1
|
Agostinelli E, Marzaro G, Gambari R, Finotti A. Potential applications of components of aged garlic extract in mitigating pro-inflammatory gene expression linked to human diseases (Review). Exp Ther Med 2025; 30:134. [PMID: 40432842 PMCID: PMC12107228 DOI: 10.3892/etm.2025.12884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/07/2025] [Indexed: 05/29/2025] Open
Abstract
In the present review, simple approaches for the screening and characterization of natural compound agents that alter pro-inflammatory gene expression are described, with a particular focus on aged garlic extract (AGE), which has been the subject of several investigations that have supported its potential application as an anti-inflammatory agent. Additionally, evidence regarding the possible effects and mechanisms of action of two major AGE components, S-allyl cysteine (SAC) and S-1-propenyl-l-cysteine (S1PC), is reviewed. The proposed molecular targets of SAC and S1PC are IKKβ kinase, the Kelch-like ECH-associated protein 1-nuclear factor erythroid 2-related factor 2 complex, peroxisome proliferator-activated receptor-γ, histone deacetylase and toll-like receptor 4 (TLR4). Targeting these molecules causes a marked reduction in NF-κB activity accompanied by a notable decrease in the transcription of NF-κB-regulated genes. Another main objective of the present review was to discuss the possibility that AGE and its bioactive components could be employed in the treatment of several human pathologies that are characterized by a hyperinflammatory state resulting from dysregulation of the TLR4 and NF-κB pathways. SAC is of interest in the treatment of lung pathologies, neurological diseases, osteoarthritis, muscular atrophy, cardiovascular diseases, diabetes and cancer. Additionally, the anti-oxidative activities of AGE, SAC and S1PC are compatible with their employment in the treatment of diseases characterized by oxidative stress, such as sickle cell disease and β-thalassemia.
Collapse
Affiliation(s)
- Enzo Agostinelli
- Department of Sensory Organs, Sapienza University of Rome, Policlinico Umberto I, I-00161 Rome, Italy
- International Polyamines Foundation ‘Ente Terzo Settore-Organizzazione Non Lucrativa di Utilità Sociale’, I-00159 Rome, Italy
| | - Giovanni Marzaro
- Department of Diagnostics and Public Health, University of Verona, I-37134 Verona, Italy
| | - Roberto Gambari
- Research Center on Innovative Therapies for Cystic Fibrosis, University of Ferrara, I-44121 Ferrara, Italy
- Department of Life Sciences and Biotechnology, Ferrara University, I-44121 Ferrara, Italy
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, Ferrara University, I-44121 Ferrara, Italy
| |
Collapse
|
2
|
Bellini G, D'Antongiovanni V, Palermo G, Antonioli L, Fornai M, Ceravolo R, Bernardini N, Derkinderen P, Pellegrini C. α-Synuclein in Parkinson's Disease: From Bench to Bedside. Med Res Rev 2025; 45:909-946. [PMID: 39704040 PMCID: PMC11976381 DOI: 10.1002/med.22091] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/24/2024] [Accepted: 11/07/2024] [Indexed: 12/21/2024]
Abstract
α-Synuclein (α-syn), a pathological hallmark of PD, is emerging as a bridging element at the crossroads between neuro/immune-inflammatory responses and neurodegeneration in PD. Several evidence show that pathological α-syn accumulates in neuronal and non-neuronal cells (i.e., neurons, microglia, macrophages, skin cells, and intestinal cells) in central and peripheral tissues since the prodromal phase of the disease, contributing to brain pathology. Indeed, pathological α-syn deposition can promote neurogenic/immune-inflammatory responses that contribute to systemic and central neuroinflammation associated with PD. After providing an overview of the structure and functions of physiological α-syn as well as its pathological forms, we review current studies about the role of neuronal and non-neuronal α-syn at the crossroads between neuroinflammation and neurodegeneration in PD. In addition, we provide an overview of the correlation between the accumulation of α-syn in central and peripheral tissues and PD, related symptoms, and neuroinflammation. Special attention was paid to discussing whether targeting α-syn can represent a suitable therapeutical approach for PD.
Collapse
Affiliation(s)
- Gabriele Bellini
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
- Department of NeurologyThe Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, NYU Langone HealthNew York CityNew YorkUSA
| | - Vanessa D'Antongiovanni
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Giovanni Palermo
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Luca Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Matteo Fornai
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Roberto Ceravolo
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Nunzia Bernardini
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Pascal Derkinderen
- Department of NeurologyNantes Université, CHU Nantes, INSERMNantesFrance
| | - Carolina Pellegrini
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| |
Collapse
|
3
|
González-Correa C, Moleón J, Miñano S, Robles-Vera I, de la Visitación N, Guerra-Hernández E, Toral M, Jiménez R, Duarte J, Romero M. Protective Effect of Dietary Fiber on Blood Pressure and Vascular Dysfunction Through Regulation of Sympathetic Tone and Immune Response in Genetic Hypertension. Phytother Res 2025; 39:1858-1875. [PMID: 40122676 DOI: 10.1002/ptr.8484] [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: 11/15/2024] [Revised: 02/17/2025] [Accepted: 02/22/2025] [Indexed: 03/25/2025]
Abstract
The mechanisms underlying the antihypertensive effect of dietary fibers remain poorly understood. This study investigates whether dietary fiber supplementation can prevent cardiovascular damage and high blood pressure in a genetic model of neurogenic hypertension. Six-week-old male spontaneously hypertensive rats (SHR) and their respective normotensive control, Wistar Kyoto rats (WKY), were divided into four groups: Untreated WKY, untreated SHR, SHR treated with resistant starch (SHR + RS), and SHR treated with inulin-type fructans (SHR + ITF) for 12 weeks. Additionally, a faecal microbiota transplantation (FMT) experiment was conducted, transferring faecal content from treated SHR donors to recipient SHRs. A diet rich in RS fiber reduced vascular oxidative stress, inflammation, and high blood pressure. These protective effects were associated with a reshaped gut microbiota, leading to increased short-chain fatty acid production, reduced endotoxemia, decreased sympathetic activity, and a restored balance between Th17 and Treg lymphocytes in mesenteric lymph nodes and aorta. Elevated plasma levels of acetate and butyrate in the SHR + RS group correlated with increased expression of aortic GPR41, GRP43 and PPARδ. Conversely, ITF treatment failed to prevent hypertension or endothelial dysfunction in SHR. FMT from the SHR + RS group to recipient SHR partially replicated these beneficial effects. This study highlights the antihypertensive benefits of dietary insoluble RS fiber, which are attributed to enhanced short-chain fatty acids production in the gut. This leads to improved gut permeability, reduced sympathetic tone, and diminished vascular T-cell accumulation. Therefore, dietary interventions with RS fiber may offer promising therapeutic strategies for preventing hypertension.
Collapse
Affiliation(s)
- Cristina González-Correa
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| | - Javier Moleón
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| | - Sofía Miñano
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Iñaki Robles-Vera
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Néstor de la Visitación
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | | | - Marta Toral
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| | - Rosario Jiménez
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Miguel Romero
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| |
Collapse
|
4
|
Sun P, Wang M, Chai X, Liu YX, Li L, Zheng W, Chen S, Zhu X, Zhao S. Disruption of tryptophan metabolism by high-fat diet-triggered maternal immune activation promotes social behavioral deficits in male mice. Nat Commun 2025; 16:2105. [PMID: 40025041 PMCID: PMC11873046 DOI: 10.1038/s41467-025-57414-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 02/20/2025] [Indexed: 03/04/2025] Open
Abstract
Diet-related maternal obesity has been implicated in neurodevelopmental disorders in progeny. Although the precise mechanisms and effective interventions remain uncertain, our research elucidates some of these complexities. We established that a prenatal high-fat diet triggered maternal immune activation (MIA), marked by elevated serum lipopolysaccharide levels and inflammatory-cytokine overproduction, which dysregulated the maternal tryptophan metabolism promoting the accumulation of neurotoxic kynurenine metabolites in the embryonic brain. Interventions aimed at mitigating MIA or blocking the kynurenine pathway effectively rescued the male mice social performance. Furthermore, excessive kynurenine metabolites initiated oxidative stress response causing neuronal migration deficits in the fetal neocortex, an effect that was mitigated by administering the glutathione synthesis precursor N-Acetylcysteine, underscoring the central role of maternal immune-metabolic homeostasis in male mice behavioral outcomes. Collectively, our study accentuated the profound influence of maternal diet-induced immuno-metabolic dysregulation on fetal brain development and provided the preventive strategies for addressing neurodevelopmental disorders.
Collapse
Affiliation(s)
- Penghao Sun
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Mengli Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xuejun Chai
- College of Basic Medicine, Xi'an Medical University, Xi'an, Shaanxi, China.
| | - Yong-Xin Liu
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, Guangdong, China
| | - Luqi Li
- Life Science Research Core Services, Northwest A&F University, Yangling, Shaanxi, China
| | - Wei Zheng
- College of Resources and Environment Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Shulin Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
5
|
Nakagawa K, Kodama K, Nagata W, Takahashi S, Satoh Y, Ishizuka T. Molecular hydrogen inhibits neuroinflammation and ameliorates depressive-like behaviors and short-term cognitive impairment in senescence-accelerated mouse prone 8 mice. Behav Brain Res 2025; 478:115330. [PMID: 39522774 DOI: 10.1016/j.bbr.2024.115330] [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: 06/04/2024] [Revised: 10/11/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND AIMS Neuroinflammation, a low-grade chronic inflammation of the central nervous system, is linked to age-related neuropsychiatric disorders such as senile depression and Alzheimer's disease. Recent studies have explored controlling neuroinflammation as a novel treatment strategy. Molecular hydrogen shows anti-inflammatory effects. However, its impacts on neuroinflammation and age-related neuropsychiatric disorders remain unelucidated. We investigated molecular hydrogen's effects on microglial activation, neuroinflammation, depressive-like behavior, and short-term cognitive decline in senescence-accelerated mouse-prone 8 (SAMP8) mice. METHODS Six-week-old SAMP8 or senescence-accelerated mouse-resistant 1 (SAMR1) mice received hydrogen-rich jelly (HRJ) or placebo jelly (PJ) from six weeks of age for 26-28 weeks. Depressive-like behavior was assessed using tail suspension and forced swimming tests, while cognitive function was evaluated using the Y-maze and object recognition tests. Brain tissues were used for immunohistochemical studies or to measure pro-inflammatory cytokine levels via enzyme-linked immunosorbent assay (ELISA). RESULTS HRJ intake reduced immobility time in both tail suspension and forced swimming tests and enhanced visual cognitive and spatial working memory in SAMP8 mice. Additionally, HRJ intake suppressed the 8-hydroxy-2'-deoxyguanosine (8-OHdG), Iba1, and cleaved caspase 3 expression levels in the medial prefrontal cortex and hippocampal dentate gyrus. Furthermore, HRJ intake significantly lowered IL-6 levels in brain tissues of SAMP8 mice. CONCLUSIONS These findings suggest that molecular hydrogen treatment may regulate neuroinflammation induced by activated microglia and improve depressive-like behavior and short-term cognitive impairment in SAMP8 mice.
Collapse
Affiliation(s)
- Keiichi Nakagawa
- Department of Pharmacology, National Defense Medical College, Tokorozawa, Saitama 359-0042, Japan
| | - Kayoko Kodama
- Department of Pharmacology, National Defense Medical College, Tokorozawa, Saitama 359-0042, Japan
| | - Wataru Nagata
- Department of Pharmacology, National Defense Medical College, Tokorozawa, Saitama 359-0042, Japan
| | - Sayaka Takahashi
- Department of Pharmacology, National Defense Medical College, Tokorozawa, Saitama 359-0042, Japan
| | - Yasushi Satoh
- Department of Biochemistry, National Defense Medical College, Tokorozawa, Saitama 359-0042, Japan
| | - Toshiaki Ishizuka
- Department of Pharmacology, National Defense Medical College, Tokorozawa, Saitama 359-0042, Japan.
| |
Collapse
|
6
|
Li L, Peng R, Wang C, Chen X, Gheyret D, Guan S, Chen B, Liu Y, Liu X, Cao Y, Han C, Xiong J, Li F, Lu T, Jia H, Li K, Wang J, Zhang X, Xu J, Wang Y, Xu X, Li T, Zhang J, Zhang S. β2 integrin regulates neutrophil trans endothelial migration following traumatic brain injury. Cell Commun Signal 2025; 23:70. [PMID: 39923080 PMCID: PMC11806581 DOI: 10.1186/s12964-025-02071-9] [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: 11/08/2024] [Accepted: 01/29/2025] [Indexed: 02/10/2025] Open
Abstract
Neutrophils are the first responders among peripheral immune cells to infiltrate the central nervous system following a traumatic brain injury (TBI), triggering neuroinflammation that can exacerbate secondary tissue damage. The precise molecular controls that dictate the inflammatory behavior of neutrophils post-TBI, however, remain largely elusive. Our comprehensive analysis of the molecular landscape surrounding the trauma in TBI mice has revealed a significant alteration in the abundance of β2 integrin (ITGB2), predominantly expressed by neutrophils and closely associated with immune responses. Using the fluid percussion injury (FPI) mouse model, we investigated the therapeutic efficacy of Rovelizumab, an agent that blocks ITGB2. The treatment has demonstrated significant improvements in neurologic function in TBI mice, attenuating blood-brain barrier permeability, mitigating oxidative stress and inflammatory mediator release, and enhancing cerebral perfusion. Moreover, ITGB2 blockade has effectively limited the adherence, migration, and infiltration of neutrophils, and has impeded the formation of neutrophil extracellular traps (NETs) upon their activation. Finally, it was demonstrated that ITGB2 mediates these effects mainly through its interaction with intercellular adhesion molecule-1 (ICAM 1) of endotheliocyte. These findings collectively illuminate ITGB2 as a crucial molecular switch that governs the adverse effects of neutrophils post-TBI and could be targeted to improve clinical outcome in patients.
Collapse
Affiliation(s)
- Lei Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Ruilong Peng
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300200, China
| | - Cong Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Xin Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Dilmurat Gheyret
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Siyu Guan
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Bo Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Yafan Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Xilei Liu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yiyao Cao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Cha Han
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jianhua Xiong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Fanjian Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Taoyuan Lu
- Xuanwu Jinan Hospital, 5106 Jingshi Road, Jinan, 250000, Shandong, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Haoran Jia
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Kaiji Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Jinchao Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Xu Zhang
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jianye Xu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Yajuan Wang
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xin Xu
- Xuanwu Jinan Hospital, 5106 Jingshi Road, Jinan, 250000, Shandong, China.
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China.
| | - Tuo Li
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300200, China.
