blood-brain barrier and the "cytokine storm", appear to be orchestrated mainly through the activation of microglial cells and communication with the neurons. The inflammatory processes prompt cellular protein dyshomeostasis. Parkinson's and Alzheimer's disease share a common feature marked by characteristic pathological hallmarks of abnormal neuronal protein accumulation. These Lewy bodies contain misfolded α-synuclein aggregates in PD or in the case of AD, they are Aβ deposits and tau-containing neurofibrillary tangles. Subsequently, these abnormal protein aggregates further elicit neurotoxic processes and events which contribute to the onset of neurodegeneration and to its progression including aggravation of neuroinflammation. However, there is a caveat for exclusively linking neuroinflammation with neurodegeneration, since it's highly unlikely that immune dysregulation is the only factor that contributes to the manifestation of many of these neurodegenerative disorders. It is unquestionably a complex interaction with other factors such as genetics, age, and environment. This endorses the "multiple hit hypothesis". Consequently, if the host has a genetic susceptibility coupled to an age-related weakened immune system, this makes them more susceptible to the virus/bacteria-related infection. This may trigger the onset of chronic cytotoxic neuroinflammatory processes leading to protein dyshomeostasis and accumulation, and finally, these events lead to neuronal destruction. Here, we differentiate "neuroinflammation" and "inflammation" with regard to the involvement of the blood-brain barrier, which seems to be intact in the case of neuroinflammation but defect in the case of inflammation. There is a neuroinflammation-inflammation continuum with regard to virus-induced brain affection. Therefore, we propose a staging of this process, which might be further developed by adding blood- and CSF parameters, their stage-dependent composition and stage-dependent severeness grade. If so, this might be suitable to optimise therapeutic strategies to fight brain neuroinflammation in its beginning and avoid inflammation at all.
Collapse
Affiliation(s)
- Jeswinder Sian-Hulsmann
- Department of Human Anatomy and Medical Physiology, University of Nairobi, P.O. Box 30197, Nairobi, 00100, Kenya
| | - Peter Riederer
- University Hospital Wuerzburg, Clinic and Policlinic for Psychiatry, Psychosomatics and Psychotherapy Margarete-Höppel-Platz 1, 97080, Würzburg, Germany.
- Department of Psychiatry, University of Southern Denmark, Winslows Vey 18, 5000, Odense, J.B, Denmark.
| |
Collapse
|
18
|
Sobue A, Komine O, Endo F, Kakimi C, Miyoshi Y, Kawade N, Watanabe S, Saito Y, Murayama S, Saido TC, Saito T, Yamanaka K. Microglial cannabinoid receptor type II stimulation improves cognitive impairment and neuroinflammation in Alzheimer's disease mice by controlling astrocyte activation. Cell Death Dis 2024; 15:858. [PMID: 39587077 PMCID: PMC11589152 DOI: 10.1038/s41419-024-07249-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/27/2024]
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is characterized by the accumulation of amyloid β (Aβ) and phosphorylated tau. Neuroinflammation, mainly mediated by glial activation, plays an important role in AD progression. Although there is growing evidence for the anti-neuroinflammatory and neuroprotective effects of the cannabinoid system modulation, the detailed mechanism remains unclear. To address these issues, we analyzed the expression levels of cannabinoid receptor type II (Cnr2/Cb2) in AppNL-G-F/NL-G-F mice and human AD precuneus, which is vulnerable to amyloid deposition in AD, and the effects of JWH 133, a selective CB2 agonist, on neuroinflammation in primary glial cells and neuroinflammation and cognitive impairment in AppNL-G-F/NL-G-F mice. The levels of Cnr2/Cb2 were upregulated in microglia isolated from the cerebral cortex of AppNL-G-F/NL-G-F mice. CNR2 expression was also increased in RNAs derived from human precuneus with advanced AD pathology. Chronic oral administration of JWH 133 significantly ameliorated the cognitive impairment of AppNL-G-F/NL-G-F mice without neuropsychiatric side effects. Microglia and astrocyte mRNAs were directly isolated from the mouse cerebral cortex by magnetic-activated cell sorting, and the gene expression was determined by quantitative PCR. JWH 133 administration significantly decreased reactive astrocyte markers and microglial C1q, an inducer for the reactive astrocytes in AppNL-G-F/NL-G-F mice. In addition, JWH133 administration inhibited the expression of p-STAT3 (signal transducer and activator of transcription 3) in astrocytes in AppNL-G-F/NL-G-F mice. Furthermore, JWH 133 administration suppressed dystrophic presynaptic terminals surrounding amyloid plaques. In conclusion, stimulation of microglial CB2 ameliorates cognitive dysfunction in AppNL-G-F/NL-G-F mice by controlling astrocyte activation and inducing beneficial neuroinflammation, and our study has implications that CB2 may represent an attractive therapeutic target for the treatment of AD and perhaps other neurodegenerative diseases involving neuroinflammation.
Collapse
Affiliation(s)
- Akira Sobue
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Aichi, 464-8601, Japan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Aichi, 466-8550, Japan
- Medical Interactive Research and Academia Industry Collaboration Center, Research Institute of Environmental Medicine, Nagoya University, Aichi, 464-8601, Japan
| | - Okiru Komine
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Aichi, 464-8601, Japan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Aichi, 466-8550, Japan
| | - Fumito Endo
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Aichi, 464-8601, Japan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Aichi, 466-8550, Japan
| | - Chihiro Kakimi
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Aichi, 464-8601, Japan
| | - Yuka Miyoshi
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Aichi, 464-8601, Japan
| | - Noe Kawade
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Aichi, 464-8601, Japan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Aichi, 466-8550, Japan
| | - Seiji Watanabe
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Aichi, 464-8601, Japan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Aichi, 466-8550, Japan
| | - Yuko Saito
- Brain Bank for Aging Research (Neuropathology), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, 173-0015, Japan
| | - Shigeo Murayama
- Brain Bank for Aging Research (Neuropathology), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, 173-0015, Japan
- Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Osaka, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, 351-0198, Japan
| | - Takashi Saito
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Aichi, 464-8601, Japan
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Aichi, 467-8601, Japan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Aichi, 464-8601, Japan.
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Aichi, 466-8550, Japan.
- Institute for Glyco-core Research (iGCORE), Nagoya University, Aichi, Japan.
- Center for One Medicine Innovative Translational Research (COMIT), Nagoya University, Aichi, Japan.
- Research Institute for Quantum and Chemical Innovation, Institutes of Innovation for Future Society, Nagoya University, Aichi, Japan.
| |
Collapse
|
19
|
Li H, Zhao Y, Dai R, Geng P, Weng D, Wu W, Yu F, Lin R, Wu Z, Li Y, Luo M. Astrocytes release ATP/ADP and glutamate in flashes via vesicular exocytosis. Mol Psychiatry 2024:10.1038/s41380-024-02851-8. [PMID: 39578520 DOI: 10.1038/s41380-024-02851-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
Astrocytes regulate brain functions through gliotransmitters like ATP/ADP and glutamate, but their release patterns and mechanisms remain controversial. Here, we visualized ATP/ADP and glutamate response following astrocyte activation and investigated their mechanisms in vivo. Employing cOpn5-mediated optogenetic stimulation, genetically encoded fluorescent sensors, and two-photon imaging, we observed ATP/ADP released as temporally prolonged and spatially extended flashes that later converted to adenosine. This release occurs via Ca2+ and VNUT-dependent vesicular exocytosis. Additionally, astrocytes also release glutamate in flashes through TeNT-sensitive exocytosis, independent of ATP/ADP release. ATP/ADP released by astrocytes triggers further ATP/ADP release from microglia through P2Y12- and VNUT-dependent mechanisms. VNUT in astrocytes and microglia also contributes to ATP/ADP release under LPS-induced brain inflammation. These findings establish Ca2+-dependent vesicular exocytosis as a key mode of action, reveal intricate astrocyte-microglia interactions, and suggest a role for gliotransmission in brain inflammation. Furthermore, the methodologies may provide valuable tools for deciphering glial physiology and pathophysiology.
Collapse
Affiliation(s)
- Heng Li
- National Institute of Biological Sciences (NIBS), Beijing, 102206, Beijing, China
- School of Life Sciences, Tsinghua University, 100084, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), 102206, Beijing, China
| | - Yuqing Zhao
- National Institute of Biological Sciences (NIBS), Beijing, 102206, Beijing, China
| | - Ruicheng Dai
- Chinese Institute for Brain Research (CIBR), Beijing, 102206, Beijing, China
| | - Peiyao Geng
- National Institute of Biological Sciences (NIBS), Beijing, 102206, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), 102206, Beijing, China
| | - Danwei Weng
- National Institute of Biological Sciences (NIBS), Beijing, 102206, Beijing, China
| | - Wenting Wu
- National Institute of Biological Sciences (NIBS), Beijing, 102206, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), 102206, Beijing, China
| | - Fengting Yu
- Chinese Institute for Brain Research (CIBR), Beijing, 102206, Beijing, China
| | - Rui Lin
- National Institute of Biological Sciences (NIBS), Beijing, 102206, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), 102206, Beijing, China
| | - Zhaofa Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, 100871, Beijing, China
- New Cornerstone Science Laboratory, 518054, Shenzhen, China
| | - Minmin Luo
- Chinese Institute for Brain Research (CIBR), Beijing, 102206, Beijing, China.
- New Cornerstone Science Laboratory, 518054, Shenzhen, China.
- Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, 100005, Beijing, China.
- Beijing Institute for Brain Research, Chinese Academy of Medical Sciences & Peking Union Medical College, 102206, Beijing, China.
| |
Collapse
|
20
|
Gao J, Okolo O, Siedlak SL, Friedland RP, Wang X. Ferritin is closely associated with microglia in amyotrophic lateral sclerosis. J Neuropathol Exp Neurol 2024; 83:917-926. [PMID: 39001793 PMCID: PMC11487107 DOI: 10.1093/jnen/nlae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/15/2024] Open
Abstract
Iron deposition is a hallmark of amyotrophic lateral sclerosis (ALS) and has been strongly implicated in its pathogenesis. As a byproduct of cellular oxidative stress, iron dysregulation modifies basal levels of the regulatory iron-binding protein ferritin. Examination of thoracic and lumbar spinal cord tissues found increased ferritin immunostaining in white matter axons that corresponded to areas of increased microgliosis in 8 ALS patients versus 8 normal subjects. Gray matter areas containing the motor neurons also demonstrated increased ferritin and microglia in ALS compared to controls but at lower levels than in the white matter. Motor neurons with or without TDP-43 inclusions did not demonstrate either increased ferritin or associated microglial activation. We also observed an association of ferritin with microglia in cerebral cortical tissue samples of ALS cases and in the spinal cord tissues of transgenic mice expressing the SOD1G93A mutation. Elevated ferritin levels were detected in the insoluble fraction from spinal cord tissues of individuals with ALS. These findings suggest that activated microglia and increased ferritin may play significant roles in ALS progression since they are found closely associated in areas of axonal and cortical degeneration.
