1
|
Mohsin M, Shams F, Li H, Alam A, Xia C, Fan L, Cao Y, Jiang W, Nasir A, Khan S, Bai Q. Nanozymes in neuropathic pain: strategies bridging oxidative stress, mitochondrial repair, and neuroimmune modulation for targeted therapy. J Neuroinflammation 2025; 22:156. [PMID: 40506712 PMCID: PMC12164103 DOI: 10.1186/s12974-025-03456-w] [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: 03/26/2025] [Accepted: 04/24/2025] [Indexed: 06/16/2025] Open
Abstract
Neuropathic pain is one of the most debilitating neurological conditions, significantly challenging to manage due to the complex interplay of oxidative stress, neuroinflammation, and mitochondrial dysfunction in its pathogenesis. Nanozyme (nanomaterials with enzyme-like activity) technology offers a promising strategy to tackle these multifaceted mechanisms. These nanozymes can scavenge reactive oxygen species (ROS), modulate inflammatory pathways, and reverse mitochondrial dysfunction, providing notable neuroprotection and pain relief for affected individuals. Additionally, nanozymes exhibit targeted delivery to the injury sites by using mechanisms such as lysosome-mediated endocytosis (e.g., SOD&Fe3O4@ZIF-8 nanozymes) and mannose receptor-mediated cellular uptake (e.g., mSPIONs nanozymes). Given the limitations of current treatment options, we underscore the advantages of nanozymes, including their multifunctional capabilities and potential to enhance therapeutic outcomes in pain management. This review focuses on the underlying mechanisms of neuropathic pain, particularly emphasizing the role of oxidative stress and its impact on disease progression. We examine the applications of nanozymes for treating neuropathic pain, highlighting their potential to scavenge ROS, relieve mitochondrial dysfunction, modulate neuroinflammatory pathways, and repair blood-spinal cord barrier integrity. Furthermore, this paper provides an overview of the current landscape of nanozyme research in neuropathic pain and future directions for their clinical translation in pain management, emphasizing their potential role in improving therapeutic outcomes.
Collapse
Grants
- NO; 82371234, NO.241111520200, NO: HNSWJW-2021001 Bai Qian
- NO; 82371234, NO.241111520200, NO: HNSWJW-2021001 Bai Qian
- NO; 82371234, NO.241111520200, NO: HNSWJW-2021001 Bai Qian
- NO; 82371234, NO.241111520200, NO: HNSWJW-2021001 Bai Qian
- NO; 82371234, NO.241111520200, NO: HNSWJW-2021001 Bai Qian
- NO; 82371234, NO.241111520200, NO: HNSWJW-2021001 Bai Qian
- NO; 82371234, NO.241111520200, NO: HNSWJW-2021001 Bai Qian
- NO; 82371234, NO.241111520200, NO: HNSWJW-2021001 Bai Qian
- NO; 82371234, NO.241111520200, NO: HNSWJW-2021001 Bai Qian
- NO; 82371234, NO.241111520200, NO: HNSWJW-2021001 Bai Qian
- NO; 82371234, NO.241111520200, NO: HNSWJW-2021001 Bai Qian
Collapse
Affiliation(s)
- Muhammad Mohsin
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
| | - Fizzah Shams
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
| | - Hong Li
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
| | - Amir Alam
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
| | - Chaoyun Xia
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
| | - Lulu Fan
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
| | - Ying Cao
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
| | - Wei Jiang
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
| | - Abdul Nasir
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
| | - Suliman Khan
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China.
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China.
| | - Qian Bai
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China.
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China.
| |
Collapse
|
2
|
Potapov KV, Platonov DN, Belyy AY, Novikov MA, Tomilov YV, Anashkina AA, Mukhina KA, Kechko OI, Solyev PN, Novikov RA, Makarov AA, Mitkevich VA. Improved Synthesis of Effective 3-(Indolin-6-yl)-4-( N-pyrazole-sulfonamide)-1 H-pyrrolo[2,3- b]pyridine-Based Inhibitors of NADPH Oxidase 2. Int J Mol Sci 2025; 26:3647. [PMID: 40332246 PMCID: PMC12026636 DOI: 10.3390/ijms26083647] [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: 02/20/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
NADPH oxidase enzymes (NOXs) are a family of enzymes generating superoxide, which form reactive oxygen species. NOX2 activity is a causative agent for the progression of many diseases: neurodegenerative, cardiovascular, immune dysregulations, and even hereditary diseases and cancer. Administering antioxidants helps in inhibiting NOX2 activity; however, the development of selective inhibitors may provide greater improvement in the therapy of diseases. Here, an optimized synthesis of two most promising NOX2 inhibitors based on the 3-(indolin-6-yl)-4-(N-pyrazole-sulfonamide)-1H-pyrrolo [2,3-b]pyridine structure, namely, GSK2795039 and NCATS-SM7270, and an isomeric derivative of the same class, IMBIOC-1, is reported. The new modified procedures simplify the isolation, reduce byproduct formation, and improve the yields in 0.1-1 g scale preparations. Molecular modeling of the structures of NOX2 complexes with inhibitors validated their binding at the same site as NADPH, with IMBIOC-1 forming the largest number of intermolecular interactions with the NOX2 active site. Testing the effects of the compounds on amyloid beta-induced oxidative stress and toxicity in HMC3 microglial cells showed that all three inhibitors completely prevented the pathological amyloid-beta effect. At the same time, NCATS-SM7270 and IMBIOC-1 provided a stronger protective effect on microglial cell survival than GSK2795039, which allowed us to assert the potential of those compounds as neuroprotective agents.
Collapse
Affiliation(s)
- Konstantin V. Potapov
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia; (K.V.P.); (M.A.N.); (A.A.A.); (K.A.M.); (O.I.K.); (A.A.M.); (V.A.M.)
- Zelinsky Institute of Organic Chemistry of the Russian Academy of Sciences, 119991 Moscow, Russia; (D.N.P.); (A.Y.B.); (Y.V.T.)
| | - Dmitry N. Platonov
- Zelinsky Institute of Organic Chemistry of the Russian Academy of Sciences, 119991 Moscow, Russia; (D.N.P.); (A.Y.B.); (Y.V.T.)
| | - Alexander Yu. Belyy
- Zelinsky Institute of Organic Chemistry of the Russian Academy of Sciences, 119991 Moscow, Russia; (D.N.P.); (A.Y.B.); (Y.V.T.)
| | - Maxim A. Novikov
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia; (K.V.P.); (M.A.N.); (A.A.A.); (K.A.M.); (O.I.K.); (A.A.M.); (V.A.M.)
- Zelinsky Institute of Organic Chemistry of the Russian Academy of Sciences, 119991 Moscow, Russia; (D.N.P.); (A.Y.B.); (Y.V.T.)
| | - Yury V. Tomilov
- Zelinsky Institute of Organic Chemistry of the Russian Academy of Sciences, 119991 Moscow, Russia; (D.N.P.); (A.Y.B.); (Y.V.T.)
| | - Anastasia A. Anashkina
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia; (K.V.P.); (M.A.N.); (A.A.A.); (K.A.M.); (O.I.K.); (A.A.M.); (V.A.M.)
| | - Kristina A. Mukhina
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia; (K.V.P.); (M.A.N.); (A.A.A.); (K.A.M.); (O.I.K.); (A.A.M.); (V.A.M.)
| | - Olga I. Kechko
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia; (K.V.P.); (M.A.N.); (A.A.A.); (K.A.M.); (O.I.K.); (A.A.M.); (V.A.M.)
| | - Pavel N. Solyev
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia; (K.V.P.); (M.A.N.); (A.A.A.); (K.A.M.); (O.I.K.); (A.A.M.); (V.A.M.)
| | - Roman A. Novikov
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia; (K.V.P.); (M.A.N.); (A.A.A.); (K.A.M.); (O.I.K.); (A.A.M.); (V.A.M.)
- Zelinsky Institute of Organic Chemistry of the Russian Academy of Sciences, 119991 Moscow, Russia; (D.N.P.); (A.Y.B.); (Y.V.T.)
| | - Alexander A. Makarov
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia; (K.V.P.); (M.A.N.); (A.A.A.); (K.A.M.); (O.I.K.); (A.A.M.); (V.A.M.)
| | - Vladimir A. Mitkevich
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia; (K.V.P.); (M.A.N.); (A.A.A.); (K.A.M.); (O.I.K.); (A.A.M.); (V.A.M.)
| |
Collapse
|
3
|
Gallagher E, Li S, Lee H, Xu H, Lee VMY, Mach RH, McManus MJ. Noninvasive Detection of Oxidative Stress in a Mouse Model of 4R Tauopathy via Positron Emission Tomography with [ 18F]ROStrace. Int J Mol Sci 2025; 26:1845. [PMID: 40076472 PMCID: PMC11899037 DOI: 10.3390/ijms26051845] [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: 01/23/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Oxidative stress, defined as the excessive production of reactive oxygen species (ROS), is a crucial factor in the pathogenesis of various neurodegenerative diseases, including the 4-repeat (4R) tauopathies. Collectively, the 4R tauopathies are characterized by the progressive aggregation of tau protein isoforms with four microtubule-binding domains in and around brain cells. The cyclical relationship between oxidative stress and 4R tau aggregation suggests that a means of imaging ROS noninvasively could be a valuable tool for the study and treatment of 4R tauopathy in both humans and animal models. To demonstrate the potential of the ROS-sensitive positron emission tomography (PET) radiotracer [18F]ROStrace as a means of filling this methodological gap, we performed [18F]ROStrace PET imaging on PS19 mice, which exhibit 4R tau aggregation similar to that seen in human 4R tauopathy. Significant increases in [18F]ROStrace signal became detectable in the hippocampus of 6-11-month-old (mo) PS19 animals and spread to the brainstem, midbrain, and thalamus of 11+ mo animals. Additionally, older PS19 mice displayed higher whole-brain average [18F]ROStrace signal compared to age-matched controls (p = 0.042), and tau pathology consistently colocalized with multiple fluorescent indicators of oxidative stress in PS19 brain samples. These results provide novel evidence that 4R tau aggregation is associated with increased oxidative stress in PS19 mouse brain and advance [18F]ROStrace as a noninvasive technology for the detection of oxidative stress in neurodegenerative diseases involving tau pathology.