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China.
| | - Shu Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China.
| |
Collapse
|
7
|
Liao W, Wang C, Wang R, Wu M, Li L, Chao P, Hu J, Chen WH. An activatable "AIE + ESIPT" fluorescent probe for dual-imaging of lipid droplets and hydrogen peroxide in drug-induced liver injury model. Anal Chim Acta 2025; 1335:343442. [PMID: 39643298 DOI: 10.1016/j.aca.2024.343442] [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: 09/27/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Drug-induced liver injury (DILI) is one of the most common liver diseases. The crucial role of lipid droplets (LDs) and hydrogen peroxide (H2O2), two important biomarkers in the pathophysiology of DILI, has spurred considerable efforts to accurately visualize H2O2 and LDs for elucidating their functions in the progression of DILI. However, construction of a single fluorescent probe that is able to simultaneously image H2O2 and LDs dynamics remains to be a challenging task. Therefore, it is of great demand to develop a novel fluorescent probe for tracking the LDs status and H2O2 fluctuation in drug-induced liver injury. RESULTS We developed an "AIE + ESIPT" fluorescent probe TPEHBT for dual-imaging of LDs and H2O2 during DILI process. TPEHBT displayed greatly enhanced fluorescent response to H2O2 by generating an excited state intramolecular proton transfer (ESIPT) fluorophore TPEHBT-OH with aggregation induced emission (AIE) properties. TPEHBT exhibits high selectivity, sensitivity (LOD = 4.73 nM) and large Stokes shift (320 nm) to H2O2. Interestingly, TPEHBT can light up LDs with high specificity. The probe was favorably applied in the detection of endogenous and exogenous H2O2 in living cells, and notably in the simultaneous real-time visualization of H2O2 generation and LDs accumulation during DILI process. Moreover, TPEHBT was able to image H2O2 generation in zebrafish animal model with APAP-induced liver injury. SIGNIFICANCE For the first time, probe TPEHBT was applied in the dual-imaging of H2O2 fluctuation and LDs status in APAP-induced liver injury model in vitro and in vivo. The present findings strongly suggest that TPEHBT is a promising tool for monitoring H2O2 and LDs dynamics in DILI progression.
Collapse
Affiliation(s)
- Wantao Liao
- School of Pharmacy and Food Engineering, Wuyi University, 529020, Jiangmen, PR China
| | - Chunzheng Wang
- School of Pharmacy and Food Engineering, Wuyi University, 529020, Jiangmen, PR China
| | - Ruiya Wang
- School of Pharmacy and Food Engineering, Wuyi University, 529020, Jiangmen, PR China
| | - Mengzhao Wu
- School of Pharmacy and Food Engineering, Wuyi University, 529020, Jiangmen, PR China
| | - Lanqing Li
- School of Pharmacy and Food Engineering, Wuyi University, 529020, Jiangmen, PR China.
| | - Pengjie Chao
- School of Applied Physics and Materials, Wuyi University, 529020, Jiangmen, PR China
| | - Jinhui Hu
- School of Pharmacy and Food Engineering, Wuyi University, 529020, Jiangmen, PR China
| | - Wen-Hua Chen
- School of Pharmacy and Food Engineering, Wuyi University, 529020, Jiangmen, PR China.
| |
Collapse
|
8
|
Silva RO, Haddad M, Counil H, Zaouter C, Patten SA, Fulop T, Ramassamy C. Exploring the potential of plasma and adipose mesenchymal stem cell-derived extracellular vesicles as novel platforms for neuroinflammation therapy. J Control Release 2025; 377:880-898. [PMID: 39617173 DOI: 10.1016/j.jconrel.2024.11.060] [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: 09/10/2024] [Revised: 11/10/2024] [Accepted: 11/22/2024] [Indexed: 12/10/2024]
Abstract
Persistent reactive oxygen species (ROS) and neuroinflammation contribute to the onset and progression of neurodegenerative diseases, underscoring the need for targeted therapeutic strategies to mitigate these effects. Extracellular vesicles (EVs) show promise in drug delivery due to their biocompatibility, ability to cross biological barriers, and specific interactions with cell and tissue receptors. In this study, we demonstrated that human plasma-derived EVs (pEVs) exhibit higher brain-targeting specificity, while adipose-derived mesenchymal stem cells EVs (ADMSC-EVs) offer regenerative and immunomodulatory properties. We further investigated the potential of these EVs as therapeutic carriers for brain-targeted drug delivery, using Donepezil (DNZ) as the model drug. DNZ, a cholinesterase inhibitor commonly used for Alzheimer's disease (AD), also has neuroprotective and anti-inflammatory properties. The size of EVs used ranged from 50 to 300 nm with a surface charge below -30 mV. Both formulations showed rapid cellular internalization, without toxicity, and the ability to cross the blood-brain barrier (BBB) in a zebrafish model. The have analyzed the anti-inflammatory and antioxidant actions of pEVs-DNZ and ADMSC-EVs-DNZ in the presence of lipopolysaccharide (LPS). ADMSC-EVs significantly reduced the inflammatory mediators released by HMC3 microglial cells while treatment with pEVs-DNZ and ADMSC-EVs-DNZ lowered both phagocytic activity and ROS levels in these cells. In vivo experiments using zebrafish larvae revealed that both EV formulations reduced microglial proliferation and exhibited antioxidant effects. Overall, this study highlights the potential of EVs loaded with DNZ as a novel approach for treating neuroinflammation underlying various neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Mohamed Haddad
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531 Boul des Prairies, Laval, QC H7V 1B7, Canada
| | - Hermine Counil
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531 Boul des Prairies, Laval, QC H7V 1B7, Canada
| | - Charlotte Zaouter
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531 Boul des Prairies, Laval, QC H7V 1B7, Canada
| | - Shunmoogum A Patten
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531 Boul des Prairies, Laval, QC H7V 1B7, Canada
| | - Tamas Fulop
- Research Center on Aging, Faculty of Medicine and Health Sciences, University Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| | - Charles Ramassamy
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531 Boul des Prairies, Laval, QC H7V 1B7, Canada.
| |
Collapse
|
9
|
McGlothen KI, Hines RM, Hines DJ. Outward depolarization of the microglia mitochondrial membrane potential following lipopolysaccharide exposure: a novel screening tool for microglia metabolomics. Front Cell Neurosci 2024; 18:1430448. [PMID: 39569069 PMCID: PMC11576292 DOI: 10.3389/fncel.2024.1430448] [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: 05/09/2024] [Accepted: 10/07/2024] [Indexed: 11/22/2024] Open
Abstract
Microglia are non-electrogenic immune cells that respond rapidly to protect the central nervous system (CNS) from infections, injuries, or other forms of damage. Microglia mitochondria are essential for providing the requisite energy resources for immune regulation. While fluctuations in energy metabolism are regulated by mitochondria and are reflected in the mitochondrial membrane potential (ΔΨm), there remains a lack of innovation in microglia-centric tools that capitalize on this. In this study, live imaging of microglia in acute slices from EGFP reporter mice expressing EGFP under the control of the fractalkine receptor (CX3CR1) promoter is combined with loading a fluorescent reporter of ΔΨm. Depolarizations in the ΔΨm were recorded after administering the well-characterized immune stimulant lipopolysaccharide (LPS). Microglia ΔΨm increased in distinctive phases with a relatively steep slope following LPS exposure. Conversely, the ΔΨm of neurons showed minimal regulation, highlighting a distinct microglia ΔΨm response to immune stimuli. Analysis of the depolarization of the microglia ΔΨm in the soma, branches, and endfeet revealed progressive changes in each subcellular domain originating in the soma and progressing outward. The inverse agonist emapunil attenuated the depolarization of the ΔΨm across states in a domain-specific manner. These findings emphasize the contribution of mitochondrial membrane dynamics in regulating microglial responses to immune stimuli. Further, this work advances a novel drug screening strategy for the therapeutic regulation of metabolic activity in inflammatory conditions of the brain.
Collapse
Affiliation(s)
- Kendra I McGlothen
- Department of Psychology, Psychological and Brain Sciences & Interdisciplinary Neuroscience Programs, College of Liberal Arts, University of Nevada, Las Vegas, NV, United States
| | - Rochelle M Hines
- Department of Psychology, Psychological and Brain Sciences & Interdisciplinary Neuroscience Programs, College of Liberal Arts, University of Nevada, Las Vegas, NV, United States
| | - Dustin J Hines
- Department of Psychology, Psychological and Brain Sciences & Interdisciplinary Neuroscience Programs, College of Liberal Arts, University of Nevada, Las Vegas, NV, United States
| |
Collapse
|
10
|
Qu H, Wu J, Pan Y, Abdulla A, Duan Z, Cheng W, Wang N, Chen H, Wang C, Yang J, Tang J, Yang C, Wu C, Xue X. Biomimetic Nanomodulator Regulates Oxidative and Inflammatory Stresses to Treat Sepsis-Associated Encephalopathy. ACS NANO 2024; 18:28228-28245. [PMID: 39367850 DOI: 10.1021/acsnano.4c08157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/07/2024]
Abstract
Sepsis-associated encephalopathy (SAE) is a devastating complication of sepsis, affecting approximately 70% of patients with sepsis in intensive care units (ICU). Although the pathophysiological mechanisms remain elusive, sepsis is typically accompanied by systemic inflammatory response syndrome (SIRS) and hyper-oxidative conditions. Here, we introduce a biomimetic nanomodulator (mAOI NP) that specifically targets inflammation site and simultaneously regulates oxidative and inflammatory stresses. mAOI NPs are constructed using metal-coordinated polyphenolic antioxidants (tannic acid) and flavonoid quercetin, which are then coated with macrophage membrane to enhance pharmacokinetics and enable SAE targeting. In a cecal ligation and puncture (CLP)-induced severe sepsis model, mAOI NPs effectively mitigate oxidative stress by purging reactive oxygen species, repairing mitochondrial damage and activating the Nrf2/HO-1 signaling pathway; while polarizing M1 macrophages or microglia toward anti-inflammatory M2 subtype. mAOI NPs potently inhibit sepsis progress, prolong overall survival from 25 to 66% and enhance learning and memory capabilities in SAE mice. Further proteomics analysis reveals that mAOI NPs modulate neurodevelopment processes related to learning and memory formation while also exerting anti-inflammatory and antioxidative effects on brain tissue responses associated with SAE pathology. This study offers significant potential for improving patient outcomes and revolutionizing the treatment landscape for this devastating complication of sepsis.
Collapse
Affiliation(s)
- Haijing Qu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jie Wu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuqing Pan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Aynur Abdulla
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhiran Duan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wei Cheng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ning Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Han Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chao Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiaojiao Yang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianguo Tang
- Department of Trauma-Emergency and Critical Care Medicine Center (TECCMC), Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Chunhui Yang
- Department of Trauma-Emergency and Critical Care Medicine Center (TECCMC), Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Chunrong Wu
- Department of Trauma-Emergency and Critical Care Medicine Center (TECCMC), Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Xiangdong Xue
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
11
|
Zhang L, Chu W, Feng X, Li J, Ren Y, Yang Y, Zheng Z, Li H. Caveolin-1 protects retinal ganglion cells in glaucoma by reducing TLR4 and activating the Akt/PTEN signaling pathway. Pathol Res Pract 2024; 262:155552. [PMID: 39180803 DOI: 10.1016/j.prp.2024.155552] [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: 04/12/2024] [Revised: 08/05/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
Glaucoma is a degenerative disease characterized by retinal ganglion cell (RGC) death and visual impairment caused by elevated intraocular pressure (IOP). Elevated IOP can activate microglia, which participate in ganglion cell injury. Based on the study of caveolin-1 (Cav-1) in glaucoma, we aimed to explore the effect and mechanism of Cav-1 on RGC apoptosis in mice with acute ocular hypertension (AOH). AOH mice were established, and Cav-1 was intravitreally injected. Retinal microglia and RGCs were isolated from neonatal mice. TUNEL staining, hematoxylin-eosin staining, immunohistochemistry, flow cytometry, PCR and western blotting were used to observe the effect of Cav-1 on RGCs and mouse retinas. The thickness of the whole retina and the inner retinal sublayer decreased significantly, retinal cell apoptosis increased after AOH injury, and Cav-1 treatment reversed the effect of AOH injury. In addition, Cav-1 treatment promoted the conversion of proinflammatory M1 microglia to anti-inflammatory M2 microglia. Microglia and RGCs were isolated from neonatal mice. Cav-1 protects RGCs from OGD/R-induced injury by changing the polarization status of retinal microglia in vitro. Further studies revealed that Cav-1 activated the Akt/PTEN signaling pathway and inhibited TLR4. Our study provides evidence that Cav-1 may be a promising therapeutic target for glaucoma.
Collapse
Affiliation(s)
- Liwei Zhang
- Department of Ophthalmology, The Affiliated Hospital of Yunnan University; The Second People's Hospital of Yunnan Province; Yunnan Eye Hospital, Kunming, Yunnan 650021, China
| | - Wen Chu
- Dental Medicine Center, The Affiliated Hospital of Yunnan University; The Second People's Hospital of Yunnan Province; Yunnan Eye Hospital, Kunming, Yunnan 650031, China
| | - Xiaoxiao Feng
- Department of Ophthalmology, The Affiliated Hospital of Yunnan University; The Second People's Hospital of Yunnan Province; Yunnan Eye Hospital, Kunming, Yunnan 650021, China
| | - Juanjuan Li
- Department of Ophthalmology, The Affiliated Hospital of Yunnan University; The Second People's Hospital of Yunnan Province; Yunnan Eye Hospital, Kunming, Yunnan 650021, China
| | - Yuling Ren
- Department of Ophthalmology, The Affiliated Hospital of Yunnan University; The Second People's Hospital of Yunnan Province; Yunnan Eye Hospital, Kunming, Yunnan 650021, China
| | - Yabin Yang
- Department of Ophthalmology, The Affiliated Hospital of Yunnan University; The Second People's Hospital of Yunnan Province; Yunnan Eye Hospital, Kunming, Yunnan 650021, China
| | - Zhikun Zheng
- Department of Ophthalmology, The Affiliated Hospital of Yunnan University; The Second People's Hospital of Yunnan Province; Yunnan Eye Hospital, Kunming, Yunnan 650021, China.
| | - Hua Li
- Department of Ophthalmology, The Affiliated Hospital of Yunnan University; The Second People's Hospital of Yunnan Province; Yunnan Eye Hospital, Kunming, Yunnan 650021, China.
| |
Collapse
|
12
|
Filipovská E, Čočková Z, Černá B, Kubištová A, Spišská V, Telenský P, Bendová Z. The role of N6-methyladenosine RNA methylation in the crosstalk of circadian clock and neuroinflammation in rodent suprachiasmatic nuclei. Eur J Neurosci 2024; 60:4586-4596. [PMID: 39007275 DOI: 10.1111/ejn.16471] [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: 02/09/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024]
Abstract
N6-methyladenosine (m6A) is the most abundant epitranscriptomic mark that regulates the fate of RNA molecules. Recent studies have revealed a bidirectional interaction between m6A modification and the circadian clock. However, the precise temporal dynamics of m6A global enrichment in the central circadian pacemaker have not been fully elucidated. Our study investigates the relationship between FTO demethylase and molecular clocks in primary cells of the suprachiasmatic nucleus (SCN). In addition, we examined the effects of lipopolysaccharide (LPS) on Fto expression and the role of FTO in LPS-induced reactive oxygen species (ROS) production in primary SCN cell culture. We observed circadian rhythmicity in the global m6A levels, which mirrored the rhythmic expression of the Fto demethylase. Silencing FTO using siRNA reduced the mesor of Per2 rhythmicity in SCN primary cells and extended the period of the PER2 rhythm in SCN primary cell cultures from PER2::LUC mice. When examining the immune response, we discovered that exposure to LPS upregulated global m6A levels while downregulating Fto expression in SCN primary cell cultures. Interestingly, we found a loss of circadian rhythmicity in Fto expression following LPS treatment, indicating that the decrease of FTO levels may contribute to m6A upregulation without directly regulating its circadian rhythm. To explore potential protective mechanisms against neurotoxic inflammation, we examined ROS production following LPS treatment in SCN primary cell cultures pretreated with FTO siRNA. We observed a time-dependent pattern of ROS induction, with significant peak at 32 h but not at 20 h after synchronization. Silencing the FTO demethylase abolished ROS induction following LPS exposure, supporting the hypothesis that FTO downregulation serves as a protective mechanism during LPS-induced neuroinflammation in SCN primary cell cultures.
