1
|
Sun Y, Gao SQ, Wang X, Li T, Han YL, Miao SH, Zhao R, Zheng XB, Qiu JY, Jin WX, Gao CC, Zhou ML. Galectin-3 activates microglia and promotes neurological impairment via NLRP3/pyroptosis pathway following traumatic brain injury. Brain Res 2025; 1855:149560. [PMID: 40074166 DOI: 10.1016/j.brainres.2025.149560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/01/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Externally caused traumatic brain injury (TBI) poses a woeful worldwide health concern, bringing about disability, death, and prolonged neurological impairment. Increased galectin-3 levels have been linked to unfavorable outcomes in several neurological conditions. This study explores the role of galectin-3 in TBI, specifically examining its contribution to neuroinflammation. METHODS BV2 microglia cells treated with lipopolysaccharide (LPS) and a mouse model of TBI were applied to investigate the impact of galectin-3 on neuroinflammation following TBI. Western blotting and immunofluorescence labeling were applied for evaluating protein levels and colocalization. Adeno-associated virus (AAV) that targets microglia was used to knock down galectin-3 in microglia. Nissl staining and the modified neurologic severity score were employed in evaluating neural survival and neurological function, and the cognitive impairment following TBI was assessed by the Y-Maze and Morri water maze test. RESULTS Galectin-3 expression was shown to rise dramatically after TBI, peaking between days five and seven. In vitro, BV2 cells treated with LPS showed reduced NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome activation when galectin-3 was inhibited. In LPS-activated microglia, galectin-3 inhibition specifically decreased the expression of Toll-like receptor 4 (TLR4), nuclear factor-κB (NF-κB), p-NF-κB, NLRP3, Apoptosis-associated speck-like protein containing a CARD (ASC), caspase-1, and Gasdermin D (GSDMD). Injection with AAV containing siRNA to knock down galectin-3 in microglia was operated on mice in vivo. Following TBI, this knockdown led to reduced NLRP3 inflammasome activation, neuronal death, neurological impairments and cognitive impairment. CONCLUSIONS Our foundings indicate that modulating microglia-derived galectin-3 following TBI to reduce neuroinflammation could serve as a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yan Sun
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Sheng-Qing Gao
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xue Wang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tao Li
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Yan-Ling Han
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shu-Hao Miao
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Ran Zhao
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiao-Bo Zheng
- Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing. China
| | - Jia-Yin Qiu
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wang-Xuan Jin
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chao-Chao Gao
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Meng-Liang Zhou
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, China; Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
2
|
Tang X, Huang L, Ma W, Huang M, Zeng Z, Yu Y, Qin N, Zhou F, Li F, Gong S, Yang H. Intestinal 8 gingerol attenuates TBI-induced neuroinflammation by inhibiting microglia NLRP3 inflammasome activation in a PINK1/Parkin-dependent manner. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156580. [PMID: 40058316 DOI: 10.1016/j.phymed.2025.156580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/23/2025] [Accepted: 02/24/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND traumatic brain injury (TBI) is irreversible brain damage, leading to inflammation and cognitive dysfunction. Microglia involved in the inflammatory response after TBI. The gut microbiota, known as the body's "second brain," regulates neurogenesis and immune responses, but its precise role in regulating TBI remains unclear. PURPOSE to investigate the effect of gut microbiota and metabolites disorder on TBI injury. STUDY DESIGN 16SrRNA and metabolomics compared gut microbiota and metabolites in sham group and TBI group, then proved that the differential metabolite 8-gingerol (8G) alleviated the microglia neuroinflammatory response after TBI. METHODS fecal microbiota transplantation explored the role of dysbiosis in TBI. LC/MS detected the content of 8-gingerol in cecum, blood, and brain. HE, Nissl, Tunel staining and mNSS score evaluated brain injury. Western blot and immunofluorescence detected the expression of inflammasome-related proteins and mitophagy-related proteins in brain tissue and BV2 cells. RNA sequencing analyzed the molecular mechanism of 8-gingerol. RESULT rats transplanted with TBI feces had worse brain injury and neurological deficits than those with normal feces. 16SrRNA and metabolomics found that TBI caused dysbiosis and decreased 8-gingerol level, leading to severe neuroinflammation. Mechanistically, 8-gingerol inhibited NLRP3 inflammasome by promoting PINK1-Parkin mediated mitophagy in microglia. Inhibition of Parkin, through either small interfering RNA or the inhibitor 3MA reversed the inhibitory effect of 8-gingerol on NLRP3 by blocking mitophagy. BV2 cells transcriptome showed that 8-gingerol significantly increased the expression of autophagy factor Wipi1, and small interfering RNA of Wipi1 abolished the effect of 8-gingerol on promoting mitophagy and the inhibitory effect on NLRP3. CONCLUSION our findings shed light on the pivotal role of gut microbes in TBI, and identify 8 gingerol as an important anti-inflammatory compound during TBI.
Collapse
Affiliation(s)
- Xuheng Tang
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, 510665, China; The Third Clinical College of Southern Medical University, China
| | - Lin Huang
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, 510665, China; The Third Clinical College of Southern Medical University, China
| | - Weiquan Ma
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, 510665, China; The Third Clinical College of Southern Medical University, China
| | - Mingxin Huang
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, 510665, China; The Third Clinical College of Southern Medical University, China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yiqin Yu
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, 510665, China; The Third Clinical College of Southern Medical University, China
| | - Na Qin
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, 510665, China; The Third Clinical College of Southern Medical University, China
| | - Fei Zhou
- Central Hospital of Guangdong Prison, Guangzhou 510430, China
| | - Fen Li
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, 510665, China; The Third Clinical College of Southern Medical University, China.
| | - Shenhai Gong
- School of Traditional Chinese Medicine, Southern Medical University, 510515, China.
| | - Hong Yang
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, 510665, China; The Third Clinical College of Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou 510515, China.
| |
Collapse
|
3
|
Zhang W, Zhang L, Fu S, Yan R, Zhang X, Song J, Lu Y. Roles of NLRC4 inflammasome in neurological disorders: Mechanisms, implications, and therapeutic potential. Pharmacol Ther 2025; 267:108803. [PMID: 39855275 DOI: 10.1016/j.pharmthera.2025.108803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 01/01/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
The nucleotide-binding oligomerization domain-like receptor family caspase recruitment domain containing 4 (NLRC4) inflammasome, a vital component of the innate immune system, is known for defending against bacterial infections. However, recent insights have revealed its significant impact on neurological disorders. This comprehensive review discussed the mechanisms underlying the activation and regulation of the NLRC4 inflammasome, highlighting the complexity of its response to cellular stress and damage signals. The biological functions of NLRC4 were explored, particularly its influence on cytokine production and the induction of pyroptosis, a form of inflammatory cell death. This review further emphasized the role of the NLRC4 inflammasome in brain injuries and neurodegenerative disorders. In the realm of brain injuries such as stroke and traumatic brain injury, as well as in neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, multiple sclerosis, and amyotrophic lateral sclerosis, the NLRC4 inflammasome played a pivotal role in modulating neuroinflammatory responses, which was crucial for understanding the progression and potential therapeutic targeting of these conditions. The emerging role of NLRC4 in psychiatric disorders and its potential impact on glioma progression were also examined. Additionally, this review presented a thorough summary of the latest research on inhibitors that impeded the assembly and activation of the NLRC4 inflammasome, pointing to new therapeutic possibilities in neurological disorders. In conclusion, by integrating current knowledge on the activation and regulation of NLRC4 with its biological functions and clinical implications, this article underscored the importance of NLRC4 inflammasome in neurological pathologies, which opened new possibilities for the treatment of challenging neurological conditions.
Collapse
Affiliation(s)
- Wen Zhang
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Li Zhang
- Beijing Key Laboratory of Polymorphic Drugs, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Shuo Fu
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Rong Yan
- Center for Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xue Zhang
- Center for Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Junke Song
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China.
| | - Yang Lu
- Beijing Key Laboratory of Polymorphic Drugs, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
4
|
Qi Y, Rajbanshi B, Hao R, Dang Y, Xu C, Lu W, Dai L, Zhang B, Zhang X. The dual role of PGAM5 in inflammation. Exp Mol Med 2025; 57:298-311. [PMID: 39930129 PMCID: PMC11873181 DOI: 10.1038/s12276-025-01391-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/04/2024] [Accepted: 11/10/2024] [Indexed: 03/04/2025] Open
Abstract
In recent years, the focus on human inflammation in research has increased, with aging-related inflammation widely recognized as a defining characteristic of aging. Inflammation is strongly correlated with mitochondrial dysfunction. Phosphoglycerate mutase family member 5 (PGAM5) is a novel modulator of mitochondrial homeostasis in response to mechanical stimulation. Here we review the structure and sublocalization of PGAM5, introduce its importance in programmed cell death and summarize its crucial roles in the development and progression of inflammatory diseases such as pneumonia, hepatitis, neuroinflammation and aging. Notably, PGAM5 has dual effects on controlling inflammation: distinct PGAM5-mediated mitochondrial functions exhibit cellular heterogeneity, leading to its dual functions in inflammation control. We therefore highlight the double-edged sword nature of PGAM5 as a potential critical regulator and innovative therapeutic target in inflammation. Finally, the challenges and future directions of the use of PGAM5, which has dual properties, as a target molecule in the clinic are discussed. This review provides crucial insights to guide the development of intelligent therapeutic strategies targeting PGAM5-specific regulation to treat intractable inflammatory conditions, as well as the potential extension of its broader application to other diseases to achieve more precise and effective treatment outcomes.
Collapse
Affiliation(s)
- Yuxin Qi
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, China
- National Facility for Translational Medicine, Shanghai, China
| | - Bhavana Rajbanshi
- Department of Dermatology and Venereology, Tongji University School of Medicine, Shanghai, China
| | - Ruihan Hao
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Facility for Translational Medicine, Shanghai, China
| | - Yifan Dang
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, China
- National Facility for Translational Medicine, Shanghai, China
| | - Churong Xu
- National Facility for Translational Medicine, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Wei Lu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Liming Dai
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Facility for Translational Medicine, Shanghai, China
| | - Bingjun Zhang
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- National Facility for Translational Medicine, Shanghai, China.
| | - Xiaoling Zhang
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, China.
- National Facility for Translational Medicine, Shanghai, China.
| |
Collapse
|
5
|
Tork MAB, Fotouhi S, Roozi P, Negah SS. Targeting NLRP3 Inflammasomes: A Trojan Horse Strategy for Intervention in Neurological Disorders. Mol Neurobiol 2025; 62:1840-1881. [PMID: 39042218 DOI: 10.1007/s12035-024-04359-2] [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: 02/05/2024] [Accepted: 07/09/2024] [Indexed: 07/24/2024]
Abstract
Recently, a growing focus has been on identifying critical mechanisms in neurological diseases that trigger a cascade of events, making it easier to target them effectively. One such mechanism is the inflammasome, an essential component of the immune response system that plays a crucial role in disease progression. The NLRP3 (nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 3) inflammasome is a subcellular multiprotein complex that is widely expressed in the central nervous system (CNS) and can be activated by a variety of external and internal stimuli. When activated, the NLRP3 inflammasome triggers the production of proinflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18) and facilitates rapid cell death by assembling the inflammasome. These cytokines initiate inflammatory responses through various downstream signaling pathways, leading to damage to neurons. Therefore, the NLRP3 inflammasome is considered a significant contributor to the development of neuroinflammation. To counter the damage caused by NLRP3 inflammasome activation, researchers have investigated various interventions such as small molecules, antibodies, and cellular and gene therapy to regulate inflammasome activity. For instance, recent studies indicate that substances like micro-RNAs (e.g., miR-29c and mR-190) and drugs such as melatonin can reduce neuronal damage and suppress neuroinflammation through NLRP3. Furthermore, the transplantation of bone marrow mesenchymal stem cells resulted in a significant reduction in the levels of pyroptosis-related proteins NLRP3, caspase-1, IL-1β, and IL-18. However, it would benefit future research to have an in-depth review of the pharmacological and biological interventions targeting inflammasome activity. Therefore, our review of current evidence demonstrates that targeting NLRP3 inflammasomes could be a pivotal approach for intervention in neurological disorders.
Collapse
Affiliation(s)
- Mohammad Amin Bayat Tork
- Clinical Research Development Unit, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soroush Fotouhi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Roozi
- Department of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sajad Sahab Negah
- Clinical Research Development Unit, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran.