Collapse
Affiliation(s)
- Evan Gallagher
- Department of Anesthesia and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA (R.H.M.)
| | - Shihong Li
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA (R.H.M.)
| | - Hsiaoju Lee
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA (R.H.M.)
| | - Hong Xu
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Virginia M.-Y. Lee
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert H. Mach
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA (R.H.M.)
| | - Meagan J. McManus
- Department of Anesthesia and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| |
Collapse
|
4
|
Alaei M, Koushki K, Taebi K, Yousefi Taba M, Keshavarz Hedayati S, Keshavarz Shahbaz S. Metal nanoparticles in neuroinflammation: impact on microglial dynamics and CNS function. RSC Adv 2025; 15:5426-5451. [PMID: 39967886 PMCID: PMC11833603 DOI: 10.1039/d4ra07798a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
Microglia, the primary immune cells of the central nervous system (CNS), are crucial in maintaining brain homeostasis and responding to pathological changes. While they play protective roles, their activation can lead to neuroinflammation and the progression of neurodegenerative diseases. Metal nanoparticles (NPs), due to their unique ability to cross the blood-brain barrier (BBB), have emerged as promising agents for drug delivery to the CNS. In this way, we aim to review the dual role of metal-containing NPs, gold (AuNPs), silver (AgNPs), iron oxide (IONPs), zinc oxide (ZnONPs), cobalt (CoNPs), titanium dioxide (TiO2NPs), and silica (SiO2NPs) in modulating microglial activity. Some NPs promote anti-inflammatory effects, while others exacerbate neuroinflammation. We examine how these NPs influence microglial activation, focusing on their potential therapeutic benefits and risks. A deeper understanding of NP-microglia interactions is crucial for developing safe and efficient treatments for neuroinflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Masood Alaei
- Student Research Committee, Qazvin University of Medical Sciences Qazvin Iran
- USERN Office, Qazvin University of Medical Science Qazvin Iran
| | - Khadijeh Koushki
- Department of Neurosurgery, University of Texas Houston Health Science Center (UTHealth) Houston TX USA
| | - Kimia Taebi
- Student Research Committee, Qazvin University of Medical Sciences Qazvin Iran
- USERN Office, Qazvin University of Medical Science Qazvin Iran
| | - Mahdieh Yousefi Taba
- Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | | | - Sanaz Keshavarz Shahbaz
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences Qazvin 34197-59811 Iran
- USERN Office, Qazvin University of Medical Science Qazvin Iran
| |
Collapse
|
5
|
Yang J, Agrawal K, Stanley J, Li R, Jacobs N, Wang H, Lu C, Qu R, Clarke D, Chen Y, Jiang Y, Bai D, Zheng S, Fox H, Ho YC, Huttner A, Gerstein M, Kluger Y, Zhang L, Spudich S. Multi-omic Characterization of HIV Effects at Single Cell Level across Human Brain Regions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636707. [PMID: 39975288 PMCID: PMC11839123 DOI: 10.1101/2025.02.05.636707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
HIV infection exerts profound and long-lasting neurodegenerative effects on the central nervous system (CNS) that can persist despite antiretroviral therapy (ART). Here, we used single-nucleus multiome sequencing to map the transcriptomic and epigenetic landscapes of postmortem human brains from 13 healthy individuals and 20 individuals with HIV who have a history of treatment with ART. Our study spanned three distinct regions-the prefrontal cortex, insular cortex, and ventral striatum-enabling a comprehensive exploration of region-specific and cross-regional perturbations. We found widespread and persistent HIV-associated transcriptional and epigenetic alterations across multiple cell types. Detailed analyses of microglia revealed state changes marked by immune activation and metabolic dysregulation, while integrative multiomic profiling of astrocytes identified multiple subpopulations, including a reactive subpopulation unique to HIV-infected brains. These findings suggest that cells from people with HIV exhibit molecular shifts that may underlie ongoing neuroinflammation and CNS dysfunction. Furthermore, cell-cell communication analyses uncovered dysregulated and pro-inflammatory interactions among glial populations, underscoring the multifaceted and enduring impact of HIV on the brain milieu. Collectively, our comprehensive atlas of HIV-associated brain changes reveals distinct glial cell states with signatures of proinflammatory signaling and metabolic dysregulation, providing a framework for developing targeted therapies for HIV-associated neurological dysfunction.
Collapse
Affiliation(s)
- Junchen Yang
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Kriti Agrawal
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Jay Stanley
- Program in Applied Mathematics, Yale University, New Haven, CT, USA
| | - Ruiqi Li
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Nicholas Jacobs
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Haowei Wang
- Department of Neurology, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale University, New Haven, CT, USA
| | - Chang Lu
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Rihao Qu
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Declan Clarke
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Yuhang Chen
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Yunzhe Jiang
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Donglu Bai
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Suchen Zheng
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Howard Fox
- Department of Neurological Sciences, University of Nebraska School of Medicine, Omaha, NB, USA
| | - Ya-chi Ho
- Department of Microbial Pathogenesis, Yale University, New Haven, CT, USA
| | - Anita Huttner
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Mark Gerstein
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Computer Science, Yale University, New Haven, CT, USA
- Department of Statistics and Data Science, Yale University, New Haven, CT, USA
- Department of Biomedical Informatics & Data Science, Yale University, New Haven, CT, USA
| | - Yuval Kluger
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Program in Applied Mathematics, Yale University, New Haven, CT, USA
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Le Zhang
- Department of Neurology, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale University, New Haven, CT, USA
| | - Serena Spudich
- Department of Neurology, Yale University, New Haven, CT, USA
- Center for Brain and Mind Health, Yale University, New Haven, CT, USA
| |
Collapse
|
6
|
Li M, Chen M, Li H, Gao D, Zhao L, Zhu M. Glial cells improve Parkinson's disease by modulating neuronal function and regulating neuronal ferroptosis. Front Cell Dev Biol 2025; 12:1510897. [PMID: 39830208 PMCID: PMC11739109 DOI: 10.3389/fcell.2024.1510897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
The main characteristics of Parkinson's disease (PD) are the loss of dopaminergic (DA) neurons and abnormal aggregation of cytosolic proteins. However, the exact pathogenesis of PD remains unclear, with ferroptosis emerging as one of the key factors driven by iron accumulation and lipid peroxidation. Glial cells, including microglia, astrocytes, and oligodendrocytes, serve as supportive cells in the central nervous system (CNS), but their abnormal activation can lead to DA neuron death and ferroptosis. This paper explores the interactions between glial cells and DA neurons, reviews the changes in glial cells during the pathological process of PD, and reports on how glial cells regulate ferroptosis in PD through iron homeostasis and lipid peroxidation. This opens up a new pathway for basic research and therapeutic strategies in Parkinson's disease.
Collapse
Affiliation(s)
- Mengzhu Li
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Mengxuan Chen
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Haiyan Li
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Da Gao
- Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lijun Zhao
- Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Meiling Zhu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
7
|
Wei J, Liu C, Qin D, Ren F, Duan J, Chen T, Wu A. Targeting inflammation and gut microbiota with antibacterial therapy: Implications for central nervous system health. Ageing Res Rev 2024; 102:102544. [PMID: 39419400 DOI: 10.1016/j.arr.2024.102544] [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: 09/02/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
The complex symbiotic relationship between inflammation, the gut microbiota, and the central nervous system (CNS) has become a pivotal focus of contemporary biomedical research. Inflammation, as a physiological defense mechanism, plays a dual role as both a protective and pathological factor, and is intricately associated with gut microbiota homeostasis, often termed the "second brain." The gutbrain axis (GBA) exemplifies this multifaceted interaction, where gut health exerts significantly regulatory effects on CNS functions. Antibacterial therapies represent both promising and challenging strategies for modulating inflammation and gut microbiota composition to confer CNS benefits. However, while such therapies may exert positive modulatory effects on the gut microbiota, they also carry the potential to disrupt microbial equilibrium, potentially exacerbating neurological dysfunction. Recent advances have provided critical insights into the therapeutic implications of antibacterial interventions; nevertheless, the application of these therapies in the context of CNS health warrants a judicious and evidence-based approach. As research progresses, deeper investigation into the microbial-neural interface is essential to fully realize the potential of therapies targeting inflammation and the gut microbiota for CNS health. Future efforts should focus on refining antibacterial interventions to modulate the gut microbiota while minimizing disruption to microbial balance, thereby reducing risks and enhancing efficacy in CNS-related conditions. In conclusion, despite challenges, a more comprehensive understanding of the GBA, along with precise modulation through targeted antibacterial therapies, offers significant promise for advancing CNS disorder treatment. Continued research in this area will lead to innovative interventions and improved patient outcomes.
Collapse
Affiliation(s)
- Jing Wei
- Eye School of Chengdu University of TCM, Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, China; School of Pharmaceutical Sciences, China-Pakistan International Science and Technology Innovation Cooperation Base for Ethnic Medicine Development in Hunan Province, Hunan University of Medicine, Huaihua 418000, China.
| | - Chunmeng Liu
- Eye School of Chengdu University of TCM, Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, China.
| | - Dalian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Department of Cardiology, the Affiliated Hospital of Southwest Medical University and Key Laboratory of Medical Electrophysiology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China.
| | - Fang Ren
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400021, China.
| | - Junguo Duan
- Eye School of Chengdu University of TCM, Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, China.
| | - Ting Chen
- School of Pharmaceutical Sciences, China-Pakistan International Science and Technology Innovation Cooperation Base for Ethnic Medicine Development in Hunan Province, Hunan University of Medicine, Huaihua 418000, China.