Collapse
Affiliation(s)
- Ju Gao
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, United States
| | - Ogoegbunam Okolo
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Sandra L Siedlak
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Robert P Friedland
- Department of Neurology, University of Louisville, Louisville, KY, United States
| | - Xinglong Wang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, United States
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
21
|
Kozina A, Herbert-Alonso G, Díaz A, Flores G, Guevara J. Effect of the aggregation state of amyloid-beta (25-35) on the brain oxidative stress in vivo. PLoS One 2024; 19:e0310258. [PMID: 39471144 PMCID: PMC11521274 DOI: 10.1371/journal.pone.0310258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/27/2024] [Indexed: 11/01/2024] Open
Abstract
Aggregation pathway of amyloid-β (25-35) in water affects the oxidative stress in the brain observed after administration of aggregated peptide in animals in vivo. Our studies on peptide aggregation ex situ prior to injection suggest that from the onset of peptide incubation in aqueous media, all samples exhibit the formation of fibril-like aggregates, characterized by a significant amount of β-sheets. This induces significant oxidative stress in vivo as observed for up to 60 min of peptide aggregation time. As the aggregation advances, the fibril-like aggregates become longer and intertwined, while the amount of β-sheets does not change significantly. An injection of such large, thick, and entangled aggregates in the animal brain results in a drastic increase in oxidative stress. This may be related to the number of activated microglia that initiate a sequence of inflammatory responses in the presence of large, highly interconnected fibrils.
Collapse
Affiliation(s)
- Anna Kozina
- Instituto de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Alfonso Díaz
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Gonzalo Flores
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Jorge Guevara
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
22
|
Herpich ME, de Oliveira Guarnieri L, de Oliveira ACP, Moraes MFD. Bacterial Lipopolysaccharide Post-Conditioning in The kainic acid animal model of Temporal Lobe epilepsy. Epilepsy Behav 2024; 161:110076. [PMID: 39467457 DOI: 10.1016/j.yebeh.2024.110076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/24/2024] [Accepted: 10/03/2024] [Indexed: 10/30/2024]
Abstract
This study used intra-hippocampal injections of Kainic Acid (KA) in Wistar rats to induce spontaneous recurrent seizures (SRS) after a 9-day latent period. A post-conditioning protocol with LPS, injected at the same site 72 h after the initial KA insult, was employed to trigger secondary competing processes. To evaluate the post-conditioning effect of LPS, 25 animals were divided into four groups: SAL-SAL (n = 6), KA-SAL (n = 6), SAL-LPS (n = 7), and KA-LPS (n = 6). SRS occurrence and seizure duration were quantified through video monitoring from days 9 to 17, along with other ictal behaviors, such as tail-chasing and wet-dog-shakes. Behavioral assessments revealed that the KA-LPS group had preserved sucrose preference and intact long-term memory in the object recognition test, indicating reduced depressive-like behavior and cognitive preservation compared to the KA-SAL group. The forced swim test showed increased depressive-like behavior in the SAL-LPS group, with LPS mitigating these effects in the KA group. The marble-burying test showed no significant differences among groups. Animals were euthanized on day 26, and hippocampal slices were analyzed using fluoro-jade staining for cell death and immunofluorescence staining for Iba-1 (microglia) and GFAP (astrocyte) labeling. The results support the hypothesis that epileptogenesis involves a cascade of plastic changes in neural networks and that precise, timely interventions can potentially interfere with this process.
Collapse
Affiliation(s)
- Mateus Eduardo Herpich
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas - Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leonardo de Oliveira Guarnieri
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas - Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Centro de Tecnologia e Pesquisa em Magneto Ressonância, Programa de Pós-Graduação em Engenharia Elétrica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Márcio Flávio Dutra Moraes
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas - Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Centro de Tecnologia e Pesquisa em Magneto Ressonância, Programa de Pós-Graduação em Engenharia Elétrica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
23
|
Chelucci E, Daniele S, Vergassola M, Ceccarelli L, Zucchi S, Boltri L, Martini C. Trazodone counteracts the response of microglial cells to inflammatory stimuli. Eur J Neurosci 2024; 60:5605-5620. [PMID: 39187397 DOI: 10.1111/ejn.16522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 07/29/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024]
Abstract
Microglia are resident brain cells that regulate neuronal development and innate immunity. Microglia activation participates in the cellular response to neuroinflammation, thus representing a possible target for pharmacological strategies aimed to counteract the onset and progression of brain disorders, including depression. Antidepressant drugs have been reported to reduce neuroinflammation by acting also on glial cells. Herein, the potential anti-inflammatory and neuroprotective effects of trazodone (TRZ) on the microglial human microglial clone 3 (HMC3) cell line were investigated. HMC3 cells were activated by a double inflammatory stimulus (lipopolysaccharide [LPS] and tumour necrosis factor-alpha [TNF-α], 24 h each), and the induction of inflammation was demonstrated by (i) the increased expression levels of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) and ionized calcium-binding adapter molecule 1 (IBA-1), and (ii) the increased release of interleukin 6 (IL-6) and transforming growth factor-beta (TGF-β). TRZ effects were evaluated by treating HMC3 cells for 24 h before (pre-treatment) and after (post-treatment) the double inflammatory stimulus. Notably, TRZ treatments significantly decreased the expression of NF-kB and IBA-1 and the release of the cytokines IL-6 and TGF-β. Moreover, TRZ prevented and reduced the release of quinolinic acid (QUIN), a known neurotoxic kynurenine metabolite. Finally, cellular supernatants collected from microglial cells pre-treated LPS-TNF-α with TRZ were able to improve neuronal-like cell viability, demonstrating a potential neuroprotective effect. Overall, this study suggests the anti-inflammatory effects of TRZ on human microglia and strives for its neuroprotective properties.
Collapse
Affiliation(s)
| | | | - Matteo Vergassola
- Angelini Pharma S.p.A. Global External Innovation & Drug Discovery, Translational Research Department, Rome, Italy
| | | | - Sara Zucchi
- Angelini Pharma S.p.A. Global R&D PLCM Preclinical Development, Ancona, Italy
| | - Luigi Boltri
- Angelini Pharma S.p.A. Global R&D PLCM Preclinical Development, Ancona, Italy
| | | |
Collapse
|
24
|
Ge TQ, Guan PP, Wang P. Complement 3a induces the synapse loss via C3aR in mitochondria-dependent NLRP3 activating mechanisms during the development and progression of Alzheimer's disease. Neurosci Biobehav Rev 2024; 165:105868. [PMID: 39218048 DOI: 10.1016/j.neubiorev.2024.105868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/08/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
As a central molecule in complement system (CS), complement (C) 3 is upregulated in the patients and animal models of Alzheimer's disease (AD). C3 will metabolize to iC3b and C3a. iC3b is responsible for clearing β-amyloid protein (Aβ). In this scenario, C3 exerts neuroprotective effects against the disease via iC3b. However, C3a will inhibit microglia to clear the Aβ, leading to the deposition of Aβ and impair the functions of synapses. To their effects on AD, activation of C3a and C3a receptor (C3aR) will impair the mitochondria, leading to the release of reactive oxygen species (ROS), which activates the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasomes. The overloading of NLRP3 inflammasomes activate microglia, leading to the formation of inflammatory environment. The inflammatory environment will facilitate the deposition of Aβ and abnormal synapse pruning, which results in the progression of AD. Therefore, the current review will decipher the mechanisms of C3a inducing the synapse loss via C3aR in mitochondria-dependent NLRP3 activating mechanisms, which facilitates the understanding the AD.
Collapse
Affiliation(s)
- Tong-Qi Ge
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, PR China; College of Life and Health Sciences, Northeastern University, Shenyang 110819, PR China
| | - Pei-Pei Guan
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, PR China.
| | - Pu Wang
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, PR China.
| |
Collapse
|
25
|
Poudel B, Bany BM, Hales DB, Cheatwood JL. Effects of Cannabidiol (CBD) on Doxorubicin-Induced Anxiety and Depression-like Behaviors and mRNA Expression of Inflammatory Markers in Rats. Brain Sci 2024; 14:999. [PMID: 39452013 PMCID: PMC11505750 DOI: 10.3390/brainsci14100999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
Background: Post-treatment side effects of chemotherapy can include cognitive deficits commonly known as Chemo-brain. The treatment of patients with Doxorubicin (DOX), one of the most widely used chemotherapeutic drugs in the treatment of cancer, can induce depression, anxiety, and impaired cognitive function. Cannabidiol (CBD) is a non-psychoactive component of Cannabis sativa that has been identified as a possible therapeutic agent against many neurodegenerative disorders, including traumatic brain injury, spinal cord injury, Tau-protein-induced neurodegeneration, and neuropathic pain. Therefore, this study aimed to assess whether oral CBD administration could reduce DOX-induced anxiety and depression-like behaviors and alter the expression of mRNA associated with neuroinflammation. Methods: Female Long Evans Hooded rats received intraperitoneal injections of DOX (6 mg/kg) or the vehicle (0.9% saline) once a week for four weeks, followed by oral administration of CBD (10 mg/kg) three times a week for the same period. Results: CBD was significantly protective against DOX-induced anxiety and depression-like behaviors, as measured by several behavioral tests. Furthermore, CBD improved DOX-induced alterations in the gene expression of biomarkers of neuroinflammation in the hippocampus and prefrontal cortex. Conclusions: This provides insights into future studies on possible mechanisms by which DOX-induced cognitive dysfunction could be alleviated by CBD.