Collapse
Affiliation(s)
- Eva Filipovská
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Zuzana Čočková
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Barbora Černá
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Aneta Kubištová
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Veronika Spišská
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Petr Telenský
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
- Dementia Research Group, International Clinical Research Center of St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Zdeňka Bendová
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
- Department of Sleep Medicine and Chronobiology, National Institute of Mental Health, Klecany, Czech Republic
| |
Collapse
|
13
|
Cai P, Li W, Xu Y, Wang H. Drp1 and neuroinflammation: Deciphering the interplay between mitochondrial dynamics imbalance and inflammation in neurodegenerative diseases. Neurobiol Dis 2024; 198:106561. [PMID: 38857809 DOI: 10.1016/j.nbd.2024.106561] [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: 03/17/2024] [Revised: 05/24/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024] Open
Abstract
Neuroinflammation and mitochondrial dysfunction are closely intertwined with the pathophysiology of neurological disorders. Recent studies have elucidated profound alterations in mitochondrial dynamics across a spectrum of neurological disorders. Dynamin-related protein 1 (DRP1) emerges as a pivotal regulator of mitochondrial fission, with its dysregulation disrupting mitochondrial homeostasis and fueling neuroinflammation, thereby exacerbating disease severity. In addition to its role in mitochondrial dynamics, DRP1 plays a crucial role in modulating inflammation-related pathways. This review synthesizes important functions of DRP1 in the central nervous system (CNS) and the impact of epigenetic modification on the progression of neurodegenerative diseases. The intricate interplay between neuroinflammation and DRP1 in microglia and astrocytes, central contributors to neuroinflammation, is expounded upon. Furthermore, the use of DRP1 inhibitors to influence the activation of microglia and astrocytes, as well as their involvement in processes such as mitophagy, mitochondrial oxidative stress, and calcium ion transport in CNS-mediated neuroinflammation, is scrutinized. The modulation of microglia to astrocyte crosstalk by DRP1 and its role in inflammatory neurodegeneration is also highlighted. Overall, targeting DRP1 presents a promising avenue for ameliorating neuroinflammation and enhancing the therapeutic management of neurological disorders.
Collapse
Affiliation(s)
- Peiyang Cai
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Wuhao Li
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Ye Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Hui Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China..
| |
Collapse
|
14
|
Siddiqui AM, Sabljic TF, Ball AK. Anatomical location of injected microglia in different activation states and time course of injury determines survival of retinal ganglion cells after optic nerve crush. Int J Neurosci 2024; 134:677-699. [PMID: 36371721 DOI: 10.1080/00207454.2022.2142579] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 10/03/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Background: Activated microglia release harmful substances to retinal ganglion cells (RGCs), but may also benefit by removing cellular debris and secreting neurotrophic factors. These paradoxical roles remain controversial because the nature and time-course of the injury that defines their role is unknown. The aim of this study was to determine if pharmacological manipulation of microglia to acquire a pro-inflammatory or pro-survival phenotype will exacerbate or enhance neuronal survival after injury.Material and methods: Treated HAP I (highly aggressively proliferating immortalized) microglia were injected into the vitreous or tail vein (T V) of female Sprague-Dawley rats. Retinas were examined at 4-14 days following optic nerve crush (ONC) and the number of surviving RGCs was determined.Results: Injection of untreated HAP I cells resulted in the greater loss of RGCs early after ONC when injected into the vitreous and later after ONC when injected into the T V. LP S activated HAP I cells injected into the vitreous resulted in greater RGC loss with and without injury. When injected into the T V with ONC there was no loss of RGCs 4 days after ONC but greater loss afterwards. Minocycline treated HAP I cells injected into the vitreous resulted in greater RGC survival than untreated HAP I cells. However, when injected into the T V with ONC there was greater loss of RGCs. These results suggest that optic nerve signals attract extrinsic microglia to the retina, resulting in a proinflammatory response.Conclusion: Neuroprotection or cytotoxicity of microglia depends on the type of activation, time course of the injury, and if they act on the axon or cell body.
Collapse
Affiliation(s)
- Ahad M Siddiqui
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Thomas F Sabljic
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Alexander K Ball
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
15
|
Saleki K, Alijanizadeh P, Javanmehr N, Rezaei N. The role of Toll-like receptors in neuropsychiatric disorders: Immunopathology, treatment, and management. Med Res Rev 2024; 44:1267-1325. [PMID: 38226452 DOI: 10.1002/med.22012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 10/20/2023] [Accepted: 12/20/2023] [Indexed: 01/17/2024]
Abstract
Neuropsychiatric disorders denote a broad range of illnesses involving neurology and psychiatry. These disorders include depressive disorders, anxiety, schizophrenia, bipolar disorder, attention deficit hyperactivity disorder, autism spectrum disorders, headaches, and epilepsy. In addition to their main neuropathology that lies in the central nervous system (CNS), lately, studies have highlighted the role of immunity and neuroinflammation in neuropsychiatric disorders. Toll-like receptors (TLRs) are innate receptors that act as a bridge between the innate and adaptive immune systems via adaptor proteins (e.g., MYD88) and downstream elements; TLRs are classified into 13 families that are involved in normal function and illnesses of the CNS. TLRs expression affects the course of neuropsychiatric disorders, and is influenced during their pharmacotherapy; For example, the expression of multiple TLRs is normalized during the major depressive disorder pharmacotherapy. Here, the role of TLRs in neuroimmunology, treatment, and management of neuropsychiatric disorders is discussed. We recommend longitudinal studies to comparatively assess the cell-type-specific expression of TLRs during treatment, illness progression, and remission. Also, further research should explore molecular insights into TLRs regulation and related pathways.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
- Department of e-Learning, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Parsa Alijanizadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Nima Javanmehr
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
16
|
Qian Z, Xia M, Zhao T, Li Y, Li G, Zhang Y, Li H, Yang L. ACOD1, rather than itaconate, facilitates p62-mediated activation of Nrf2 in microglia post spinal cord contusion. Clin Transl Med 2024; 14:e1661. [PMID: 38644791 PMCID: PMC11033726 DOI: 10.1002/ctm2.1661] [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: 02/02/2024] [Revised: 03/31/2024] [Accepted: 04/02/2024] [Indexed: 04/23/2024] Open
Abstract
BACKGROUND Spinal cord injury (SCI)-induced neuroinflammation and oxidative stress (OS) are crucial events causing neurological dysfunction. Aconitate decarboxylase 1 (ACOD1) and its metabolite itaconate (Ita) inhibit inflammation and OS by promoting alkylation of Keap1 to induce Nrf2 expression; however, it is unclear whether there is another pathway regulating their effects in inflammation-activated microglia after SCI. METHODS Adult male C57BL/6 ACOD1-/- mice and their wild-type (WT) littermates were subjected to a moderate thoracic spinal cord contusion. The degree of neuroinflammation and OS in the injured spinal cord were assessed using qPCR, western blot, flow cytometry, immunofluorescence, and trans-well assay. We then employed immunoprecipitation-western blot, chromatin immunoprecipitation (ChIP)-PCR, dual-luciferase assay, and immunofluorescence-confocal imaging to examine the molecular mechanisms of ACOD1. Finally, the locomotor function was evaluated with the Basso Mouse Scale and footprint assay. RESULTS Both in vitro and in vivo, microglia with transcriptional blockage of ACOD1 exhibited more severe levels of neuroinflammation and OS, in which the expression of p62/Keap1/Nrf2 was down-regulated. Furthermore, silencing ACOD1 exacerbated neurological dysfunction in SCI mice. Administration of exogenous Ita or 4-octyl itaconate reduced p62 phosphorylation. Besides, ACOD1 was capable of interacting with phosphorylated p62 to enhance Nrf2 activation, which in turn further promoted transcription of ACOD1. CONCLUSIONS Here, we identified an unreported ACOD1-p62-Nrf2-ACOD1 feedback loop exerting anti-inflammatory and anti-OS in inflammatory microglia, and demonstrated the neuroprotective role of ACOD1 after SCI, which was different from that of endogenous and exogenous Ita. The present study extends the functions of ACOD1 and uncovers marked property differences between endogenous and exogenous Ita. KEY POINTS ACOD1 attenuated neuroinflammation and oxidative stress after spinal cord injury. ACOD1, not itaconate, interacted with p-p62 to facilitate Nrf2 expression and nuclear translocation. Nrf2 was capable of promoting ACOD1 transcription in microglia.
Collapse
Affiliation(s)
- Zhanyang Qian
- Department of OrthopedicsTaizhou School of Clinical MedicineTaizhou People's Hospital of Nanjing Medical University, Nanjing Medical UniversityTaizhouChina
| | - Mingjie Xia
- Department of Spine SurgeryNantong First People's HospitalThe Second Affiliated Hospital of Nantong UniversityNantongChina
| | - Tianyu Zhao
- Department of OrthopedicsTaizhou School of Clinical MedicineTaizhou People's Hospital of Nanjing Medical University, Nanjing Medical UniversityTaizhouChina
- Postgraduate SchoolDalian Medical UniversityDalianChina
| | - You Li
- Department of Trauma and Reconstructive SurgeryRWTH Aachen University HospitalAachenGermany
| | - Guangshen Li
- Department of OrthopedicsTaizhou School of Clinical MedicineTaizhou People's Hospital of Nanjing Medical University, Nanjing Medical UniversityTaizhouChina
| | - Yanan Zhang
- Department of OrthopedicsTaizhou School of Clinical MedicineTaizhou People's Hospital of Nanjing Medical University, Nanjing Medical UniversityTaizhouChina
- Postgraduate SchoolDalian Medical UniversityDalianChina
| | - Haijun Li
- Department of OrthopedicsTaizhou School of Clinical MedicineTaizhou People's Hospital of Nanjing Medical University, Nanjing Medical UniversityTaizhouChina
| | - Lei Yang
- Department of OrthopedicsTaizhou School of Clinical MedicineTaizhou People's Hospital of Nanjing Medical University, Nanjing Medical UniversityTaizhouChina
| |
Collapse
|
17
|
von Bernhardi R, Eugenín J. Aging Microglia and Their Impact in the Nervous System. ADVANCES IN NEUROBIOLOGY 2024; 37:379-395. [PMID: 39207703 DOI: 10.1007/978-3-031-55529-9_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Aging is the greatest risk factor for neurodegenerative diseases. Microglia are the resident immune cells in the central nervous system (CNS), playing key roles in its normal functioning, and as mediators for age-dependent changes of the CNS, condition at which they generate a hostile environment for neurons. Transforming Growth Factor β1 (TGFβ1) is a regulatory cytokine involved in immuneregulation and neuroprotection, affecting glial cell inflammatory activation, neuronal survival, and function. TGFβ1 signaling undergoes age-dependent changes affecting the regulation of microglial cells and can contribute to the pathophysiology of neurodegenerative diseases. This chapter focuses on assessing the role of age-related changes on the regulation of microglial cells and their impact on neuroinflammation and neuronal function, for understanding age-dependent changes of the nervous system.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Faculty of Odontology and Rehabilitation Sciences, Universidad San Sebastian, Santiago, Chile.
| | - Jaime Eugenín
- Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
18
|
Castro R, Lopes M, De Biase L, Valdez G. Aging spinal cord microglia become phenotypically heterogeneous and preferentially target motor neurons and their synapses. Glia 2024; 72:206-221. [PMID: 37737058 PMCID: PMC10773989 DOI: 10.1002/glia.24470] [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: 06/01/2023] [Revised: 08/07/2023] [Accepted: 09/03/2023] [Indexed: 09/23/2023]
Abstract
Microglia have been found to acquire unique region-dependent deleterious features with age and diseases that contribute to neuronal dysfunction and degeneration in the brain. However, it remains unknown whether microglia exhibit similar phenotypic heterogeneity in the spinal cord. Here, we performed a regional analysis of spinal cord microglia in 3-, 16-, 23-, and 30-month-old mice. Using light and electron microscopy, we discovered that spinal cord microglia acquire an increasingly activated phenotype during the course of aging regardless of regional location. However, aging causes microglia in the ventral but not dorsal horn to lose their spatial organization. Aged ventral horn microglia also aggregate around the somata of motor neurons and increase their contacts with motor synapses, which have been shown to be lost with age. These findings suggest that microglia may affect the ability of motor neurons to receive and relay motor commands during aging. To generate additional insights about aging spinal cord microglia, we performed RNA-sequencing on FACS-isolated microglia from 3-, 18-, 22-, and 29-month-old mice. We found that spinal cord microglia acquire a similar transcriptional identity as those in the brain during aging that includes altered expression of genes with roles in microglia-neuron interactions and inflammation. By 29 months of age, spinal cord microglia exhibit additional and unique transcriptional changes known and predicted to cause senescence and to alter lysosomal and ribosomal regulation. Altogether, this work provides the foundation to target microglia to ameliorate aged-related changes in the spinal cord, and particularly on the motor circuit.
Collapse
Affiliation(s)
- Ryan Castro
- Neuroscience Graduate Program, Brown University, Providence, Rhode Island, USA
| | - Mikayla Lopes
- Molecular Biology, Cell Biology & Biochemistry Graduate Program, Brown University, Providence, Rhode Island, USA
| | - Lindsay De Biase
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Gregorio Valdez
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, Rhode Island, USA
- Department of Neurology, Warren Alpert Medical School of Medicine, Brown University, Providence, USA
| |
Collapse
|
19
|
Strogulski NR, Portela LV, Polster BM, Loane DJ. Fundamental Neurochemistry Review: Microglial immunometabolism in traumatic brain injury. J Neurochem 2023; 167:129-153. [PMID: 37759406 PMCID: PMC10655864 DOI: 10.1111/jnc.15959] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023]
Abstract
Traumatic brain injury (TBI) is a devastating neurological disorder caused by a physical impact to the brain that promotes diffuse damage and chronic neurodegeneration. Key mechanisms believed to support secondary brain injury include mitochondrial dysfunction and chronic neuroinflammation. Microglia and brain-infiltrating macrophages are responsible for neuroinflammatory cytokine and reactive oxygen species (ROS) production after TBI. Their production is associated with loss of homeostatic microglial functions such as immunosurveillance, phagocytosis, and immune resolution. Beyond providing energy support, mitochondrial metabolic pathways reprogram the pro- and anti-inflammatory machinery in immune cells, providing a critical immunometabolic axis capable of regulating immunologic response to noxious stimuli. In the brain, the capacity to adapt to different environmental stimuli derives, in part, from microglia's ability to recognize and respond to changes in extracellular and intracellular metabolite levels. This capacity is met by an equally plastic metabolism, capable of altering immune function. Microglial pro-inflammatory activation is associated with decreased mitochondrial respiration, whereas anti-inflammatory microglial polarization is supported by increased oxidative metabolism. These metabolic adaptations contribute to neuroimmune responses, placing mitochondria as a central regulator of post-traumatic neuroinflammation. Although it is established that profound neurometabolic changes occur following TBI, key questions related to metabolic shifts in microglia remain unresolved. These include (a) the nature of microglial mitochondrial dysfunction after TBI, (b) the hierarchical positions of different metabolic pathways such as glycolysis, pentose phosphate pathway, glutaminolysis, and lipid oxidation during secondary injury and recovery, and (c) how immunometabolism alters microglial phenotypes, culminating in chronic non-resolving neuroinflammation. In this basic neurochemistry review article, we describe the contributions of immunometabolism to TBI, detail primary evidence of mitochondrial dysfunction and metabolic impairments in microglia and macrophages, discuss how major metabolic pathways contribute to post-traumatic neuroinflammation, and set out future directions toward advancing immunometabolic phenotyping in TBI.