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Department of Cardiology, the Affiliated Hospital of Southwest Medical University and Key Laboratory of Medical Electrophysiology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
8
|
Kardam S, Ambasta RK, Kumar P. Overview of pro-inflammatory and pro-survival components in neuroinflammatory signalling and neurodegeneration. Ageing Res Rev 2024; 100:102465. [PMID: 39187022 DOI: 10.1016/j.arr.2024.102465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/07/2024] [Accepted: 08/18/2024] [Indexed: 08/28/2024]
Abstract
Neurodegenerative diseases (NDDs) are identified by the progressive deterioration of neurons and a subsequent decline in cognitive function, creating an enormous burden on the healthcare system globally. Neuroinflammation is an intricate procedure that initiates the immune response in the central nervous system (CNS) and significantly impacts the expansion of NDDs. This study scrutinizes the complicated interaction between neuronal degeneration and neuroinflammation, with an appropriate emphasis on their reciprocal impacts. It also describes how neuroinflammatory reactions in NDDs are controlled by activating certain pro-inflammatory transcription factors, including p38 MAPK, FAF1, Toll-like receptors (TLRs), and STAT3. Alternatively, it evaluates the impact of pro-survival transcription factors, such as the SOCS pathway, YY1, SIRT1, and MEF2, which provide neuroprotective protection against damage triggered by neuroinflammation. Moreover, we study the feasibility of accommodating drug repositioning as a therapeutic approach for treating neuroinflammatory disorders. This suggests the use of existing medications for novel utilization in the treatment of NDDs. Furthermore, the study intends to reveal novel biomarkers of neuroinflammation that contribute fundamental observation for the initial detection and diagnosis of these disorders. This study aims to strengthen therapy interference and augment patient outcomes by combining ongoing data and evaluating novel therapeutic and diagnostic approaches. The goal is to devote the growth of an effective strategy to reducing the impact of neuroinflammation on neuronal protection in NDDs.
Collapse
Affiliation(s)
- Shefali Kardam
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Rashmi K Ambasta
- Department of Biotechnology and Microbiology, SRM University, Sonepat, India; Department of Medicine, Vanderbilt University Medical Centre, Nashville, Tennessee, USA
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India.
| |
Collapse
|
9
|
Marino AL, Rex TS, Harrison FE. Modulation of microglia activation by the ascorbic acid transporter SVCT2. Brain Behav Immun 2024; 120:557-570. [PMID: 38972487 PMCID: PMC11458066 DOI: 10.1016/j.bbi.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/04/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024] Open
Abstract
Neuroinflammation is a major characteristic of pathology in several neurodegenerative diseases. Microglia, the brain's resident myeloid cells, shift between activation states under neuroinflammatory conditions, both responding to, but also driving damage in the brain. Vitamin C (ascorbate) is an essential antioxidant for central nervous system function that may have a specific role in the neuroinflammatory response. Uptake of ascorbate throughout the central nervous system is facilitated by the sodium-dependent vitamin C transporter 2 (SVCT2). SVCT2 transports the reduced form of ascorbate into neurons and microglia, however the contribution of altered SVCT2 expression to the neuroinflammatory response in microglia is not well understood. In this study we demonstrate that SVCT2 expression modifies microglial response, as shown through changes in cell morphology and mRNA expression, following a mild traumatic brain injury (mTBI) in mice with decreased or increased expression of SVCT2. Results were supported by in vitro studies in an immortalized microglial cell line and in primary microglial cultures derived from SVCT2-heterozygous and transgenic animals. Overall, this work demonstrates the importance of SVCT2 and ascorbate in modulating the microglial response to mTBI and suggests a potential role for both in response to neuroinflammatory challenges.
Collapse
Affiliation(s)
- Amanda L Marino
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States
| | - Tonia S Rex
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States; Division of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Fiona E Harrison
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States; Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States.
| |
Collapse
|
10
|
Demos-Davies K, Lawrence J, Coffey J, Morgan A, Ferreira C, Hoeppner LH, Seelig D. Longitudinal Neuropathological Consequences of Extracranial Radiation Therapy in Mice. Int J Mol Sci 2024; 25:5731. [PMID: 38891920 PMCID: PMC11171684 DOI: 10.3390/ijms25115731] [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: 04/18/2024] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Cancer-related cognitive impairment (CRCI) is a consequence of chemotherapy and extracranial radiation therapy (ECRT). Our prior work demonstrated gliosis in the brain following ECRT in SKH1 mice. The signals that induce gliosis were unclear. Right hindlimb skin from SKH1 mice was treated with 20 Gy or 30 Gy to induce subclinical or clinical dermatitis, respectively. Mice were euthanized at 6 h, 24 h, 5 days, 12 days, and 25 days post irradiation, and the brain, thoracic spinal cord, and skin were collected. The brains were harvested for spatial proteomics, immunohistochemistry, Nanostring nCounter® glial profiling, and neuroinflammation gene panels. The thoracic spinal cords were evaluated by immunohistochemistry. Radiation injury to the skin was evaluated by histology. The genes associated with neurotransmission, glial cell activation, innate immune signaling, cell signal transduction, and cancer were differentially expressed in the brains from mice treated with ECRT compared to the controls. Dose-dependent increases in neuroinflammatory-associated and neurodegenerative-disease-associated proteins were measured in the brains from ECRT-treated mice. Histologic changes in the ECRT-treated mice included acute dermatitis within the irradiated skin of the hindlimb and astrocyte activation within the thoracic spinal cord. Collectively, these findings highlight indirect neuronal transmission and glial cell activation in the pathogenesis of ECRT-related CRCI, providing possible signaling pathways for mitigation strategies.
Collapse
Affiliation(s)
- Kimberly Demos-Davies
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, Saint Paul, MN 55108, USA; (J.L.); (J.C.); (A.M.); (D.S.)
| | - Jessica Lawrence
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, Saint Paul, MN 55108, USA; (J.L.); (J.C.); (A.M.); (D.S.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA;
- Department of Radiation Oncology, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Jessica Coffey
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, Saint Paul, MN 55108, USA; (J.L.); (J.C.); (A.M.); (D.S.)
| | - Amy Morgan
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, Saint Paul, MN 55108, USA; (J.L.); (J.C.); (A.M.); (D.S.)
| | - Clara Ferreira
- Department of Radiation Oncology, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Luke H. Hoeppner
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA;
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN 55912, USA
| | - Davis Seelig
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, Saint Paul, MN 55108, USA; (J.L.); (J.C.); (A.M.); (D.S.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
11
|
Khan Z, Mehan S, Gupta GD, Narula AS. Immune System Dysregulation in the Progression of Multiple Sclerosis: Molecular Insights and Therapeutic Implications. Neuroscience 2024; 548:9-26. [PMID: 38692349 DOI: 10.1016/j.neuroscience.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 05/03/2024]
Abstract
Multiple sclerosis (MS), a prevalent neurological disorder, predominantly affects young adults and is characterized by chronic autoimmune activity. The study explores the immune system dysregulation in MS, highlighting the crucial roles of immune and non-neuronal cells in the disease's progression. This review examines the dual role of cytokines, with some like IL-6, TNF-α, and interferon-gamma (IFN-γ) promoting inflammation and CNS tissue injury, and others such as IL-4, IL-10, IL-37, and TGF-β fostering remyelination and protecting against MS. Elevated chemokine levels in the cerebrospinal fluid (CSF), including CCL2, CCL5, CXCL10, CXCL13, and fractalkine, are analyzed for their role in facilitating immune cell migration across the blood-brain barrier (BBB), worsening inflammation and neurodegeneration. The study also delves into the impact of auto-antibodies targeting myelin components like MOG and AQP4, which activate complement cascades leading to further myelin destruction. The article discusses how compromised BBB integrity allows immune cells and inflammatory mediators to infiltrate the CNS, intensifying MS symptoms. It also examines the involvement of astrocytes, microglia, and oligodendrocytes in the disease's progression. Additionally, the effectiveness of immunomodulatory drugs such as IFN-β and CD20-targeting monoclonal antibodies (e.g., rituximab) in modulating immune responses is reviewed, highlighting their potential to reduce relapse rates and delaying MS progression. These insights emphasize the importance of immune system dysfunction in MS development and progression, guiding the development of new therapeutic strategies. The study underscores recent advancements in understanding MS's molecular pathways, opening avenues for more targeted and effective treatments.
Collapse
Affiliation(s)
- Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India), Moga 142001, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India), Moga 142001, Punjab, India.
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India), Moga, Punjab, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| |
Collapse
|
12
|
Liu X, Gu J, Wang C, Peng M, Zhou J, Fei X, Zhong Z, Li B. Ginsenoside Rg3 attenuates neuroinflammation and hippocampal neuronal damage after traumatic brain injury in mice by inactivating the NF-kB pathway via SIRT1 activation. Cell Cycle 2024; 23:662-681. [PMID: 38796716 PMCID: PMC11229723 DOI: 10.1080/15384101.2024.2355008] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/24/2024] [Accepted: 05/07/2024] [Indexed: 05/28/2024] Open
Abstract
This investigation examined the potential of ginsenoside Rg3 in addressing traumatic brain injury (TBI). A TBI mouse model underwent treatment with ginsenoside Rg3 and nicotinamide (NAM). Neurological and motor functions were assessed using modified neurological severity score and rotarod tests. Brain water content in mice was detected. Primary mouse microglia were exposed to lipopolysaccharide (LPS), ginsenoside Rg3, and NAM. Nissl and immunofluorescence staining were utilized to investigate hippocampal damage, and localization of P65, Iba1 and INOS in microglia. Hippocampal neurons were grown in a culture medium derived from microglia. CCK-8 and TUNEL assays were employed to evaluate the viability and apoptosis of hippocampal neurons. Proinflammatory factors and proteins were tested using ELISA, western blot and immunofluorescence staining. As a result, ginsenoside Rg3 enhanced neurological and motor functions in mice post-TBI, reduced brain water content, alleviated hippocampal neuronal neuroinflammation and damage, activated SIRT1, and deactivated the NF-kB pathway. In LPS-stimulated microglia, ginsenoside Rg3 diminished inflammation, activated SIRT1, deactivated the NF-kB pathway, and facilitated nuclear localization of P65 and co-localization of Iba1 and INOS. The effects of ginsenoside Rg3 were countered by NAM in both TBI mice and LPS-stimulated microglia. Hippocampal neurons cultured in a medium containing LPS, ginsenoside Rg3, and NAM-treated microglia showed improved viability and reduced apoptosis compared to those cultured in a medium with LPS and ginsenoside Rg3-treated microglia alone. Ginsenoside Rg3 was effective in reducing neuroinflammation and damage in hippocampal neurons following TBI by modulating the SIRT1/NF-kB pathway, suggesting its potential as a therapeutic agent for TBI.