Collapse
Affiliation(s)
| | | | | | - Joseph L. Cheatwood
- Department of Biomedical Sciences, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA; (B.P.)
| |
Collapse
|
26
|
Thant MT, Hasriadi H, Poldorn P, Jungsuttiwong S, Rojsitthisak P, Böttcher C, Towiwat P, Sritularak B. New phenanthrenequinones from Cymbidium ensifolium roots and their anti-inflammatory activity on lipopolysaccharide-activated BV2 microglial cells. RSC Adv 2024; 14:28390-28400. [PMID: 39239286 PMCID: PMC11375963 DOI: 10.1039/d4ra04761c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024] Open
Abstract
The roots of Cymbidium ensifolium yielded a total of 17 compounds, comprising two new compounds (1-2), one new natural product (3), and 14 known compounds (4-17). The structures of new compounds were determined through the analysis of their spectroscopic data, including NMR, MS, UV, FT-IR, optical rotation, and CD. The anti-inflammatory activity of the isolated pure compounds was assessed using lipopolysaccharide-activated BV2 microglial cells. Compounds 1, 3, 6, 12, 14, and 16 showed the ability to reduce LPS induced NO release in BV2 microglial cells, with IC50 values of 9.95 ± 2.13, 8.77 ± 3.78, 2.39 ± 0.91, 6.69 ± 2.94, 2.96 ± 1.38, 8.42 ± 2.99 μM, respectively and reduced the secretion of proinflammatory mediators (TNF-α, IL-6, MCP-1) in a concentration-dependent manner. Furthermore, the mechanistic role of the compound 3 was determined, which demonstrated its ability to inhibit the nuclear factor-κB (NF-κB) pathway through decreasing phosphorylation of p65 subunits.
Collapse
Affiliation(s)
- May Thazin Thant
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University Bangkok 10330 Thailand
| | - Hasriadi Hasriadi
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University Bangkok 10330 Thailand
| | | | - Siriporn Jungsuttiwong
- Department of Chemistry, Center of Excellence for Innovation in Chemistry, Faculty of Science, Ubon Ratchathani University Ubon Ratchathani 34190 Thailand
| | - Pornchai Rojsitthisak
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Chulalongkorn University Bangkok 10330 Thailand
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University Bangkok 10330 Thailand
| | - Chotima Böttcher
- Experimental and Clinical Research Center, A Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité - Universitätsmedizin Berlin Berlin 13125 Germany
| | - Pasarapa Towiwat
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University Bangkok 10330 Thailand
- Animal Models of Chronic Inflammation-associated Diseases for Drug Discovery Research Unit, Chulalongkorn University Bangkok 10330 Thailand
| | - Boonchoo Sritularak
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University Bangkok 10330 Thailand
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Chulalongkorn University Bangkok 10330 Thailand
| |
Collapse
|
27
|
Theophanous S, Sargiannidou I, Kleopa KA. Glial Cells as Key Regulators in Neuroinflammatory Mechanisms Associated with Multiple Sclerosis. Int J Mol Sci 2024; 25:9588. [PMID: 39273535 PMCID: PMC11395575 DOI: 10.3390/ijms25179588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Even though several highly effective treatments have been developed for multiple sclerosis (MS), the underlying pathological mechanisms and drivers of the disease have not been fully elucidated. In recent years, there has been a growing interest in studying neuroinflammation in the context of glial cell involvement as there is increasing evidence of their central role in disease progression. Although glial cell communication and proper function underlies brain homeostasis and maintenance, their multiple effects in an MS brain remain complex and controversial. In this review, we aim to provide an overview of the contribution of glial cells, oligodendrocytes, astrocytes, and microglia in the pathology of MS during both the activation and orchestration of inflammatory mechanisms, as well as of their synergistic effects during the repair and restoration of function. Additionally, we discuss how the understanding of glial cell involvement in MS may provide new therapeutic targets either to limit disease progression or to facilitate repair.
Collapse
Affiliation(s)
- Styliani Theophanous
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Irene Sargiannidou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Kleopas A Kleopa
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
- Center for Multiple Sclerosis and Related Disorders, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| |
Collapse
|
28
|
Yin Y, Ju T, Zeng D, Duan F, Zhu Y, Liu J, Li Y, Lu W. "Inflamed" depression: A review of the interactions between depression and inflammation and current anti-inflammatory strategies for depression. Pharmacol Res 2024; 207:107322. [PMID: 39038630 DOI: 10.1016/j.phrs.2024.107322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/13/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Depression is a common mental disorder, the effective treatment of which remains a challenging issue worldwide. The clinical pathogenesis of depression has been deeply explored, leading to the formulation of various pathogenic hypotheses. Among these, the monoamine neurotransmitter hypothesis holds a prominent position, yet it has significant limitations as more than one-third of patients do not respond to conventional treatments targeting monoamine transmission disturbances. Over the past few decades, a growing body of research has highlighted the link between inflammation and depression as a potential key factor in the pathophysiology of depression. In this review, we first summarize the relationship between inflammation and depression, with a focus on the pathophysiological changes mediated by inflammation in depression. The mechanisms linking inflammation to depression as well as multiple anti-inflammatory strategies are also discussed, and their efficacy and safety are assessed. This review broadens the perspective on specific aspects of using anti-inflammatory strategies for treating depression, laying the groundwork for advancing precision medicine for individuals suffering from "inflamed" depression.
Collapse
Affiliation(s)
- Yishu Yin
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Ting Ju
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Deyong Zeng
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Fangyuan Duan
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Yuanbing Zhu
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Junlian Liu
- China Astronaut Research and Training Center, Beijing 100094, China
| | - Yongzhi Li
- China Astronaut Research and Training Center, Beijing 100094, China.
| | - Weihong Lu
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China.
| |
Collapse
|
29
|
Kim K, Cho HR, Kim BY, Kim J, Park D, Kwon RJ, Son Y. Oxysterol Induces Expression of 60 kDa Chaperone Protein on Cell Surface of Microglia. Int J Mol Sci 2024; 25:9073. [PMID: 39201760 PMCID: PMC11354638 DOI: 10.3390/ijms25169073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
Microglia, essential immune cells in the brain, play crucial roles in neuroinflammation by performing various functions such as neurogenesis, synaptic pruning, and pathogen defense. These cells are activated by inflammatory factors like β-amyloid (Aβ) and oxysterols, leading to morphological and functional changes, including the secretion of inflammatory cytokines and the upregulation of MHC class II molecules. This study focused on identifying specific markers for microglial activation, with a particular emphasis on the roles of oxysterols in this process. We used the HMC3 human microglial cell line to investigate the induction of heat shock protein 60 (HSP60), a chaperonin protein by oxysterols, specifically in the presence of 25-hydroxycholesterol (25OHChol) and 27-hydroxycholesterol (27OHChol). Our findings obtained by the proteomics approach revealed that these oxysterols significantly increased HSP60 expression on microglial cells. This induction was further confirmed using Western blot analysis and immunofluorescence microscopy. Additionally, Aβ1-42 also promoted HSP60 expression, indicating its role as a microglial activator. HSP60 involved in protein folding and immune modulation was identified as a potential marker for microglial activation. This study underscores the importance of HSP60 in the inflammatory response of microglia, suggesting its utility as a target for new therapeutic approaches in neuroinflammatory diseases such as Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (K.K.); (J.K.); (D.P.)
| | - Hyok-rae Cho
- Department of Neurosurgery, College of Medicine, Kosin University, Busan 49267, Republic of Korea;
| | - Bo-young Kim
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea;
| | - Jaesung Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (K.K.); (J.K.); (D.P.)
| | - Dongha Park
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (K.K.); (J.K.); (D.P.)
| | - Ryuk Jun Kwon
- Family Medicine Clinic and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Department of Family Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Yonghae Son
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (K.K.); (J.K.); (D.P.)
| |
Collapse
|
30
|
Saadh MJ, Muhammad FA, Singh A, Mustafa MA, Al Zuhairi RAH, Ghildiyal P, Hashim G, Alsaikhan F, Khalilollah S, Akhavan-Sigari R. MicroRNAs Modulating Neuroinflammation in Parkinson's disease. Inflammation 2024:10.1007/s10753-024-02125-z. [PMID: 39162871 DOI: 10.1007/s10753-024-02125-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/20/2024] [Accepted: 08/06/2024] [Indexed: 08/21/2024]
Abstract
Parkinson's disease (PD) is one of the most frequent age-associated neurodegenerative disorder. Presence of α-synuclein-containing aggregates in the substantia nigra pars compacta (SNpc) and loss of dopaminergic (DA) neurons are among the characteristic of PD. One of the hallmarks of PD pathophysiology is chronic neuroinflammation. Activation of glial cells and elevated levels of pro-inflammatory factors are confirmed as frequent features of the PD brain. Chronic secretion of pro-inflammatory cytokines by activated astrocytes and microglia exacerbates DA neuron degeneration in the SNpc. MicroRNAs (miRNAs) are among endogenous non-coding small RNA with the ability to perform post-transcriptional regulation in target genes. In that regard, the capability of miRNAs for modulating inflammatory signaling is the center of attention in many investigations. MiRNAs could enhance or limit inflammatory signaling, exacerbating or ameliorating the pathological consequences of extreme neuroinflammation. This review summarizes the importance of inflammation in the pathophysiology of PD. Besides, we discuss the role of miRNAs in promoting or protecting neural cell injury in the PD model by controlling the inflammatory pathway. Modifying the neuroinflammation by miRNAs could be considered a primary therapeutic strategy for PD.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | - Anamika Singh
- Department of Biotechnology and Genetics, Jain (Deemed-to-Be) University, Bengaluru, Karnataka, 560069, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Mohammed Ahmed Mustafa
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh, 247341, India
- Department of Pharmacy, Arka Jain University, Jamshedpur,, Jamshedpur,, India, Jharkhand, 831001
| | | | - Pallavi Ghildiyal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ghassan Hashim
- Department of Nursing, Al-Zahrawi University College, Karbala, Iraq
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
- School of Pharmacy, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia.