Collapse
Affiliation(s)
- Nathan R. Strogulski
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Luis V. Portela
- Neurotrauma and Biomarkers Laboratory, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Brian M. Polster
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - David J. Loane
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
20
|
You Y, Liu Y, Ma C, Xu J, Xie L, Tong S, Sun Y, Ma F, Huang Y, Liu J, Xiao W, Dai C, Li S, Lei J, Mei Q, Gao X, Chen J. Surface-tethered ROS-responsive micelle backpacks for boosting mesenchymal stem cell vitality and modulating inflammation in ischemic stroke treatment. J Control Release 2023; 362:210-224. [PMID: 37619863 DOI: 10.1016/j.jconrel.2023.08.039] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/15/2023] [Accepted: 08/20/2023] [Indexed: 08/26/2023]
Abstract
Mesenchymal stem cells (MSCs) exhibited remarkable therapeutic potential in ischemic stroke due to their exceptional immunomodulatory ability and paracrine effect; they have also been regarded as excellent neuroprotectant delivery vehicles with inflammatory tropism. However, the presence of high levels of reactive oxygen species (ROS) and an oxidative stress environment at the lesion site inhibits cell survival and further therapeutic effects. Using bioorthogonal click chemistry, ROS-responsive luteolin-loaded micelles were tethered to the surface of MSCs. As MSCs migrated to the ischemic brain, the micelles would achieve ROS-responsive release of luteolin to protect MSCs from excessive oxidative damage while inhibiting neuroinflammation and scavenging ROS to ameliorate ischemic stroke. This study provided an effective and prospective therapeutic strategy for ischemic stroke and a framework for a stem cell-based therapeutic system to treat inflammatory cerebral diseases.
Collapse
Affiliation(s)
- Yang You
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China
| | - Yipu Liu
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China
| | - Chuchu Ma
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China
| | - Jianpei Xu
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China
| | - Laozhi Xie
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China
| | - Shiqiang Tong
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China
| | - Yinzhe Sun
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China
| | - Fenfen Ma
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China
| | - Yukun Huang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Junbin Liu
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai 200003, China
| | - Wenze Xiao
- Department of Rheumatology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Shanghai 201399, China
| | - Chengxiang Dai
- Daxing Research Institute, University of Science and Technology Beijing, 41 Yongda Road, Biomedical Industry Base, Zhongguancun Science and Technology Park, Daxing District, Beijing 102600, China; Cellular Biomedicine Group, Inc., 85 Faladi Road, Building 3, Zhangjiang, Pudong New Area, Shanghai 201210, China
| | - Suke Li
- Cellular Biomedicine Group, Inc., 85 Faladi Road, Building 3, Zhangjiang, Pudong New Area, Shanghai 201210, China
| | - Jigang Lei
- Cellular Biomedicine Group, Inc., 85 Faladi Road, Building 3, Zhangjiang, Pudong New Area, Shanghai 201210, China
| | - Qiyong Mei
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai 200003, China.
| | - Xiaoling Gao
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| | - Jun Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China.
| |
Collapse
|
21
|
Eugenín J, Eugenín-von Bernhardi L, von Bernhardi R. Age-dependent changes on fractalkine forms and their contribution to neurodegenerative diseases. Front Mol Neurosci 2023; 16:1249320. [PMID: 37818457 PMCID: PMC10561274 DOI: 10.3389/fnmol.2023.1249320] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/06/2023] [Indexed: 10/12/2023] Open
Abstract
The chemokine fractalkine (FKN, CX3CL1), a member of the CX3C subfamily, contributes to neuron-glia interaction and the regulation of microglial cell activation. Fractalkine is expressed by neurons as a membrane-bound protein (mCX3CL1) that can be cleaved by extracellular proteases generating several sCX3CL1 forms. sCX3CL1, containing the chemokine domain, and mCX3CL1 have high affinity by their unique receptor (CX3CR1) which, physiologically, is only found in microglia, a resident immune cell of the CNS. The activation of CX3CR1contributes to survival and maturation of the neural network during development, glutamatergic synaptic transmission, synaptic plasticity, cognition, neuropathic pain, and inflammatory regulation in the adult brain. Indeed, the various CX3CL1 forms appear in some cases to serve an anti-inflammatory role of microglia, whereas in others, they have a pro-inflammatory role, aggravating neurological disorders. In the last decade, evidence points to the fact that sCX3CL1 and mCX3CL1 exhibit selective and differential effects on their targets. Thus, the balance in their level and activity will impact on neuron-microglia interaction. This review is focused on the description of factors determining the emergence of distinct fractalkine forms, their age-dependent changes, and how they contribute to neuroinflammation and neurodegenerative diseases. Changes in the balance among various fractalkine forms may be one of the mechanisms on which converge aging, chronic CNS inflammation, and neurodegeneration.
Collapse
Affiliation(s)
- Jaime Eugenín
- Facultad de Química y Biología, Departamento de Biología, Universidad de Santiago de Chile, USACH, Santiago, Chile
| | | | - Rommy von Bernhardi
- Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Santiago, Chile
| |
Collapse
|
22
|
Vershinina YS, Krasnov GS, Garbuz DG, Shaposhnikov MV, Fedorova MS, Pudova EA, Katunina IV, Kornev AB, Zemskaya NV, Kudryavtsev AA, Bulavkina EV, Matveeva AA, Ulyasheva NS, Guvatova ZG, Anurov AA, Moskalev AA, Kudryavtseva AV. Transcriptomic Analysis of the Effect of Torin-2 on the Central Nervous System of Drosophila melanogaster. Int J Mol Sci 2023; 24:ijms24109095. [PMID: 37240439 DOI: 10.3390/ijms24109095] [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: 02/02/2023] [Revised: 04/24/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Torin-2, a synthetic compound, is a highly selective inhibitor of both TORC1 and TORC2 (target of rapamycin) complexes as an alternative to the well-known immunosuppressor, geroprotector, and potential anti-cancer natural compound rapamycin. Torin-2 is effective at hundreds of times lower concentrations and prevents some negative side effects of rapamycin. Moreover, it inhibits the rapamycin-resistant TORC2 complex. In this work, we evaluated transcriptomic changes in D. melanogaster heads induced with lifetime diets containing Torin-2 and suggested possible neuroprotective mechanisms of Torin-2. The analysis included D. melanogaster of three ages (2, 4, and 6 weeks old), separately for males and females. Torin-2, taken at the lowest concentration being tested (0.5 μM per 1 L of nutrient paste), had a slight positive effect on the lifespan of D. melanogaster males (+4% on the average) and no positive effect on females. At the same time, RNA-Seq analysis revealed interesting and previously undiscussed effects of Torin-2, which differed between sexes as well as in flies of different ages. Among the cellular pathways mostly altered by Torin-2 at the gene expression level, we identified immune response, protein folding (heat shock proteins), histone modification, actin cytoskeleton organization, phototransduction and sexual behavior. Additionally, we revealed that Torin-2 predominantly reduced the expression of Srr gene responsible for the conversion of L-serine to D-serine and thus regulating activity of NMDA receptor. Via western blot analysis, we showed than in old males Torin-2 tends to increase the ratio of the active phosphorylated form of ERK, the lowest node of the MAPK cascade, which may play a significant role in neuroprotection. Thus, the complex effect of Torin-2 may be due to the interplay of the immune system, hormonal background, and metabolism. Our work is of interest for further research in the field of NMDA-mediated neurodegeneration.
Collapse
Affiliation(s)
- Yulia S Vershinina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - George S Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - David G Garbuz
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | | | - Maria S Fedorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Elena A Pudova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Irina V Katunina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alexey B Kornev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Nadezhda V Zemskaya
- Institute of Biology, Komi Science Center, Ural Branch of RAS, 167000 Syktyvkar, Russia
| | - Alexander A Kudryavtsev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Elizaveta V Bulavkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anna A Matveeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Natalia S Ulyasheva
- Institute of Biology, Komi Science Center, Ural Branch of RAS, 167000 Syktyvkar, Russia
| | - Zulfiya G Guvatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Artemiy A Anurov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alexey A Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anna V Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
23
|
Huang Y, Zhang X, Zou Y, Yuan Q, Xian YF, Lin ZX. Quercetin Ameliorates Neuropathic Pain after Brachial Plexus Avulsion via Suppressing Oxidative Damage through Inhibition of PKC/MAPK/ NOX Pathway. Curr Neuropharmacol 2023; 21:2343-2361. [PMID: 37533160 PMCID: PMC10556381 DOI: 10.2174/1570159x21666230802144940] [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: 11/07/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 08/04/2023] Open
Abstract
BACKGROUND Brachial plexus avulsion (BPA) animally involves the separation of spinal nerve roots themselves and the correlative spinal cord segment, leading to formidable neuropathic pain of the upper limb. METHODS The right seventh cervical (C7) ventral and dorsal roots were avulsed to establish a neuropathic pain model in rats. After operation, rats were treated with quercetin (QCN) by intragastric administration for 1 week. The effects of QCN were evaluated using mechanical allodynia tests and biochemical assay kits. RESULTS QCN treatment significantly attenuated the avulsion-provoked mechanical allodynia, elevated the levels of catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GPx) and total antioxidant capacity (TAC) in the C7 spinal dorsal horn. In addition, QCN administration inhibited the activations of macrophages, microglia and astrocytes in the C6 dorsal root ganglion (DRG) and C6-8 spinal dorsal horn, as well as attenuated the release of purinergic 2X (P2X) receptors in C6 DRG. The molecular mechanism underlying the above alterations was found to be related to the suppression of the PKC/MAPK/NOX signal pathway. To further study the anti-oxidative effects of QCN, we applied QCN on the H2O2-induced BV-2 cells in vitro, and the results attested that QCN significantly ameliorated the H2O2-induced ROS production in BV-2 cells, inhibited the H2O2-induced activation of PKC/MAPK/NOX pathway. CONCLUSION Our study for the first time provided evidence that QCN was able to attenuate pain hypersensitivity following the C7 spinal root avulsion in rats, and the molecular mechanisms involve the reduction of both neuro-inflammatory infiltration and oxidative stress via suppression of P2X receptors and inhibition of the activation of PKC/MAPK/NOX pathway. The results indicate that QCN is a natural compound with great promise worthy of further development into a novel therapeutic method for the treatment of BPA-induced neuropathic pain.
Collapse
Affiliation(s)
- Yanfeng Huang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Xie Zhang
- Research Center for Integrative Medicine of Guangzhou University of Chinese Medicine, Key Laboratory of Chinese Medicine Pathogenesis and Therapy Research, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong. P.R. China
- Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong. P.R. China
| | - Yidan Zou
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Qiuju Yuan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Shatin, N.T., Hong Kong SAR, China
| | - Yan-Fang Xian
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
24
|
Javanmehr N, Saleki K, Alijanizadeh P, Rezaei N. Microglia dynamics in aging-related neurobehavioral and neuroinflammatory diseases. J Neuroinflammation 2022; 19:273. [PMID: 36397116 PMCID: PMC9669544 DOI: 10.1186/s12974-022-02637-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022] Open
Abstract
Microglia represent the first line of immune feedback in the brain. Beyond immune surveillance, they are essential for maintaining brain homeostasis. Recent research has revealed the microglial cells' spatiotemporal heterogeneity based on their local and time-based functions in brain trauma or disease when homeostasis is disrupted. Distinct "microglial signatures" have been recorded in physiological states and brain injuries, with discrete or sometimes overlapping pro- and anti-inflammatory functions. Microglia are involved in the neurological repair processes, such as neurovascular unit restoration and synaptic plasticity, and manage the extent of the damage due to their phenotype switching. The versatility of cellular phenotypes beyond the classical M1/M2 classification, as well as the double-edge actions of microglia in neurodegeneration, indicate the need for further exploration of microglial cell dynamics and their contribution to neurodegenerative processes. This review discusses the homeostatic functions of different microglial subsets focusing on neuropathological conditions. Also, we address the feasibility of targeting microglia as a therapeutic strategy in neurodegenerative diseases.
Collapse
Affiliation(s)
- Nima Javanmehr
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Parsa Alijanizadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
25
|
Smith AN, Shaughness M, Collier S, Hopkins D, Byrnes KR. Therapeutic targeting of microglia mediated oxidative stress after neurotrauma. Front Med (Lausanne) 2022; 9:1034692. [PMID: 36405593 PMCID: PMC9671221 DOI: 10.3389/fmed.2022.1034692] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/12/2022] [Indexed: 10/06/2023] Open
Abstract
Inflammation is a primary component of the central nervous system injury response. Traumatic brain and spinal cord injury are characterized by a pronounced microglial response to damage, including alterations in microglial morphology and increased production of reactive oxygen species (ROS). The acute activity of microglia may be beneficial to recovery, but continued inflammation and ROS production is deleterious to the health and function of other cells. Microglial nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX), mitochondria, and changes in iron levels are three of the most common sources of ROS. All three play a significant role in post-traumatic brain and spinal cord injury ROS production and the resultant oxidative stress. This review will evaluate the current state of therapeutics used to target these avenues of microglia-mediated oxidative stress after injury and suggest avenues for future research.