Collapse
Affiliation(s)
- Xi Liu
- Department of Neurosurgery, Changsha of Traditional Chinese Medicine Hospital, Changsha, Hunan, China
| | - Jia Gu
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Cheng Wang
- Department of Neurosurgery, Changsha of Traditional Chinese Medicine Hospital, Changsha, Hunan, China
| | - Min Peng
- Department of Neurosurgery, Changsha of Traditional Chinese Medicine Hospital, Changsha, Hunan, China
| | - Jilin Zhou
- Department of Neurosurgery, Changsha of Traditional Chinese Medicine Hospital, Changsha, Hunan, China
| | - Xiyun Fei
- Department of Neurosurgery, Changsha of Traditional Chinese Medicine Hospital, Changsha, Hunan, China
| | - Zhijun Zhong
- Department of Neurosurgery, Changsha of Traditional Chinese Medicine Hospital, Changsha, Hunan, China
| | - Bo Li
- Department of Thoracic Surgery, Changsha of Traditional Chinese Medicine Hospital, Changsha, Hunan, China
| |
Collapse
|
13
|
Boland R, Kokiko-Cochran ON. Deplete and repeat: microglial CSF1R inhibition and traumatic brain injury. Front Cell Neurosci 2024; 18:1352790. [PMID: 38450286 PMCID: PMC10915023 DOI: 10.3389/fncel.2024.1352790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/25/2024] [Indexed: 03/08/2024] Open
Abstract
Traumatic brain injury (TBI) is a public health burden affecting millions of people. Sustained neuroinflammation after TBI is often associated with poor outcome. As a result, increased attention has been placed on the role of immune cells in post-injury recovery. Microglia are highly dynamic after TBI and play a key role in the post-injury neuroinflammatory response. Therefore, microglia represent a malleable post-injury target that could substantially influence long-term outcome after TBI. This review highlights the cell specific role of microglia in TBI pathophysiology. Microglia have been manipulated via genetic deletion, drug inhibition, and pharmacological depletion in various pre-clinical TBI models. Notably, colony stimulating factor 1 (CSF1) and its receptor (CSF1R) have gained much traction in recent years as a pharmacological target on microglia. CSF1R is a transmembrane tyrosine kinase receptor that is essential for microglia proliferation, differentiation, and survival. Small molecule inhibitors targeting CSF1R result in a swift and effective depletion of microglia in rodents. Moreover, discontinuation of the inhibitors is sufficient for microglia repopulation. Attention is placed on summarizing studies that incorporate CSF1R inhibition of microglia. Indeed, microglia depletion affects multiple aspects of TBI pathophysiology, including neuroinflammation, oxidative stress, and functional recovery with measurable influence on astrocytes, peripheral immune cells, and neurons. Taken together, the data highlight an important role for microglia in sustaining neuroinflammation and increasing risk of oxidative stress, which lends to neuronal damage and behavioral deficits chronically after TBI. Ultimately, the insights gained from CSF1R depletion of microglia are critical for understanding the temporospatial role that microglia develop in mediating TBI pathophysiology and recovery.
Collapse
Affiliation(s)
- Rebecca Boland
- Department of Neuroscience, College of Medicine, Chronic Brain Injury Program, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| | - Olga N Kokiko-Cochran
- Department of Neuroscience, College of Medicine, Chronic Brain Injury Program, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
14
|
Kocanci FG, Erol AYG, Yildiz F, Eciroglu H. Pimecrolimus protects neuron-like SH-SY5Y cells against anti-inflammatory and anti-oxidant effects of both microglial secretome and hydrogen peroxide. Scand J Immunol 2024; 99:e13328. [PMID: 38441277 DOI: 10.1111/sji.13328] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/13/2023] [Accepted: 08/18/2023] [Indexed: 03/07/2024]
Abstract
Calcineurin inhibitors have been found to exhibit a preventive role against neuroinflammation, which represents a crucial underlying mechanism in neurodegenerative diseases (ND). Additionally, they possess suppressive effects on the activation of apoptotic pathways, which constitute another mechanism underlying such diseases. Given that pimecrolimus, a calcineurin inhibitor, impedes the synthesis of pro-inflammatory cytokines, such as interleukin (IL)-2, IL-4, and IL-10, and influences apoptotic processes, it is noteworthy to test its potential neuroprotective properties. Thus, the objective of this investigation was to assess the potential protective effects of pimecrolimus against the degenerative consequences of both microglial secretomes and hydrogen peroxide (H2O2), an oxidant agent. The survival rates of HMC3 microglia cells, neuron-like differentiated SH-SY5Y (d-SH-SY5Y) cells, and their co-culture were determined using the 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT) method. Furthermore, the levels of pro-inflammatory cytokines IL-1β and IL-6, and anti-inflammatory cytokine IL-10 were measured using ELISA kits, besides total antioxidant and oxidant capacities in conditioned media of cells. Additionally, the effect of pimecrolimus on neurite length in these cell groups was evaluated through morphological observations. This study revealed, for the first time, that pimecrolimus exerts preventive effects on neurodegenerative processes by virtue of its anti-inflammatory and -antioxidant activities. It holds promise as a potential treatment option for ND.
Collapse
Affiliation(s)
- Fatma Gonca Kocanci
- Department of Medical Laboratory Techniques, Vocational High School of Health Services, Alanya Alaaddin Keykubat University, Alanya, Turkey
| | - Azize Yasemin Goksu Erol
- Faculty of Medicine, Department of Gene and Cell Therapy, Akdeniz University, Antalya, Turkey
- Faculty of Medicine, Department of Histology and Embryology, Akdeniz University, Antalya, Turkey
| | - Fatma Yildiz
- Department of Medical Laboratory Techniques, Vocational High School of Health Services, Alanya Alaaddin Keykubat University, Alanya, Turkey
| | - Hamiyet Eciroglu
- Department of Medical Laboratory Techniques, Vocational High School of Health Services, Alanya Alaaddin Keykubat University, Alanya, Turkey
| |
Collapse
|
15
|
Spisni E, Valerii MC, Massimino ML. Essential Oil Molecules Can Break the Loop of Oxidative Stress in Neurodegenerative Diseases. BIOLOGY 2023; 12:1504. [PMID: 38132330 PMCID: PMC10740714 DOI: 10.3390/biology12121504] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/30/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023]
Abstract
Essential oils (EOs) are mixtures of volatile compounds, extracted from aromatic plants, with multiple activities including antioxidant and anti-inflammatory ones. EOs are complex mixtures easy to find on the market and with low costs. In this mini narrative review, we have collected the results of in vitro and in vivo studies, which tested these EOs on validated models of neurodegeneration and in particular of the two main neurodegenerative diseases (NDs) that afflict humans: Alzheimer's and Parkinson's. Since EO compositions can vary greatly, depending on the environmental conditions, plant cultivar, and extraction methods, we focused our attention to studies involving single EO molecules, and in particular those that have demonstrated the ability to cross the blood-brain barrier. These single EO molecules, alone or in defined mixtures, could be interesting new therapies to prevent or slow down oxidative and inflammatory processes which are common mechanisms that contribute to neuronal death in all NDs.
Collapse
Affiliation(s)
- Enzo Spisni
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, 40126 Bologna, Italy;
- CIRI Life Sciences and Health Technologies, University of Bologna, 40126 Bologna, Italy
| | - Maria Chiara Valerii
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, 40126 Bologna, Italy;
- CIRI Life Sciences and Health Technologies, University of Bologna, 40126 Bologna, Italy
| | - Maria Lina Massimino
- Neuroscience Institute, Italian National Research Council (CNR), 35131 Padova, Italy
| |
Collapse
|
16
|
Chen Y, Zhang M, Li W, Wang X, Chen X, Wu Y, Zhang H, Yang L, Han B, Tang J. Drug repurposing based on the similarity gene expression signatures to explore for potential indications of quercetin: a case study of multiple sclerosis. Front Chem 2023; 11:1250043. [PMID: 37744058 PMCID: PMC10514366 DOI: 10.3389/fchem.2023.1250043] [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: 06/29/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
Quercetin (QR) is a natural flavonol compound widely distributed in the plant kingdom with extensive pharmacological effects. To find the potential clinical indications of QR, 156 differentially expressed genes (DEGs) regulated by QR were obtained from the Gene Expression Omnibus database, and new potential pharmacological effects and clinical indications of QR were repurposed by integrating compounds with similar gene perturbation signatures and associated-disease signatures to QR based on the Connectivity Map and Coexpedia platforms. The results suggested QR has mainly potential therapeutic effects on multiple sclerosis (MS), osteoarthritis, type 2 diabetes mellitus, and acute leukemia. Then, MS was selected for subsequent animal experiments as a representative potential indication, and it found that QR significantly delays the onset time of classical MS model animal mice and ameliorates the inflammatory infiltration and demyelination in the central nervous system. Combined with network pharmacology technology, the therapeutic mechanism of QR on MS was further demonstrated to be related to the inhibition of the expression of inflammatory cytokines (TNF-α, IL-6, IL-1β, IFN-γ, IL-17A, and IL-2) related to TNF-α/TNFR1 signaling pathway. In conclusion, this study expanded the clinical indications of QR and preliminarily confirmed the therapeutic effect and potential mechanism of QR on MS.
Collapse
Affiliation(s)
- Yulong Chen
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Mingliang Zhang
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Weixia Li
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaoyan Wang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaofei Chen
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Yali Wu
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Hui Zhang
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Liuqing Yang
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Bing Han
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jinfa Tang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
17
|
Ravanelli F, Musazzi L, Barbieri SS, Rovati G, Popoli M, Barbon A, Ieraci A. Differential Epigenetic Changes in the Dorsal Hippocampus of Male and Female SAMP8 Mice: A Preliminary Study. Int J Mol Sci 2023; 24:13084. [PMID: 37685895 PMCID: PMC10488283 DOI: 10.3390/ijms241713084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/16/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Alzheimer's disease (AD) is the most common age-related neurodegenerative disease characterized by memory loss and cognitive impairment. The causes of the disease are not well understood, as it involves a complex interaction between genetic, environmental, and epigenetic factors. SAMP8 mice have been proposed as a model for studying late-onset AD, since they show age-related learning and memory deficits as well as several features of AD pathogenesis. Epigenetic changes have been described in SAMP8 mice, although sex differences have never been evaluated. Here we used western blot and qPCR analyses to investigate whether epigenetic markers are differentially altered in the dorsal hippocampus, a region important for the regulation of learning and memory, of 9-month-old male and female SAMP8 mice. We found that H3Ac was selectively reduced in male SAMP8 mice compared to male SAMR1 control mice, but not in female mice, whereas H3K27me3 was reduced overall in SAMP8 mice. Moreover, the levels of HDAC2 and JmjD3 were increased, whereas the levels of HDAC4 and Dnmt3a were reduced in SAMP8 mice compared to SAMR1. In addition, levels of HDAC1 were reduced, whereas Utx and Jmjd3 were selectively increased in females compared to males. Although our results are preliminary, they suggest that epigenetic mechanisms in the dorsal hippocampus are differentially regulated in male and female SAMP8 mice.