| | - Shayan Khalilollah
- Department of Neurosurgery, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center, Tuebingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Warszawa, Poland
| |
Collapse
|
31
|
Bian Y, Qiao N, Han S, Gao J, Lv X, Yuan L, Zhang L, Wei Z. Anti-Neuroinflammatory Effect of Ombuin from Rhamnus erythroxylon Pall. Leaves in LPS-Induced BV-2 Microglia by Targeting Src and Suppressing the PI3K-AKT/NF-κB Signaling Pathway. Int J Mol Sci 2024; 25:8789. [PMID: 39201475 PMCID: PMC11354356 DOI: 10.3390/ijms25168789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
The leaves of Rhamnus erythroxylon Pall. are widely used as tea substitutes in northwest China for their fragrant aroma, anti-irritability, and digestion-enhancing properties. Ombuin, a main flavonoid compound found in the leaves, exhibited notable anti-inflammatory and antioxidant effects. However, its potential role in treating neuroinflammatory-related diseases remains unexplored. Thus, this study aims to evaluate the anti-neuroinflammatory effects of ombuin and to explore the underlying molecular mechanisms. According to our findings, ombuin dramatically reduced the release of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), IL-1β, nitric oxide (NO), and reactive oxygen species (ROS) in lipopolysaccharide (LPS)-stimulated BV-2 microglia. Further analysis, including transcriptomics, network pharmacology, molecular docking, and cellular heat transfer assays, revealed that Src was a direct target of ombuin. Western blot analysis showed that ombuin effectively suppressed Src phosphorylation and inhibited the downstream expressions of p-PI3K p85, p-AKT1, p-IKKα/β, p-IκBα, and nuclear factor κB (NF-κB). Meanwhile, the repression of Src significantly reversed the anti-neuroinflammatory activity of ombuin. Our results identified Src as a direct target of ombuin and implied that ombuin exerted an anti-neuroinflammatory effect by inhibiting Src phosphorylation and suppressing the activation of the PI3K-AKT and NF-κB pathways, which might provide an alternative therapeutic strategy for neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Linjing Zhang
- School of Life Science, Shanxi Normal University, Taiyuan 030006, China
| | - Zuofu Wei
- School of Life Science, Shanxi Normal University, Taiyuan 030006, China
| |
Collapse
|
32
|
Gabele L, Bochow I, Rieke N, Sieben C, Michaelsen-Preusse K, Hosseini S, Korte M. H7N7 viral infection elicits pronounced, sex-specific neuroinflammatory responses in vitro. Front Cell Neurosci 2024; 18:1444876. [PMID: 39171200 PMCID: PMC11335524 DOI: 10.3389/fncel.2024.1444876] [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: 06/06/2024] [Accepted: 07/18/2024] [Indexed: 08/23/2024] Open
Abstract
Influenza A virus (IAV) infection can increase the risk of neuroinflammation, and subsequent neurodegenerative diseases. Certain IAV strains, such as avian H7N7 subtype, possess neurotropic properties, enabling them to directly invade the brain parenchyma and infect neurons and glia cells. Host sex significantly influences the severity of IAV infections. Studies indicate that females of the reproductive age exhibit stronger innate and adaptive immune responses to IAVs compared to males. This heightened immune response correlates with increased morbidity and mortality, and potential neuronal damage in females. Understanding the sex-specific neurotropism of IAV and associated mechanisms leading to adverse neurological outcomes is essential. Our study reveals that primary hippocampal cultures from female mice show heightened interferon-β and pro-inflammatory chemokine secretion following neurotropic IAV infection. We observed sex-specific differences in microglia activation: both sexes showed a transition into a hyper-ramified state, but only male-derived microglia exhibited an increase in amoeboid-shaped cells. These disparities extended to alterations in neuronal morphology. Neurons derived from female mice displayed increased spine density within 24 h post-infection, while no significant change was observed in male cultures. This aligns with sex-specific differences in microglial synaptic pruning. Data suggest that amoeboid-shaped microglia preferentially target postsynaptic terminals, potentially reducing neuronal hyperexcitability. Conversely, hyper-ramified microglia may focus on presynaptic terminals, potentially limiting viral spread. In conclusion, our findings underscore the utility of primary hippocampal cultures, incorporating microglia, as an effective model to study sex-specific, virus-induced effects on brain-resident cells.
Collapse
Affiliation(s)
- Lea Gabele
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
- Helmholtz Centre for Infection Research, Research Group Neuroinflammation and Neurodegeneration, Braunschweig, Germany
| | - Isabell Bochow
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Nele Rieke
- Helmholtz Centre for Infection Research, Nanoscale Infection Biology Group, Braunschweig, Germany
| | - Christian Sieben
- Helmholtz Centre for Infection Research, Nanoscale Infection Biology Group, Braunschweig, Germany
- Institute of Genetics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Kristin Michaelsen-Preusse
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Shirin Hosseini
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
- Helmholtz Centre for Infection Research, Research Group Neuroinflammation and Neurodegeneration, Braunschweig, Germany
| | - Martin Korte
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
- Helmholtz Centre for Infection Research, Research Group Neuroinflammation and Neurodegeneration, Braunschweig, Germany
| |
Collapse
|
33
|
Hoyer-Kimura C, Hay M, Konhilas JP, Morrison HW, Methajit M, Strom J, Polt R, Salcedo V, Fricks JP, Kalya A, Pires PW. PNA5, A Novel Mas Receptor Agonist, Improves Neurovascular and Blood-Brain-Barrier Function in a Mouse Model of Vascular Cognitive Impairment and Dementia. Aging Dis 2024; 15:1927-1951. [PMID: 37815905 PMCID: PMC11272189 DOI: 10.14336/ad.2023.0928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 09/28/2023] [Indexed: 10/12/2023] Open
Abstract
It is well established that decreased brain blood flow, increased reactive oxygen species production (ROS), and pro-inflammatory mechanisms accelerate neurodegenerative disease progressions, including vascular cognitive impairment and dementia (VCID). Previous studies in our laboratory have shown that our novel glycosylated Angiotensin-(1-7) Mas receptor agonist PNA5 reverses cognitive deficits, decreases ROS production, and inhibits inflammatory cytokine production in our preclinical mouse model of VCID that is induced by chronic heart failure (VCID-HF). In the present study, the effects of VCID-HF and treatment with PNA5 on microglia activation, blood-brain-barrier (BBB) integrity, and neurovascular coupling were assessed in our mouse model of VCID-HF. Three-month-old male C57BL/6J mice were subjected to myocardial infarction (MI) to induce heart failure for four weeks and then treated with subcutaneous injections of extended-release PNA5. Microglia activation, BBB permeability, cerebral perfusion, and neurovascular coupling were assessed. Results show that in our VCID-HF model, there was an increase in microglial activation and recruitment within the CA1 and CA3 regions of the hippocampus, a disruption in BBB integrity, and a decrease in neurovascular coupling. Treatment with PNA5 reversed these neuropathological effects of VCID-HF, suggesting that PNA5 may be an effective disease-modifying therapy to treat and prevent VCID. This study identifies potential mechanisms by which heart failure may induce VCID and highlights the possible mechanisms by which treatment with our novel glycosylated Angiotensin-(1-7) Mas receptor agonist, PNA5, may protect cognitive function in our model of VCID.
Collapse
Affiliation(s)
| | - Meredith Hay
- Department of Physiology, The University of Arizona, Tucson, AZ 85724, USA.
- Evelyn F. McKnight Brain Institute, The University of Arizona, Tucson, AZ 85724, USA.
- ProNeurogen, Inc, Tucson, AZ, USA
| | - John P Konhilas
- Department of Physiology, The University of Arizona, Tucson, AZ 85724, USA.
| | - Helena W Morrison
- College of Nursing, The University of Arizona, Tucson, AZ 85724, USA.
| | - Methawasin Methajit
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85724, USA.
| | - Joshua Strom
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85724, USA.
| | - Robin Polt
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ 85724, USA.
| | - Victoria Salcedo
- Department of Physiology, The University of Arizona, Tucson, AZ 85724, USA.
| | | | - Anjna Kalya
- Department of Physiology, The University of Arizona, Tucson, AZ 85724, USA.
| | - Paulo W Pires
- Department of Physiology, The University of Arizona, Tucson, AZ 85724, USA.
| |
Collapse
|
34
|
Alonso M, Petit AC, Lledo PM. The impact of adult neurogenesis on affective functions: of mice and men. Mol Psychiatry 2024; 29:2527-2542. [PMID: 38499657 DOI: 10.1038/s41380-024-02504-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/20/2024]
Abstract
In most mammals, new neurons are not only produced during embryogenesis but also after birth. Soon after adult neurogenesis was discovered, the influence of recruiting new neurons on cognitive functions, especially on memory, was documented. Likewise, the late process of neuronal production also contributes to affective functions, but this outcome was recognized with more difficulty. This review covers hypes and hopes of discovering the influence of newly-generated neurons on brain circuits devoted to affective functions. If the possibility of integrating new neurons into the adult brain is a commonly accepted faculty in the realm of mammals, the reluctance is strong when it comes to translating this concept to humans. Compiling data suggest now that new neurons are derived not only from stem cells, but also from a population of neuroblasts displaying a protracted maturation and ready to be engaged in adult brain circuits, under specific signals. Here, we discuss the significance of recruiting new neurons in the adult brain circuits, specifically in the context of affective outcomes. We also discuss the fact that adult neurogenesis could be the ultimate cellular process that integrates elements from both the internal and external environment to adjust brain functions. While we must be critical and beware of the unreal promises that Science could generate sometimes, it is important to continue exploring the potential of neural recruitment in adult primates. Reporting adult neurogenesis in humankind contributes to a new vision of humans as mammals whose brain continues to develop throughout life. This peculiar faculty could one day become the target of treatment for mental health, cognitive disorders, and elderly-associated diseases. The vision of an adult brain which never stops integrating new neurons is a real game changer for designing new therapeutic interventions to treat mental disorders associated with substantial morbidity, mortality, and social costs.