Collapse
Affiliation(s)
- Austin N. Smith
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Michael Shaughness
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Sean Collier
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Deanna Hopkins
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Kimberly R. Byrnes
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
26
|
Yamamoto Y, Kadoya K, Terkawi MA, Endo T, Konno K, Watanabe M, Ichihara S, Hara A, Kaneko K, Iwasaki N, Ishijima M. Neutrophils delay repair process in Wallerian degeneration by releasing NETs outside the parenchyma. Life Sci Alliance 2022; 5:e202201399. [PMID: 35961782 PMCID: PMC9375156 DOI: 10.26508/lsa.202201399] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 12/05/2022] Open
Abstract
Although inflammation is indispensable for the repair process in Wallerian degeneration (WD), the role of neutrophils in the WD repair process remains unclear. After peripheral nerve injury, neutrophils accumulate at the epineurium but not the parenchyma in the WD region because of the blood-nerve barrier. An increase or decrease in the number of neutrophils delayed or promoted macrophage infiltration from the epineurium into the parenchyma and the repair process in WD. Abundant neutrophil extracellular traps (NETs) were formed around neutrophils, and its inhibition dramatically increased macrophage infiltration into the parenchyma. Furthermore, inhibition of either MIF or its receptor, CXCR4, in neutrophils decreased NET formation, resulting in enhanced macrophage infiltration into the parenchyma. Moreover, inhibiting MIF for just 2 h after peripheral nerve injury promoted the repair process. These findings indicate that neutrophils delay the repair process in WD from outside the parenchyma by inhibiting macrophage infiltration via NET formation and that neutrophils, NETs, MIF, and CXCR4 are therapeutic targets for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Yasuhiro Yamamoto
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Department of Medicine for Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ken Kadoya
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Mohamad Alaa Terkawi
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takeshi Endo
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kohtarou Konno
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Satoshi Ichihara
- Department of Orthopaedic Surgery, Juntendo University Urayasu Hospital, Urayasu, Japan
| | - Akira Hara
- Department of Orthopaedic Surgery, Juntendo University Urayasu Hospital, Urayasu, Japan
| | - Kazuo Kaneko
- Department of Medicine for Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Norimasa Iwasaki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Muneaki Ishijima
- Department of Medicine for Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
27
|
Illias AM, Yu KJ, Hwang SH, Solis J, Zhang H, Velasquez JF, Cata JP, Dougherty PM. Dorsal root ganglion toll-like receptor 4 signaling contributes to oxaliplatin-induced peripheral neuropathy. Pain 2022; 163:923-935. [PMID: 34490849 DOI: 10.1097/j.pain.0000000000002454] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 08/10/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Activation of toll-like receptor 4 (TLR4) in the dorsal root ganglion (DRG) and spinal cord contributes to the generation of paclitaxel-related chemotherapy-induced peripheral neuropathy (CIPN). Generalizability of TLR4 signaling in oxaliplatin-induced CIPN was tested here. Mechanical hypersensitivity developed in male SD rats by day 1 after oxaliplatin treatment, reached maximum intensity by day 14, and persisted through day 35. Western blot revealed an increase in TLR4 expression in the DRG of oxaliplatin at days 1 and 7 after oxaliplatin treatment. Cotreatment of rats with the TLR4 antagonist lipopolysaccharide derived from Rhodobacter sphaeroides ultrapure or with the nonspecific immunosuppressive minocycline with oxaliplatin resulted in significantly attenuated hyperalgesia on day 7 and 14 compared with rats that received oxaliplatin plus saline vehicle. Immunostaining of DRGs revealed an increase in the number of neurons expressing TLR4, its canonical downstream signal molecules myeloid differentiation primary response gene 88 (MyD88) and TIR-domain-containing adapter-inducing interferon-β, at both day 7 and day 14 after oxaliplatin treatment. These increases were blocked by cotreatment with either lipopolysaccharide derived from Rhodobacter sphaeroides or minocycline. Double staining showed the localization of TLR4, MyD88, and TIR-domain-containing adapter-inducing interferon-β in subsets of DRG neurons. Finally, there was no significant difference in oxaliplatin-induced mechanical hypersensitivity between male and female rats when observed for 2 weeks. Furthermore, upregulation of TLR4 was detected in both sexes when tested 14 days after treatment with oxaliplatin. These findings suggest that the activation of TLR4 signaling in DRG neurons is a common mechanism in CIPN induced by multiple cancer chemotherapy agents.
Collapse
Affiliation(s)
- Amina M Illias
- Department of Anesthesiology and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kai-Jie Yu
- Department of Urology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Seon-Hee Hwang
- Department of Anesthesiology and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Jacob Solis
- Department of Anesthesiology and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Hongmei Zhang
- Department of Anesthesiology and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Jose F Velasquez
- Department of Anesthesiology and Perioperative Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Juan P Cata
- Department of Anesthesiology and Perioperative Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Patrick M Dougherty
- Department of Anesthesiology and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
28
|
Pang QM, Chen SY, Xu QJ, Zhang M, Liang DF, Fu SP, Yu J, Liu ZL, Zhang Q, Zhang T. Effects of astrocytes and microglia on neuroinflammation after spinal cord injury and related immunomodulatory strategies. Int Immunopharmacol 2022; 108:108754. [PMID: 35397392 DOI: 10.1016/j.intimp.2022.108754] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/14/2022] [Accepted: 03/31/2022] [Indexed: 12/12/2022]
Abstract
Spinal cord injury (SCI) is a catastrophic event which is still without adequate therapies. Neuroinflammation is the main pathogenesis of secondary damage post-SCI, leading to tissue loss and neurological dysfunction. Previous studies have shown that microglia and astrocytes are the major immune cells in the central nervous system (CNS) and play a crucial role in modulating neuroinflammatory responses. In this study, we mainly review the effects of neuroinflammation in SCI, focusing on the contributions of microglia and astrocytes and their cross-talk. Furthermore, we will also discuss therapeutic strategies on how to regulate their immunophenotype to suppress robust inflammation and facilitate injury prognosis.
Collapse
Affiliation(s)
- Qi-Ming Pang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China; Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Si-Yu Chen
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Qi-Jing Xu
- Department of Human Anatomy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Meng Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Da-Fei Liang
- Department of Human Anatomy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Sheng-Ping Fu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China; Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Jiang Yu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Zu-Lin Liu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Qian Zhang
- Department of Human Anatomy, Zunyi Medical University, Zunyi, Guizhou, China.
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China; Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
29
|
Chen G, Yan F, Wei W, Wang F, Wang Z, Nie J, Jin M, Pang Y, Qin M, Wang L, Zhang X. CD38 deficiency protects the retina from ischaemia/reperfusion injury partly via suppression of TLR4/MyD88/NF-κB signalling. Exp Eye Res 2022; 219:109058. [PMID: 35364100 DOI: 10.1016/j.exer.2022.109058] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/14/2022] [Accepted: 03/25/2022] [Indexed: 12/22/2022]
Abstract
PURPOSE This study aimed to explore cellular localisation of CD38 in the retina and evaluate the role and potential mechanism of CD38 deficiency in retinal ischaemia/reperfusion (I/R) injury. METHODS Six-to eight-week-old male CD38 knockout (KO) and wild-type mice in C57BL/6 background were used. Immunostaining was performed to determine the cellular localisation of CD38 in the retina. Haematoxylin and eosin staining and immunostaining of Brn3a were used to evaluate the retinal I/R injury. Western blotting was performed to detect toll-like receptor 4 (TLR4), myeloid differentiation primary response 88 (MyD88), p-p65, ionised calcium-binding adapter molecule 1, Sirtuin1 (Sirt1), Ac-p65, and pro-inflammatory cytokines protein expression. RESULTS CD38 was highly expressed in mouse retinal microglia and astrocytes/Müller cells. CD38 deficiency reduced I/R-induced retinal damage and retinal ganglion cell death. Following retinal I/R injury, TLR4, MyD88, nuclear factor-κB p-p65 (NF-κB p-p65), pro-inflammatory cytokines and CD38 protein levels were also upregulated. After I/R injury, retinal inflammation factors IL-1β, IL-6, and TNF-α mRNA and protein levels were increased. IL-1β, IL-6, and TNF-α were reduced in CD38 KO mice after I/R injury. Retinal I/R injury induced the activation of microglia, but this effect was also suppressed by KO of CD38. Additionally, retinal I/R induced a significant increase in Ac-p65 protein levels and decrease in Sirt1 protein levels, while this effect was greatly attenuated by KO of CD38. CONCLUSION CD38 deficiency protects the retina from I/R injury by suppressing microglial activation partly via activating Sirt1-mediated suppression of TLR4/MyD88/NF-κB signalling.
Collapse
Affiliation(s)
- Guiping Chen
- Affiliated Eye Hospital of Nanchang University, Jiangxi Clinical Research Center of Ophthalmic Disease, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China
| | - Feng Yan
- Affiliated Eye Hospital of Nanchang University, Jiangxi Clinical Research Center of Ophthalmic Disease, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China; School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Wei Wei
- Affiliated Eye Hospital of Nanchang University, Jiangxi Clinical Research Center of Ophthalmic Disease, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China
| | - Feifei Wang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Clinical Research Center of Ophthalmic Disease, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China
| | - Zhiruo Wang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Clinical Research Center of Ophthalmic Disease, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China
| | - Jiahe Nie
- Affiliated Eye Hospital of Nanchang University, Jiangxi Clinical Research Center of Ophthalmic Disease, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China
| | - Ming Jin
- Affiliated Eye Hospital of Nanchang University, Jiangxi Clinical Research Center of Ophthalmic Disease, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China
| | - Yulian Pang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Clinical Research Center of Ophthalmic Disease, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China
| | - Mengqi Qin
- Affiliated Eye Hospital of Nanchang University, Jiangxi Clinical Research Center of Ophthalmic Disease, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China
| | - Lingfang Wang
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Xu Zhang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Clinical Research Center of Ophthalmic Disease, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China.
| |
Collapse
|
30
|
Kuo HC, Lee KF, Chen SL, Chiu SC, Lee LY, Chen WP, Chen CC, Chu CH. Neuron–Microglia Contacts Govern the PGE2 Tolerance through TLR4-Mediated de Novo Protein Synthesis. Biomedicines 2022; 10:biomedicines10020419. [PMID: 35203628 PMCID: PMC8962342 DOI: 10.3390/biomedicines10020419] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 02/02/2022] [Accepted: 02/07/2022] [Indexed: 02/05/2023] Open
Abstract
Cellular and molecular mechanisms of the peripheral immune system (e.g., macrophage and monocyte) in programming endotoxin tolerance (ET) have been well studied. However, regulatory mechanism in development of brain immune tolerance remains unclear. The inducible COX-2/PGE2 axis in microglia, the primary innate immune cells of the brain, is a pivotal feature in causing inflammation and neuronal injury, both in acute excitotoxic insults and chronic neurodegenerative diseases. This present study investigated the regulatory mechanism of PGE2 tolerance in microglia. Multiple reconstituted primary brain cells cultures, including neuron–glial (NG), mixed glial (MG), neuron-enriched, and microglia-enriched cultures, were performed and consequently applied to a treatment regimen for ET induction. Our results revealed that the levels of COX-2 mRNA and supernatant PGE2 in NG cultures, but not in microglia-enriched and MG cultures, were drastically reduced in response to the ET challenge, suggesting that the presence of neurons, rather than astroglia, is required for PGE2 tolerance in microglia. Furthermore, our data showed that neural contact, instead of its soluble factors, is sufficient for developing microglial PGE2 tolerance. Simultaneously, this finding determined how neurons regulated microglial PGE2 tolerance. Moreover, by inhibiting TLR4 activation and de novo protein synthesis by LPS-binding protein (LBP) manipulation and cycloheximide, our data showed that the TLR4 signal and de novo protein synthesis are necessary for microglia to develop PGE2 tolerance in NG cells under the ET challenge. Altogether, our findings demonstrated that neuron–microglia contacts are indispensable in emerging PGE2 tolerance through the regulation of TLR4-mediated de novo protein synthesis.
Collapse
Affiliation(s)
- Hsing-Chun Kuo
- Division of Basic Medical Sciences, Department of Nursing, Chang Gung University of Science and Technology, Chiayi 61363, Taiwan;
- Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan
- Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Chiayi 61363, Taiwan
| | - Kam-Fai Lee
- Department of Pathology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan;
| | - Shiou-Lan Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University (KMU), Kaohsiung 80708, Taiwan;
| | - Shu-Chen Chiu
- National Laboratory Animal Center (NLAC), NARLabs, Tainan 74147, Taiwan;
| | - Li-Ya Lee
- Grape King Biotechnology Inc (Grape King Bio Ltd.), Zhong-Li, Taoyuan 32542, Taiwan; (L.-Y.L.); (W.-P.C.); (C.-C.C.)
| | - Wan-Ping Chen
- Grape King Biotechnology Inc (Grape King Bio Ltd.), Zhong-Li, Taoyuan 32542, Taiwan; (L.-Y.L.); (W.-P.C.); (C.-C.C.)
| | - Chin-Chu Chen
- Grape King Biotechnology Inc (Grape King Bio Ltd.), Zhong-Li, Taoyuan 32542, Taiwan; (L.-Y.L.); (W.-P.C.); (C.-C.C.)
| | - Chun-Hsien Chu
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan 70456, Taiwan
- Correspondence: or ; Tel.: +886-6-235-3535 (ext. 3592); Fax: +886-6-209-5845
| |
Collapse
|
31
|
Markoutsa E, Mayilsamy K, Gulick D, Mohapatra SS, Mohapatra S. Extracellular vesicles derived from inflammatory-educated stem cells reverse brain inflammation-implication of miRNAs. Mol Ther 2022; 30:816-830. [PMID: 34371179 PMCID: PMC8821927 DOI: 10.1016/j.ymthe.2021.08.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 07/04/2021] [Accepted: 07/29/2021] [Indexed: 02/04/2023] Open
Abstract
Inflammation plays a key role in the development of age-related diseases. In Alzheimer's disease, neuronal cell death is attributed to amyloidbeta oligomers that trigger microglial activation. Stem cells have shown promise as therapies for inflammatory diseases- because of their paracrine activity combined with their ability to respond to the inflammatory environment. However, the mechanisms underlying stem cell-promoted neurological recovery are poorly understood. To elucidate these mechanisms, we first primed stem cells with the secretome of lipopolysaccharide- or amyloidbeta-activated microglia. Then, we compared the immunomodulatory effects of extracellular vesicles (EVs) secreted from primed and non-primed stem cells. Our results demonstrate that EVs from primed cells are more effective in inhibiting microglia and astrocyte activation, amyloid deposition, demyelination, memory loss and motor and anxiety-like behavioral dysfunction, compared to EVs from non-primed cells. MicroRNA (miRNA) profiling revealed the upregulation of at least 19 miRNAs on primed-stem cell EVs. The miRNA targets were identified, and KEGG pathway analysis showed that the overexpressed miRNAs target key genes on the toll-like receptor-4 (TLR4) signaling pathway. Overall, our results demonstrate that priming mesenchymal stem cells (MSCs) with the secretome of activated microglia results in the release of miRNAs from EVs with enhanced immune regulatory potential able to fight neuroinflammation.
Collapse
Affiliation(s)
- Eleni Markoutsa
- James A. Haley VA Hospital, Tampa, FL 33612, USA,Division of Translational Medicine and Center for Research and Education in Nanobio-engineering, Department of Internal Medicine, University of South Florida Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA,College of Pharmacy Graduate Programs, University of South Florida, Tampa, FL, USA,Corresponding author: Eleni Markoutsa, Division of Translational Medicine and Center for Research and Education in Nanobio-engineering, Department of Internal Medicine, University of South Florida Mrsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Karthick Mayilsamy
- James A. Haley VA Hospital, Tampa, FL 33612, USA,Department of Molecular Medicine, University of South Florida Mrsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, USA
| | - Dannielle Gulick
- Department of Molecular Medicine, University of South Florida Mrsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, USA
| | - Shyam S. Mohapatra
- James A. Haley VA Hospital, Tampa, FL 33612, USA,Division of Translational Medicine and Center for Research and Education in Nanobio-engineering, Department of Internal Medicine, University of South Florida Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA,College of Pharmacy Graduate Programs, University of South Florida, Tampa, FL, USA
| | - Subhra Mohapatra
- James A. Haley VA Hospital, Tampa, FL 33612, USA,Department of Molecular Medicine, University of South Florida Mrsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, USA,Corresponding author: Subhra Mohapatra, Department of Molecular Medicine, University of South Florida Mrsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
32
|
Nisa FY, Rahman MA, Hossen MA, Khan MF, Khan MAN, Majid M, Sultana F, Haque MA. Role of neurotoxicants in the pathogenesis of Alzheimer's disease: a mechanistic insight. Ann Med 2021; 53:1476-1501. [PMID: 34433343 PMCID: PMC8405119 DOI: 10.1080/07853890.2021.1966088] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/04/2021] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most conspicuous chronic neurodegenerative syndrome, which has become a significant challenge for the global healthcare system. Multiple studies have corroborated a clear association of neurotoxicants with AD pathogenicity, such as Amyloid beta (Aβ) proteins and neurofibrillary tangles (NFTs), signalling pathway modifications, cellular stress, cognitive dysfunctions, neuronal apoptosis, neuroinflammation, epigenetic modification, and so on. This review, therefore, aimed to address several essential mechanisms and signalling cascades, including Wnt (wingless and int.) signalling pathway, autophagy, mammalian target of rapamycin (mTOR), protein kinase C (PKC) signalling cascades, cellular redox status, energy metabolism, glutamatergic neurotransmissions, immune cell stimulations (e.g. microglia, astrocytes) as well as an amyloid precursor protein (APP), presenilin-1 (PSEN1), presenilin-2 (PSEN2) and other AD-related gene expressions that have been pretentious and modulated by the various neurotoxicants. This review concluded that neurotoxicants play a momentous role in developing AD through modulating various signalling cascades. Nevertheless, comprehension of this risk agent-induced neurotoxicity is far too little. More in-depth epidemiological and systematic investigations are needed to understand the potential mechanisms better to address these neurotoxicants and improve approaches to their risk exposure that aid in AD pathogenesis.Key messagesInevitable cascade mechanisms of how Alzheimer's Disease-related (AD-related) gene expressions are modulated by neurotoxicants have been discussed.Involvement of the neurotoxicants-induced pathways caused an extended risk of AD is explicited.Integration of cell culture, animals and population-based analysis on the clinical severity of AD is addressed.