Collapse
Affiliation(s)
- Federico Ravanelli
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy; (F.R.); (G.R.); (M.P.)
| | - Laura Musazzi
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| | - Silvia Stella Barbieri
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy;
| | - Gianenrico Rovati
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy; (F.R.); (G.R.); (M.P.)
| | - Maurizio Popoli
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy; (F.R.); (G.R.); (M.P.)
| | - Alessandro Barbon
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy;
| | - Alessandro Ieraci
- Department of Theoretical and Applied Sciences, eCampus University, 22060 Novedrate, Italy
| |
Collapse
|
18
|
Rey F, Berardo C, Maghraby E, Mauri A, Messa L, Esposito L, Casili G, Ottolenghi S, Bonaventura E, Cuzzocrea S, Zuccotti G, Tonduti D, Esposito E, Paterniti I, Cereda C, Carelli S. Redox Imbalance in Neurological Disorders in Adults and Children. Antioxidants (Basel) 2023; 12:antiox12040965. [PMID: 37107340 PMCID: PMC10135575 DOI: 10.3390/antiox12040965] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/03/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Oxygen is a central molecule for numerous metabolic and cytophysiological processes, and, indeed, its imbalance can lead to numerous pathological consequences. In the human body, the brain is an aerobic organ and for this reason, it is very sensitive to oxygen equilibrium. The consequences of oxygen imbalance are especially devastating when occurring in this organ. Indeed, oxygen imbalance can lead to hypoxia, hyperoxia, protein misfolding, mitochondria dysfunction, alterations in heme metabolism and neuroinflammation. Consequently, these dysfunctions can cause numerous neurological alterations, both in the pediatric life and in the adult ages. These disorders share numerous common pathways, most of which are consequent to redox imbalance. In this review, we will focus on the dysfunctions present in neurodegenerative disorders (specifically Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis) and pediatric neurological disorders (X-adrenoleukodystrophies, spinal muscular atrophy, mucopolysaccharidoses and Pelizaeus-Merzbacher Disease), highlighting their underlining dysfunction in redox and identifying potential therapeutic strategies.
Collapse
Affiliation(s)
- Federica Rey
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, University of Milano, 20157 Milano, Italy
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children's Hospital, 20154 Milano, Italy
| | - Clarissa Berardo
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, University of Milano, 20157 Milano, Italy
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children's Hospital, 20154 Milano, Italy
| | - Erika Maghraby
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, University of Milano, 20157 Milano, Italy
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Alessia Mauri
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, University of Milano, 20157 Milano, Italy
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children's Hospital, 20154 Milano, Italy
| | - Letizia Messa
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children's Hospital, 20154 Milano, Italy
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, 20133 Milano, Italy
| | - Letizia Esposito
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, University of Milano, 20157 Milano, Italy
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children's Hospital, 20154 Milano, Italy
| | - Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Sara Ottolenghi
- Department of Medicine and Surgery, University of Milano Bicocca, 20126 Milano, Italy
| | - Eleonora Bonaventura
- Child Neurology Unit, Buzzi Children's Hospital, 20154 Milano, Italy
- Center for Diagnosis and Treatment of Leukodystrophies and Genetic Leukoencephalopathies (COALA), Buzzi Children's Hospital, 20154 Milano, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Gianvincenzo Zuccotti
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, University of Milano, 20157 Milano, Italy
- Department of Pediatrics, Buzzi Children's Hospital, 20154 Milano, Italy
| | - Davide Tonduti
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, University of Milano, 20157 Milano, Italy
- Child Neurology Unit, Buzzi Children's Hospital, 20154 Milano, Italy
- Center for Diagnosis and Treatment of Leukodystrophies and Genetic Leukoencephalopathies (COALA), Buzzi Children's Hospital, 20154 Milano, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Cristina Cereda
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children's Hospital, 20154 Milano, Italy
| | - Stephana Carelli
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, University of Milano, 20157 Milano, Italy
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children's Hospital, 20154 Milano, Italy
| |
Collapse
|
19
|
Wijeweera G, Wijekoon N, Gonawala L, Imran Y, Mohan C, De Silva KRD. Therapeutic Implications of Some Natural Products for Neuroimmune Diseases: A Narrative of Clinical Studies Review. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2023; 2023:5583996. [PMID: 37089709 PMCID: PMC10118888 DOI: 10.1155/2023/5583996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 01/24/2023] [Accepted: 02/11/2023] [Indexed: 04/25/2023]
Abstract
Neuroimmune diseases are a group of disorders that occur due to the dysregulation of both the nervous and immune systems, and these illnesses impact tens of millions of people worldwide. However, patients who suffer from these debilitating conditions have very few FDA-approved treatment options. Neuroimmune crosstalk is important for controlling the immune system both centrally and peripherally to maintain tissue homeostasis. This review aims to provide readers with information on how natural products modulate neuroimmune crosstalk and the therapeutic implications of natural products, including curcumin, epigallocatechin-3-gallate (EGCG), ginkgo special extract, ashwagandha, Centella asiatica, Bacopa monnieri, ginseng, and cannabis to mitigate the progression of neuroimmune diseases, such as Alzheimer's disease, multiple sclerosis, amyotrophic lateral sclerosis, Parkinson's disease, depression, and anxiety disorders. The majority of the natural products based clinical studies mentioned in this study have yielded positive results. To achieve the expected results from natural products based clinical studies, researchers should focus on enhancing bioavailability and determining the synergistic mechanisms of herbal compounds and extracts, which will lead to the discovery of more effective phytomedicines while averting the probable negative effects of natural product extracts. Therefore, future studies developing nutraceuticals to mitigate neuroimmune diseases that incorporate phytochemicals to produce synergistic effects must analyse efficacy, bioavailability, gut-brain axis function safety, chemical modifications, and encapsulation with nanoparticles.
Collapse
Affiliation(s)
- Gayathri Wijeweera
- Institute for Combinatorial Advanced Research and Education (KDU-CARE), General Sir John Kotelawala Defense University, Sri Lanka
- Interdisciplinary Centre for Innovation in Biotechnology and Neurosciences, Faculty of Medical Sciences, University of Sri Jayewardenepura, Sri Lanka
| | - Nalaka Wijekoon
- Institute for Combinatorial Advanced Research and Education (KDU-CARE), General Sir John Kotelawala Defense University, Sri Lanka
- Interdisciplinary Centre for Innovation in Biotechnology and Neurosciences, Faculty of Medical Sciences, University of Sri Jayewardenepura, Sri Lanka
- Department of Cellular Neuroscience, Faculty of Health, Medicine & Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Lakmal Gonawala
- Institute for Combinatorial Advanced Research and Education (KDU-CARE), General Sir John Kotelawala Defense University, Sri Lanka
- Interdisciplinary Centre for Innovation in Biotechnology and Neurosciences, Faculty of Medical Sciences, University of Sri Jayewardenepura, Sri Lanka
- Department of Cellular Neuroscience, Faculty of Health, Medicine & Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Yoonus Imran
- Interdisciplinary Centre for Innovation in Biotechnology and Neurosciences, Faculty of Medical Sciences, University of Sri Jayewardenepura, Sri Lanka
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - K. Ranil D. De Silva
- Institute for Combinatorial Advanced Research and Education (KDU-CARE), General Sir John Kotelawala Defense University, Sri Lanka
- Interdisciplinary Centre for Innovation in Biotechnology and Neurosciences, Faculty of Medical Sciences, University of Sri Jayewardenepura, Sri Lanka
| |
Collapse
|
20
|
Bai R, Song C, Lv S, Chang L, Hua W, Weng W, Wu H, Dai L. Role of microglia in HIV-1 infection. AIDS Res Ther 2023; 20:16. [PMID: 36927791 PMCID: PMC10018946 DOI: 10.1186/s12981-023-00511-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 03/10/2023] [Indexed: 03/18/2023] Open
Abstract
The usage of antiretroviral treatment (ART) has considerably decreased the morbidity and mortality related to HIV-1 (human immunodeficiency virus type 1) infection. However, ART is ineffective in eradicating the virus from the persistent cell reservoirs (e.g., microglia), noticeably hindering the cure for HIV-1. Microglia participate in the progression of neuroinflammation, brain aging, and HIV-1-associated neurocognitive disorder (HAND). Some methods have currently been studied as fundamental strategies targeting microglia. The purpose of this study was to comprehend microglia biology and its functions in HIV-1 infection, as well as to look into potential therapeutic approaches targeting microglia.
Collapse
Affiliation(s)
- Ruojing Bai
- Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Chengcheng Song
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Shiyun Lv
- Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Linlin Chang
- Department of Dermatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Wei Hua
- Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Wenjia Weng
- Department of Dermatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| | - Hao Wu
- Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| | - Lili Dai
- Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
21
|
Ishikawa M, Izumi Y, Sato K, Sato T, Zorumski CF, Kunikata H, Nakazawa T. Glaucoma and microglia-induced neuroinflammation. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1132011. [PMID: 38983051 PMCID: PMC11182182 DOI: 10.3389/fopht.2023.1132011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 02/15/2023] [Indexed: 07/11/2024]
Abstract
Glaucoma is a multifactorial neurodegenerative disease characterized by a progressive optic neuropathy resulting in visual field defects. Elevated intraocular pressure (IOP) is the greatest risk factor for the development of glaucoma, and IOP reduction therapy is the only treatment currently available. However, there are many cases in which retinal degeneration progresses despite sufficient control of IOP. Therefore, it is important to elucidate the pathophysiology of glaucoma that is resistant to current IOP lowering therapies. Experiments using animal glaucoma models show the relationships between microglial neuroinflammatory responses and damage of retinal ganglion cells (RGCs). Inhibition of neuroinflammatory pathways associated with microglial activation appears to be neuroprotective, indicating that microglia may be an important therapeutic target for RGC protection. In this review, we will focus on microglia-induced neuroinflammation in the pathogenesis of glaucoma to offer new insights into the possibility of developing novel neuroprotective therapies targeting microglia.