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Pardis Campus, Azadi Square, Kalantari Blvd., Mashhad, Iran.
| |
Collapse
|
6
|
Chen Y, Sun W, Mei H, Zhu S. Partially hydrolyzed guar gum alleviates neurological deficits and gastrointestinal dysfunction in mice with traumatic brain injury. Neurosurg Rev 2025; 48:103. [PMID: 39883194 DOI: 10.1007/s10143-024-03161-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/06/2024] [Accepted: 12/23/2024] [Indexed: 01/31/2025]
Abstract
Traumatic brain injury (TBI)-associated neuroinflammation and neurotoxicity can induce gastrointestinal dysfunction through the brain-gut axis. Partially hydrolyzed guar gum (PHGG) was demonstrated to exert beneficial health effects by altering gut microbiota and short-chain fatty acids (SCFAs) production. Our study aimed to explore the effects of PHGG on gastrointestinal dysfunction in TBI mouse models. Controlled cortical impact (CCI)-induced TBI mouse models were administrated with PHGG (600 mg/kg/d) for 21 consecutive days. Behavioral tests (modified neurological severity score and beam walk test) and Y‑maze assay were performed to evaluate neurological functions and cognitive impairment. Enzyme-linked immunosorbent assay, reverse transcription-quantitative polymerase chain reaction, and western blotting examined the levels of inflammatory cytokines, intestinal mucosal damage markers, intestinal tight junction proteins, and NLRP3 inflammasome-related molecules in the serum, cerebral cortex, and colon tissues. The histological changes in the cerebral cortex and colon tissues were observed through hematoxylin and eosin and Nissl staining. Liquid chromatography/mass spectrometry analyzed SCFA amounts in the cecum contents and bile acid levels in the serum. PHGG administration alleviated neurological deficits and cognitive perturbations, reduced neuroinflammation, and attenuated cortical tissue damage and neuron loss in TBI mice. PHGG ameliorated intestinal barrier impairment, upregulated intestinal production of SCFAs, and elevated serum bile acid levels in TBI mice. Besides, PHGG treatment repressed NLRP3 inflammasome activation in TBI mice. Overexpressing NLRP3 reversed the beneficial effects of PHGG against TBI in mice. PHGG ameliorates neuroinflammation and gastrointestinal dysfunction in TBI murine models by inhibiting NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Yao Chen
- Department of Infection Control, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu Province, 225300, China
| | - Wenbin Sun
- Department of Critical Care Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Zhou shan hui shui Community,199 Hailing South Road, Taizhou, Jiangsu Province, 225300, China
| | - Haifeng Mei
- Department of Critical Care Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Zhou shan hui shui Community,199 Hailing South Road, Taizhou, Jiangsu Province, 225300, China
| | - Shang Zhu
- Department of Critical Care Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Zhou shan hui shui Community,199 Hailing South Road, Taizhou, Jiangsu Province, 225300, China.
| |
Collapse
|
7
|
Chang Y, He Y, Wang D, Zhang K, Zhang Y, Li Z, Zeng S, Xiao S, Pan S, Huang K. ROS-regulated SUR1-TRPM4 drives persistent activation of NLRP3 inflammasome in microglia after whole-brain radiation. Acta Neuropathol Commun 2025; 13:16. [PMID: 39871308 PMCID: PMC11771008 DOI: 10.1186/s40478-025-01932-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 01/18/2025] [Indexed: 01/29/2025] Open
Abstract
Delayed radiation-induced brain injury (RIBI) characterized by progressive cognitive decline significantly impacts patient outcomes after radiotherapy. The activation of NLRP3 inflammasome within microglia after brain radiation is involved in the progression of RIBI by mediating inflammatory responses. We have previously shown that sulfonylurea receptor 1-transient receptor potential M4 (SUR1-TRPM4) mediates microglial NLRP3-related inflammation following global brain ischemia. However, the role of SUR1-TRPM4 in RIBI remains unclear. Here, we found that whole-brain radiation induced up-regulation and assembly of SUR1-TRPM4, which further activated the NLRP3 inflammasome in microglia and caused persistent neuroinflammation in mice. Blocking SUR1-TRPM4 by glibenclamide or gene deletion of Trpm4 effectively prevented NLRP3-mediated neuroinflammation and alleviated RIBI. Utilizing the mouse model of RIBI and irradiated BV2 cells, we further demonstrated that irradiation caused mitochondrial damage to microglia, leading to violent release of reactive oxygen species (ROS), which enhanced the transcription of SUR1, TRPM4, and NLRP3 inflammasome-related molecules. Moreover, ROS up-regulated ten-eleven translocation 2 (TET2) to enhance TRPM4 expression by mediating the demethylation of the gene promoter, thereby facilitating the assembly of SUR1-TRPM4 in microglia. In summary, this study deciphers that SUR1-TRPM4 crucially mediates the persistent activation of microglial NLRP3 inflammasome under the action of ROS after whole-brain radiation, offering novel therapeutic strategies for delayed RIBI as well as other NLRP3-related neurological disorders involving excessive ROS production.
Collapse
Affiliation(s)
- Yuan Chang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yihua He
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Di Wang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Kunxue Zhang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuzhen Zhang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhentong Li
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shuxin Zeng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Sheng Xiao
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Suyue Pan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Kaibin Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Department of Neurology, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, China.
| |
Collapse
|
8
|
Xing Y, Wang MM, Zhang F, Xin T, Wang X, Chen R, Sui Z, Dong Y, Xu D, Qian X, Lu Q, Li Q, Cai W, Hu M, Wang Y, Cao JL, Cui D, Qi J, Wang W. Lysosomes finely control macrophage inflammatory function via regulating the release of lysosomal Fe 2+ through TRPML1 channel. Nat Commun 2025; 16:985. [PMID: 39856099 PMCID: PMC11760952 DOI: 10.1038/s41467-025-56403-x] [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: 01/10/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Lysosomes are best known for their roles in inflammatory responses by engaging in autophagy to remove inflammasomes. Here, we describe an unrecognized role for the lysosome, showing that it finely controls macrophage inflammatory function by manipulating the lysosomal Fe2+-prolyl hydroxylase domain enzymes (PHDs)-NF-κB-interleukin 1 beta (IL1B) transcription pathway that directly links lysosomes with inflammatory responses. TRPML1, a lysosomal cationic channel, is activated secondarily to ROS elevation upon inflammatory stimuli, which in turn suppresses IL1B transcription, thus limiting the excessive production of IL-1β in macrophages. Mechanistically, the suppression of IL1B transcription caused by TRPML1 activation results from its modulation on the release of lysosomal Fe2+, which subsequently activates PHDs. The activated PHDs then represses transcriptional activity of NF-κB, ultimately resulting in suppressed IL1B transcription. More importantly, in vivo stimulation of TRPML1 ameliorates multiple clinical signs of Dextran sulfate sodium-induced colitis in mice, suggesting TRPML1 has potential in treating inflammatory bowel disease.
Collapse
Affiliation(s)
- Yanhong Xing
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Meng-Meng Wang
- Department of Otolaryngology and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Feifei Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Tianli Xin
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xinyan Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Rong Chen
- The First People's Hospital of Yancheng, Yancheng, China
| | - Zhongheng Sui
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, University of Hong Kong, Hong Kong, China
| | - Yawei Dong
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dongxue Xu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xingyu Qian
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qixia Lu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qingqing Li
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Weijie Cai
- New Cornerstone Science Laboratory, Liangzhu Laboratory & School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Meiqin Hu
- New Cornerstone Science Laboratory, Liangzhu Laboratory & School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yuqing Wang
- Department of Medicine and Biosystemic Science, Faculty of Medicine, Kyushu University, Fukuoka, Kyushu, Japan
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Derong Cui
- Department of Anesthesiology, The Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jiansong Qi
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Wuyang Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
9
|
Zhang Y, Bai Y, Hou X, Yang Y, Ma H, Wang G, Li Y. Neuroprotective effects of hypidone hydrochloride (YL-0919) after traumatic brain injury in mice. Chin Med J (Engl) 2025:00029330-990000000-01391. [PMID: 39809708 DOI: 10.1097/cm9.0000000000003400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Neurological dysfunction is a common complication of traumatic brain injury (TBI), and early treatments are critical for the long-term prognosis. This study aimed to investigate whether hypidone hydrochloride (YL-0919) improves neurological function impairment in mice with TBI. METHODS TBI was induced in adult male C57BL/6J mice using the controlled cortical impact (CCI) method. First, the modified neurological severity score (mNSS), rotarod test, and Morris water maze (MWM) test were conducted to assess the impact of YL-0919 on neurological function in mice with TBI. Next, immunofluorescence and laser speckle contrast imaging were utilized to measure the number and activation of microglia and cerebral blood flow (CBF) after TBI. Enzyme-linked immunosorbent assays (ELISAs) were employed to assess the inflammatory factors. Finally, Western blotting was performed to measure the expression of proteins. Golgi-Cox staining was utilized to investigate the structure of pyramidal neurons. RESULTS YL-0919 significantly alleviated neurological dysfunction in TBI+YL-0919 mice compared with TBI+Vehicle mice, increased the time spent on the rotarod (F = 1.297, P <0.05), and partially relieved cognitive dysfunction in TBI mice (for mNSS, F = 5.540, P <0.01; for MWM test, F = 30.78, P <0.05). Additionally, YL-0919 effectively inhibited the proliferation and activation of microglia (both P <0.01), promoted the recovery of CBF around the brain injury site and inhibited the expression of tumor necrosis factor-α (F = 9.142, P <0.05) and IL-1β (F = 4.662, P <0.05), and increased the concentration of IL-4 (F = 5.172, P <0.05). Furthermore, continuous gavage of YL-0919 (2.5 mg/kg) for seven days effectively increased the protein expression of brain-derived neurotrophic factor (BDNF), promoted the phosphorylation of mammalian target of rapamycin (mTOR), increased postsynaptic density protein 95 (PSD95) and synapsin1 levels, and increased the neuronal dendritic complexity and the dendritic spine density around the brain injury site (all P <0.05). CONCLUSIONS Our findings indicated that YL-0919 can ameliorate neurological dysfunction in mice after TBI through the suppression of inflammation and the stimulation of the BDNF-mTOR signaling pathway. These findings provide an insightful perspective on the potential pharmacological mechanism involved in the neuroprotective effect of YL-0919.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yafan Bai
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Xiaojuan Hou
- Hebei North University, Zhangjiakou, Hebei 075000, China
| | - Yixin Yang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Hui Ma
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Guyan Wang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Yunfeng Li
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing 100850, China
| |
Collapse
|
10
|
Sun M, Symons GF, Spitz G, O'Brien WT, Baker TL, Fan J, Martins BD, Allen J, Giesler LP, Mychasiuk R, van Donkelaar P, Brand J, Christie B, O'Brien TJ, O'Sullivan MJ, Mitra B, Wellington C, McDonald SJ, Shultz SR. Pathophysiology, blood biomarkers, and functional deficits after intimate partner violence-related brain injury: Insights from emergency department patients and a new rat model. Brain Behav Immun 2025; 123:383-396. [PMID: 39349286 DOI: 10.1016/j.bbi.2024.09.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 08/22/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024] Open
Abstract
Intimate partner violence is a serious, but underappreciated, issue that predominantly affects women and often results in concussion (i.e., mild traumatic brain injury). However, concussion in intimate partner violence is unique because it often involves a concomitant strangulation which may exacerbate or alter the physiology and clinical presentation of the brain injury. Therefore, here we conducted human and rodent studies to provide insight into knowledge gaps related to the detection, pathophysiology, and functional consequences of intimate partner violence-related brain injury. We conducted the first study to analyze blood biomarkers and symptoms of brain injury in intimate partner violence patients presenting to an emergency department within 72 h of concussion. Intimate partner violence concussion patients, some of whom had also experienced a concomitant strangulation, had elevated serum neurofilament light and worse brain injury symptoms compared to healthy control, orthopedic trauma, and non-intimate partner violence concussion groups. We also developed the first rat model of non-fatal strangulation and examined the consequences of strangulation and concussion in isolation and in combination on pathophysiology, blood biomarkers, and behavior at 2 h and 1wk post-injury. Rats exposed to combined strangulation and concussion had exacerbated motor and cognitive deficits, neuroinflammation, and serum glial fibrillary acidic protein levels compared with either injury in isolation. Taken together, these rodent findings demonstrate that a concomitant strangulation modifies and exacerbates concussion pathophysiology, biomarkers, and functional consequences. Overall, these findings provide novel insights into intimate partner violence-related brain injury and provides a foundation for future translational studies.