Collapse
Affiliation(s)
- Fatema Yasmin Nisa
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Md. Atiar Rahman
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Md. Amjad Hossen
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
| | - Mohammad Forhad Khan
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
| | - Md. Asif Nadim Khan
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Mumtahina Majid
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Farjana Sultana
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Md. Areeful Haque
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
33
|
Su R, Zhou T. Alpha-Synuclein Induced Immune Cells Activation and Associated Therapy in Parkinson's Disease. Front Aging Neurosci 2021; 13:769506. [PMID: 34803660 PMCID: PMC8602361 DOI: 10.3389/fnagi.2021.769506] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/04/2021] [Indexed: 12/30/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder closely related to immunity. An important aspect of the pathogenesis of PD is the interaction between α-synuclein and a series of immune cells. Studies have shown that accumulation of α-synuclein can induce an autoimmune response that accelerates the progression of PD. This study discusses the mechanisms underlying the interaction between α-synuclein and the immune system. During the development of PD, abnormally accumulated α-synuclein becomes an autoimmune antigen that binds to Toll-like receptors (TLRs) that activate microglia, which differentiate into the microglia type 1 (M1) subtype. The microglia activate intracellular inflammatory pathways, induce the release of proinflammatory cytokines, and promote the differentiation of cluster of differentiation 4 + (CD4 +) T cells into proinflammatory T helper type 1 (Th1) and T helper type 17 (Th17) subtypes. Given the important role of α-synuclein in the immune system of the patients with PD, identifying potential targets of immunotherapy related to α-synuclein is critical for slowing disease progression. An enhanced understanding of immune-associated mechanisms in PD can guide the development of associated therapeutic strategies in the future.
Collapse
Affiliation(s)
- Ruichen Su
- Queen Mary School of Nanchang University, Nanchang University, Nanchang, China
| | - Tian Zhou
- School of Basic Medical Science, Nanchang University, Nanchang, China
| |
Collapse
|
34
|
Tirunavalli SK, Gourishetti K, Kotipalli RSS, Kuncha M, Kathirvel M, Kaur R, Jerald MK, Sistla R, Andugulapati SB. Dehydrozingerone ameliorates Lipopolysaccharide induced acute respiratory distress syndrome by inhibiting cytokine storm, oxidative stress via modulating the MAPK/NF-κB pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 92:153729. [PMID: 34517257 PMCID: PMC8390101 DOI: 10.1016/j.phymed.2021.153729] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/12/2021] [Accepted: 08/23/2021] [Indexed: 06/01/2023]
Abstract
BACKGROUND Inflammation-mediated lung injury is a major cause of health problems in many countries and has been the leading cause of morbidity/mortality in intensive care units. In the current COVID-19 pandemic, the majority of the patients experienced serious pneumonia resulting from inflammation (Acute respiratory distress syndrome/ARDS). Pathogenic infections cause cytokine release syndrome (CRS) by hyperactivation of immune cells, which in turn release excessive cytokines causing ARDS. Currently, there are no standard therapies for viral, bacterial or pathogen-mediated CRS. PURPOSE This study aimed to investigate and validate the protective effects of Dehydrozingerone (DHZ) against LPS induced lung cell injury by in-vitro and in-vivo models and to gain insights into the molecular mechanisms that mediate these therapeutic effects. METHODS The therapeutic activity of DHZ was determined in in-vitro models by pre-treating the cells with DHZ and exposed to LPS to stimulate the inflammatory cascade of events. We analysed the effect of DHZ on LPS induced inflammatory cytokines, chemokines and cell damage markers expression/levels using various cell lines. We performed gene expression, ELISA, and western blot analysis to elucidate the effect of DHZ on inflammation and its modulation of MAPK and NF-κB pathways. Further, the prophylactic and therapeutic effect of DHZ was evaluated against the LPS induced ARDS model in rats. RESULTS DHZ significantly (p < 0.01) attenuated the LPS induced ROS, inflammatory cytokine, chemokine gene expression and protein release in macrophages. Similarly, DHZ treatment protected the lung epithelial and endothelial cells by mitigating the LPS induced inflammatory events in a dose-dependent manner. In vivo analysis showed that DHZ treatment significantly (p < 0.001) mitigated the LPS induced ARDS pathophysiology of increase in the inflammatory cells in BALF, inflammatory cytokine and chemokines in lung tissues. LPS stimulated neutrophil-mediated events, apoptosis, alveolar wall thickening and alveolar inflammation were profoundly reduced by DHZ treatment in a rat model. CONCLUSION This study demonstrates for the first time that DHZ has the potential to ameliorate LPS induced ARDS by inhibiting cytokine storm and oxidative through modulating the MAPK and NF-κB pathways. This data provides pre-clinical support to develop DHZ as a potential therapeutic agent against ARDS.
Collapse
Affiliation(s)
- Satya Krishna Tirunavalli
- CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India; Centre for Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| | - Karthik Gourishetti
- CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India
| | | | - Madusudhana Kuncha
- CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India
| | | | - Rajwinder Kaur
- CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India
| | - Mahesh Kumar Jerald
- CSIR - Centre for Cellular & Molecular Biology (CCMB), Hyderabad 500 007, India
| | - Ramakrishna Sistla
- CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India; Centre for Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| | - Sai Balaji Andugulapati
- CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India; Centre for Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India.
| |
Collapse
|
35
|
Domínguez-Rivas E, Ávila-Muñoz E, Schwarzacher SW, Zepeda A. Adult hippocampal neurogenesis in the context of lipopolysaccharide-induced neuroinflammation: A molecular, cellular and behavioral review. Brain Behav Immun 2021; 97:286-302. [PMID: 34174334 DOI: 10.1016/j.bbi.2021.06.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 06/17/2021] [Accepted: 06/19/2021] [Indexed: 12/17/2022] Open
Abstract
The continuous generation of new neurons occurs in at least two well-defined niches in the adult rodent brain. One of these areas is the subgranular zone of the dentate gyrus (DG) in the hippocampus. While the DG is associated with contextual and spatial learning and memory, hippocampal neurogenesis is necessary for pattern separation. Hippocampal neurogenesis begins with the activation of neural stem cells and culminates with the maturation and functional integration of a portion of the newly generated glutamatergic neurons into the hippocampal circuits. The neurogenic process is continuously modulated by intrinsic factors, one of which is neuroinflammation. The administration of lipopolysaccharide (LPS) has been widely used as a model of neuroinflammation and has yielded a body of evidence for unveiling the detrimental impact of inflammation upon the neurogenic process. This work aims to provide a comprehensive overview of the current knowledge on the effects of the systemic and central administration of LPS upon the different stages of neurogenesis and discuss their effects at the molecular, cellular, and behavioral levels.
Collapse
Affiliation(s)
- Eduardo Domínguez-Rivas
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Evangelina Ávila-Muñoz
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Stephan W Schwarzacher
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Angélica Zepeda
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico; Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
36
|
Morphine promotes microglial activation by upregulating the EGFR/ERK signaling pathway. PLoS One 2021; 16:e0256870. [PMID: 34520454 PMCID: PMC8439491 DOI: 10.1371/journal.pone.0256870] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/17/2021] [Indexed: 11/20/2022] Open
Abstract
Although they represent the cornerstone of analgesic therapy, opioids, such as morphine, are limited in efficacy by drug tolerance, hyperalgesia and other side effects. Activation of microglia and the consequent production of proinflammatory cytokines play a key pathogenic role in morphine tolerance, but the exact mechanisms are not well understood. This study aimed to investigate the regulatory mechanism of epidermal growth factor receptor (EGFR) on microglial activation induced by morphine in mouse microglial BV-2 cells. In this research, BV-2 cells were stimulated with morphine or pretreated with AG1478 (an inhibitor of EGFR). Expression levels of cluster of differentiation molecule 11b (CD11b), EGFR, and phospho-EGFR were detected by immunofluorescence staining. Cell signaling was assayed by Western blot. The migration ability of BV-2 cells was tested by Transwell assay. The production of interleukin-1beta (IL-1β) and tumor necrosis factor-alpha (TNF-α) in the cell supernatant was determined by ELISA. We observed that the expression of CD11b induced by morphine was increased in a dose- and time- dependent manner in BV-2 cells. Phosphorylation levels of EGFR and ERK1/2, migration of BV-2 cells, and production of IL-1β and TNFα were markedly enhanced by morphine treatment. The activation, migration, and production of proinflammatory cytokines in BV-2 cells were inhibited by blocking the EGFR signaling pathway with AG1478. The present study demonstrated that the EGFR/ERK signaling pathway may represent a novel pharmacological strategy to suppress morphine tolerance through attenuation of microglial activation.
Collapse
|
37
|
Kose O, Altin A, Kurt Bayrakdar S, Bostan SA, Mercantepe T, Akyildiz K, Tumkaya L, Yilmaz A, Kose S, Yemenoglu H, Turker Sener L, Kuluslu G. Influences of periodontitis on hippocampal inflammation, oxidative stress, and apoptosis in rats. J Periodontal Res 2021; 56:1154-1162. [PMID: 34486732 DOI: 10.1111/jre.12929] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/26/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIM The hippocampus, which has a central role in cognitive and behavioral activities, is one of the most sensitive parts of the brain to systemic inflammatory diseases. This animal study aims to comprehensively investigate the possible inflammatory, oxidative, and apoptotic effects of periodontitis on the hippocampus. METHODS Sixteen male Sprague-Dawley rats were randomly assigned to two groups: control and experimental periodontitis (Ep). In the Ep group, periodontitis was induced by placing 3.0 sutures sub-paramarginally around the necks of right and left mandibular first molars and maintaining the ligatures in place for 5 weeks. Following the euthanasia, mandibula and hippocampus samples were collected bilaterally. Alveolar bone loss was measured histomorphometrically and radiologically on the right and left mandibles. On the right hippocampal sections histological (Caspase-3, TNF-α, and 8-OHdG) and the left hippocampal sections, biochemical (IL-1β, Aβ1-42 , MDA, GSH, and TAS levels) evaluations were performed. RESULTS Histopathological changes associated with periodontitis were limited (p > .05). A slight increase in caspase-3 positive neuron density in EP rats showed that apoptotic changes were also limited (p > .05). 8-OHdG activity, on the other hand, was significantly higher compared to controls (p < .05). In biochemical analysis, there was a significant increase in IL-1β levels and oxidative membrane damage (MDA) (p < .05) whereas Aβ1-42 and antioxidant marker (GSH and TAS) levels were slightly increased (p > .05). CONCLUSION Periodontitis causes marked increases in IL-1β levels and oxidative stress in the hippocampus, but limited degenerative and apoptotic changes.
Collapse
Affiliation(s)
- Oğuz Kose
- Department of Periodontology, School of Dentistry, Recep Tayyip Erdogan University, Rize, Turkey
| | - Ahmet Altin
- Department of Periodontology, School of Dentistry, Recep Tayyip Erdogan University, Rize, Turkey
| | - Sevda Kurt Bayrakdar
- Department of Periodontology, School of Dentistry, Eskişehir Osmangazi University, Eskisehir, Turkey
| | - Semih Alperen Bostan
- Department of Periodontology, School of Dentistry, Recep Tayyip Erdogan University, Rize, Turkey
| | - Tolga Mercantepe
- Department of Histology and Embryology, School of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Kerimali Akyildiz
- Department of Medical Services and Techniques, School of Health Care Services Vocational, Recep Tayyip Erdogan University, Rize, Turkey
| | - Levent Tumkaya
- Department of Histology and Embryology, School of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Adnan Yilmaz
- Department of Biochemistry, School of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Sefanur Kose
- Department of Psychiatry, School of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Hatice Yemenoglu
- Department of Periodontology, School of Dentistry, Recep Tayyip Erdogan University, Rize, Turkey
| | - Leyla Turker Sener
- Department of Biophysics, School of Medicine, Istanbul University, Istanbul, Turkey
| | - Göker Kuluslu
- 3D Medical and Industrial Design Laboratory, Istanbul University, Istanbul, Turkey
| |
Collapse
|
38
|
Ferreira Junior NC, dos Santos Pereira M, Francis N, Ramirez P, Martorell P, González-Lizarraga F, Figadère B, Chehin R, Del Bel E, Raisman-Vozari R, Michel PP. The Chemically-Modified Tetracycline COL-3 and Its Parent Compound Doxycycline Prevent Microglial Inflammatory Responses by Reducing Glucose-Mediated Oxidative Stress. Cells 2021; 10:cells10082163. [PMID: 34440932 PMCID: PMC8392055 DOI: 10.3390/cells10082163] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 12/31/2022] Open
Abstract
We used mouse microglial cells in culture activated by lipopolysaccharide (LPS) or α-synuclein amyloid aggregates (αSa) to study the anti-inflammatory effects of COL-3, a tetracycline derivative without antimicrobial activity. Under LPS or αSa stimulation, COL-3 (10, 20 µM) efficiently repressed the induction of the microglial activation marker protein Iba-1 and the stimulated-release of the pro-inflammatory cytokine TNF-α. COL-3′s inhibitory effects on TNF-α were reproduced by the tetracycline antibiotic doxycycline (DOX; 50 µM), the glucocorticoid dexamethasone, and apocynin (APO), an inhibitor of the superoxide-producing enzyme NADPH oxidase. This last observation suggested that COL-3 and DOX might also operate themselves by restraining oxidative stress-mediated signaling events. Quantitative measurement of intracellular reactive oxygen species (ROS) levels revealed that COL-3 and DOX were indeed as effective as APO in reducing oxidative stress and TNF-α release in activated microglia. ROS inhibition with COL-3 or DOX occurred together with a reduction of microglial glucose accumulation and NADPH synthesis. This suggested that COL-3 and DOX might reduce microglial oxidative burst activity by limiting the glucose-dependent synthesis of NADPH, the requisite substrate for NADPH oxidase. Coherent with this possibility, the glycolysis inhibitor 2-deoxy-D-glucose reproduced the immunosuppressive action of COL-3 and DOX in activated microglia. Overall, we propose that COL-3 and its parent compound DOX exert anti-inflammatory effects in microglial cells by inhibiting glucose-dependent ROS production. These effects might be strengthened by the intrinsic antioxidant properties of DOX and COL-3 in a self-reinforcing manner.