Collapse
Affiliation(s)
- Makoto Ishikawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yukitoshi Izumi
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, United States
- Center for Brain Research in Mood Disorders, Washington University School of Medicine, St. Louis, MO, United States
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Kota Sato
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Taimu Sato
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Charles F. Zorumski
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, United States
- Center for Brain Research in Mood Disorders, Washington University School of Medicine, St. Louis, MO, United States
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Hiroshi Kunikata
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
22
|
Ni W, Ramalingam M, Li Y, Park JH, Dashnyam K, Lee JH, Bloise N, Fassina L, Visai L, De Angelis MGC, Pedraz JL, Kim HW, Hu J. Immunomodulatory and Anti-inflammatory effect of Neural Stem/Progenitor Cells in the Central Nervous System. Stem Cell Rev Rep 2023; 19:866-885. [PMID: 36650367 DOI: 10.1007/s12015-022-10501-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2022] [Indexed: 01/19/2023]
Abstract
Neuroinflammation is a critical event that responds to disturbed homeostasis and governs various neurological diseases in the central nervous system (CNS). The excessive inflammatory microenvironment in the CNS can adversely affect endogenous neural stem cells, thereby impeding neural self-repair. Therapies with neural stem/progenitor cells (NSPCs) have shown significant inhibitory effects on inflammation, which is mainly achieved through intercellular contact and paracrine signalings. The intercellular contact between NSPCs and immune cells, the activated CNS- resident microglia, and astrocyte plays a critical role in the therapeutic NSPCs homing and immunomodulatory effects. Moreover, the paracrine effect mainly regulates infiltrating innate and adaptive immune cells, activated microglia, and astrocyte through the secretion of bioactive molecules and extracellular vesicles. However, the molecular mechanism involved in the immunomodulatory effect of NSPCs is not well discussed. This article provides a systematic analysis of the immunomodulatory mechanism of NSPCs, discusses efficient ways to enhance its immunomodulatory ability, and gives suggestions on clinical therapy.
Collapse
Affiliation(s)
- Wei Ni
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China.,Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Murugan Ramalingam
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea. .,Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea. .,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea. .,School of Basic Medical Sciences, Chengdu University, Chengdu, 610106, People's Republic of China.
| | - Yumeng Li
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea.,Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jeong-Hui Park
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea.,Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Khandmaa Dashnyam
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea.,Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Nora Bloise
- Department of Molecular Medicine, Centre for Health Technologies (CHT), INSTM UdR of Pavia, University of Pavia, 27100, Pavia, Italy.,Medicina Clinica-Specialistica, UOR5 Laboratorio di Nanotecnologie, ICS Maugeri, IRCCS, 27100, Pavia, Italy
| | - Lorenzo Fassina
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, 27100, Pavia, Italy
| | - Livia Visai
- Department of Molecular Medicine, Centre for Health Technologies (CHT), INSTM UdR of Pavia, University of Pavia, 27100, Pavia, Italy.,Medicina Clinica-Specialistica, UOR5 Laboratorio di Nanotecnologie, ICS Maugeri, IRCCS, 27100, Pavia, Italy
| | | | - Jose Luis Pedraz
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006, Vitoria-Gasteiz, Spain.,Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, 28029, Madrid, Spain
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea. .,Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea. .,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea.
| | - Jiabo Hu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
23
|
Maurya SK, Gupta S, Mishra R. Transcriptional and epigenetic regulation of microglia in maintenance of brain homeostasis and neurodegeneration. Front Mol Neurosci 2023; 15:1072046. [PMID: 36698776 PMCID: PMC9870594 DOI: 10.3389/fnmol.2022.1072046] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/15/2022] [Indexed: 01/12/2023] Open
Abstract
The emerging role of microglia in brain homeostasis, neurodegeneration, and neurodevelopmental disorders has attracted considerable interest. In addition, recent developments in microglial functions and associated pathways have shed new light on their fundamental role in the immunological surveillance of the brain. Understanding the interconnections between microglia, neurons, and non-neuronal cells have opened up additional avenues for research in this evolving field. Furthermore, the study of microglia at the transcriptional and epigenetic levels has enhanced our knowledge of these native brain immune cells. Moreover, exploring various facets of microglia biology will facilitate the early detection, treatment, and management of neurological disorders. Consequently, the present review aimed to provide comprehensive insight on microglia biology and its influence on brain development, homeostasis, management of disease, and highlights microglia as potential therapeutic targets in neurodegenerative and neurodevelopmental diseases.
Collapse
Affiliation(s)
- Shashank Kumar Maurya
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India,*Correspondence: Shashank Kumar Maurya, ;
| | - Suchi Gupta
- Tech Cell Innovations Private Limited, Centre for Medical Innovation and Entrepreneurship (CMIE), All India Institute of Medical Sciences, New Delhi, India
| | - Rajnikant Mishra
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, India
| |
Collapse
|
24
|
Kamaruzzaman MA, Romli MH, Abas R, Vidyadaran S, Hidayat Baharuldin MT, Nasaruddin ML, Thirupathirao V, Sura S, Warsito K, Mohd Nor NH, Azwaruddin MA, Alshawsh MA, Mohd Moklas MA. Regulatory role of the endocannabinoid system on glial cells toward cognitive function in Alzheimer's disease: A systematic review and meta-analysis of animal studies. Front Pharmacol 2023; 14:1053680. [PMID: 36959856 PMCID: PMC10028478 DOI: 10.3389/fphar.2023.1053680] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 02/08/2023] [Indexed: 03/06/2023] Open
Abstract
Objective: Over the last decade, researchers have sought to develop novel medications against dementia. One potential agent under investigation is cannabinoids. This review systematically appraised and meta-analyzed published pre-clinical research on the mechanism of endocannabinoid system modulation in glial cells and their effects on cognitive function in animal models of Alzheimer's disease (AD). Methods: A systematic review complying with PRISMA guidelines was conducted. Six databases were searched: EBSCOHost, Scopus, PubMed, CINAHL, Cochrane, and Web of Science, using the keywords AD, cannabinoid, glial cells, and cognition. The methodological quality of each selected pre-clinical study was evaluated using the SYRCLE risk of bias tool. A random-effects model was applied to analyze the data and calculate the effect size, while I2 and p-values were used to assess heterogeneity. Results: The analysis included 26 original articles describing (1050 rodents) with AD-like symptoms. Rodents treated with cannabinoid agonists showed significant reductions in escape latency (standard mean difference [SMD] = -1.26; 95% confidence interval [CI]: -1.77 to -0.76, p < 0.00001) and ability to discriminate novel objects (SMD = 1.40; 95% CI: 1.04 to 1.76, p < 0.00001) compared to the control group. Furthermore, a significant decrease in Aβ plaques (SMD = -0.91; 95% CI: -1.55 to -0.27, p = 0.006) was observed in the endocannabinoid-treated group compared to the control group. Trends were observed toward neuroprotection, as represented by decreased levels of glial cell markers including glial fibrillary acid protein (SMD = -1.47; 95% CI: -2.56 to -0.38, p = 0.008) and Iba1 (SMD = -1.67; 95% CI: -2.56 to -0.79, p = 0.0002). Studies on the wild-type mice demonstrated significantly decreased levels of pro-inflammatory markers TNF-α, IL-1, and IL-6 (SMD = -2.28; 95% CI: -3.15 to -1.41, p = 0.00001). Despite the non-significant decrease in pro-inflammatory marker levels in transgenic mice (SMD = -0.47; 95% CI: -1.03 to 0.08, p = 0.09), the result favored the endocannabinoid-treated group over the control group. Conclusion: The revised data suggested that endocannabinoid stimulation promotes cognitive function via modulation of glial cells by decreasing pro-inflammatory markers in AD-like rodent models. Thus, cannabinoid agents may be required to modulate the downstream chain of effect to enhance cognitive stability against concurrent neuroinflammation in AD. Population-based studies and well-designed clinical trials are required to characterize the acceptability and real-world effectiveness of cannabinoid agents. Systematic Review Registration: [https://inplasy.com/inplasy-2022-8-0094/], identifier [Inplasy Protocol 3770].