Collapse
Affiliation(s)
- Mujun Sun
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Georgia F Symons
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Centre for Trauma & Mental Health Research, Vancouver Island University, Nanaimo, BC, Canada
| | - Gershon Spitz
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Monash-Epworth Rehabilitation Research Centre, School of Psychological Sciences, Monash University, Melbourne, VIC, Australia
| | - William T O'Brien
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Tamara L Baker
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Jianjia Fan
- Department of Pathology & Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Beatriz D Martins
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Josh Allen
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Centre for Trauma & Mental Health Research, Vancouver Island University, Nanaimo, BC, Canada; Neuroscience Program, Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Lauren P Giesler
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Paul van Donkelaar
- Health & Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Justin Brand
- Neuroscience Program, Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Brian Christie
- Neuroscience Program, Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Terence J O'Brien
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Michael J O'Sullivan
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Biswadev Mitra
- Emergency Services, Alfred Health, Melbourne, VIC, Australia; School of Public Health & Preventative Medicine, Monash University, Melbourne, VIC, Australia
| | - Cheryl Wellington
- Department of Pathology & Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Stuart J McDonald
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Centre for Trauma & Mental Health Research, Vancouver Island University, Nanaimo, BC, Canada; Neuroscience Program, Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
11
|
Henry RJ, Loane DJ. Unraveling the complexity of microglial responses in traumatic brain and spinal cord injury. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:113-132. [PMID: 40148040 DOI: 10.1016/b978-0-443-19102-2.00015-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Microglia, the resident innate immune cells of the central nervous system (CNS), play an important role in neuroimmune signaling, neuroprotection, and neuroinflammation. In the healthy CNS, microglia adopt a surveillant and antiinflammatory phenotype characterized by a ramified scanning morphology that maintains CNS homeostasis. In response to acquired insults, such as traumatic brain injury (TBI) or spinal cord injury (SCI), microglia undergo a dramatic morphologic and functional switch to that of a reactive state. This microglial switch is initially protective and supports the return of the injured tissue to a physiologic homeostatic state. However, there is now a significant body of evidence that both TBI and SCI can result in a chronic state of microglial activation, which contributes to neurodegeneration and impairments in long-term neurologic outcomes in humans and animal models. In this review, we discuss the complex role of microglia in the pathophysiology of TBI and SCI, and recent advancements in knowledge of microglial phenotypic states in the injured CNS. Furthermore, we highlight novel therapeutic strategies targeting chronic microglial responses in experimental models and discuss how they may ultimately be translated to the clinic for human brain and SCI.
Collapse
Affiliation(s)
- Rebecca J Henry
- Department of Pharmacology, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.
| | - David J Loane
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
12
|
Valero-Hernandez E, Tremoleda JL, Michael-Titus AT. Omega-3 Fatty Acids and Traumatic Injury in the Adult and Immature Brain. Nutrients 2024; 16:4175. [PMID: 39683568 DOI: 10.3390/nu16234175] [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: 10/31/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives: Traumatic brain injury (TBI) can lead to substantial disability and health loss. Despite its importance and impact worldwide, no treatment options are currently available to help protect or preserve brain structure and function following injury. In this review, we discuss the potential benefits of using omega-3 polyunsaturated fatty acids (O3 PUFAs) as therapeutic agents in the context of TBI in the paediatric and adult populations. Methods: Preclinical and clinical research reports investigating the effects of O3 PUFA-based interventions on the consequences of TBI were retrieved and reviewed, and the evidence presented and discussed. Results: A range of animal models of TBI, types of injury, and O3 PUFA dosing regimens and administration protocols have been used in different strategies to investigate the effects of O3 PUFAs in TBI. Most evidence comes from preclinical studies, with limited clinical data available thus far. Overall, research indicates that high O3 PUFA levels help lessen the harmful effects of TBI by reducing tissue damage and cell loss, decreasing associated neuroinflammation and the immune response, which in turn moderates the severity of the associated neurological dysfunction. Conclusions: Data from the studies reviewed here indicate that O3 PUFAs could substantially alleviate the impact of traumatic injuries in the central nervous system, protect structure and help restore function in both the immature and adult brains.
Collapse
Affiliation(s)
- Ester Valero-Hernandez
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Jordi L Tremoleda
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Adina T Michael-Titus
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| |
Collapse
|
13
|
Yuan Z, Lu B, Zhang M, Lu Y, Wang Z, Zhang W, Cheng H, Wu Z, Ji Q. Effect of NLRP3 inflammasome induced astrocyte phenotype alteration in morphine tolerance. Front Pharmacol 2024; 15:1434295. [PMID: 39600361 PMCID: PMC11588488 DOI: 10.3389/fphar.2024.1434295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
Introduction Morphine is a widely used analgesic, but its prolonged use often leads to tolerance, limiting its therapeutic efficacy. Research implicates the NLRP3 inflammasome and reactive astrocytes in the development of morphine tolerance, with reactive astrocytes classified into A1 neurotoxic and A2 neuroprotective phenotypes. This study explores the role of the NLRP3 inflammasome and the transformation of astrocyte phenotypes in the progression of morphine tolerance. Methods A model of morphine tolerance was established by administering morphine intrathecally for seven consecutive days. To inhibit NLRP3 inflammasome activation, we coadministered MCC950, a selective NLRP3 inhibitor. Thermal withdrawal latency was used to assess tolerance development. Protein and mRNA levels of GFAP, IL-18, NLRP3, C3 (A1 marker), and S100A10 (A2 marker) in the spinal cord were measured using Western blotting (WB) and real-time quantitative polymerase chain reaction (RT-qPCR). Immunofluorescence was employed to assess the colocalization of C3 and GFAP. Results Seven days of morphine administration induced tolerance, which was associated with increased levels of GFAP, IL-18, NLRP3, and C3, and a decreased level of S100A10. Coadministration of morphine and MCC950 significantly slowed the development of morphine tolerance and reversed changes in NLRP3, IL-18, GFAP, C3, and S100A10 protein levels. Discussion Our findings indicate a significant link between NLRP3 inflammasome activation and morphine tolerance, suggesting that NLRP3 contributes to the transformation of astrocytes to the A1 phenotype. Inhibiting NLRP3 inflammasome activation holds promise in reversing astrocyte phenotype changes, potentially mitigating morphine tolerance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhifang Wu
- Department of Anesthesiology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qing Ji
- Department of Anesthesiology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
14
|
de Almeida Chuffa LG, Seiva FRF, Silveira HS, Cesário RC, da Silva Tonon K, Simão VA, Zuccari DAPC, Reiter RJ. Melatonin regulates endoplasmic reticulum stress in diverse pathophysiological contexts: A comprehensive mechanistic review. J Cell Physiol 2024; 239:e31383. [PMID: 39039752 DOI: 10.1002/jcp.31383] [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: 04/10/2024] [Revised: 06/13/2024] [Accepted: 07/05/2024] [Indexed: 07/24/2024]
Abstract
The endoplasmic reticulum (ER) is crucial for protein quality control, and disruptions in its function can lead to various diseases. ER stress triggers an adaptive response called the unfolded protein response (UPR), which can either restore cellular homeostasis or induce cell death. Melatonin, a safe and multifunctional compound, shows promise in controlling ER stress and could be a valuable therapeutic agent for managing the UPR. By regulating ER and mitochondrial functions, melatonin helps maintain cellular homeostasis via reduction of oxidative stress, inflammation, and apoptosis. Melatonin can directly or indirectly interfere with ER-associated sensors and downstream targets of the UPR, impacting cell death, autophagy, inflammation, molecular repair, among others. Crucially, this review explores the mechanistic role of melatonin on ER stress in various diseases including liver damage, neurodegeneration, reproductive disorders, pulmonary disease, cardiomyopathy, insulin resistance, renal dysfunction, and cancer. Interestingly, while it alleviates the burden of ER stress in most pathological contexts, it can paradoxically stimulate ER stress in cancer cells, highlighting its intricate involvement in cellular homeostasis. With numerous successful studies using in vivo and in vitro models, the continuation of clinical trials is imperative to fully explore melatonin's therapeutic potential in these conditions.
Collapse
Affiliation(s)
- Luiz Gustavo de Almeida Chuffa
- Department of Structural and Functional Biology, Institute of Bioscences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Fábio Rodrigues Ferreira Seiva
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Henrique S Silveira
- Department of Structural and Functional Biology, Institute of Bioscences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Roberta Carvalho Cesário
- Department of Structural and Functional Biology, Institute of Bioscences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Karolina da Silva Tonon
- Department of Structural and Functional Biology, Institute of Bioscences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Vinicius Augusto Simão
- Department of Structural and Functional Biology, Institute of Bioscences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Debora Aparecida P C Zuccari
- Department of Molecular Biology, Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto, São Paulo, Brazil
| | - Russel J Reiter
- Department of Cellular and Structural Biology, UTHealth, San Antonio, Texas, USA
| |
Collapse
|
15
|
Wu J, Ren R, Chen T, Su LD, Tang T. Neuroimmune and neuroinflammation response for traumatic brain injury. Brain Res Bull 2024; 217:111066. [PMID: 39241894 DOI: 10.1016/j.brainresbull.2024.111066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/18/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Traumatic brain injury (TBI) is one of the major diseases leading to mortality and disability, causing a serious disease burden on individuals' ordinary lives as well as socioeconomics. In primary injury, neuroimmune and neuroinflammation are both responsible for the TBI. Besides, extensive and sustained injury induced by neuroimmune and neuroinflammation also prolongs the course and worsens prognosis of TBI. Therefore, this review aims to explore the role of neuroimmune, neuroinflammation and factors associated them in TBI as well as the therapies for TBI. Thus, we conducted by searching PubMed, Scopus, and Web of Science databases for articles published between 2010 and 2023. Keywords included "traumatic brain injury," "neuroimmune response," "neuroinflammation," "astrocytes," "microglia," and "NLRP3." Articles were selected based on relevance and quality of evidence. On this basis, we provide the cellular and molecular mechanisms of TBI-induced both neuroimmune and neuroinflammation response, as well as the different factors affecting them, are introduced based on physiology of TBI, which supply a clear overview in TBI-induced chain-reacting, for a better understanding of TBI and to offer more thoughts on the future therapies for TBI.
Collapse
Affiliation(s)
- Junyun Wu
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Reng Ren
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Tao Chen
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Li-Da Su
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China.
| | - Tianchi Tang
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China.
| |
Collapse
|
16
|
Chen W, Wang Z, Ye G, Zhu G, Li S, Chen P, Wang H, Zou S, Chen M. Changes of NLRP3 in serum and cerebrospinal fluid of patients after moderate to severe traumatic brain injury and their predictive values for prognosis. CNS Neurosci Ther 2024; 30:e70009. [PMID: 39302033 PMCID: PMC11413909 DOI: 10.1111/cns.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 07/25/2024] [Accepted: 08/10/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) remains a major concern for global health. Recent studies have suggested the role of NOD-like receptor pyrin domain-containing protein 3 (NLRP3), an inflammatory marker, in the cerebrospinal fluid (CSF) and serum as potential indicators of TBI prognosis. The objective of the study was to characterize NLRP3 as a clinically applicable tool for predicting the outcomes of TBI patients. METHODS A total of 270 patients with moderate to severe TBI were included in this retrospective analysis. Serum and CSF samples were collected at 1-, 3-, 7-, and 21-day post-injury to measure NLRP3 levels. The prognosis of patients was evaluated after 3 months using the Glasgow Outcome Scale (GOS). Patients were categorized into good prognosis (GOS score >3) and poor prognosis (GOS score ≤3) groups. The relationship between NLRP3 levels and prognosis was analyzed. RESULTS Patients with poor prognosis had significantly elevated NLRP3 levels in their serum on days 1 and 3 post-injury compared with those with a good prognosis. The difference was more pronounced during these early days compared with days 7 and 21. However, NLRP3 levels in CSF consistently showed a large difference between the two groups throughout the observation period. Receiver operating characteristic analysis revealed that the level of NLRP3 in the CSF on day 3 post-injury had the highest predictive value for prognosis, with an area under the curve of 0.83, followed by the level of NLRP3 in the serum on day 3 post-injury. CONCLUSIONS The levels of NLRP3, especially in the CSF on day 3 post-injury, can serve as a potential biomarker for predicting prognosis in moderate to severe TBI patients. Early measurement of NLRP3 levels can provide valuable insights into patient outcomes and guide therapeutic strategies.