Collapse
Affiliation(s)
- Nilson Carlos Ferreira Junior
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Av. Café, s/no, Ribeirão Preto 14040-904, Brazil;
- USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), São Paulo 05508-220, Brazil
| | - Maurício dos Santos Pereira
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Av. Café, s/no, Ribeirão Preto 14040-904, Brazil;
- USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), São Paulo 05508-220, Brazil
| | - Nour Francis
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
| | - Paola Ramirez
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
| | - Paula Martorell
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
| | - Florencia González-Lizarraga
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), CP 4000 Tucumán, Argentina; (F.G.-L.); (R.C.)
| | - Bruno Figadère
- BioCIS, CNRS, Université Paris-Saclay, 92290 Châtenay-Malabry, France;
| | - Rosana Chehin
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), CP 4000 Tucumán, Argentina; (F.G.-L.); (R.C.)
| | - Elaine Del Bel
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Av. Café, s/no, Ribeirão Preto 14040-904, Brazil;
- USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), São Paulo 05508-220, Brazil
| | - Rita Raisman-Vozari
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
- Correspondence: (R.R.-V.); (P.P.M.); Tel.: +33-(0)157274550 (R.R.-V.); +33-(0)157274534 (P.P.M.)
| | - Patrick Pierre Michel
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
- Correspondence: (R.R.-V.); (P.P.M.); Tel.: +33-(0)157274550 (R.R.-V.); +33-(0)157274534 (P.P.M.)
| |
Collapse
|
39
|
Réus GZ, Giridharan VV, de Moura AB, Borba LA, Botelho MEM, Behenck JP, Generoso JS, Selvaraj S, Bhatti G, Barichello T, Quevedo J. The impact of early life stress and immune challenge on behavior and glia cells alteration in late adolescent rats. Int J Dev Neurosci 2021; 81:407-415. [PMID: 33788296 DOI: 10.1002/jdn.10108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/08/2021] [Accepted: 03/25/2021] [Indexed: 01/06/2023] Open
Abstract
Maternal deprivation (MD) is known to be related to long-term changes that could influence the onset of psychiatric disorders. Studies have demonstrated that early life stress makes the cells in the brain more susceptible to subsequent stressors. To test it, we used an animal model of MD conducted from postnatal day (PND) 1 to 10. Deprived and non-deprived rats (control) were randomized to receive or not lipopolysaccharide (LPS) at 5 mg/kg on PND 50. The behavior and glial cells activation were evaluated in all groups from 51 to 53 PND. There was an increase in the immobility time in the MD and MD+LPS groups. The spontaneous locomotor activity was not changed between groups. We found elevated ionized calcium-binding adapter molecule 1 (Iba-1)-positive cells levels in the control+LPS and MD+LPS groups. In the MD+LPS group, it was found an increase in Iba-positive cells compared to the MD+sal group. The glial fibrillary acidic protein (GFAP)-positive cells were also increased in the MD+LPS, compared to control+sal, control+LPS, and MD+sal groups. Immune challenge by LPS in late adolescence, which was subjected to MD, did not influence the depressive-like behavior but exerted a pronounced effect in the microglial activation and astrocyte atrophy.
Collapse
Affiliation(s)
- Gislaine Z Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Vijayasree V Giridharan
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Airam B de Moura
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Laura A Borba
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Maria Eduarda M Botelho
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - João Paulo Behenck
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Jaqueline S Generoso
- Laboratory of Experimental Microbiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Sudhakar Selvaraj
- Louis Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Gursimrat Bhatti
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Tatiana Barichello
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Laboratory of Experimental Microbiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| |
Collapse
|
40
|
RKC-B1 Blocks Activation of NF-κB and NLRP3 Signaling Pathways to Suppress Neuroinflammation in LPS-Stimulated Mice. Mar Drugs 2021; 19:md19080429. [PMID: 34436268 PMCID: PMC8398414 DOI: 10.3390/md19080429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 12/24/2022] Open
Abstract
RKC-B1 is a novel fermentation product obtained from the marine micromonospora FIM02-523A. Thus far, there have been few reports about the pharmacological activity of RKC-B1. In our present study, we investigated the anti-neuroinflammatory effects and the possible mechanism of RKC-B1 in LPS-stimulated mice. After treatment with RKC-B1, RNA-seq transcriptome of the cerebral cortex tissue was conducted to find the differentially expressed genes (DEGs). Inflammatory cytokines and proteins were evaluated by ELISA and WB. In RNA-seq analysis, there were 193 genes screened as core genes of RKC-B1 for treatment with neuroinflammation. The significant KEGG enrichment signaling pathways of these core genes were mainly included TNF signaling pathway, IL-17 signaling pathway, NOD-like receptor signaling pathway, NF-κB signaling pathway and others. The corresponding top five KEGG enrichment pathways of three main clusters in PPI network of core genes were closely related to human immune system and immune disease. The results showed that RKC-B1 reduced the levels of pro-inflammatory factors (IL-6, IL-1β, MCP-1, and ICAM-1) and the expression of COX2 in cerebral cortex tissue. Additionally, we found that the anti-neuroinflammation activity of RKC-B1 might be related to suppress activating of NF-κB and NLRP3/cleaved caspase-1 signaling pathways. The current findings suggested that RKC-B1 might be a promising anti-neuroinflammatory agent.
Collapse
|
41
|
Wang X, Liu Z, Wang F. MicroRNA-93 Blocks Signal Transducers and Activator of Transcription 3 to Reduce Neuronal Damage in Parkinson's Disease. Neurochem Res 2021; 46:1859-1868. [PMID: 33900518 DOI: 10.1007/s11064-021-03333-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 04/18/2021] [Accepted: 04/20/2021] [Indexed: 11/27/2022]
Abstract
MicroRNA-93 (miR-93) is an oncogene that promotes tumor growth and angiogenesis. However, its role in Parkinson's disease (PD) remains unknown. This study aimed at investigating the role of miR-93 in PD and the molecular mechanisms involved. 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-induced PD mouse model and lipopolysaccharide (LPS)-exposed BV2 cells were constructed. Real-time quantitative PCR was used to detect the mRNA expression of miR-93, iNOS, IL-6, IL-10, TNF-α and TGF-β1. Bioinformatics analysis and luciferase reporter assay were used to predict and confirm the interaction between miR-93 and STAT3. Flow cytometry was used to detect cell apoptosis. Western blotting was used to detect the protein expression of STAT3. Immunohistochemistry was used to analyze the Iba1-positive and TH positive cells. It was found that the expression of miR-93 was down-regulated in LPS-exposed BV2 cells. Overexpression of miR-93 inhibited the expression of iNOS, IL-6 and TNF-α, while enhanced the expression of TGF-β1 and IL-10. The expression of transcriptional activator 3 (STAT3) was found to be up-regulated in LPS-exposed BV2 cells. Knockdown of STAT3 inhibited the expression of iNOS, IL-6 and TNF-α, while enhanced the expression of TGF-β1 and IL-10. Moreover, STAT3 was found to be a direct target of miR-93, and miR-93 overexpression inhibited the expression of STAT3. Furthermore, both miR-93 overexpression and STAT3 knockdown reduced LPS-induced BV2 cell apoptosis, whereas STAT3 overexpression eliminated the inhibitory effect of miR-93 on LPS-induced BV2 cell apoptosis. In addition, miR-93 overexpression inhibited MPTP-induced STAT3 expression, microglial activation and inflammatory reaction and reduced the loss of tyrosine hydroxylase in the substantia nigra of mice. In conclusion, we demonstrate that miR-93 may be involved in PD by regulating the expression of STAT3.
Collapse
Affiliation(s)
- Xiufeng Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhijun Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fang Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
42
|
Han X, Shao J, Ren X, Li Y, Yu W, Lin C, Li L, Sun Y, Xu B, Luo H, Zhu C, Cao J, Li Z. The different mechanisms of peripheral and central TLR4 on chronic postsurgical pain in rats. J Anat 2021; 239:111-124. [PMID: 33730389 PMCID: PMC8197940 DOI: 10.1111/joa.13406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 12/30/2022] Open
Abstract
Chronic postsurgical pain (CPSP) is a common complication after surgery; however, the underlying mechanisms of CPSP are poorly understood. As one of the most important inflammatory pathways, the Toll-like receptor 4/nuclear factor-kappa B (TLR4/NF-κB) signaling pathway plays an important role in chronic pain. However, the precise role of the TLR4/NF-κB signaling pathway in CPSP remains unclear. In the present study, we established a rat model of CPSP induced by skin/muscle incision and retraction (SMIR) and verified the effects and mechanisms of central and peripheral TLR4 and NF-κB on hyperalgesia in SMIR rats. The results showed that TLR4 expression was increased in both the spinal dorsal horn and dorsal root ganglia (DRGs) of SMIR rats. However, the TLR4 expression pattern in the spinal cord was different from that in DRGs. In the spinal cord, TLR4 was expressed in both neurons and microglia, whereas it was expressed in neurons but not in satellite glial cells in DRGs. Further results demonstrate that the central and peripheral TLR4/NF-κB signaling pathway is involved in the SMIR-induced CPSP by different mechanisms. In the peripheral nervous system, we revealed that the TLR4/NF-κB signaling pathway induced upregulation of voltage-gated sodium channel 1.7 (Nav1.7) in DRGs, triggering peripheral hyperalgesia in SMIR-induced CPSP. In the central nervous system, the TLR4/NF-κB signaling pathway participated in SMIR-induced CPSP by activating microglia in the spinal cord. Ultimately, our findings demonstrated that activation of the peripheral and central TLR4/NF-κB signaling pathway involved in the development of SMIR-induced CPSP.
Collapse
Affiliation(s)
- Xuemin Han
- The Second Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Children’s Hospital of Soochow UniversitySoochowChina
| | - Jinping Shao
- Department of Human AnatomySchool of Basic MedicineZhengzhou UniversityZhengzhouChina
- Institute of NeuroscienceZhengzhou UniversityZhengzhouChina
| | - Xiuhua Ren
- Department of Human AnatomySchool of Basic MedicineZhengzhou UniversityZhengzhouChina
- Institute of NeuroscienceZhengzhou UniversityZhengzhouChina
| | - Yaru Li
- The Second Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Wenli Yu
- Department of Human AnatomySchool of Basic MedicineZhengzhou UniversityZhengzhouChina
- Institute of NeuroscienceZhengzhou UniversityZhengzhouChina
| | - Caihong Lin
- Department of Human AnatomySchool of Basic MedicineZhengzhou UniversityZhengzhouChina
- Institute of NeuroscienceZhengzhou UniversityZhengzhouChina
| | - Lei Li
- Department of Human AnatomySchool of Basic MedicineZhengzhou UniversityZhengzhouChina
- Institute of NeuroscienceZhengzhou UniversityZhengzhouChina
| | - Yanyan Sun
- Department of Human AnatomySchool of Basic MedicineZhengzhou UniversityZhengzhouChina
- Institute of NeuroscienceZhengzhou UniversityZhengzhouChina
| | - Bo Xu
- Department of AnesthesiologyGeneral Hospital of Southern Theatre Command of PLAGuangzhouChina
| | - Huan Luo
- Klinik für AugenheilkundeCharité–Universitätsmedizin Berlin, corporate member of Freie Universität BerlinHumboldt‐Universität zu Berlin, and Berlin Institute of HealthGermany
| | - Changlian Zhu
- Center for Brain Repair and RehabilitationInstitute of Neuroscience and PhysiologyGothenburg UniversityGothenburgSweden
| | - Jing Cao
- Department of Human AnatomySchool of Basic MedicineZhengzhou UniversityZhengzhouChina
- Institute of NeuroscienceZhengzhou UniversityZhengzhouChina
| | - Zhisong Li
- The Second Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
43
|
Liu M, Zhang SS, Liu DN, Yang YL, Wang YH, Du GH. Chrysomycin A Attenuates Neuroinflammation by Down-Regulating NLRP3/Cleaved Caspase-1 Signaling Pathway in LPS-Stimulated Mice and BV2 Cells. Int J Mol Sci 2021; 22:ijms22136799. [PMID: 34202695 PMCID: PMC8268846 DOI: 10.3390/ijms22136799] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 01/08/2023] Open
Abstract
Chrysomycin A (Chr-A), an antibiotic chrysomycin, was discovered in 1955 and is used to treat cancer and tuberculosis. In the present study, the anti-neuroinflammatory effects and possible mechanism of Chr-A in BALB/c mice and in BV2 microglia cells stimulated by lipopolysaccharide (LPS) were investigated. Firstly, the cortex tissues of mice were analyzed by RNA-seq transcriptome to identify differentially expressed genes (DEGs) regulated by Chr-A in LPS-stimulated mice. Inflammatory cytokines and inflammatory proteins were detected by enzyme-linked immunosorbent assay and Western blot. In RNAseq detection, 639 differential up-regulated genes between the control group and LPS model group and 113 differential down-regulated genes between the LPS model group and Chr-A treatment group were found, and 70 overlapping genes were identified as key genes for Chr-A against neuroinflammation. Subsequent GO biological process enrichment analysis showed that the anti-neuroinflammatory effect of Chr-A might be related to the response to cytokine, cellular response to cytokine stimulus, and regulation of immune system process. The significant signaling pathways of KEGG enrichment analysis were mainly involved in TNF signaling pathway, cytokine-cytokine receptor interaction, NF-κB signaling pathway, IL-17 signaling pathway and NOD-like receptor signaling pathway. Our results of in vivo or in vitro experiments showed that the levels of pro-inflammatory factors including NO, IL-6, IL-1β, IL-17, TNF-α, MCP-1, CXCL12, GM-CSF and COX2 in the LPS-stimulated group were higher than those in the control group, while Chr-A reversed those conditions. Furthermore, the Western blot analysis showed that its anti-neuroinflammation appeared to be related to the down-regulation of NLRP3/cleaved caspase-1 signaling pathway. The current findings provide new insights into the activity and molecular mechanisms of Chr-A for the treatment of neuroinflammation.
Collapse
Affiliation(s)
- Man Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (M.L.); (S.-S.Z.); (D.-N.L.); (Y.-L.Y.)
- Beijing Key Laboratory of Drug Target Identification and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Shan-Shan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (M.L.); (S.-S.Z.); (D.-N.L.); (Y.-L.Y.)
- Beijing Key Laboratory of Drug Target Identification and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Dong-Ni Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (M.L.); (S.-S.Z.); (D.-N.L.); (Y.-L.Y.)
- Beijing Key Laboratory of Drug Target Identification and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ying-Lin Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (M.L.); (S.-S.Z.); (D.-N.L.); (Y.-L.Y.)
- Beijing Key Laboratory of Drug Target Identification and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yue-Hua Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (M.L.); (S.-S.Z.); (D.-N.L.); (Y.-L.Y.)
- Beijing Key Laboratory of Drug Target Identification and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Correspondence: (Y.-H.W.); (G.-H.D.)
| | - Guan-Hua Du
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (M.L.); (S.-S.Z.); (D.-N.L.); (Y.-L.Y.)