Collapse
Affiliation(s)
- Mohd Amir Kamaruzzaman
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Department of Human Anatomy, Faculty of Medicine and Health Science, Universiti Putra Malaysia, Kuala Lumpur, Malaysia
| | - Muhammad Hibatullah Romli
- Department of Nursing and Rehabilitation, Faculty of Medicine and Health Science, Universiti Putra Malaysia, Kuala Lumpur, Malaysia
| | - Razif Abas
- Department of Human Anatomy, Faculty of Medicine and Health Science, Universiti Putra Malaysia, Kuala Lumpur, Malaysia
| | - Sharmili Vidyadaran
- Department of Pathology, Faculty of Medicine and Health Science, Universiti Putra Malaysia, Kuala Lumpur, Malaysia
| | | | | | | | - Sreenivasulu Sura
- Department of Human Anatomy, Faculty of Medicine and Health Science, Universiti Putra Malaysia, Kuala Lumpur, Malaysia
- Department of Preclinical Sciences, Faculty of Medicine and Health Sciences, University Tunku Abdul Rahman, Kampar, Malaysia
| | - Kabul Warsito
- Department of Agrotechnology, Faculty of Science and Technology, University of Pembangunan Panca Budi, Medan, Indonesia
| | - Nurul Huda Mohd Nor
- Department of Human Anatomy, Faculty of Medicine and Health Science, Universiti Putra Malaysia, Kuala Lumpur, Malaysia
| | - Muhammad Amsyar Azwaruddin
- Department of Human Anatomy, Faculty of Medicine and Health Science, Universiti Putra Malaysia, Kuala Lumpur, Malaysia
| | - Mohammed Abdullah Alshawsh
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- Department of Paediatrics, School of Clinical Sciences, Faculty of Medicine, Nursing and Health Science, Monash University, Clayton, VIC, Australia
- *Correspondence: Mohamad Aris Mohd Moklas, ; Mohammed Abdullah Alshawsh,
| | - Mohamad Aris Mohd Moklas
- Department of Human Anatomy, Faculty of Medicine and Health Science, Universiti Putra Malaysia, Kuala Lumpur, Malaysia
- *Correspondence: Mohamad Aris Mohd Moklas, ; Mohammed Abdullah Alshawsh,
| |
Collapse
|
25
|
Silva P, Portillo MP, Fernández-Quintela A. Resveratrol and Wine: An Overview of Thirty Years in the Digital News. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:15815. [PMID: 36497888 PMCID: PMC9740773 DOI: 10.3390/ijerph192315815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Resveratrol's health benefits have received wide media coverage. Since resveratrol is usually associated with wine, informative texts about it should be prepared very carefully, since inaccurate website content could easily change people's wine consumption behavior. This study aimed to assess the quality of informative texts related to resveratrol on science journalism websites. METHODS We analyzed 125 resveratrol posts on Science Daily, WebMD, and EurekAlert! published between 1990 and 2020. RESULTS A higher number of posts was published in the years in which the number of people looking for information on the internet also increased. The increase can also be related to David Sinclair's notoriety, a fact that we called the "Sinclair effect". Most of the posts are replications of universities' press releases, mainly reporting resveratrol's health benefits, which resulted from preclinical studies and cannot be translated to humans. Most of them mention wine in the text and some in the title. CONCLUSIONS Wine is usually mentioned in headline resveratrol news, which could potentially influence wine consumption behavior. Scientists must intensify their efforts to communicate with the public to increase people's health literacy. Online news portals should have science journalists skilled in exploring scientific data and their translation into a simple and accurate language.
Collapse
Affiliation(s)
- Paula Silva
- Laboratory of Histology and Embryology, Institute of Biomedical Sciences Abel Salazar (ICBAS), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- ICNOVA—NOVA Institute of Communication, NOVA School of Social Sciences and Humanities, Universidade NOVA de Lisboa, 1069-061 Lisbon, Portugal
| | - María P. Portillo
- Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain
- Lucio Lascaray Research Institute, 01006 Vitoria-Gasteiz, Spain
- BIOARABA Health Research Institute, 01006 Vitoria-Gasteiz, Spain
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, 01006 Vitoria-Gasteiz, Spain
| | - Alfredo Fernández-Quintela
- Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain
- Lucio Lascaray Research Institute, 01006 Vitoria-Gasteiz, Spain
- BIOARABA Health Research Institute, 01006 Vitoria-Gasteiz, Spain
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, 01006 Vitoria-Gasteiz, Spain
| |
Collapse
|
26
|
Park J, Kim Y, Lee C, Kim YT. 3,5-Dicaffeoylquinic acid attenuates microglial activation-mediated inflammatory pain by enhancing autophagy through the suppression of MCP3/JAK2/STAT3 signaling. Biomed Pharmacother 2022; 153:113549. [DOI: 10.1016/j.biopha.2022.113549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/02/2022] Open
|
27
|
Kang N, Shi Y, Song J, Gao F, Fan M, Jin W, Gao Y, Lv P. Resveratrol reduces inflammatory response and detrimental effects in chronic cerebral hypoperfusion by down-regulating stimulator of interferon genes/TANK-binding kinase 1/interferon regulatory factor 3 signaling. Front Aging Neurosci 2022; 14:868484. [PMID: 35936778 PMCID: PMC9354401 DOI: 10.3389/fnagi.2022.868484] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 06/27/2022] [Indexed: 11/29/2022] Open
Abstract
Inflammatory responses induced by chronic cerebral hypoperfusion (CCH) play a critical role in the progression of vascular dementia. Stimulator of interferon genes (STING) signaling function as a key mediator of inflammation and immunological responses in the central nervous system (CNS), and resveratrol (RES) exerts potent anti-inflammatory effects. However, the role of STING signaling and the relationship between RES and STING signaling in persistent hypoperfusion-induced cerebral inflammation remain unclear. In this study, Sprague–Dawley rats were subjected to either Sham or bilateral common carotid artery occlusion (2VO) surgery and received RES or vehicle daily by intraperitoneal injection for 4 or 8 weeks. Morris’s water maze was used for the analysis of cognitive function. The neuroinflammatory responses in white matter and hippocampus of the rat brain were assessed by Western blot, Immunofluorescence staining, and qRT-PCR analyses. Myelin integrity, neutrophil infiltration, and microglia proliferation were assessed by Immunohistochemistry and histologic analysis. We demonstrated that after CCH, neurons, microglia, and astrocyte under endoplasmic reticulum (ER) stress upregulated the expression of STING, TANK-binding kinase 1 (TBK1), and the transcription factor interferon regulatory factor 3 (IRF3), as well as translocation of IRF3 into the nucleus. These were accompanied by infiltration of neutrophils, activation of microglia, and overproduction of proinflammatory mediators. Improvements in cognitive deficits were related to reduced hippocampal neuronal cell death and increased myelin integrity in RES-treated rats. The neuroprotective effects of RES were associated with suppression of the expression of tumor necrosis factor-alpha (TNF-α), intercellular adhesion molecule 1 (ICAM-1), VCAM-1, interferon-β (IFN-β), and IL-1β, likely through mitigation of the STING/TBK1/IRF3 pathway. These inhibitory effects exerted by RES also inhibited the levels of myeloperoxidase, reduced excess expression of reactive astrocytes, and activated microglia. In conclusion, the STING/TBK1/IRF3 axis may be critical for proinflammatory responses in cerebral tissue with persistent hypoperfusion, and RES exerts its anti-inflammatory effects by suppressing STING/TBK1/IRF3 signaling.
Collapse
Affiliation(s)
- Ning Kang
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Yuanyuan Shi
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Jiaxi Song
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Fei Gao
- Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Mingyue Fan
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Wei Jin
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Yaran Gao
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Peiyuan Lv
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
- *Correspondence: Peiyuan Lv,
| |
Collapse
|
28
|
Maurya SK, Gupta S, Bakshi A, Kaur H, Jain A, Senapati S, Baghel MS. Targeting mitochondria in the regulation of neurodegenerative diseases: A comprehensive review. J Neurosci Res 2022; 100:1845-1861. [PMID: 35856508 DOI: 10.1002/jnr.25110] [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: 03/20/2022] [Revised: 06/21/2022] [Accepted: 07/09/2022] [Indexed: 11/09/2022]
Abstract
Mitochondria are one of the essential cellular organelles. Apart from being considered as the powerhouse of the cell, mitochondria have been widely known to regulate redox reaction, inflammation, cell survival, cell death, metabolism, etc., and are implicated in the progression of numerous disease conditions including neurodegenerative diseases. Since brain is an energy-demanding organ, mitochondria and their functions are important for maintaining normal brain homeostasis. Alterations in mitochondrial gene expression, mutations, and epigenetic modification contribute to inflammation and neurodegeneration. Dysregulation of reactive oxygen species production by mitochondria and aggregation of proteins in neurons leads to alteration in mitochondria functions which further causes neuronal death and progression of neurodegeneration. Pharmacological studies have prioritized mitochondria as a possible drug target in the regulation of neurodegenerative diseases. Therefore, the present review article has been intended to provide a comprehensive understanding of mitochondrial role in the development and progression of neurodegenerative diseases mainly Alzheimer's, Parkinson's, multiple sclerosis, and amyotrophic lateral sclerosis followed by possible intervention and future treatment strategies to combat mitochondrial-mediated neurodegeneration.
Collapse
Affiliation(s)
| | - Suchi Gupta
- Stem Cell Facility, All India Institute of Medical Sciences, Delhi, India
| | - Amrita Bakshi
- Department of Zoology, University of Delhi, Delhi, India
| | - Harpreet Kaur
- Department of Zoology, University of Delhi, Delhi, India.,Division of Infectious Disease, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Arushi Jain
- Immunogenomics Laboratory, Department of Human Genetics & Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Sabyasachi Senapati
- Immunogenomics Laboratory, Department of Human Genetics & Molecular Medicine, Central University of Punjab, Bathinda, India
| | | |
Collapse
|
29
|
Cai Y, Liu J, Wang B, Sun M, Yang H. Microglia in the Neuroinflammatory Pathogenesis of Alzheimer's Disease and Related Therapeutic Targets. Front Immunol 2022; 13:856376. [PMID: 35558075 PMCID: PMC9086828 DOI: 10.3389/fimmu.2022.856376] [Citation(s) in RCA: 116] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease worldwide, characterized by progressive neuron degeneration or loss due to excessive accumulation of β-amyloid (Aβ) peptides, formation of neurofibrillary tangles (NFTs), and hyperphosphorylated tau. The treatment of AD has been only partially successful as the majority of the pharmacotherapies on the market may alleviate some of the symptoms. In the occurrence of AD, increasing attention has been paid to neurodegeneration, while the resident glial cells, like microglia are also observed. Microglia, a kind of crucial glial cells associated with the innate immune response, functions as double-edge sword role in CNS. They exert a beneficial or detrimental influence on the adjacent neurons through secretion of both pro-inflammatory cytokines as well as neurotrophic factors. In addition, their endocytosis of debris and toxic protein like Aβ and tau ensures homeostasis of the neuronal microenvironment. In this review, we will systematically summarize recent research regarding the roles of microglia in AD pathology and latest microglia-associated therapeutic targets mainly including pro-inflammatory genes, anti-inflammatory genes and phagocytosis at length, some of which are contradictory and controversial and warrant to further be investigated.