Collapse
Affiliation(s)
- Wei Chen
- Department of NeurosurgeryThe Affiliated Lihuili Hospital of Ningbo UniversityNingboZhejiangChina
- Department of Neurosurgery, Shanghai East Hospital, School of MedicineTongji UniversityShanghaiChina
- Department of NeurosurgeryThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxiChina
| | - Zhigang Wang
- Department of Neurosurgery, Shanghai East Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Gengfan Ye
- Department of NeurosurgeryThe Affiliated Lihuili Hospital of Ningbo UniversityNingboZhejiangChina
| | - Guangyao Zhu
- Department of NeurosurgeryThe Affiliated Lihuili Hospital of Ningbo UniversityNingboZhejiangChina
| | - Shiwei Li
- Department of NeurosurgeryThe Affiliated Lihuili Hospital of Ningbo UniversityNingboZhejiangChina
| | - Pandi Chen
- Department of NeurosurgeryThe Affiliated Lihuili Hospital of Ningbo UniversityNingboZhejiangChina
| | - Hongcai Wang
- Department of NeurosurgeryThe Affiliated Lihuili Hospital of Ningbo UniversityNingboZhejiangChina
| | - Shufeng Zou
- Department of NeurosurgeryThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxiChina
| | - Maosong Chen
- Department of NeurosurgeryThe Affiliated Lihuili Hospital of Ningbo UniversityNingboZhejiangChina
| |
Collapse
|
17
|
Navabi SP, Badreh F, Khombi Shooshtari M, Hajipour S, Moradi Vastegani S, Khoshnam SE. Microglia-induced neuroinflammation in hippocampal neurogenesis following traumatic brain injury. Heliyon 2024; 10:e35869. [PMID: 39220913 PMCID: PMC11365414 DOI: 10.1016/j.heliyon.2024.e35869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Traumatic brain injury (TBI) is one of the most causes of death and disability among people, leading to a wide range of neurological deficits. The important process of neurogenesis in the hippocampus, which includes the production, maturation and integration of new neurons, is affected by TBI due to microglia activation and the inflammatory response. During brain development, microglia are involved in forming or removing synapses, regulating the number of neurons, and repairing damage. However, in response to injury, activated microglia release a variety of pro-inflammatory cytokines, chemokines and other neurotoxic mediators that exacerbate post-TBI injury. These microglia-related changes can negatively affect hippocampal neurogenesis and disrupt learning and memory processes. To date, the intracellular signaling pathways that trigger microglia activation following TBI, as well as the effects of microglia on hippocampal neurogenesis, are poorly understood. In this review article, we discuss the effects of microglia-induced neuroinflammation on hippocampal neurogenesis following TBI, as well as the intracellular signaling pathways of microglia activation.
Collapse
Affiliation(s)
- Seyedeh Parisa Navabi
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Maryam Khombi Shooshtari
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Somayeh Hajipour
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sadegh Moradi Vastegani
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
18
|
Yang X, Sun A, Kong L, Yang X, Zhao X, Wang S. Inhibition of NLRP3 inflammasome alleviates cognitive deficits in a mouse model of anti-NMDAR encephalitis induced by active immunization. Int Immunopharmacol 2024; 137:112374. [PMID: 38851162 DOI: 10.1016/j.intimp.2024.112374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
Anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis is a neurological disorder, characterized by cognitive deficits as one of its vital features. The nucleotide-binding oligomerization domain-like receptor (NLRP3) inflammasome is a key contributor to neuroinflammation and cognitive deficits in neurological diseases. However, the underlying mechanism of anti-NMDAR encephalitis remains unclear, and the biological function of the NLRP3 inflammasome in this condition has not been elucidated. In this study, a mouse model of anti-NMDAR encephalitis was induced by active immunization with the GluN1356-385 peptide (NEA model). The NLRP3 inflammasome in the hippocampus and temporal cortex was investigated using real-time quantitative PCR (RT-qPCR), western blotting, and immunofluorescence staining. The impact of MCC950 on cognitive function and NLRP3 inflammation was assessed. Confocal immunofluorescence staining and Sholl analysis were employed to examine the function and morphology of microglia. In the current study, we discovered overactivation of the NLRP3 inflammasome and an enhanced inflammatory response in the NEA model, particularly in the hippocampus and temporal cortex. Furthermore, significant cognitive dysfunction was observed in the NEA model. While, MCC950, a selective inhibitor of the NLRP3 inflammasome, sharply attenuated the inflammatory response in mice, leading to mitigated cognitive deficits of mice and more regular arrangements of neurons and reduced number of hyperchromatic cells were also observed in the hippocampus area. In addition, we found that the excess elevation of NLRP3 inflammasome was mainly expressed in microglia accompanied with the overactivation of microglia, while MCC950 treatment significantly inhibited the increased number and activated morphological changes of microglia in the NEA model. Altogether, our study reveals the vital role of overactivated NLRP3 signaling pathway in aggravating the inflammatory response and cognitive deficits and the potential protective effect of MCC950 in anti-NMDAR encephalitis. Thus, MCC950 represents a promising strategy for anti-inflammation in anti-NMDAR encephalitis and our study lays a theoretical foundation for it to become a clinically targeted drug.
Collapse
Affiliation(s)
- Xiaxin Yang
- Department of Neurology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, China
| | - Anqi Sun
- Department of Neurology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, China
| | - Liangbo Kong
- Department of Neurology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, China
| | - Xue Yang
- Department of Neurology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, China
| | - Xiuhe Zhao
- Department of Neurology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, China.
| | - Shengjun Wang
- Department of Neurology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, China.
| |
Collapse
|
19
|
Jiang Y, Xu Y, Wang Q, Chen Z, Liu C. Significance of serum NLRP3 as a potential predictor of 5-year death in hemodialysis patients: A prospective observational cohort study. Medicine (Baltimore) 2024; 103:e39185. [PMID: 39093762 PMCID: PMC11296485 DOI: 10.1097/md.0000000000039185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024] Open
Abstract
Nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) is involved in inflammatory response. This study was done to explore the role of serum NLRP3 as a predictive biomarker of death after hemodialysis. In this prospective observational study of 331 patients undergoing maintenance hemodialysis, serum NLRP3 levels were measured. Univariate analysis and multivariate analysis were sequentially performed to determine predictors of 5-year death after hemodialysis. Age, major adverse cardiac and cerebral events (MACCE), and serum NLRP3 levels independently predicted 5-year mortality and overall survival (all P < .05). No interactions were found between serum NLRP3 levels and other variables, such as age, gender, hypertension, diabetes mellitus, primary renal diseases, and MACCE (all P interaction > .05). Serum NLRP3 levels were linearly correlated with risk of death and overall survival under restricted cubic spline (both P > .05) and substantially discriminated patients at risk of death under receiver operating characteristic curve (P < .001). Two models, in which age, MACCE, and serum NLRP3 were combined, were built to predict 5-year mortality and overall survival. The mortality prediction model had significantly higher predictive ability than age, AMCCE, and serum NLRP3 alone under receiver operating characteristic curve (all P < .05). The models, which were graphically represented by nomograms, performed well under calibration curve and decision curve. Serum NLRP3 levels are independently related to 5-year mortality and overall survival of patients after hemodialysis, suggesting that serum NLRP3 may be a potential prognostic biomarker of hemodialysis patients.
Collapse
Affiliation(s)
- Yi Jiang
- Department of Nephrology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Yandan Xu
- Department of Nephrology, Quzhou KeCheng People’s Hospital, Quzhou, China
| | - Qiuli Wang
- Traditional Chinese Medicine Department, Quzhou Hospital of Zhejiang Medical and Health Group, Quzhou, China
| | - Zhiwei Chen
- Department of Nephrology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Chunya Liu
- Department of Nephrology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| |
Collapse
|
20
|
Xu A, Huang F, Chen E, Zhang Z, He Y, Yu X, He G. Hyperbaric oxygen therapy attenuates heatstroke-induced hippocampal injury by inhibiting microglial pyroptosis. Int J Hyperthermia 2024; 41:2382162. [PMID: 39043380 DOI: 10.1080/02656736.2024.2382162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024] Open
Abstract
Background: Central nervous system (CNS) injury is the most prominent feature of heatstroke and the hippocampus is prone to damage. However, the mechanisms underlying the heatstroke-induced hippocampal injury remain unclear. Hyperbaric oxygen (HBO) therapy prevents CNS injury in heatstroke mice. However, the underlying mechanisms of HBO in heatstroke-induced hippocampal injury remain unclear. This study aimed to elucidate the protective effects of HBO against hippocampal injury and its potential role in microglial pyroptosis in heatstroke rats.Methods: A rat heatstroke model and a heat stress model with a mouse microglial cell line (BV2) were, respectively, used to illustrate the effect of HBO on heat-induced microglial pyroptosis in vivo and in vitro. We used a combination of molecular and histological methods to assess microglial pyroptosis and neuroinflammation both in vivo and in vitro.Results: The results revealed that HBO improved heatstroke-induced survival outcomes, hippocampal injury, and neurological dysfunction in rats. In addition, HBO mitigates microglial pyroptosis and reduces the expression of pro-inflammatory cytokines in the hippocampus of heatstroke rats. In vitro experiments showed that HBO attenuated BV2 cell injury under heat stress. Furthermore, HBO prevented heat-induced pyroptosis of BV2 cells, and the expression of pro-inflammatory cytokines IL-18 and IL-1β was reduced. Mechanistically, HBO alleviates heatstroke-induced neuroinflammation and hippocampal injury by preventing microglial pyroptosis. Conclusions: In conclusion, HBO attenuates heatstroke-induced neuroinflammation and hippocampal injury by inhibiting microglial pyroptosis.
Collapse
Affiliation(s)
- Ancong Xu
- Department of Intensive Care Unit, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fan Huang
- Wenzhou Medical University, Wenzhou, China
| | - Er Chen
- Department of Intensive Care Unit, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | | | - Yanxuan He
- Wenzhou Medical University, Wenzhou, China
| | - Xichong Yu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Guoxin He
- Department of Intensive Care Unit, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
21
|
Chen S, Yu Z, Wen W, Chen J, Lu K. NLRP3 Expression and Its Predictive Role in Heart Failure with Preserved Ejection Fraction among Non-Valvular Atrial Fibrillation Patients. Cardiology 2024; 150:72-78. [PMID: 38964309 DOI: 10.1159/000540204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/29/2024] [Indexed: 07/06/2024]
Abstract
INTRODUCTION The aim of this study was to investigate the expression and predictive value of NOD-like receptor thermal protein domain-related protein 3 (NLRP3) in patients with non-valvular atrial fibrillation (NVAF) with heart failure with preserved ejection fraction (HFpEF). METHODS This was a retrospective analysis of 121 patients diagnosed with NVAF. According to the occurrence of HFpEF, 81 patients were assigned to the NVAF group and 40 patients to the NVAF/HFpEF group. The levels of NLRP3, B natriuretic peptide (BNP), and interleukin-1β (IL-1β) were determined using ELISA. Independent predictors for HFpEF in NVAF were determined using logistic regression. The receiver operating characteristic (ROC) curve was used to evaluate the predictive value of each factor. RESULTS Expression levels of NLRP3, BNP, and IL-1β in the NVAF/HFpEF group, as well as the H2FPEF score were significantly higher than those in the NVAF group. Pearson analysis showed that NLRP3, BNP, and IL-1β expression levels in NVAF patients and the H2FPEF score was positively correlated (r = 0.409, r = 0.244, r = 0.299, p < 0.001). Multivariate logistic regression analysis showed that the NLRP3, BNP, or H2FPEF score can be used as independent factor for predicting the occurrence of HFpEF in NVAF. ROC curves showed that the areas under the curve of NLRP3, BNP, and H2FPEF scores for predicting the occurrence of HFpEF in NVAF patients were 0.856, 0.831, and 0.811, respectively. CONCLUSION The NLRP3 level is elevated in the peripheral blood of NVAF patients with HFpEF and is positively correlated with the H2FPEF score. NLRP3 may serve as a potential predictor of HFpEF in patients with NVAF.
Collapse
Affiliation(s)
- Shijian Chen
- Department of Cardiovascular Medicine, Huzhou Central Hospital, Huzhou, China
| | - Ziheng Yu
- Department of Cardiovascular Medicine, Huzhou Central Hospital, Huzhou, China
| | - Wen Wen
- Department of Cardiovascular Medicine, Huzhou Central Hospital, Huzhou, China
| | - Jiming Chen
- Department of Cardiovascular Medicine, Huzhou Central Hospital, Huzhou, China
| | - Kongjie Lu
- Department of Cardiovascular Medicine, Huzhou Central Hospital, Huzhou, China
| |
Collapse
|
22
|
Banderwal R, Kadian M, Garg S, Kumar A. 'Comprehensive review of emerging drug targets in traumatic brain injury (TBI): challenges and future scope. Inflammopharmacology 2024:10.1007/s10787-024-01524-w. [PMID: 39023681 DOI: 10.1007/s10787-024-01524-w] [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: 08/03/2023] [Accepted: 02/12/2024] [Indexed: 07/20/2024]
Abstract
Traumatic brain injury (TBI) is a complex brain problem that causes significant morbidity and mortality among people of all age groups. The complex pathophysiology, varied symptoms, and inadequate treatment further precipitate the problem. Further, TBI produces several psychiatric problems and other related complications in post-TBI survival patients, which are often treated symptomatically or inadequately. Several approaches, including neuroprotective agents targeting several pathways of oxidative stress, neuroinflammation, cytokines, immune system GABA, glutamatergic, microglia, and astrocytes, are being tried by researchers to develop effective treatments or magic bullets to manage the condition effectively. The problem of TBI is therefore treated as a challenge among pharmaceutical scientists or researchers to develop drugs for the effective management of this problem. The goal of the present comprehensive review is to provide an overview of the several pharmacological targets, processes, and cellular pathways that researchers are focusing on, along with an update on their current state.