- Beijing Key Laboratory of Drug Target Identification and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Correspondence: (Y.-H.W.); (G.-H.D.)
| |
Collapse
|
44
|
A Novel 1,8-Naphthyridine-2-Carboxamide Derivative Attenuates Inflammatory Responses and Cell Migration in LPS-Treated BV2 Cells via the Suppression of ROS Generation and TLR4/Myd88/NF-κB Signaling Pathway. Int J Mol Sci 2021; 22:ijms22052527. [PMID: 33802409 PMCID: PMC7959294 DOI: 10.3390/ijms22052527] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/22/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023] Open
Abstract
Novel 1,8-naphthyridine-2-carboxamide derivatives with various substituents (HSR2101-HSR2113) were synthesized and evaluated for their effects on the production of pro-inflammatory mediators and cell migration in lipopolysaccharide (LPS)-treated BV2 microglial cells. Among the tested compounds, HSR2104 exhibited the most potent inhibitory effects on the LPS-stimulated production of inflammatory mediators, including nitric oxide (NO), tumor necrosis factor-α, and interleukin-6. Therefore, this compound was chosen for further investigation. We found that HSR2104 attenuated levels of inducible NO synthase and cyclooxygenase 2 in LPS-treated BV2 cells. In addition, it markedly suppressed LPS-induced cell migration as well as the generation of intracellular reactive oxygen species (ROS). Moreover, HSR2104 abated the LPS-triggered nuclear translocation of nuclear factor-κB (NF-κB) through inhibition of inhibitor kappa Bα phosphorylation. Furthermore, it reduced the expressions of Toll-like receptor 4 (TLR4) and myeloid differentiation factor 88 (MyD88) in LPS-treated BV2 cells. Similar results were observed with TAK242, a specific inhibitor of TLR4, suggesting that TLR4 is an upstream regulator of NF-κB signaling in BV2 cells. Collectively, our findings demonstrate that HSR2104 exhibits anti-inflammatory and anti-migratory activities in LPS-treated BV2 cells via the suppression of ROS and TLR4/MyD88/NF-κB signaling pathway. Based on our observations, HSR2104 may have a beneficial impact on inflammatory responses and microglial cell migration involved in the pathogenesis of various neurodegenerative disorders.
Collapse
|
45
|
Heinz R, Brandenburg S, Nieminen-Kelhä M, Kremenetskaia I, Boehm-Sturm P, Vajkoczy P, Schneider UC. Microglia as target for anti-inflammatory approaches to prevent secondary brain injury after subarachnoid hemorrhage (SAH). J Neuroinflammation 2021; 18:36. [PMID: 33516246 PMCID: PMC7847606 DOI: 10.1186/s12974-021-02085-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/14/2021] [Indexed: 12/11/2022] Open
Abstract
Background Microglia-driven cerebral spreading inflammation is a key contributor to secondary brain injury after SAH. Genetic depletion or deactivation of microglia has been shown to ameliorate neuronal cell death. Therefore, clinically feasible anti-inflammatory approaches counteracting microglia accumulation or activation are interesting targets for SAH treatment. Here, we tested two different methods of interference with microglia-driven cerebral inflammation in a murine SAH model: (i) inflammatory preconditioning and (ii) pharmacological deactivation. Methods 7T-MRI-controlled SAH was induced by endovascular perforation in four groups of C57Bl/6 mice: (i) Sham-operation, (ii) SAH naïve, (iii) SAH followed by inflammatory preconditioning (LPS intraperitoneally), and (iv) SAH followed by pharmacological microglia deactivation (colony-stimulating factor-1 receptor-antagonist PLX3397 intraperitoneally). Microglia accumulation and neuronal cell death (immuno-fluorescence), as well as activation status (RT-PCR for inflammation-associated molecules from isolated microglia) were recorded at day 4 and 14. Toll-like receptor4 (TLR4) status was analyzed using FACS. Results Following SAH, significant cerebral spreading inflammation occurred. Microglia accumulation and pro-inflammatory gene expression were accompanied by neuronal cell death with a maximum on day 14 after SAH. Inflammatory preconditioning as well as PLX3397-treatment resulted in significantly reduced microglia accumulation and activation as well as neuronal cell death. TLR4 surface expression in preconditioned animals was diminished as a sign for receptor activation and internalization. Conclusions Microglia-driven cerebral spreading inflammation following SAH contributes to secondary brain injury. Two microglia-focused treatment strategies, (i) inflammatory preconditioning with LPS and (ii) pharmacological deactivation with PLX3397, led to significant reduction of neuronal cell death. Increased internalization of inflammation-driving TLR4 after preconditioning leaves less receptor molecules on the cell surface, providing a probable explanation for significantly reduced microglia activation. Our findings support microglia-focused treatment strategies to overcome secondary brain injury after SAH. Delayed inflammation onset provides a valuable clinical window of opportunity. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02085-3.
Collapse
Affiliation(s)
- Rebecca Heinz
- Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Susan Brandenburg
- Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Melina Nieminen-Kelhä
- Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Irina Kremenetskaia
- Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Philipp Boehm-Sturm
- Department of Experimental Neurology and Center for Stroke Research, Charité - Universitätsmedizin Berlin, Berlin, Germany.,NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Peter Vajkoczy
- Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology and Center for Stroke Research, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Neurosurgery, Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Ulf C Schneider
- Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany. .,Department of Neurosurgery, Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany.
| |
Collapse
|
46
|
Lipopolysaccharide-induced sepsis-like state compromises post-ischemic neurological recovery, brain tissue survival and remodeling via mechanisms involving microvascular thrombosis and brain T cell infiltration. Brain Behav Immun 2021; 91:627-638. [PMID: 33122024 DOI: 10.1016/j.bbi.2020.10.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 09/04/2020] [Accepted: 10/13/2020] [Indexed: 02/03/2023] Open
Abstract
Sepsis predisposes for poor stroke outcome. This association suggests that sepsis disturbs post-ischemic tissue survival and brain remodeling. To elucidate this link, we herein exposed mice to 30 min intraluminal middle cerebral artery occlusion (MCAO) and induced a sepsis-like state at 72 h post-ischemia by intraperitoneal delivery of Escherichia coli lipopolysaccharide (LPS; three doses of 0.1 or 1 mg/kg, separated by 6 h), a major component of the bacterium's outer membrane. Neurological recovery, ischemic injury, brain remodeling and immune responses were evaluated over up to 56 days post-sepsis (dps) by behavioral tests, immunohistochemistry and flow cytometry. Delivery of 1 mg/kg but not 0.1 mg/kg LPS reduced rectal temperature over 48 h by up to 3.4 ± 3.1 °C, increased general and focal neurological deficits in the Clark score over 72 h and increased motor-coordination deficits in the tight rope test over up to 21 days. Notably, 1 mg/kg, but not 0.1 mg/kg LPS increased intercellular adhesion molecule-1 abundance on ischemic microvessels, increased microvascular thrombosis and increased patrolling monocyte and T cell infiltrates in ischemic brain tissue at 3 dps. Infarct volume was increased by 1 mg/kg, but not 0.1 mg/kg LPS at 3 dps (that is, 6 days post-MCAO), as was brain atrophy at 28 and 56 dps. Microglial activation in ischemic brain tissue, evaluated by morphology analysis of Iba-1 immunostainings, was transiently increased by 0.1 and 1 mg/kg LPS at 3 dps. Our data provide evidence that neurological recovery and brain remodeling are profoundly compromised in the ischemic brain post-sepsis as a consequence of cerebral thromboinflammation.
Collapse
|
47
|
Lenz M, Eichler A, Kruse P, Strehl A, Rodriguez-Rozada S, Goren I, Yogev N, Frank S, Waisman A, Deller T, Jung S, Maggio N, Vlachos A. Interleukin 10 Restores Lipopolysaccharide-Induced Alterations in Synaptic Plasticity Probed by Repetitive Magnetic Stimulation. Front Immunol 2020; 11:614509. [PMID: 33391287 PMCID: PMC7772211 DOI: 10.3389/fimmu.2020.614509] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/16/2020] [Indexed: 12/26/2022] Open
Abstract
Systemic inflammation is associated with alterations in complex brain functions such as learning and memory. However, diagnostic approaches to functionally assess and quantify inflammation-associated alterations in synaptic plasticity are not well-established. In previous work, we demonstrated that bacterial lipopolysaccharide (LPS)-induced systemic inflammation alters the ability of hippocampal neurons to express synaptic plasticity, i.e., the long-term potentiation (LTP) of excitatory neurotransmission. Here, we tested whether synaptic plasticity induced by repetitive magnetic stimulation (rMS), a non-invasive brain stimulation technique used in clinical practice, is affected by LPS-induced inflammation. Specifically, we explored brain tissue cultures to learn more about the direct effects of LPS on neural tissue, and we tested for the plasticity-restoring effects of the anti-inflammatory cytokine interleukin 10 (IL10). As shown previously, 10 Hz repetitive magnetic stimulation (rMS) of organotypic entorhino-hippocampal tissue cultures induced a robust increase in excitatory neurotransmission onto CA1 pyramidal neurons. Furthermore, LPS-treated tissue cultures did not express rMS-induced synaptic plasticity. Live-cell microscopy in tissue cultures prepared from a novel transgenic reporter mouse line [C57BL/6-Tg(TNFa-eGFP)] confirms that ex vivo LPS administration triggers microglial tumor necrosis factor alpha (TNFα) expression, which is ameliorated in the presence of IL10. Consistent with this observation, IL10 hampers the LPS-induced increase in TNFα, IL6, IL1β, and IFNγ and restores the ability of neurons to express rMS-induced synaptic plasticity in the presence of LPS. These findings establish organotypic tissue cultures as a suitable model for studying inflammation-induced alterations in synaptic plasticity, thus providing a biological basis for the diagnostic use of transcranial magnetic stimulation in the context of brain inflammation.
Collapse
Affiliation(s)
- Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Amelie Eichler
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Pia Kruse
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas Strehl
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt, Germany
| | - Silvia Rodriguez-Rozada
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt, Germany
| | - Itamar Goren
- Pharmazentrum Frankfurt/ZAFES, Goethe-University Frankfurt, Frankfurt, Germany
| | - Nir Yogev
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Department of Dermatology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Stefan Frank
- Pharmazentrum Frankfurt/ZAFES, Goethe-University Frankfurt, Frankfurt, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt, Germany
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Nicola Maggio
- Department of Neurology and Sagol Center for Neurosciences, Talpiot Medical Leadership Program, The Chaim Sheba Medical Center, Tel HaShomer, Israel
- Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center Brain Links Brain Tools, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
48
|
Inflammatory Response, a Key Pathophysiological Mechanism of Obesity-Induced Depression. Mediators Inflamm 2020; 2020:8893892. [PMID: 33299381 PMCID: PMC7707993 DOI: 10.1155/2020/8893892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022] Open
Abstract
In recent years, with the acceleration of life rhythm and the increase of social competition, the incidence of obesity and depression has been increasing, which has seriously affected the quality of life and health of people. Obesity and depression, two seemingly unrelated physical and psychological diseases, in fact, are closely related: obese people are more likely to have depression than nonobese ones. We have reviewed and analyzed the relevant research literature and found that the inflammatory response plays a key role in obesity-induced depression. This article will discuss in detail the inflammatory mechanisms by which obesity induces depression.
Collapse
|
49
|
Halder A, Yadav K, Aggarwal A, Singhal N, Sandhir R. Activation of TNFR1 and TLR4 following oxygen glucose deprivation promotes mitochondrial fission in C6 astroglial cells. Cell Signal 2020; 75:109714. [PMID: 32693013 DOI: 10.1016/j.cellsig.2020.109714] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 06/30/2020] [Accepted: 07/12/2020] [Indexed: 12/14/2022]
Abstract
Astrocytes have emerged as active players in the innate immune response triggered by various types of insults. Recent literature suggests that mitochondria are key participants in innate immunity. The present study investigates the role of ischemia-induced innate immune response on p65/PGC-1α mediated mitochondrial dynamics in C6 astroglial cells. OGD conditions induced astroglial differentiation in C6 cells and increased the expression of hypoxia markers; HIF-1α, HO-1 and Cox4i2. OGD conditions resulted in induction of innate immune response in terms of expression of TNFR1 and TLR4 along with increase in IL-6 and TNF-α levels. OGD conditions resulted in decreased expression of I-κB with a concomitant increase in phos-p65 levels. The expression of PGC-1α, a key regulator of mitochondrial biogenesis, was also increased. Immunochemical staining suggested that phos-p65 and PGC-1α was co-localized. Studies on mitochondrial fusion (Mfn-1) and fission (DRP1) markers revealed shift toward fission. In addition, mitochondrial membrane potential decreased with increased DNA degradation and apoptosis confirming mitochondrial fission under OGD conditions. However, inhibition of phos-p65 by MG132 reduced the co-localization of phos-p65/ PGC-1α and significantly increased the Mfn-1 expression. The findings demonstrate the involvement of TNFR1 and TLR4 mediated immune response followed by interaction between phos-p65 and PGC-1α in promoting fission in C6 cells under hypoxic condition.
Collapse
Affiliation(s)
- Avishek Halder
- Department of Biochemistry, Basic Medical Science Block II, Panjab University, Chandigarh, India
| | - Kamalendra Yadav
- National Agri-Food Biotechnology Institute, Sector 81, Mohali, Punjab, India
| | - Aanchal Aggarwal
- National Agri-Food Biotechnology Institute, Sector 81, Mohali, Punjab, India
| | - Nitin Singhal
- National Agri-Food Biotechnology Institute, Sector 81, Mohali, Punjab, India
| | - Rajat Sandhir
- Department of Biochemistry, Basic Medical Science Block II, Panjab University, Chandigarh, India.
| |
Collapse
|
50
|
Wang H, Song X, Li M, Wang X, Tao Y, Xiya X, Liu H, Zhao Y, Chang D, Sha Q. The role of TLR4/NF-κB signaling pathway in activated microglia of rats with chronic high intraocular pressure and vitro scratch injury-induced microglia. Int Immunopharmacol 2020; 83:106395. [PMID: 32199351 DOI: 10.1016/j.intimp.2020.106395] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/12/2020] [Accepted: 03/09/2020] [Indexed: 12/15/2022]
Abstract
Glaucoma is a kind of blind-causing disease with structural damages of optic nerve and defection of visual field. It is believed that the death of retinal ganglion cell (RGC) is a consequential event of over-reactive immune orchestral cells such as microglia. Previous evidences in animal and clinical studies show the innate immunity plays a pivotal role in neuro-inflammation of glaucoma. Toll-like receptor 4 (TLR4) is expressed on microglia and mediates many neuroinflammatory diseases. We aimed to explore the impacts of high intraocular pressure (IOP) on rat microglia in retina and the regulation of TLR4/NF-κB signaling pathway in scratched microglia cells. In our study, we successfully established chronic high IOP rat model by episcleral vein cauterization (EVC) which behaved like the chronic glaucoma. Besides, we set up an in vitro scratch-induced injury model in rat microglia cells. We found the level of activated microglia cells were significantly increased in the retina of chronic high IOP groups. Moreover, the inhibition of TLR4/NF-κB signaling pathway suppressed the expression of TLR4 protein and mRNA levels of P50, IL-6 and TNF-α. Our original study provided a theoretical basis on targeting TLR4/NF-κB to suppress pro-inflammatory factors releasing in activated microglia and it might be a good treatment target to prevent glaucoma from progressing.
Collapse
Affiliation(s)
- Hongjun Wang
- Department of Medical, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Xiangyuan Song
- Department of Ophthalmology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Mingzhe Li
- Department of Ophthalmology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Xuefei Wang
- Department of Ophthalmology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Yi Tao
- Department of Ophthalmology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Xiamu Xiya
- Department of Ophthalmology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Hui Liu
- Department of Ophthalmology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Yini Zhao
- Department of Ophthalmology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Dong Chang
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Qian Sha
- Department of Ophthalmology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.
| |
Collapse
|