Collapse
Affiliation(s)
- Mariana Alonso
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Perception and Action Unit, F-75015, Paris, France
| | - Anne-Cécile Petit
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Perception and Action Unit, F-75015, Paris, France
- Pôle Hospitalo-Universitaire Psychiatrie Paris 15, GHU Paris Psychiatry and Neurosciences, Hôpital Sainte-Anne, Paris, France
| | - Pierre-Marie Lledo
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Perception and Action Unit, F-75015, Paris, France.
| |
Collapse
|
35
|
Li HL, Shao LH, Chen X, Wang M, Qin QJ, Yang YL, Zhang GR, Hai Y, Tian YH. Anti-inflammatory and DNA Repair Effects of Astragaloside IV on PC12 Cells Damaged by Lipopolysaccharide. Curr Med Sci 2024; 44:854-863. [PMID: 39112916 DOI: 10.1007/s11596-024-2912-0] [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: 07/13/2023] [Accepted: 06/06/2024] [Indexed: 08/23/2024]
Abstract
OBJECTIVE This study aimed to establish a neural cell injury model in vitro by stimulating PC12 cells with lipopolysaccharide (LPS) and to examine the effects of astragaloside IV on key targets using high-throughput sequence technology and bioinformatics analyses. METHODS PC12 cells in the logarithmic growth phase were treated with LPS at final concentrations of 0.25, 0.5, 0.75, 1, and 1.25 mg/mL for 24 h. Cell morphology was evaluated, and cell survival rates were calculated. A neurocyte inflammatory model was established with LPS treatment, which reached a 50% cell survival rate. PC12 cells were treated with 0.01, 0.1, 1, 10, or 100 µmol/L astragaloside IV for 24 h. The concentration of astragaloside IV that did not affect the cell survival rate was selected as the treatment group for subsequent experiments. NOS activity was detected by colorimetry; the expression levels of ERCC2, XRCC4, XRCC2, TNF-α, IL-1β, TLR4, NOS and COX-2 mRNA and protein were detected by RT-qPCR and Western blotting. The differentially expressed genes (DEGs) between the groups were screened using a second-generation sequence (fold change>2, P<0.05) with the following KEGG enrichment analysis, RT-qPCR and Western blotting were used to detect the mRNA and protein expression of DEGs related to the IL-17 pathway in different groups of PC12 cells. RESULTS The viability of PC12 cells was not altered by treatment with 0.01, 0.1, or 1 µmol/L astragaloside IV for 24 h (P>0.05). However, after treatment with 0.5, 0.75, 1, or 1.25 mg/mL LPS for 24 h, the viability steadily decreased (P<0.01). The mRNA and protein expression levels of ERCC2, XRCC4, XRCC2, TNF-α, IL-1β, TLR4, NOS, and COX-2 were significantly increased after PC12 cells were treated with 1 mg/mL LPS for 24 h (P<0.01); however, these changes were reversed when PC12 cells were pretreated with 0.01, 0.1, or 1 µmol/L astragaloside IV in PC12 cells and then treated with 1 mg/mL LPS for 24 h (P<0.05). Second-generation sequencing revealed that 1026 genes were upregulated, while 1287 genes were downregulated. The DEGs were associated with autophagy, TNF-α, interleukin-17, MAPK, P53, Toll-like receptor, and NOD-like receptor signaling pathways. Furthermore, PC12 cells treated with a 1 mg/mL LPS for 24 h exhibited increased mRNA and protein expression of CCL2, CCL11, CCL7, MMP3, and MMP10, which are associated with the IL-17 pathway. RT-qPCR and Western blotting analyses confirmed that the DEGs listed above corresponded to the sequence assay results. CONCLUSION LPS can damage PC12 cells and cause inflammatory reactions in nerve cells and DNA damage. astragaloside IV plays an anti-inflammatory and DNA damage protective role and inhibits the IL-17 signaling pathway to exert a neuroprotective effect in vitro.
Collapse
Affiliation(s)
- Hai-Long Li
- Department of Geriatrics, The Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, China
- Key Laboratory for Mining, Innovation and Transformation of Traditional Chinese Medicine in Gansu Province and the New Product Creation Engineering Laboratory of Traditional Chinese Medicine in Gansu Province, Lanzhou, 730000, China
| | - Li-Hua Shao
- Key Laboratory for Mining, Innovation and Transformation of Traditional Chinese Medicine in Gansu Province and the New Product Creation Engineering Laboratory of Traditional Chinese Medicine in Gansu Province, Lanzhou, 730000, China
| | - Xi Chen
- Key Laboratory for Mining, Innovation and Transformation of Traditional Chinese Medicine in Gansu Province and the New Product Creation Engineering Laboratory of Traditional Chinese Medicine in Gansu Province, Lanzhou, 730000, China
| | - Meng Wang
- Key Laboratory for Mining, Innovation and Transformation of Traditional Chinese Medicine in Gansu Province and the New Product Creation Engineering Laboratory of Traditional Chinese Medicine in Gansu Province, Lanzhou, 730000, China
| | - Qi-Jie Qin
- Department of Neurology, the First People's Hospital of Lanzhou, Lanzhou, Gansu, 730000, China
| | - Ya-Li Yang
- Key Laboratory for Mining, Innovation and Transformation of Traditional Chinese Medicine in Gansu Province and the New Product Creation Engineering Laboratory of Traditional Chinese Medicine in Gansu Province, Lanzhou, 730000, China
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | | | - Yang Hai
- Scientific Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| | - Yi-Hong Tian
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
- Scientific Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| |
Collapse
|
36
|
Lista S, Imbimbo BP, Grasso M, Fidilio A, Emanuele E, Minoretti P, López-Ortiz S, Martín-Hernández J, Gabelle A, Caruso G, Malaguti M, Melchiorri D, Santos-Lozano A, Imbimbo C, Heneka MT, Caraci F. Tracking neuroinflammatory biomarkers in Alzheimer's disease: a strategy for individualized therapeutic approaches? J Neuroinflammation 2024; 21:187. [PMID: 39080712 PMCID: PMC11289964 DOI: 10.1186/s12974-024-03163-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Recent trials of anti-amyloid-β (Aβ) monoclonal antibodies, including lecanemab and donanemab, in early Alzheimer disease (AD) showed that these drugs have limited clinical benefits and their use comes with a significant risk of serious adverse events. Thus, it seems crucial to explore complementary therapeutic approaches. Genome-wide association studies identified robust associations between AD and several AD risk genes related to immune response, including but not restricted to CD33 and TREM2. Here, we critically reviewed the current knowledge on candidate neuroinflammatory biomarkers and their role in characterizing the pathophysiology of AD. MAIN BODY Neuroinflammation is recognized to be a crucial and contributing component of AD pathogenesis. The fact that neuroinflammation is most likely present from earliest pre-stages of AD and co-occurs with the deposition of Aβ reinforces the need to precisely define the sequence and nature of neuroinflammatory events. Numerous clinical trials involving anti-inflammatory drugs previously yielded unfavorable outcomes in early and mild-to-moderate AD. Although the reasons behind these failures remain unclear, these may include the time and the target selected for intervention. Indeed, in our review, we observed a stage-dependent neuroinflammatory process in the AD brain. While the initial activation of glial cells counteracts early brain Aβ deposition, the downregulation in the functional state of microglia occurs at more advanced disease stages. To address this issue, personalized neuroinflammatory modulation therapy is required. The emergence of reliable blood-based neuroinflammatory biomarkers, particularly glial fibrillary acidic protein, a marker of reactive astrocytes, may facilitate the classification of AD patients based on the ATI(N) biomarker framework. This expands upon the traditional classification of Aβ ("A"), tau ("T"), and neurodegeneration ("N"), by incorporating a novel inflammatory component ("I"). CONCLUSIONS The present review outlines the current knowledge on potential neuroinflammatory biomarkers and, importantly, emphasizes the role of longitudinal analyses, which are needed to accurately monitor the dynamics of cerebral inflammation. Such a precise information on time and place will be required before anti-inflammatory therapeutic interventions can be considered for clinical evaluation. We propose that an effective anti-neuroinflammatory therapy should specifically target microglia and astrocytes, while considering the individual ATI(N) status of patients.
Collapse
Affiliation(s)
- Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012, Valladolid, Spain.
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, 43122, Parma, Italy
| | | | | | | | | | - Susana López-Ortiz
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012, Valladolid, Spain
| | - Juan Martín-Hernández
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012, Valladolid, Spain
| | - Audrey Gabelle
- CMRR, Memory Resources and Research Center, Montpellier University of Excellence i-site, 34295, Montpellier, France
| | - Giuseppe Caruso
- Oasi Research Institute-IRCCS, 94018, Troina, Italy
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
| | - Marco Malaguti
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, 40126, Bologna, Italy
| | - Daniela Melchiorri
- Department of Physiology and Pharmacology, Sapienza University, 00185, Rome, Italy
| | - Alejandro Santos-Lozano
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012, Valladolid, Spain
- Physical Activity and Health Research Group (PaHerg), Research Institute of the Hospital, 12 de Octubre ('imas12'), 28041, Madrid, Spain
| | - Camillo Imbimbo
- Department of Brain and Behavioral Sciences, University of Pavia, 27100, Pavia, Italy
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4367, Esch-Belval, Luxembourg.
| | - Filippo Caraci
- Oasi Research Institute-IRCCS, 94018, Troina, Italy.
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy.
| |
Collapse
|
37
|
Wang Z, Liu J, Han J, Zhang T, Li S, Hou Y, Su H, Han F, Zhang C. Herpes simplex virus 1 accelerates the progression of Alzheimer's disease by modulating microglial phagocytosis and activating NLRP3 pathway. J Neuroinflammation 2024; 21:176. [PMID: 39026249 PMCID: PMC11264637 DOI: 10.1186/s12974-024-03166-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/03/2024] [Indexed: 07/20/2024] Open
Abstract
Accumulating evidence implicates that herpes simplex virus type 1 (HSV-1) has been linked to the development and progression of Alzheimer's disease (AD). HSV-1 infection induces β-amyloid (Aβ) deposition in vitro and in vivo, but the effect and precise mechanism remain elusive. Here, we show that HSV-1 infection of the brains of transgenic 5xFAD mice resulted in accelerated Aβ deposition, gliosis, and cognitive dysfunction. We demonstrate that HSV-1 infection induced the recruitment of microglia to the viral core to trigger microglial phagocytosis of HSV-GFP-positive neuronal cells. In addition, we reveal that the NLRP3 inflammasome pathway induced by HSV-1 infection played a crucial role in Aβ deposition and the progression of AD caused by HSV-1 infection. Blockade of the NLRP3 inflammasome signaling reduces Aβ deposition and alleviates cognitive decline in 5xFAD mice after HSV-1 infection. Our findings support the notion that HSV-1 infection is a key factor in the etiology of AD, demonstrating that NLRP3 inflammasome activation functions in the interface of HSV-1 infection and Aβ deposition in AD.