Collapse
Affiliation(s)
- Rittu Banderwal
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC- Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Monika Kadian
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC- Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Sukant Garg
- Department of General Pathology, Dr HS Judge Institute of Dental Sciences and Hospital, Panjab University, Chandigarh, 160014, India
| | - Anil Kumar
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC- Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
23
|
Javalgekar M, Jupp B, Vivash L, O'Brien TJ, Wright DK, Jones NC, Ali I. Inflammasomes at the crossroads of traumatic brain injury and post-traumatic epilepsy. J Neuroinflammation 2024; 21:172. [PMID: 39014496 PMCID: PMC11250980 DOI: 10.1186/s12974-024-03167-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 07/05/2024] [Indexed: 07/18/2024] Open
Abstract
Post-traumatic epilepsy (PTE) is one of the most debilitating consequences of traumatic brain injury (TBI) and is one of the most drug-resistant forms of epilepsy. Novel therapeutic treatment options are an urgent unmet clinical need. The current focus in healthcare has been shifting to disease prevention, rather than treatment, though, not much progress has been made due to a limited understanding of the disease pathogenesis. Neuroinflammation has been implicated in the pathophysiology of traumatic brain injury and may impact neurological sequelae following TBI including functional behavior and post-traumatic epilepsy development. Inflammasome signaling is one of the major components of the neuroinflammatory response, which is increasingly being explored for its contribution to the epileptogenic mechanisms and a novel therapeutic target against epilepsy. This review discusses the role of inflammasomes as a possible connecting link between TBI and PTE with a particular focus on clinical and preclinical evidence of therapeutic inflammasome targeting and its downstream effector molecules for their contribution to epileptogenesis. Finally, we also discuss emerging evidence indicating the potential of evaluating inflammasome proteins in biofluids and the brain by non-invasive neuroimaging, as potential biomarkers for predicting PTE development.
Collapse
Affiliation(s)
- Mohit Javalgekar
- The Department of Neuroscience, School of Translational Medicine, Monash University, 99, Commercial Road, Melbourne, Australia
- Department of Neurology, The Alfred Hospital, 99 commercial road, Melbourne, Australia
| | - Bianca Jupp
- The Department of Neuroscience, School of Translational Medicine, Monash University, 99, Commercial Road, Melbourne, Australia
- Department of Neurology, The Alfred Hospital, 99 commercial road, Melbourne, Australia
| | - Lucy Vivash
- The Department of Neuroscience, School of Translational Medicine, Monash University, 99, Commercial Road, Melbourne, Australia
- Department of Neurology, The Alfred Hospital, 99 commercial road, Melbourne, Australia
- The University of Melbourne, Parkville, Australia
| | - Terence J O'Brien
- The Department of Neuroscience, School of Translational Medicine, Monash University, 99, Commercial Road, Melbourne, Australia
- Department of Neurology, The Alfred Hospital, 99 commercial road, Melbourne, Australia
- The University of Melbourne, Parkville, Australia
| | - David K Wright
- The Department of Neuroscience, School of Translational Medicine, Monash University, 99, Commercial Road, Melbourne, Australia
- Department of Neurology, The Alfred Hospital, 99 commercial road, Melbourne, Australia
| | - Nigel C Jones
- The Department of Neuroscience, School of Translational Medicine, Monash University, 99, Commercial Road, Melbourne, Australia.
- Department of Neurology, The Alfred Hospital, 99 commercial road, Melbourne, Australia.
- The University of Melbourne, Parkville, Australia.
| | - Idrish Ali
- The Department of Neuroscience, School of Translational Medicine, Monash University, 99, Commercial Road, Melbourne, Australia.
- Department of Neurology, The Alfred Hospital, 99 commercial road, Melbourne, Australia.
- The University of Melbourne, Parkville, Australia.
| |
Collapse
|
24
|
Amelimojarad M, Amelimojarad M, Cui X. The emerging role of brain neuroinflammatory responses in Alzheimer's disease. Front Aging Neurosci 2024; 16:1391517. [PMID: 39021707 PMCID: PMC11253199 DOI: 10.3389/fnagi.2024.1391517] [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: 02/26/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
As the most common cause of dementia, Alzheimer's disease (AD) is characterized by neurodegeneration and synaptic loss with an increasing prevalence in the elderly. Increased inflammatory responses triggers brain cells to produce pro-inflammatory cytokines and accelerates the Aβ accumulation, tau protein hyper-phosphorylation leading to neurodegeneration. Therefore, in this paper, we discuss the current understanding of how inflammation affects brain activity to induce AD pathology, the inflammatory biomarkers and possible therapies that combat inflammation for AD.
Collapse
Affiliation(s)
| | | | - Xiaonan Cui
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
25
|
Singh AA, Yadav D, Khan F, Song M. Indole-3-Carbinol and Its Derivatives as Neuroprotective Modulators. Brain Sci 2024; 14:674. [PMID: 39061415 PMCID: PMC11274471 DOI: 10.3390/brainsci14070674] [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: 06/06/2024] [Revised: 06/26/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and its downstream tropomyosin receptor kinase B (TrkB) signaling pathway play pivotal roles in the resilience and action of antidepressant drugs, making them prominent targets in psychiatric research. Oxidative stress (OS) contributes to various neurological disorders, including neurodegenerative diseases, stroke, and mental illnesses, and exacerbates the aging process. The nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant responsive element (ARE) serves as the primary cellular defense mechanism against OS-induced brain damage. Thus, Nrf2 activation may confer endogenous neuroprotection against OS-related cellular damage; notably, the TrkB/phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway, stimulated by BDNF-dependent TrkB signaling, activates Nrf2 and promotes its nuclear translocation. However, insufficient neurotrophin support often leads to the downregulation of the TrkB signaling pathway in brain diseases. Thus, targeting TrkB activation and the Nrf2-ARE system is a promising therapeutic strategy for treating neurodegenerative diseases. Phytochemicals, including indole-3-carbinol (I3C) and its metabolite, diindolylmethane (DIM), exhibit neuroprotective effects through BDNF's mimetic activity; Akt phosphorylation is induced, and the antioxidant defense mechanism is activated by blocking the Nrf2-kelch-like ECH-associated protein 1 (Keap1) complex. This review emphasizes the therapeutic potential of I3C and its derivatives for concurrently activating neuronal defense mechanisms in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Alka Ashok Singh
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| | - Dhananjay Yadav
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| | - Fazlurrahman Khan
- Institute of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea;
- International Graduate Program of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| |
Collapse
|
26
|
Kang J, Zhou Y, Xiong Q, Dong X. Trigeminal nerve electrical stimulation attenuates early traumatic brain injury through the TLR4/NF-κB/NLRP3 signaling pathway mediated by orexin-A/OX1R system. Aging (Albany NY) 2024; 16:7946-7960. [PMID: 38713160 PMCID: PMC11131994 DOI: 10.18632/aging.205795] [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: 12/20/2023] [Accepted: 04/09/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND Traumatic brain injury (TBI) is a significant contributor to global mortality and disability, and emerging evidence indicates that trigeminal nerve electrical stimulation (TNS) is a promising therapeutic intervention for neurological impairment following TBI. However, the precise mechanisms underlying the neuroprotective effects of TNS in TBI are poorly understood. Thus, the objective of this study was to investigate the potential involvement of the orexin-A (OX-A)/orexin receptor 1 (OX1R) mediated TLR4/NF-κB/NLRP3 signaling pathway in the neuroprotective effects of TNS in rats with TBI. METHODS Sprague-Dawley rats were randomly assigned to four groups: sham, TBI, TBI+TNS+SB334867, and TBI+TNS. TBI was induced using a modified Feeney's method, and subsequent behavioral assessments were conducted to evaluate neurological function. The trigeminal nerve trunk was isolated, and TNS was administered following the establishment of the TBI model. The levels of neuroinflammation, brain tissue damage, and proteins associated with the OX1R/TLR4/NF-κB/NLRP3 signaling pathway were assessed using hematoxylin-eosin staining, Nissl staining, western blot analysis, quantitative real-time polymerase chain reaction, and immunofluorescence techniques. RESULTS The findings of our study indicate that TNS effectively mitigated tissue damage, reduced brain edema, and alleviated neurological deficits in rats with TBI. Furthermore, TNS demonstrated the ability to attenuate neuroinflammation levels and inhibit the expression of proteins associated with the TLR4/NF-κB/NLRP3 signaling pathway. However, it is important to note that the aforementioned effects of TNS were reversible upon intracerebroventricular injection of an OX1R antagonist. CONCLUSION TNS may prevent brain damage and relieve neurological deficits after a TBI by inhibiting inflammation, possibly via the TLR4/NF-κB/NLRP3 signaling pathway mediated by OX-A/OX1R.
Collapse
Affiliation(s)
- Junwei Kang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Yifan Zhou
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Qi Xiong
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Xiaoyang Dong
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| |
Collapse
|
27
|
Xu L, An T, Jia B, Wu Q, Shen J, Jin J, Liu J, Li C. Histone deacetylase 3-specific inhibitor RGFP966 attenuates oxidative stress and inflammation after traumatic brain injury by activating the Nrf2 pathway. BURNS & TRAUMA 2024; 12:tkad062. [PMID: 38708192 PMCID: PMC11069425 DOI: 10.1093/burnst/tkad062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 05/07/2024]
Abstract
Background Oxidative stress (OS) and inflammatory reactions play pivotal roles in secondary brain injury after traumatic brain injury (TBI). Histone deacetylase 3 (HDAC3) controls the acetylation of histones and non-histones, which has a significant impact on the central nervous system's reaction to damage. This research determined the implications of RGFP966, a new and specific inhibitor of HDAC3, for the antioxidant (AO) systems mediated by nuclear factor erythroid2-related factor 2 (Nrf2) and the Nod-like receptor protein 3 (NLRP3) inflammasome in TBI. The study also studied the underlying mechanisms of RGFP966's actions. Our objective was to examine the impacts and underlying RGFP966 mechanisms in TBI. Methods In vitro, a rat cortical neuron OS model was induced by H2O2, followed by the addition of RGFP966 to the culture medium. Neurons were collected after 24 h for western blot (WB), terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and 2'-7'-dichlorodihydrofluorescein diacetate staining. In vivo, RGFP966 (10 mg/kg) was administered post-TBI. Brain tissue water content and modified neurological severity scores were assessed 72 h post-injury. Cortical tissues surrounding the focal injury were subjected to western blot, TUNEL staining, Nissl staining and immunofluorescence/immunohistochemistry staining, and malondialdehyde level, hindered glutathione content and superoxide dismutase activity were measured. Serum was collected for the enzyme-linked immunosorbent assay. Nrf2-specific shRNA lentivirus was injected into the lateral ventricle of rats for 7 days, and cerebral cortex tissue was analyzed by WB and real-time polymerase chain reaction. Results During in vitro and in vivo experiments, RGFP966 suppressed HDAC3 expression, promoted Nrf2 nuclear translocation, activated downstream AO enzymes, mitigated excessive reactive oxygen species production and alleviated nerve cell apoptosis. RGFP966 effectively reduced brain edema and histological damage and enhanced neurological and cognitive function in rats with TBI. RGFP966 markedly inhibited NLRP3 inflammasome activation mediated by high-mobility group box 1 (HMGB1)/toll-like receptor 4 (TLR4). Nrf2 knockdown in TBI rats attenuated the AO and anti-inflammatory, neuroprotective impacts of RGFP966. Conclusions Overall, our findings demonstrate that RGFP966 can mitigate the first brain damage and neurological impairments in TBI. The underlying mechanism involves triggering the Nrf2-mediated AO system and negatively regulating the HMGB1/TLR4-mediated NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Lanjuan Xu
- Department of Critical Care Medicine, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Tingting An
- Department of Critical Care Medicine, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Baohui Jia
- Department of Critical Care Medicine, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Qiong Wu
- Department of Critical Care Medicine, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Jinggui Shen
- Department of Critical Care Medicine, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Jie Jin
- Department of Critical Care Medicine, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Jing Liu
- Department of Critical Care Medicine, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Chengjian Li
- Department of Critical Care Medicine, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan Province 450001, China
| |
Collapse
|
28
|
Zhao Y, Zhou YG, Chen JF. Targeting the adenosine A 2A receptor for neuroprotection and cognitive improvement in traumatic brain injury and Parkinson's disease. Chin J Traumatol 2024; 27:125-133. [PMID: 37679245 PMCID: PMC11138351 DOI: 10.1016/j.cjtee.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 07/25/2023] [Accepted: 08/16/2023] [Indexed: 09/09/2023] Open
Abstract
Adenosine exerts its dual functions of homeostasis and neuromodulation in the brain by acting at mainly 2 G-protein coupled receptors, called A1 and A2A receptors. The adenosine A2A receptor (A2AR) antagonists have been clinically pursued for the last 2 decades, leading to final approval of the istradefylline, an A2AR antagonist, for the treatment of OFF-Parkinson's disease (PD) patients. The approval paves the way to develop novel therapeutic methods for A2AR antagonists to address 2 major unmet medical needs in PD and traumatic brain injury (TBI), namely neuroprotection or improving cognition. In this review, we first consider the evidence for aberrantly increased adenosine signaling in PD and TBI and the sufficiency of the increased A2AR signaling to trigger neurotoxicity and cognitive impairment. We further discuss the increasing preclinical data on the reversal of cognitive deficits in PD and TBI by A2AR antagonists through control of degenerative proteins and synaptotoxicity, and on protection against TBI and PD pathologies by A2AR antagonists through control of neuroinflammation. Moreover, we provide the supporting evidence from multiple human prospective epidemiological studies which revealed an inverse relation between the consumption of caffeine and the risk of developing PD and cognitive decline in aging population and Alzheimer's disease patients. Collectively, the convergence of clinical, epidemiological and experimental evidence supports the validity of A2AR as a new therapeutic target and facilitates the design of A2AR antagonists in clinical trials for disease-modifying and cognitive benefit in PD and TBI patients.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yuan-Guo Zhou
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jiang-Fan Chen
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang Province, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, 325035, Zhejiang Province, China.