Collapse
Affiliation(s)
| | | | | | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hao Yang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
30
|
Sahebkar A, Sathyapalan T, Guest PC, Barreto GE. Identification of difluorinated curcumin molecular targets linked to traumatic brain injury pathophysiology. Biomed Pharmacother 2022; 148:112770. [PMID: 35278853 DOI: 10.1016/j.biopha.2022.112770] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/18/2022] [Accepted: 02/27/2022] [Indexed: 11/02/2022] Open
Abstract
Traumatic brain injury (TBI) affects approximately 50% of the world population at some point in their lifetime. To date, there are no effective treatments as most of the damage occurs due to secondary effects through a variety of pathophysiological pathways. The phytoceutical curcumin has been traditionally used as a natural remedy for numerous conditions including diabetes, inflammatory diseases, and neurological and neurodegenerative disorders. We have carried out a system pharmacology study to identify potential targets of a difluorinated curcumin analogue (CDF) that overlap with those involved in the pathophysiological mechanisms of TBI. This resulted in identification of 312 targets which are mostly involved in G protein-coupled receptor activity and cellular signalling. These include adrenergic, serotonergic, opioid and cannabinoid receptor families, which have been implicated in regulation of pain, inflammation, mood, learning and cognition pathways. We conclude that further studies should be performed to validate curcumin as a potential novel treatment to ameliorate the effects of TBI.
Collapse
Affiliation(s)
- Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, Australia; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, United Kingdom
| | - Paul C Guest
- Department of Biochemistry and Tissue Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.
| |
Collapse
|
31
|
Salas-Venegas V, Flores-Torres RP, Rodríguez-Cortés YM, Rodríguez-Retana D, Ramírez-Carreto RJ, Concepción-Carrillo LE, Pérez-Flores LJ, Alarcón-Aguilar A, López-Díazguerrero NE, Gómez-González B, Chavarría A, Konigsberg M. The Obese Brain: Mechanisms of Systemic and Local Inflammation, and Interventions to Reverse the Cognitive Deficit. Front Integr Neurosci 2022; 16:798995. [PMID: 35422689 PMCID: PMC9002268 DOI: 10.3389/fnint.2022.798995] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Overweight and obesity are now considered a worldwide pandemic and a growing public health problem with severe economic and social consequences. Adipose tissue is an organ with neuroimmune-endocrine functions, which participates in homeostasis. So, adipocyte hypertrophy and hyperplasia induce a state of chronic inflammation that causes changes in the brain and induce neuroinflammation. Studies with obese animal models and obese patients have shown a relationship between diet and cognitive decline, especially working memory and learning deficiencies. Here we analyze how obesity-related peripheral inflammation can affect central nervous system physiology, generating neuroinflammation. Given that the blood-brain barrier is an interface between the periphery and the central nervous system, its altered physiology in obesity may mediate the consequences on various cognitive processes. Finally, several interventions, and the use of natural compounds and exercise to prevent the adverse effects of obesity in the brain are also discussed.
Collapse
Affiliation(s)
- Verónica Salas-Venegas
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana - Unidad Iztapalapa, Mexico City, Mexico
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
| | - Rosa Pamela Flores-Torres
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana - Unidad Iztapalapa, Mexico City, Mexico
- Departamento de Biología de la Reproducción, DCBS, Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México (CDMX), Mexico City, Mexico
| | - Yesica María Rodríguez-Cortés
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Diego Rodríguez-Retana
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Ricardo Jair Ramírez-Carreto
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Luis Edgar Concepción-Carrillo
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Laura Josefina Pérez-Flores
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
| | - Adriana Alarcón-Aguilar
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
| | - Norma Edith López-Díazguerrero
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
| | - Beatriz Gómez-González
- Departamento de Biología de la Reproducción, DCBS, Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México (CDMX), Mexico City, Mexico
| | - Anahí Chavarría
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Mina Konigsberg
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
- *Correspondence: Mina Konigsberg,
| |
Collapse
|
32
|
Maurya SK, Mishra R. Molecular docking studies of natural immunomodulators indicate their interactions with the CD40L of CD40/CD40L pathway and CSF1R kinase domain of microglia. J Mol Model 2022; 28:101. [PMID: 35325302 DOI: 10.1007/s00894-022-05084-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/08/2022] [Indexed: 10/18/2022]
Abstract
Natural products have proved beneficial in reducing neuroinflammation in neurological diseases. Their impacts have also been associated with the activities of microglia, responsible for brain-specific immunity. Recent studies have shown the involvement of the number of microglia-specific proteins in the regulation of brain-specific immunity. However, molecular targets of natural products and their mechanism of interaction with microglia-specific proteins are elusive. Since the genetic signature of microglia offers many potential targets for drug discovery, molecular docking followed by molecular dynamics (MD) simulations of cluster of differentiation 40 ligand (CD40L) and colony-stimulating factor 1 receptor (CSF1R) kinase domain protein with some known neuro-immunomodulators (Curcumin, Cannabidiol, Ginsenoside Rg1, Resveratrol, and Sulforaphane) has been evaluated. Curcumin and cannabidiol were observed likely to modulate CD40L and expression of cytokines and entry of inflammatory cells. Resveratrol and cannabidiol may affect the CSF1R kinase domain and activation of microglia. Our finding suggests that curcumin, cannabidiol, and resveratrol may serve specific drug ligands in regulating microglia-mediated brain immunity.
Collapse
Affiliation(s)
- Shashank Kumar Maurya
- Department of Zoology, Ramjas College, University of Delhi, 110007, Delhi, India.,Department of Zoology, School of Sciences, Cluster University of Jammu, 180001, Jammu, India.,Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, 221005, Varanasi, India
| | - Rajnikant Mishra
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, 221005, Varanasi, India.
| |
Collapse
|
33
|
Ikram M, Jo MH, Choe K, Khan A, Ahmad S, Saeed K, Kim MW, Kim MO. Cycloastragenol, a Triterpenoid Saponin, Regulates Oxidative Stress, Neurotrophic Dysfunctions, Neuroinflammation and Apoptotic Cell Death in Neurodegenerative Conditions. Cells 2021; 10:2719. [PMID: 34685699 PMCID: PMC8534642 DOI: 10.3390/cells10102719] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/24/2022] Open
Abstract
Here, we have unveiled the effects of cycloastragenol against Aβ (Amyloid-beta)-induced oxidative stress, neurogenic dysfunction, activated mitogen-activated protein (MAP) kinases, and mitochondrial apoptosis in an Aβ-induced mouse model of Alzheimer's disease (AD). The Aβ-induced mouse model was developed by the stereotaxic injection of amyloid-beta (5 μg/mouse/intracerebroventricular), and cycloastragenol was given at a dose of 20 mg/kg/day/p.o for 6 weeks daily. For the biochemical analysis, we used immunofluorescence and Western blotting. Our findings showed that the injection of Aβ elevated oxidative stress and reduced the expression of neurogenic markers, as shown by the reduced expression of brain-derived neurotrophic factor (BDNF) and the phosphorylation of its specific receptor tropomyosin receptor kinase B (p-TrKB). In addition, there was a marked reduction in the expression of NeuN (neuronal nuclear protein) in the Aβ-injected mice brains (cortex and hippocampus). Interestingly, the expression of Nrf2 (nuclear factor erythroid 2-related factor 2), HO-1 (heme oxygenase-1), p-TrKB, BDNF, and NeuN was markedly enhanced in the Aβ + Cycloastragenol co-treated mice brains. We have also evaluated the expressions of MAP kinases such as phospho c-Jun-N-terminal kinase (p-JNK), p-38, and phospho-extracellular signal-related kinase (ERK1/2) in the experimental groups, which suggested that the expression of p-JNK, p-P-38, and p-Erk were significantly upregulated in the Aβ-injected mice brains; interestingly, these markers were downregulated in the Aβ + Cycloastragenol co-treated mice brains. We also checked the expression of activated microglia and inflammatory cytokines, which showed that cycloastragenol reduced the activated microglia and inflammatory cytokines. Moreover, we evaluated the effects of cycloastragenol against mitochondrial apoptosis and memory dysfunctions in the experimental groups. The findings showed significant regulatory effects against apoptosis and memory dysfunction as revealed by the Morris water maze (MWM) test. Collectively, the findings suggested that cycloastragenol regulates oxidative stress, neurotrophic processes, neuroinflammation, apoptotic cell death, and memory impairment in the mouse model of AD.
Collapse
Affiliation(s)
- Muhammad Ikram
- Division of Life Science and Applied Life Science (BK21 Four), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (M.I.); (M.H.J.); (A.K.); (S.A.); (K.S.); (M.W.K.)
| | - Myeung Hoon Jo
- Division of Life Science and Applied Life Science (BK21 Four), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (M.I.); (M.H.J.); (A.K.); (S.A.); (K.S.); (M.W.K.)
| | - Kyonghwan Choe
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6211 LK Maastricht, The Netherlands;
| | - Amjad Khan
- Division of Life Science and Applied Life Science (BK21 Four), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (M.I.); (M.H.J.); (A.K.); (S.A.); (K.S.); (M.W.K.)
| | - Sareer Ahmad
- Division of Life Science and Applied Life Science (BK21 Four), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (M.I.); (M.H.J.); (A.K.); (S.A.); (K.S.); (M.W.K.)
| | - Kamran Saeed
- Division of Life Science and Applied Life Science (BK21 Four), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (M.I.); (M.H.J.); (A.K.); (S.A.); (K.S.); (M.W.K.)
| | - Min Woo Kim
- Division of Life Science and Applied Life Science (BK21 Four), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (M.I.); (M.H.J.); (A.K.); (S.A.); (K.S.); (M.W.K.)
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 Four), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (M.I.); (M.H.J.); (A.K.); (S.A.); (K.S.); (M.W.K.)
- Alz-Dementia Korea Co., Jinju 52828, Korea
| |
Collapse
|
34
|
The Influence of Virus Infection on Microglia and Accelerated Brain Aging. Cells 2021; 10:cells10071836. [PMID: 34360004 PMCID: PMC8303900 DOI: 10.3390/cells10071836] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
Microglia are the resident immune cells of the central nervous system contributing substantially to health and disease. There is increasing evidence that inflammatory microglia may induce or accelerate brain aging, by interfering with physiological repair and remodeling processes. Many viral infections affect the brain and interfere with microglia functions, including human immune deficiency virus, flaviviruses, SARS-CoV-2, influenza, and human herpes viruses. Especially chronic viral infections causing low-grade neuroinflammation may contribute to brain aging. This review elucidates the potential role of various neurotropic viruses in microglia-driven neurocognitive deficiencies and possibly accelerated brain aging.
Collapse
|