Collapse
Affiliation(s)
- Zhimeng Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center of Biological Structure, SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Tsinghua University, Beijing, 100084, China
| | - Jing Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jing Han
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center of Biological Structure, SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Tsinghua University, Beijing, 100084, China
| | - Tianyi Zhang
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Shangjin Li
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yanfei Hou
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Huili Su
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Fangping Han
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Conggang Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center of Biological Structure, SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
38
|
Leterrier S, Goutal S, Hugon G, Goislard M, Saba W, Hosten B, Specklin S, Winkeler A, Tournier N. Imaging quantitative changes in blood-brain barrier permeability using [ 18F]2-fluoro-2-deoxy-sorbitol ([ 18F]FDS) PET in relation to glial cell recruitment in a mouse model of endotoxemia. J Cereb Blood Flow Metab 2024; 44:1117-1127. [PMID: 38441006 PMCID: PMC11179610 DOI: 10.1177/0271678x241236755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 03/06/2024]
Abstract
The quantitative relationship between the disruption of the blood-brain barrier (BBB) and the recruitment of glial cells was explored in a mouse model of endotoxemia. [18F]2-Fluoro-2-deoxy-sorbitol ([18F]FDS) PET imaging was used as a paracellular marker for quantitative monitoring of BBB permeability after i.v injection of increasing doses of lipopolysaccharide (LPS) or vehicle (saline, n = 5). The brain distribution of [18F]FDS (VT, mL.cm-3) was estimated using kinetic modeling. LPS dose-dependently increased the brain VT of [18F]FDS after injection of LPS 4 mg/kg (5.2 ± 2.4-fold, n = 4, p < 0.01) or 5 mg/kg (9.0 ± 9.1-fold, n = 4, p < 0.01) but not 3 mg/kg (p > 0.05, n = 7). In 12 individuals belonging to the different groups, changes in BBB permeability were compared with expression of markers of astrocyte (GFAP) and microglial cell (CD11b) using ex vivo immunohistochemistry. Increased expression of CD11b and GFAP expression was observed in mice injected with 3 mg/kg of LPS, which did not increase with higher LPS doses. Quantitative [18F]FDS PET imaging can capture different levels of BBB permeability in vivo. A biphasic effect was observed with the lowest dose of LPS that triggered neuroinflammation without disruptive changes in BBB permeability, and higher LPS doses that increased BBB permeability without additional recruitment of glial cells.
Collapse
Affiliation(s)
- Sarah Leterrier
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Sébastien Goutal
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Gaëlle Hugon
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Maud Goislard
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Wadad Saba
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Benoit Hosten
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Simon Specklin
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Alexandra Winkeler
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Nicolas Tournier
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| |
Collapse
|
39
|
Khoshnavay Foumani M, Amirshahrokhi K, Namjoo Z, Niapour A. Carvedilol attenuates inflammatory reactions of lipopolysaccharide-stimulated BV2 cells and modulates M1/M2 polarization of microglia via regulating NLRP3, Notch, and PPAR-γ signaling pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:4727-4736. [PMID: 38133658 DOI: 10.1007/s00210-023-02914-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
Microglial cells coordinate immune responses in the central nervous system. Carvedilol (CVL) is a non-selective β-blocker with anti-inflammatory and anti-oxidant effects. This study aims to investigate the anti-inflammatory effects and the underlying mechanisms of CVL on lipopolysaccharide (LPS)-induced inflammation in microglial BV2 cells. BV2 cells were stimulated with LPS, and the protective effects of CVL were investigated via measurement of cell viability, reactive oxygen species (ROS), and interleukin (IL)-1β liberation. The protein levels of some inflammatory cascade, Notch, and peroxisome proliferator-activated receptor (PPAR)-γ pathways and relative markers of M1/M2 microglial phenotypes were assessed. Neuroblastoma SH-SY5Y cells were cultured with a BV2-conditioned medium (CM), and the capacity of CVL to protect cell viability was evaluated. CVL displayed a protective effect against LPS stress through reducing ROS and down-regulating of nuclear factor kappa B (NF-κB) p65, NLR family pyrin domain containing-3 (NLRP3), and IL-1β proteins. LPS treatment significantly increased the levels of the M1 microglial marker inducible nitric oxide synthase (iNOS) and M1-associated cleaved-NOTCH1 and hairy and enhancer of split-1 (HES1) proteins. Conversely, LPS treatment reduced the levels of the M2 marker arginase-1 (Arg-1) and PPAR-γ proteins. CVL pre-treatment reduced the protein levels of iNOS, cleaved-NOTCH1, and HES1, while increased Arg-1 and PPAR-γ. CM of CVL-primed BV2 cells significantly improved SH-SY5Y cell viability as compared with the LPS-induced cells. CVL suppressed ROS production and alleviated the expression of inflammatory markers in LPS-stimulated BV2 cells. Our results demonstrated that targeting Notch and PPAR-γ pathways as well as directing BV2 cell polarization toward the M2 phenotype may provide a therapeutic strategy to suppress neuroinflammation by CVL.
Collapse
Affiliation(s)
- Mohammadjavad Khoshnavay Foumani
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Keyvan Amirshahrokhi
- Department of Pharmacology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Zeinab Namjoo
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Ali Niapour
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
40
|
Hillari L, Frank P, Cadar D. Systemic inflammation, lifestyle behaviours and dementia: A 10-year follow-up investigation. Brain Behav Immun Health 2024; 38:100776. [PMID: 38706574 PMCID: PMC11068506 DOI: 10.1016/j.bbih.2024.100776] [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: 09/29/2023] [Revised: 03/17/2024] [Accepted: 04/16/2024] [Indexed: 05/07/2024] Open
Abstract
Objectives Lifestyle behaviours have been linked to dementia incidence, but their cumulative impact on dementia and the underlying mechanisms remain poorly understood. This study investigated the association of co-occurring lifestyle behaviours with dementia incidence and the mediating role of systemic inflammation in this association. Methods The sample comprised 3131 participants (55.2% female) from the English Longitudinal Study of Ageing aged 52-92 years at baseline (2008/09). Self-reported baseline lifestyle behaviours (alcohol intake, fruit and vegetable consumption, smoking, physical activity, sleep duration, social engagement, and cognitive activity) were summed to derive an index of lifestyle behaviours, ranging from 0 to 7, with higher scores denoting a higher number of health-risk behaviours. Incident dementia cases (n = 130, 4.2%) were identified through doctor-diagnosed dementia, informant interviews, and health records between 2014/15 and 2018/19. Systemic inflammation was measured through fasting plasma concentrations of C-reactive protein in 2012/13. Results Binary logistic regression models indicated that the odds of subsequent dementia increased by 1.19 for each additional health-risk behaviour (95% confidence intervals: 1.04, 1.37, p = 0.014) after adjusting for age, sex, ethnicity, wealth, education, marital status, body mass index, coronary heart disease, hypertension, stroke, and depression. However, this association was not mediated by C-reactive protein. Conclusions Co-occurring health-risk behaviours were associated with higher dementia incidence up to 10 years later, underscoring the importance of modifying health-risk behaviours for the prevention of dementia. Systemic inflammation did not explain the association between behaviours and dementia.
Collapse
Affiliation(s)
- Leah Hillari
- Behavioural Science and Health, University College London, London, UK
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Philipp Frank
- Behavioural Science and Health, University College London, London, UK
- UCL Brain Sciences, University College London, London, UK
| | - Dorina Cadar
- Behavioural Science and Health, University College London, London, UK
- Centre for Dementia Studies, Brighton and Sussex Medical School, Brighton, UK
| |
Collapse
|
41
|
Dar W. Aspartame-induced cognitive dysfunction: Unveiling role of microglia-mediated neuroinflammation and molecular remediation. Int Immunopharmacol 2024; 135:112295. [PMID: 38776852 DOI: 10.1016/j.intimp.2024.112295] [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: 03/04/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
Aspartame, an artificial sweetener, is consumed by millions of people globally. There are multiple reports of aspartame and its metabolites affecting cognitive functions in animal models and humans, which include learning problems, headaches, seizures, migraines, irritable moods, anxiety, depression, and insomnia. These cognitive deficits and associated symptoms are partly attributed to dysregulated excitatory and inhibitory neurotransmitter balance due to aspartate released from aspartame, resulting in an excitotoxic effect in neurons, leading to neuronal damage. However, microglia, a central immunocompetent cell type in brain tissue and a significant player in inflammation can contribute to the impact. Microglia rapidly respond to changes in CNS homeostasis. Aspartame consumption might affect the microglia phenotype directly via methanol-induced toxic effects and indirectly via aspartic acid-mediated excitotoxicity, exacerbating symptoms of cognitive decline. Long-term oral consumption of aspartame thus might change microglia's phenotype from ramified to activated, resulting in chronic or sustained activation, releasing excess pro-inflammatory molecules. This pro-inflammatory surge might lead to the degeneration of healthy neurons and other glial cells, impairing cognition. This review will deliberate on possible links and research gaps that need to be explored concerning aspartame consumption, ecotoxicity and microglia-mediated inflammatory cognitive impairment. The study covers a comprehensive analysis of the impact of aspartame consumption on cognitive function, considering both direct and indirect effects, including the involvement of microglia-mediated neuroinflammation. We also propose a novel intervention strategy involving tryptophan supplementation to mitigate cognitive decline symptoms in individuals with prolonged aspartame consumption, providing a potential solution to address the adverse effects of aspartame on cognitive function.
Collapse
Affiliation(s)
- Waseem Dar
- Translational Neurobiology and Disease Modelling Laboratory, Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence, Greater Noida, 201314, India.
| |
Collapse
|
42
|
Lu C, Hyde DR. Cytokines IL-1β and IL-10 are required for Müller glia proliferation following light damage in the adult zebrafish retina. Front Cell Dev Biol 2024; 12:1406330. [PMID: 38938553 PMCID: PMC11208712 DOI: 10.3389/fcell.2024.1406330] [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: 03/24/2024] [Accepted: 05/16/2024] [Indexed: 06/29/2024] Open
Abstract
Zebrafish possess the ability to regenerate dying neurons in response to retinal injury, with both Müller glia and microglia playing integral roles in this response. Resident Müller glia respond to damage by reprogramming and undergoing an asymmetric cell division to generate a neuronal progenitor cell, which continues to proliferate and differentiate into the lost neurons. In contrast, microglia become reactive, phagocytose dying cells, and release inflammatory signals into the surrounding tissue following damage. In recent years, there has been increased attention on elucidating the role that microglia play in regulating retinal regeneration. Here we demonstrate that inflammatory cytokines are differentially expressed during retinal regeneration, with the expression of a subset of pro-inflammatory cytokine genes upregulated shortly after light damage and the expression of a different subset of cytokine genes subsequently increasing. We demonstrate that both cytokine IL-1β and IL-10 are essential for Müller glia proliferation in the light-damaged retina. While IL-1β is sufficient to induce Müller glia proliferation in an undamaged retina, expression of IL-10 in undamaged retinas only induces Müller glia to express gliotic markers. Together, these findings demonstrate the essential role of inflammatory cytokines IL-1β and IL-10 on Müller glia proliferation following light damage in adult zebrafish.