| |
Collapse
|
29
|
Goodman GW, Devlin P, West BE, Ritzel RM. The emerging importance of skull-brain interactions in traumatic brain injury. Front Immunol 2024; 15:1353513. [PMID: 38680490 PMCID: PMC11047125 DOI: 10.3389/fimmu.2024.1353513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 04/01/2024] [Indexed: 05/01/2024] Open
Abstract
The recent identification of skull bone marrow as a reactive hematopoietic niche that can contribute to and direct leukocyte trafficking into the meninges and brain has transformed our view of this bone structure from a solid, protective casing to a living, dynamic tissue poised to modulate brain homeostasis and neuroinflammation. This emerging concept may be highly relevant to injuries that directly impact the skull such as in traumatic brain injury (TBI). From mild concussion to severe contusion with skull fracturing, the bone marrow response of this local myeloid cell reservoir has the potential to impact not just the acute inflammatory response in the brain, but also the remodeling of the calvarium itself, influencing its response to future head impacts. If we borrow understanding from recent discoveries in other CNS immunological niches and extend them to this nascent, but growing, subfield of neuroimmunology, it is not unreasonable to consider the hematopoietic compartment in the skull may similarly play an important role in health, aging, and neurodegenerative disease following TBI. This literature review briefly summarizes the traditional role of the skull in TBI and offers some additional insights into skull-brain interactions and their potential role in affecting secondary neuroinflammation and injury outcomes.
Collapse
Affiliation(s)
| | | | | | - Rodney M. Ritzel
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
30
|
Qin T, Guo L, Wang X, Zhou G, Liu L, Zhang Z, Ding G. Repetitive transcranial magnetic stimulation ameliorates cognitive deficits in mice with radiation-induced brain injury by attenuating microglial pyroptosis and promoting neurogenesis via BDNF pathway. Cell Commun Signal 2024; 22:216. [PMID: 38570868 PMCID: PMC10988892 DOI: 10.1186/s12964-024-01591-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/23/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Radiation-induced brain injury (RIBI) is a common and severe complication during radiotherapy for head and neck tumor. Repetitive transcranial magnetic stimulation (rTMS) is a novel and non-invasive method of brain stimulation, which has been applied in various neurological diseases. rTMS has been proved to be effective for treatment of RIBI, while its mechanisms have not been well understood. METHODS RIBI mouse model was established by cranial irradiation, K252a was daily injected intraperitoneally to block BDNF pathway. Immunofluorescence staining, immunohistochemistry and western blotting were performed to examine the microglial pyroptosis and hippocampal neurogenesis. Behavioral tests were used to assess the cognitive function and emotionality of mice. Golgi staining was applied to observe the structure of dendritic spine in hippocampus. RESULTS rTMS significantly promoted hippocampal neurogenesis and mitigated neuroinflammation, with ameliorating pyroptosis in microglia, as well as downregulation of the protein expression level of NLRP3 inflammasome and key pyroptosis factor Gasdermin D (GSDMD). BDNF signaling pathway might be involved in it. After blocking BDNF pathway by K252a, a specific BDNF pathway inhibitor, the neuroprotective effect of rTMS was markedly reversed. Evaluated by behavioral tests, the cognitive dysfunction and anxiety-like behavior were found aggravated with the comparison of mice in rTMS intervention group. Moreover, the level of hippocampal neurogenesis was found to be attenuated, the pyroptosis of microglia as well as the levels of GSDMD, NLRP3 inflammasome and IL-1β were upregulated. CONCLUSION Our study indicated that rTMS notably ameliorated RIBI-induced cognitive disorders, by mitigating pyroptosis in microglia and promoting hippocampal neurogenesis via mediating BDNF pathway.
Collapse
Affiliation(s)
- Tongzhou Qin
- Department of radiation protection medicine, School of Preventive Medicine, Fourth Military Medical University, Xi'an, 710032, China
- Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Xi'an, China
| | - Ling Guo
- Department of radiation protection medicine, School of Preventive Medicine, Fourth Military Medical University, Xi'an, 710032, China
- Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Xi'an, China
| | - Xing Wang
- Department of radiation protection medicine, School of Preventive Medicine, Fourth Military Medical University, Xi'an, 710032, China
- Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Xi'an, China
| | - Guiqiang Zhou
- Department of radiation protection medicine, School of Preventive Medicine, Fourth Military Medical University, Xi'an, 710032, China
- Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Xi'an, China
- Department of occupational & environmental health, School of Public Health, Weifang Medical University, Weifang, 261021, China
| | - Liyuan Liu
- Department of radiation protection medicine, School of Preventive Medicine, Fourth Military Medical University, Xi'an, 710032, China
- Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Xi'an, China
| | - Zhaowen Zhang
- Department of radiation protection medicine, School of Preventive Medicine, Fourth Military Medical University, Xi'an, 710032, China
- Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Xi'an, China
| | - Guirong Ding
- Department of radiation protection medicine, School of Preventive Medicine, Fourth Military Medical University, Xi'an, 710032, China.
- Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Xi'an, China.
| |
Collapse
|
31
|
Gandasasmita N, Li J, Loane DJ, Semple BD. Experimental Models of Hospital-Acquired Infections After Traumatic Brain Injury: Challenges and Opportunities. J Neurotrauma 2024; 41:752-770. [PMID: 37885226 DOI: 10.1089/neu.2023.0453] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023] Open
Abstract
Patients hospitalized after a moderate or severe traumatic brain injury (TBI) are at increased risk of nosocomial infections, including bacterial pneumonia and other upper respiratory tract infections. Infections represent a secondary immune challenge for vulnerable TBI patients that can lead to increased morbidity and poorer long-term prognosis. This review first describes the clinical significance of infections after TBI, delving into the known mechanisms by which a TBI can alter systemic immunological responses towards an immunosuppressive state, leading to promotion of increased vulnerability to infections. Pulmonary dysfunction resulting from respiratory tract infections is considered in the context of neurotrauma, including the bidirectional relationship between the brain and lungs. Turning to pre-clinical modeling, current laboratory approaches to study experimental TBI and lung infections are reviewed, to highlight findings from the limited key studies to date that have incorporated both insults. Then, practical decisions for the experimental design of animal studies of post-injury infections are discussed. Variables associated with the host animal, the infectious agent (e.g., species, strain, dose, and administration route), as well as the timing of the infection relative to the injury model are important considerations for model development. Together, the purpose of this review is to highlight the significant clinical need for increased pre-clinical research into the two-hit insult of a hospital-acquired infection after TBI to encourage further scientific enquiry in the field.
Collapse
Affiliation(s)
| | - Jian Li
- Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
- Department of Microbiology, Monash University, Melbourne, Victoria, Australia
| | - David J Loane
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Bridgette D Semple
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Prahran, Victoria, Australia
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
32
|
Luo J, Luo Y, Chen J, Gao Y, Tan J, Yang Y, Yang C, Jiang N, Luo Y. Intestinal metabolite UroB alleviates cerebral ischemia/reperfusion injury by promoting competition between TRIM65 and TXNIP for binding to NLRP3 inflammasome in response to neuroinflammation. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167056. [PMID: 38360072 DOI: 10.1016/j.bbadis.2024.167056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 01/28/2024] [Accepted: 01/31/2024] [Indexed: 02/17/2024]
Abstract
Our previous research suggests that targeting NLRP3 inflammasomes holds promise for mitigating cerebral ischemia/reperfusion injury. The gut metabolite Urolithin B (UroB) has been shown to inhibit the neuroinflammation. However, the specific role of UroB in cerebral ischemia/reperfusion injury and its potential impact on NLRP3 inflammasome remain unclear. In this study, acute stroke was simulated using the MCAO model in male Sprague-Dawley rats. UroB was intraperitoneally administered after 1 h of reperfusion. The effects of UroB on brain tissue were evaluated, including infarct volume, brain edema, and neurobehavioral changes. Western blotting and immunofluorescence were performed to investigate the effect of UroB on inflammation-related proteins. Furthermore, TRIM65 knockdown and TXNIP overexpression experiments elucidated the role of UroB in NLRP3 inflammasome activation. The ( demonstrate the neuroprotective effect of UroB in acute stroke, reducing brain tissue damage and improving motor function. Mechanistically, UroB modulated neuroinflammation by influencing TXNIP and TRIM65 protein expression, as well as competitive binding to the NLRP3 inflammasome, attenuating cerebral ischemia/reperfusion injury. In conclusion, the potential of UroB as a protective agent against cerebral ischemia/reperfusion injury in acute stroke stands out as it regulates TRIM65 and TXNIP competitive binding to the NLRP3 inflammasome. These findings suggest that UroB is a promising drug candidate for the treatment of acute stroke.
Collapse
Affiliation(s)
- Jing Luo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Pathology, Chongqing Medical University, Chongqing, China; Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China; Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yujia Luo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jialei Chen
- Molecular Medicine and Cancer Research Center, College of Basic Medical Sciences, Chongqing Medical University, Chongqing, China; Department of Pathology and Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Yu Gao
- Department of Pathology, Chongqing Medical University, Chongqing, China; Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China; Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Junyi Tan
- Department of Pathology and Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Yongkang Yang
- Department of Clinical Medicine, Clinical Medical College of Chengdu University, Chengdu, China
| | - Changhong Yang
- Department of Bioinformatics, Chongqing Medical University, Chongqing, China
| | - Ning Jiang
- Department of Pathology, Chongqing Medical University, Chongqing, China; Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China; Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Yong Luo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
33
|
Amlerova Z, Chmelova M, Anderova M, Vargova L. Reactive gliosis in traumatic brain injury: a comprehensive review. Front Cell Neurosci 2024; 18:1335849. [PMID: 38481632 PMCID: PMC10933082 DOI: 10.3389/fncel.2024.1335849] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/01/2024] [Indexed: 01/03/2025] Open
Abstract
Traumatic brain injury (TBI) is one of the most common pathological conditions impacting the central nervous system (CNS). A neurological deficit associated with TBI results from a complex of pathogenetic mechanisms including glutamate excitotoxicity, inflammation, demyelination, programmed cell death, or the development of edema. The critical components contributing to CNS response, damage control, and regeneration after TBI are glial cells-in reaction to tissue damage, their activation, hypertrophy, and proliferation occur, followed by the formation of a glial scar. The glial scar creates a barrier in damaged tissue and helps protect the CNS in the acute phase post-injury. However, this process prevents complete tissue recovery in the late/chronic phase by producing permanent scarring, which significantly impacts brain function. Various glial cell types participate in the scar formation, but this process is mostly attributed to reactive astrocytes and microglia, which play important roles in several brain pathologies. Novel technologies including whole-genome transcriptomic and epigenomic analyses, and unbiased proteomics, show that both astrocytes and microglia represent groups of heterogenic cell subpopulations with different genomic and functional characteristics, that are responsible for their role in neurodegeneration, neuroprotection and regeneration. Depending on the representation of distinct glia subpopulations, the tissue damage as well as the regenerative processes or delayed neurodegeneration after TBI may thus differ in nearby or remote areas or in different brain structures. This review summarizes TBI as a complex process, where the resultant effect is severity-, region- and time-dependent and determined by the model of the CNS injury and the distance of the explored area from the lesion site. Here, we also discuss findings concerning intercellular signaling, long-term impacts of TBI and the possibilities of novel therapeutical approaches. We believe that a comprehensive study with an emphasis on glial cells, involved in tissue post-injury processes, may be helpful for further research of TBI and be the decisive factor when choosing a TBI model.
Collapse
Affiliation(s)
- Zuzana Amlerova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Martina Chmelova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Miroslava Anderova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Lydia Vargova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
34
|
Byun DJ, Lee J, Ko K, Hyun YM. NLRP3 exacerbates EAE severity through ROS-dependent NET formation in the mouse brain. Cell Commun Signal 2024; 22:96. [PMID: 38308301 PMCID: PMC10835891 DOI: 10.1186/s12964-023-01447-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/19/2023] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Neutrophil extracellular trap (NET) has been implicated in the pathology of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE). However, the specific contributions of NLRP3, a NET-associated molecule, to EAE pathogenesis and its regulatory role in NET formation remain unknown. METHODS To investigate the detrimental effect of NETs supported by NLRP3 in MS pathogenesis, we induced EAE in WT and NLRP3 KO mice and monitored the disease severity. At the peak of the disease, NET formation was assessed by flow cytometry, immunoblotting, and immunofluorescence staining. To further identify the propensity of infiltrated neutrophils, NET-related chemokine receptors, degranulation, ROS production, and PAD4 expression levels were evaluated by flow cytometry. In some experiments, mice were injected with DNase-1 to eliminate the formed NETs. RESULTS Our data revealed that neutrophils significantly infiltrate the brain and spinal cord and form NETs during EAE pathogenesis. NLRP3 significantly elevates NET formation, primarily in the brain. NLRP3 also modulated the phenotypes of brain-infiltrated and circulating neutrophils, augmenting CXCR2 and CXCR4 expression, thereby potentially enhancing NET formation. NLRP3 facilitates NET formation in a ROS-dependent and PAD4-independent manner in brain-infiltrated neutrophils. Finally, NLRP3-supported NET formation exacerbates disease severity, triggering Th1 and Th17 cells recruitment. CONCLUSIONS Collectively, our findings suggest that NLRP3-supported NETs may be an etiological factor in EAE pathogenesis, primarily in the brain. This study provides evidence that targeting NLRP3 could be a potential therapeutic strategy for MS, specifically by attenuating NET formation.
Collapse
Affiliation(s)
- Da Jeong Byun
- Department of Anatomy and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jaeho Lee
- Department of Anatomy and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyungryung Ko
- Department of Anatomy and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young-Min Hyun
- Department of Anatomy and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
35
|
Sun X, Guo P, Wang N, Shi Y, Li Y. A refined therapeutic plan based on the machine-learning prognostic model of liver hepatocellular carcinoma. Comput Biol Med 2024; 169:107907. [PMID: 38184863 DOI: 10.1016/j.compbiomed.2023.107907] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/08/2023] [Accepted: 12/24/2023] [Indexed: 01/09/2024]
Abstract
To deeply explore new strategy of the individual therapy for the patients with liver hepatocellular carcinoma (LIHC), we observed gene expression profile in patients with LIHC and made a comprehensive analysis of the inflammation-related phenotypes, we detected a set of characteristic genes associated with the biological activities of tumor cells, among which 3 genes and 2 lncRNAs are tagged on the LIHC prognosis. Then we constructed a novel prognostic model by machine learning, called Inf-PR model, and evaluated the drug sensitivity and immune targets by a series of bioinformatics tools. Ten-fold cross-validation testified that the model achieved excellent performance on prediction and classification of prognostic risks, which was not only able to get more reliable prognosis information than the age, gender, grade and stage, but also exceeded those previously reported similar models. Accordingly, drug sensitivity was detected in different prognostic risk groups, the result displayed that 10 FDA-approved small molecular drugs including lovastatin and sorafenib had higher sensitivities and perturbativities in the high-risk group, and other 15 drugs including doxorubicin and lenvatinib had better sensitivities and perturbativities in the low-risk group. Moreover, it suggested the patients with high risk would better combine with immunotherapy than those with low risk. Taken together, this study presents a new individual precision strategy about drug and target selection to treat LIHC based on this evaluation model, which is a powerful supplement for current anti-tumor therapy.
Collapse
Affiliation(s)
- Xiangcheng Sun
- West China Biopharm Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, China
| | - Peng Guo
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Ning Wang
- West China Biopharm Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yun Shi
- West China Biopharm Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yan Li
- West China Biopharm Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
36
|
Choi BJ, Park MH, Jin HK, Bae JS. Acid sphingomyelinase as a pathological and therapeutic target in neurological disorders: focus on Alzheimer's disease. Exp Mol Med 2024; 56:301-310. [PMID: 38337058 PMCID: PMC10907607 DOI: 10.1038/s12276-024-01176-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 02/12/2024] Open
Abstract
Over the past decade, numerous studies have highlighted the importance of acid sphingomyelinase (ASM) in disease treatment in humans. This enzyme functions primarily to generate ceramide, maintain the cellular membrane, and regulate cellular function. However, in the blood and brain of patients with neurological disorders, including major depression, ischemic stroke, amyotrophic lateral sclerosis, multiple sclerosis, and Alzheimer's disease (AD), elevated ASM levels significantly suggest disease onset or progression. In these diseases, increased ASM is profoundly involved in neuronal death, abnormal autophagy, neuroinflammation, blood-brain barrier disruption, hippocampal neurogenesis loss, and immune cell dysfunction. Moreover, genetic and pharmacological inhibition of ASM can prevent or ameliorate various diseases. The therapeutic effects of ASM inhibition have prompted the urgent need to develop ASM inhibitors, and several ASM inhibitors have been identified. In this review, we summarize the current knowledge on the critical roles and mechanisms of ASM in brain cells and blood that are associated with different neuropathological features, especially those observed in AD. Furthermore, we elucidate the potential possibility and limitations of existing ASM-targeting drugs according to experimental studies in neurological disorder mouse models.
Collapse
Affiliation(s)
- Byung Jo Choi
- KNU Alzheimer's Disease Research Institute, Kyungpook National University, Daegu, 41566, South Korea
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea
| | - Min Hee Park
- KNU Alzheimer's Disease Research Institute, Kyungpook National University, Daegu, 41566, South Korea
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea
| | - Hee Kyung Jin
- KNU Alzheimer's Disease Research Institute, Kyungpook National University, Daegu, 41566, South Korea
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, South Korea
| | - Jae-Sung Bae
- KNU Alzheimer's Disease Research Institute, Kyungpook National University, Daegu, 41566, South Korea.
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea.
| |
Collapse
|
37
|
Mendoza-Mari Y, Rai V, Radwan MM, Brazdzionis J, Connett DA, Miulli DE, Agrawal DK. Modulation of Inflammatory Response by Electromagnetic Field Stimulation in Traumatic Brain Injury in Yucatan Swine. JOURNAL OF SURGERY AND RESEARCH 2024; 7:20-40. [PMID: 38389906 PMCID: PMC10883333 DOI: 10.26502/jsr.10020338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Traumatic brain injury is a leading cause of disability and death worldwide and represents a high economic burden for families and national health systems. After mechanical impact to the head, the first stage of the damage comprising edema, physical damage, and cell loss gives rise to a second phase characterized by glial activation, increased oxidative stress and excitotoxicity, mitochondrial damage, and exacerbated neuroinflammatory state, among other molecular calamities. Inflammation strongly influences the molecular events involved in the pathogenesis of TBI. Therefore, several components of the inflammatory cascade have been targeted in experimental therapies. Application of Electromagnetic Field (EMF) stimulation has been found to be effective in some inflammatory conditions. However, its effect in the neuronal recovery after TBI is not known. In this pilot study, Yucatan miniswine were subjected to TBI using controlled cortical impact approach. EMF stimulation via a helmet was applied immediately or two days after mechanical impact. Three weeks later, inflammatory markers were assessed in the brain tissues of injured and contralateral non-injured areas of control and EMF-treated animals by histomorphometry, immunohistochemistry, RT-qPCR, Western blot, and ELISA. Our results revealed that EMF stimulation induced beneficial effect with the preservation of neuronal tissue morphology as well as the reduction of inflammatory markers at the transcriptional and translational levels. Immediate EMF application showed better resolution of inflammation. Although further studies are warranted, our findings contribute to the notion that EMF stimulation could be an effective therapeutic approach in TBI patients.
Collapse
Affiliation(s)
- Yssel Mendoza-Mari
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766
| | - Vikrant Rai
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766
| | - Mohamed M Radwan
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766
| | - James Brazdzionis
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766
| | - David A Connett
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766
| | - Dan E Miulli
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766
| |
Collapse
|
38
|
Xu Y, Xu Y, Biby S, Kaur B, Liu Y, Bagdasarian FA, Wey HY, Tanzi R, Zhang C, Wang C, Zhang S. Design and Discovery of Novel NLRP3 Inhibitors and PET Imaging Radiotracers Based on a 1,2,3-Triazole-Bearing Scaffold. J Med Chem 2024; 67:555-571. [PMID: 38150705 PMCID: PMC11002996 DOI: 10.1021/acs.jmedchem.3c01782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
The NOD-like receptor (NLR) family pyrin-domain-containing 3 (NLRP3) inflammasome, an essential component of the innate immune system, has been emerging as a viable drug target and a potential biomarker for human diseases. In our efforts to develop novel small molecule NLRP3 inhibitors, a 1-(5-chloro-2-methoxybenzyl)-4-phenyl-1H-1,2,3-triazole scaffold was designed via a rational approach based on our previous leads. Structure-activity relationship studies and biophysical studies identified a new lead compound 8 as a potent (IC50: 0.55 ± 0.16 μM), selective, and direct NLRP3 inhibitor. Positron emission tomography (PET) imaging studies of [11C]8 demonstrated its rapid and high brain uptake as well as fast washout in mice and rhesus macaque. Notably, plasma kinetic analysis of this radiotracer from the PET/magnetic resonance imaging studies in rhesus macaque suggested radiometabolic stability. Collectively, our data not only encourage further studies of this lead compound but also warrant further optimization to generate additional novel NLRP3 inhibitors and suitable central nervous system PET radioligands with translational promise.
Collapse
Affiliation(s)
- Yiming Xu
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Yulong Xu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Savannah Biby
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Baljit Kaur
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Yan Liu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Frederick Andrew Bagdasarian
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Hsiao-Ying Wey
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Rudolph Tanzi
- Genetics and Aging Research Unit, McCane Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Can Zhang
- Genetics and Aging Research Unit, McCane Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Changning Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Shijun Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| |
Collapse
|
39
|
Zhang Z, Zhang Y, Zhang M, Yu C, Yang P, Xu M, Ling J, Wu Y, Zhu Z, Chen Y, Shi A, Liu X, Zhang J, Yu P, Zhang D. Food-derived peptides as novel therapeutic strategies for NLRP3 inflammasome-related diseases: a systematic review. Crit Rev Food Sci Nutr 2023; 65:1433-1464. [PMID: 38153262 DOI: 10.1080/10408398.2023.2294164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3), a member of the nucleotide-binding domain (NOD) and leucine-rich repeat sequence (LRR) protein (NLR) family, plays an essential role in the inflammation initiation and inflammatory mediator secretion, and thus is also associated with many disease progressions. Food-derived bioactive peptides (FDBP) exhibit excellent anti-inflammatory activity in both in vivo and in vitro models. They are encrypted in plant, meat, and milk proteins and can be released under enzymatic hydrolysis or fermentation conditions, thereby hindering the progression of hyperuricemia, inflammatory bowel disease, chronic liver disease, neurological disorders, lung injury and periodontitis by inactivating the NLRP3. However, there is a lack of systematic review around FDBP, NLRP3, and NLRP3-related diseases. Therefore, this review summarized FDBP that exert inhibiting effects on NLRP3 inflammasome from different protein sources and detailed their preparation and purification methods. Additionally, this paper also compiled the possible inhibitory mechanisms of FDBP on NLRP3 inflammasomes and its regulatory role in NLRP3 inflammasome-related diseases. Finally, the progress of cutting-edge technologies, including nanoparticle, computer-aided screening strategy and recombinant DNA technology, in the acquisition or encapsulation of NLRP3 inhibitory FDBP was discussed. This review provides a scientific basis for understanding the anti-inflammatory mechanism of FDBP through the regulation of the NLRP3 inflammasome and also provides guidance for the development of therapeutic adjuvants or functional foods enriched with these FDBP.