Collapse
Affiliation(s)
| | - David R. Hyde
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, and Center for Zebrafish Research, Galvin Life Sciences Building, University of Notre Dame, Notre Dame, IN, United States
| |
Collapse
|
43
|
You JE, Kim EJ, Kim HW, Kim JS, Kim K, Kim PH. Exploring the Role of Guanylate-Binding Protein-2 in Activated Microglia-Mediated Neuroinflammation and Neuronal Damage. Biomedicines 2024; 12:1130. [PMID: 38791092 PMCID: PMC11117630 DOI: 10.3390/biomedicines12051130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
Neuron damage by microglia, which act as macrophage cells in the brain, can result in various brain diseases. However, the function of pro-inflammatory or anti-inflammatory microglia in the neurons remains controversial. Guanylate-binding protein-2 (GBP2) is expressed and activated in the microglia in the early phase of the inflammatory response and plays an important role in controlling immune responses. In this study, we evaluated whether GBP2 initially reduces the immune response induced by microglia, and whether microglia induce pro-inflammatory functions in neurons via GBP2 expression. In lipopolysaccharide (LPS)-stimulated microglia, we assessed the expression of GBP2 and how it affects neurons via activated microglia. The biological functions of microglia due to the downregulation of the GBP2 gene were examined using short hairpin RNA (shRNA)-RNA-GBP2. Downregulated GBP2 affected the function of mitochondria in the microglia and showed reduced neuronal damage when compared to the control group in the co-culture system. Furthermore, this protein was observed to be highly expressed in the brains of dementia mice. Our results are the first to report that the downregulation of GBP2 in activated microglia has an anti-inflammatory function. This study suggests that the GBP2 gene can be used as a therapeutic target biomarker for inflammation-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Ji-Eun You
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Republic of Korea; (J.-E.Y.); (E.-J.K.)
| | - Eun-Ji Kim
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Republic of Korea; (J.-E.Y.); (E.-J.K.)
| | - Ho Won Kim
- Myunggok Medical Research Institute, College of Medical School, Konyang University, Daejeon 35365, Republic of Korea; (H.W.K.); (J.-S.K.)
| | - Jong-Seok Kim
- Myunggok Medical Research Institute, College of Medical School, Konyang University, Daejeon 35365, Republic of Korea; (H.W.K.); (J.-S.K.)
| | - Kyunggon Kim
- Department of Digital Medicine, College of Medicine, University of Ulsan, Seoul 05505, Republic of Korea;
- Department of Convergence Medicine, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Pyung-Hwan Kim
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Republic of Korea; (J.-E.Y.); (E.-J.K.)
| |
Collapse
|
44
|
Gao F, Zhang Z, Xue N, Ma Y, Jiao J, Wang C, Zhang K, Lin Y, Li S, Guo Z, An J, Wang P, Xu B, Lei H. Identification of a novel oligopeptide from defatted walnut meal hydrolysate as a potential neuroprotective agent. Food Funct 2024; 15:5566-5578. [PMID: 38712886 DOI: 10.1039/d3fo05501a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Free radical damage and oxidative stress are thought to play a crucial role in the development of neurodegenerative diseases. Walnut peptides, especially walnut oligopeptides, have been shown to protect nerve cells from oxidative stress and inflammatory damage, as well as improve memory function. In this study, walnut peptides were obtained from walnut meal through enzymatic hydrolysis, ultrafiltration, and gel filtration chromatography. A novel oligopeptide called AQ was successfully isolated and its chemical structure was identified as AASCDQ using ESI-MS/MS. AQ demonstrated remarkable scavenging activity against O2- free radicals (81.00%), DPPH free radicals (79.40%), and ABTS free radicals (67.09%) at a concentration of 1 mg mL-1. Furthermore, AQ exhibited strong neuroprotective effects against hydrogen peroxide-induced damage in SH-SY5Y cells, reducing cell injury and apoptosis. AQ also effectively inhibited the secretion of pro-inflammatory factors NO (IC50 = 46.03 ± 0.32 μM) and suppressed the expression of IL-6 and TNF-α in RAW264.7 cells stimulated by LPS. In vivo experiments demonstrated that AQ promoted angiogenesis in the quail chick chorioallantoic membrane assay and reduced ROS accumulation in Caenorhabditis elegans, thereby extending its lifespan. The anti-inflammatory mechanism of AQ was further confirmed by western blotting. In summary, the novel oligopeptide AQ possesses potential neuroprotective effects, including antioxidant, anti-inflammatory, angiogenic, and anti-aging properties, making it a promising candidate for the development of functional foods and pharmaceutical products.
Collapse
Affiliation(s)
- Feng Gao
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China.
| | - Zixuan Zhang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China.
| | - Nannan Xue
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China.
| | - Yunnan Ma
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China.
| | - Jingyi Jiao
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China.
| | - Cheng Wang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China.
| | - Keyi Zhang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China.
| | - Yixuan Lin
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China.
| | - Shanlan Li
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China.
| | - Zhuoqian Guo
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China.
| | - Jin An
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China.
| | - Penglong Wang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China.
| | - Bing Xu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China.
| | - Haimin Lei
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China.
| |
Collapse
|
45
|
He Q, Qi Q, Ibeanu GC, Li PA. B355252 Suppresses LPS-Induced Neuroinflammation in the Mouse Brain. Brain Sci 2024; 14:467. [PMID: 38790446 PMCID: PMC11119117 DOI: 10.3390/brainsci14050467] [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: 03/28/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
B355252 is a small molecular compound known for potentiating neural growth factor and protecting against neuronal cell death induced by glutamate in vitro and cerebral ischemia in vivo. However, its other biological functions remain unclear. This study aims to investigate whether B355252 suppresses neuroinflammatory responses and cell death in the brain. C57BL/6j mice were intraperitoneally injected with a single dosage of lipopolysaccharide (LPS, 1 mg/kg) to induce inflammation. B355252 (1 mg/kg) intervention was started two days prior to the LPS injection. The animal behavioral changes were assessed pre- and post-LPS injections. The animal brains were harvested at 4 and 24 h post-LPS injection, and histological, biochemical, and cytokine array outcomes were examined. Results showed that B355252 improved LPS-induced behavioral deterioration, mitigated brain tissue damage, and suppressed the activation of microglial and astrocytes. Furthermore, B355252 reduced the protein levels of key pyroptotic markers TLR4, NLRP3, and caspase-1 and inhibited the LPS-induced increases in IL-1β, IL-18, and cytokines. In conclusion, B355252 demonstrates a potent anti-neuroinflammatory effect in vivo, suggesting that its potential therapeutic value warrants further investigation.
Collapse
Affiliation(s)
- Qingping He
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), College of Health and Sciences, North Carolina Central University, Durham, NC 27707, USA; (Q.H.); (G.C.I.)
| | - Qi Qi
- Human Vaccine Institute, Department of Surgery, Duke University Medical Center, Durham, NC 27707, USA;
| | - Gordon C. Ibeanu
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), College of Health and Sciences, North Carolina Central University, Durham, NC 27707, USA; (Q.H.); (G.C.I.)
| | - P. Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), College of Health and Sciences, North Carolina Central University, Durham, NC 27707, USA; (Q.H.); (G.C.I.)
| |
Collapse
|
46
|
Savant R, Pradhan RK, Bhagat S, Mythri RB, Varghese AM, Vengalil S, Nalini A, Sathyaprabha TN, Raju TR, Vijayalakshmi K. Enhanced levels of fractalkine and HSP60 in cerebrospinal fluid of sporadic amyotrophic lateral sclerosis patients. Int J Neurosci 2024:1-11. [PMID: 38625841 DOI: 10.1080/00207454.2024.2344581] [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: 04/07/2023] [Accepted: 04/07/2024] [Indexed: 04/18/2024]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a multifactorial neurodegenerative disorder with a significant contribution of non-cell autonomous mechanisms to motor neuronal degeneration. Amongst a plethora of molecules, fractalkine (C-X3-C motif chemokine ligand 1), and Heat Shock Protein 60 (HSP60), are key modulators of microglial activation. The contribution of these molecules in Sporadic ALS (SALS) remains unexplored. To investigate this, fractalkine levels were estimated in Cerebrospinal fluid (CSF) of SALS patients (ALS-CSF; n = 44) by Enzyme-linked Immunosorbent Assay (ELISA) and correlated with clinical parameters including disease severity and duration. CSF HSP60 levels were estimated by Western blotting (ALS-CSF; n = 19). Also, CSF levels of Chitotriosidase-1 (CHIT-1), a microglia-specific neuroinflammatory molecule, were measured and its association, if any, with fractalkine and HSP60 was investigated. Both fractalkine and HSP60 levels were significantly elevated in ALS-CSF. Similar to our earlier observation, CHIT-1 levels were also upregulated. Fractalkine showed a moderate negative correlation with the ALS-Functional Rating Scale (ALSFRS) score indicating its significant rise in mild cases which plateaued in cases with high disease severity. However, no obvious correlation was found between fractalkine, HSP60, and CHIT-1. Our study hints that high fractalkine levels in mild cases might be conferring neuroprotection by combating microglial activation and highlights its importance as a novel therapeutic target for SALS. On the other hand, significantly enhanced levels of HSP60, a pro-inflammatory molecule, hint towards its role in accentuating microgliosis, although, it doesn't act synergistically with CHIT-1. Our study suggests that fractalkine and HSP60 act independently of CHIT-1 to suppress and accentuate neuroinflammation, respectively.
Collapse
Affiliation(s)
- Rashmi Savant
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences, Bangalore, Karnataka, India
| | - Raj Kumar Pradhan
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences, Bangalore, Karnataka, India
| | - Savita Bhagat
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences, Bangalore, Karnataka, India
| | - Rajeswara Babu Mythri
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences, Bangalore, Karnataka, India
| | - Anu Mary Varghese
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences, Bangalore, Karnataka, India
| | - Seena Vengalil
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bangalore, Karnataka, India
| | - Atchayaram Nalini
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bangalore, Karnataka, India
| | - Talakad N Sathyaprabha
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences, Bangalore, Karnataka, India
| | - Trichur R Raju
- A.S. Paintal Distinguished Scientific Chair National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - K Vijayalakshmi
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences, Bangalore, Karnataka, India
| |
Collapse
|
47
|
Kim DW, Lee TK, Ahn JH, Yang SR, Shin MC, Cho JH, Won MH, Kang IJ, Park JH. Porphyran Attenuates Neuronal Loss in the Hippocampal CA1 Subregion Induced by Ischemia and Reperfusion in Gerbils by Inhibiting NLRP3 Inflammasome-Mediated Neuroinflammation. Mar Drugs 2024; 22:170. [PMID: 38667787 PMCID: PMC11050983 DOI: 10.3390/md22040170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Porphyran, a sulfated polysaccharide found in various species of marine red algae, has been demonstrated to exhibit diverse bioactivities, including anti-inflammatory effects. However, the protective effects of porphyran against cerebral ischemia and reperfusion (IR) injury have not been investigated. The aim of this study was to examine the neuroprotective effects of porphyran against brain IR injury and its underlying mechanisms using a gerbil model of transient forebrain ischemia (IR in the forebrain), which results in pyramidal cell (principal neuron) loss in the cornu ammonis 1 (CA1) subregion of the hippocampus on day 4 after IR. Porphyran (25 and 50 mg/kg) was orally administered daily for one week prior to IR. Pretreatment with 50 mg/kg of porphyran, but not 25 mg/kg, significantly attenuated locomotor hyperactivity and protected pyramidal cells located in the CA1 area from IR injury. The pretreatment with 50 mg/kg of porphyran significantly suppressed the IR-induced activation and proliferation of microglia in the CA1 subregion. Additionally, the pretreatment significantly inhibited the overexpressions of nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing protein-3 (NLRP3) inflammasome complex, and pro-inflammatory cytokines (interleukin 1 beta and interleukin 18) induced by IR in the CA1 subregion. Overall, our findings suggest that porphyran exerts neuroprotective effects against brain IR injury, potentially by reducing the reaction (activation) and proliferation of microglia and reducing NLRP3 inflammasome-mediated neuroinflammation.
Collapse
Affiliation(s)
- Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea;
| | - Tae-Kyeong Lee
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea;
| | - Ji Hyeon Ahn
- Department of Physical Therapy, College of Health Science, Youngsan University, Yangsan 50510, Republic of Korea;
| | - Se-Ran Yang
- Department of Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea;
| | - Myoung Cheol Shin
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon 24289, Republic of Korea; (M.C.S.); (J.H.C.); (M.-H.W.)
| | - Jun Hwi Cho
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon 24289, Republic of Korea; (M.C.S.); (J.H.C.); (M.-H.W.)
| | - Moo-Ho Won
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon 24289, Republic of Korea; (M.C.S.); (J.H.C.); (M.-H.W.)
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea;
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea
| |
Collapse
|
48
|
Deng W, Yi P, Xiong Y, Ying J, Lin Y, Dong Y, Wei G, Wang X, Hua F. Gut Metabolites Acting on the Gut-Brain Axis: Regulating the Functional State of Microglia. Aging Dis 2024; 15:480-502. [PMID: 37548933 PMCID: PMC10917527 DOI: 10.14336/ad.2023.0727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/27/2023] [Indexed: 08/08/2023] Open
Abstract
The gut-brain axis is a communication channel that mediates a complex interplay of intestinal flora with the neural, endocrine, and immune systems, linking gut and brain functions. Gut metabolites, a group of small molecules produced or consumed by biochemical processes in the gut, are involved in central nervous system regulation via the highly interconnected gut-brain axis affecting microglia indirectly by influencing the structure of the gut-brain axis or directly affecting microglia function and activity. Accordingly, pathological changes in the central nervous system are connected with changes in intestinal metabolite levels as well as altered microglia function and activity, which may contribute to the pathological process of each neuroinflammatory condition. Here, we discuss the mechanisms by which gut metabolites, for instance, the bile acids, short-chain fatty acids, and tryptophan metabolites, regulate the structure of each component of the gut-brain axis, and explore the important roles of gut metabolites in the central nervous system from the perspective of microglia. At the same time, we highlight the roles of gut metabolites affecting microglia in the pathogenesis of neurodegenerative diseases and neurodevelopmental disorders. Understanding the relationship between microglia, gut microbiota, neuroinflammation, and neurodevelopmental disorders will help us identify new strategies for treating neuropsychiatric disorders.
Collapse
Affiliation(s)
- Wenze Deng
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi, China.
| | - Pengcheng Yi
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi, China.
| | - Yanhong Xiong
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi, China.
| | - Jun Ying
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi, China.
| | - Yue Lin
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi, China.
| | - Yao Dong
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi, China.
| | - Gen Wei
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi, China.
| | - Xifeng Wang
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| | - Fuzhou Hua
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi, China.
| |
Collapse
|
49
|
Mingo YB, Gabele L, Lonnemann N, Brône B, Korte M, Hosseini S. The effects of urolithin A on poly I:C-induced microglial activation. Front Cell Neurosci 2024; 18:1343562. [PMID: 38577490 PMCID: PMC10993698 DOI: 10.3389/fncel.2024.1343562] [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: 11/23/2023] [Accepted: 03/11/2024] [Indexed: 04/06/2024] Open
Abstract
Neuroinflammation can be triggered by various stimuli, including viral infections. Viruses can directly invade the brain and infect neuronal cells or indirectly trigger a "cytokine storm" in the periphery that eventually leads to microglial activation in the brain. While this initial activation of microglial cells is important for viral clearance, chronic activation leads to excessive inflammation and oxidative stress, which can be neurotoxic. Remarkebly, recent studies have shown that certain viruses such as influenza A virus, coronavirus, herpes virus and Epstein-Barr virus may be involved in the development of neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, and multiple sclerosis. Therefore, it is important to find therapeutic strategies against chronic neuroinflammation triggered by viral infections. Here, we investigated the effects of urolithin A (UA) on microglial activation in vitro induced by a viral mimetic, poly I:C, in a triple co-culture system of neurons, astrocytes and microglial cells. Immunocytochemistry was used to perform a comprehensive single-cell analysis of the morphological changes of microglia as an indicator of their reactive state. Treatment with UA significantly prevented the poly I:C-induced reactive state of microglia, which was characterized by increased expression of the microglial activation markers CD68 and IBA-1. UA restored the poly I:C-induced morphology by restoring microglial ramification. In addition, UA was able to reduce the release of the pro-inflammatory mediators CCL2, TNF-α, and IL-1β and showed a trend toward attenuation of cellular ROS production in poly I:C-treated cultures. Overall, this study suggests that UA as a component of a healthy diet may help prevent virus-induced neuroinflammation and may have therapeutic potential for future studies to prevent or treat neurodegenerative diseases by targeting the associated neuroinflammatory processes.
Collapse
Affiliation(s)
- Yakum Benard Mingo
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
- Laboratory for Neurophysiology, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Lea Gabele
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
- Helmholtz Centre for Infection Research, Research Group Neuroinflammation and Neurodegeneration, Braunschweig, Germany
| | - Niklas Lonnemann
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Bert Brône
- Laboratory for Neurophysiology, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Martin Korte
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
- Helmholtz Centre for Infection Research, Research Group Neuroinflammation and Neurodegeneration, Braunschweig, Germany
| | - Shirin Hosseini
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
- Helmholtz Centre for Infection Research, Research Group Neuroinflammation and Neurodegeneration, Braunschweig, Germany
| |
Collapse
|
50
|
Kim H, Le B, Goshi N, Zhu K, Grodzki AC, Lein PJ, Zhao M, Seker E. Rat primary cortical cell tri-culture to study effects of amyloid-beta on microglia function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.584736. [PMID: 38558989 PMCID: PMC10979983 DOI: 10.1101/2024.03.15.584736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Introduction The etiology and progression of sporadic Alzheimer's Disease (AD) have been studied for decades. One proposed mechanism is that amyloid-beta (Aβ) proteins induce neuroinflammation, synapse loss, and neuronal cell death. Microglia play an especially important role in Aβ clearance, and alterations in microglial function due to aging or disease may result in Aβ accumulation and deleterious effects on neuronal function. However, studying these complex factors in vivo , where numerous confounding processes exist, is challenging, and until recently, in vitro models have not allowed sustained culture of microglia, astrocytes and neurons in the same culture. Here, we employ a tri-culture model of rat primary neurons, astrocytes, and microglia and compare it to co-culture (neurons and astrocytes) and mono-culture enriched for microglia to study microglial function (i.e., motility and Aβ clearance) and proteomic response to exogenous Aβ. Methods We established cortical co-culture (neurons and astrocytes), tri-culture (neurons, astrocytes, and microglia), and mono-culture (microglia) from perinatal rat pups. On days in vitro (DIV) 7 - 14, the cultures were exposed to fluorescently-labeled Aβ (FITC-Aβ) particles for varying durations. Images were analyzed to determine the number of FITC-Aβ particles after specific lengths of exposure. A group of cells were stained for βIII-tubulin, GFAP, and Iba1 for morphological analysis via quantitative fluorescence microscopy. Cytokine profiles from conditioned media were obtained. Live-cell imaging with images acquired every 5 minutes for 4 hours was employed to extract microglia motility parameters (e.g., Euclidean distance, migration speed, directionality ratio). Results and discussion FITC-Aβ particles were more effectively cleared in the tri-culture compared to the co-culture. This was attributed to microglia engulfing FITC-Aβ particles, as confirmed via epifluorescence and confocal microscopy. Adding FITC-Aβ significantly increased the size of microglia, but had no significant effect on neuronal surface coverage or astrocyte size. Analysis of the cytokine profile upon FITC-Aβ addition revealed a significant increase in proinflammatory cytokines (TNF-α, IL-1α, IL-1β, IL-6) in tri-culture, but not co-culture. In addition, Aβ addition altered microglia motility marked by swarming-like motion with decreased Euclidean distance yet unaltered speed. These results highlight the importance of cell-cell communication in microglia function (e.g., motility and Aβ clearance) and the utility of the tri-culture model to further investigate microglia dysfunction in AD.
Collapse
|