Collapse
Affiliation(s)
- Ziqi Zhang
- The Second Clinical Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Jiangxi, China
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuan Zhang
- School of Public Health, Nanchang University, Jiangxi, China
| | - Meiying Zhang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Chenfeng Yu
- Huankui College, Nanchang University, Jiangxi, China
| | - Pingping Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Minxuan Xu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Yuting Wu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Zicheng Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yixuan Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ao Shi
- School of Medicine, St. George University of London, London, UK
| | - Xiao Liu
- Cardiology Department, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Deju Zhang
- The Second Clinical Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Jiangxi, China
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Hong Kong
| |
Collapse
|
40
|
Wang J, Ru QM, Yu XH, Wang C, Li K, Han CZY, Li N, Zhao J, Wood JN, Liu X, Wang R, Wang Y. Direct inhibition of microglial activation by a μ receptor selective agonist alleviates inflammatory-induced pain hypersensitivity. Eur J Pharmacol 2023; 961:176182. [PMID: 37951488 DOI: 10.1016/j.ejphar.2023.176182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 11/14/2023]
Abstract
Opioids are widely used in the treatment of moderate and severe pain. Nociceptive stimulation has been reported to potentially promote microglial activation and neuroinflammation, which also causes chronic pain sensitization. The aim of this study was to demonstrate whether the novel μ receptor agonist MEL-0614 could inhibit activated microglia directly and the associated signaling pathway. Mice were administered lipopolysaccharide and formalin to induce allodynia. Von Frey test was used to detect the anti-allodynia effect of MEL-0614 before and after LPS and formalin injection. In the spinal cord, the levels of proinflammatory cytokines and microglial activation were determined after MEL-0614 administration. BV2 and primary microglia were cultured to further explore the effect of MEL-0614 on LPS-induced microglial activation and key signaling pathways involved. MEL-0614 partially prevented and reversed allodynia induced by LPS and formalin in vivo, which was not inhibited by the μ receptor antagonist CTAP. Minocycline was effective in reversing the established allodynia. MEL-0614 also downregulated the activation of microglia and related proinflammatory cytokines in the spinal cord. Additionally, in BV2 and primary microglia, MEL-0614 inhibited the LPS-induced upregulation of proinflammatory factors, which was unaffected by CTAP. The NLR family pyrin domain containing 3 (NLRP3) related signaling pathway may be involved in the interaction between MEL-0614 and microglia. The opioid agonist MEL-0614 inhibited the activation of microglia and the subsequent upregulation of proinflammatory factors both in vivo and in vitro. Notably, this effect is partially mediated by the μ receptor.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pharmacology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Qiao-Min Ru
- Department of Pharmacology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiao-Hui Yu
- Department of Pharmacology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Changlong Wang
- Department of Pharmacology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Kai Li
- Department of Pharmacology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Chao-Zhen-Yi Han
- Department of Pharmacology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Na Li
- Department of Pharmacology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jing Zhao
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Xin Liu
- Department of Pharmacology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| | - Rui Wang
- Department of Pharmacology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, China.
| | - Yuan Wang
- Department of Pharmacology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| |
Collapse
|
41
|
Ayyubova G, Kodali M, Upadhya R, Madhu LN, Attaluri S, Somayaji Y, Shuai B, Rao S, Shankar G, Shetty AK. Extracellular vesicles from hiPSC-NSCs can prevent peripheral inflammation-induced cognitive dysfunction with inflammasome inhibition and improved neurogenesis in the hippocampus. J Neuroinflammation 2023; 20:297. [PMID: 38087314 PMCID: PMC10717852 DOI: 10.1186/s12974-023-02971-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
Extracellular vesicles (EVs) released by human induced pluripotent stem cell-derived neural stem cells (hiPSC-NSCs) are enriched with miRNAs and proteins capable of mediating robust antiinflammatory activity. The lack of tumorigenic and immunogenic properties and ability to permeate the entire brain to incorporate into microglia following intranasal (IN) administrations makes them an attractive biologic for curtailing chronic neuroinflammation in neurodegenerative disorders. We tested the hypothesis that IN administrations of hiPSC-NSC-EVs can alleviate chronic neuroinflammation and cognitive impairments induced by the peripheral lipopolysaccharide (LPS) challenge. Adult male, C57BL/6J mice received intraperitoneal injections of LPS (0.75 mg/kg) for seven consecutive days. Then, the mice received either vehicle (VEH) or hiPSC-NSC-EVs (~ 10 × 109 EVs/administration, thrice over 6 days). A month later, mice in all groups were investigated for cognitive function with behavioral tests and euthanized for histological and biochemical studies. Mice receiving VEH after LPS displayed deficits in associative recognition memory, temporal pattern processing, and pattern separation. Such impairments were associated with an increased incidence of activated microglia presenting NOD-, LRR-, and pyrin domain containing 3 (NLRP3) inflammasomes, elevated levels of NLRP3 inflammasome mediators and end products, and decreased neurogenesis in the hippocampus. In contrast, the various cognitive measures in mice receiving hiPSC-NSC-EVs after LPS were closer to naive mice. Significantly, these mice displayed diminished microglial activation, NLRP3 inflammasomes, proinflammatory cytokines, and a level of neurogenesis matching age-matched naïve controls. Thus, IN administrations of hiPSC-NSC-EVs are an efficacious approach to reducing chronic neuroinflammation-induced cognitive impairments.
Collapse
Affiliation(s)
- Gunel Ayyubova
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Leelavathi N Madhu
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Yogish Somayaji
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Shama Rao
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Goutham Shankar
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA.
| |
Collapse
|
42
|
Zeb MA, Ding L, Wang MR, Tu WC, Li XL, Zhang XJ, Xiao WL. Three New Clerodane Diterpenoids and Their NLRP3 Inflammasome Activation Inhibitory Activity from Callicarpa arborea Roxb. Chem Biodivers 2023; 20:e202301676. [PMID: 37971960 DOI: 10.1002/cbdv.202301676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 11/19/2023]
Abstract
Three new compounds callicarpenoids A-C (1-3), were isolated from the stems of Callicarpa arborea Roxb together with fifteen known compounds (4-18). The structures of these compounds were elucidated using advanced spectroscopic techniques, including 1D and 2D NMR, UV, IR, HR-ESI-MS, ECD, ORD, and quantum chemical calculations. Compound 3, a rare rearranged diterpenoid with a fused 5/6-ring system demonstrated strong potential as an inhibitor of the NLRP3 inflammasome activation with an IC50 value of 3.153 μM. It effectively reduced GSDMD-NT production, inhibited caspase-1 activation, and suppressed IL-1β secretion, thereby mitigating NLRP3 inflammasome-induced pyroptosis in J774A.1 cells. These findings suggest that compound 3 warrants further research and development as a promising NLRP3 inflammasome inhibitor.
Collapse
Affiliation(s)
- Muhammad Aurang Zeb
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Centre for Natural Products, School of Chemical Science and Technology and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, 650500, P. R. China
| | - Ling Ding
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Centre for Natural Products, School of Chemical Science and Technology and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, 650500, P. R. China
| | - Meng-Ru Wang
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Centre for Natural Products, School of Chemical Science and Technology and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, 650500, P. R. China
| | - Wen-Chao Tu
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Centre for Natural Products, School of Chemical Science and Technology and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, 650500, P. R. China
| | - Xiao-Li Li
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Centre for Natural Products, School of Chemical Science and Technology and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, 650500, P. R. China
| | - Xing-Jie Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Centre for Natural Products, School of Chemical Science and Technology and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, 650500, P. R. China
| | - Wei-Lie Xiao
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Centre for Natural Products, School of Chemical Science and Technology and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, 650500, P. R. China
| |
Collapse
|
43
|
Shaik MG, Joshi SV, Akunuri R, Rana P, Rahman Z, Polomoni A, Yaddanapudi VM, Dandekar MP, Srinivas N. Small molecule inhibitors of NLRP3 inflammasome and GSK-3β in the management of traumatic brain injury: A review. Eur J Med Chem 2023; 259:115718. [PMID: 37573828 DOI: 10.1016/j.ejmech.2023.115718] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 07/28/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023]
Abstract
Traumatic brain injury (TBI) is a debilitating mental condition which causes physical disability and morbidity worldwide. TBI may damage the brain by direct injury that subsequently triggers a series of neuroinflammatory events. The activation of NLRP3 inflammasome and dysregulated host immune system has been documented in various neurological disorders such as TBI, ischemic stroke and multiple sclerosis. The activation of NLRP3 post-TBI increases the production of pro-inflammatory cytokines and caspase-1, which are major drivers of neuroinflammation and apoptosis. Similarly, GSK-3β regulates apoptosis through tyrosine kinase and canonical Wnt signalling pathways. Thus, therapeutic targeting of NLRP3 inflammasome and GSK-3β has emerged as promising strategies for regulating the post-TBI neuroinflammation and neurobehavioral disturbances. In this review, we discuss the identification & development of several structurally diverse and pharmacologically interesting small molecule inhibitors for targeting the NLRP3 inflammasome and GSK-3β in the management of TBI.
Collapse
Affiliation(s)
- Mahammad Ghouse Shaik
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Swanand Vinayak Joshi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Ravikumar Akunuri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India; Ellen and Ronald Caplan Cancer Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Preeti Rana
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Ziaur Rahman
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500 037, India
| | - Anusha Polomoni
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Venkata Madhavi Yaddanapudi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Manoj P Dandekar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500 037, India.
| | - Nanduri Srinivas
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India.
| |
Collapse
|
44
|
Du X, Amin N, Xu L, Botchway BOA, Zhang B, Fang M. Pharmacological intervention of curcumin via the NLRP3 inflammasome in ischemic stroke. Front Pharmacol 2023; 14:1249644. [PMID: 37915409 PMCID: PMC10616488 DOI: 10.3389/fphar.2023.1249644] [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: 09/26/2023] [Indexed: 11/03/2023] Open
Abstract
Ischemic-induced neuronal injury arises due to low oxygen/nutrient levels and an inflammatory response that exacerbates neuronal loss. NOD-like receptor family pyrin domain-containing 3 (NLRP3) is an important regulator of inflammation after ischemic stroke, with its inhibition being involved in nerve regeneration. Curcumin, a main active ingredient in Chinese herbs, plays a positive role in neuronal repair and neuroprotection by regulating the NLRP3 signaling pathway. Nevertheless, the signaling mechanisms relating to how curcumin regulates NLRP3 inflammasome in inflammation and neural restoration following ischemic stroke are unknown. In this report, we summarize the main biological functions of the NLRP3 inflammasome along with the neuroprotective effects and underlying mechanisms of curcumin via impairment of the NLRP3 pathway in ischemic brain injury. We also discuss the role of medicinal interventions that target the NLRP3 and potential pathways, as well as possible directions for curcumin therapy to penetrate the blood-brain barrier (BBB) and hinder inflammation in ischemic stroke. This report conclusively demonstrates that curcumin has neuroprotective properties that inhibit inflammation and prevent nerve cell loss, thereby delaying the progression of ischemic brain damage.
Collapse
Affiliation(s)
- Xiaoxue Du
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Nashwa Amin
- Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Zoology, Faculty of Science, Aswan University, Aswan, Egypt
| | - Linhao Xu
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cardiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Benson O. A. Botchway
- Department of Neurology, Children’s Hospital of Zhejiang University School of Medicine, National Clinical Research Centre for Child Health, Hangzhou, China
- Pharmacy Department, Bupa Cromwell Hospital, London, United Kingdom
| | - Bo Zhang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Marong Fang
- Department of Neurology, Children’s Hospital of Zhejiang University School of Medicine, National Clinical Research Centre for Child Health, Hangzhou, China
| |
Collapse
|
45
|
Muslimov A, Tereshchenko V, Shevyrev D, Rogova A, Lepik K, Reshetnikov V, Ivanov R. The Dual Role of the Innate Immune System in the Effectiveness of mRNA Therapeutics. Int J Mol Sci 2023; 24:14820. [PMID: 37834268 PMCID: PMC10573212 DOI: 10.3390/ijms241914820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/24/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Advances in molecular biology have revolutionized the use of messenger RNA (mRNA) as a therapeutic. The concept of nucleic acid therapy with mRNA originated in 1990 when Wolff et al. reported successful expression of proteins in target organs by direct injection of either plasmid DNA or mRNA. It took decades to bring the transfection efficiency of mRNA closer to that of DNA. The next few decades were dedicated to turning in vitro-transcribed (IVT) mRNA from a promising delivery tool for gene therapy into a full-blown therapeutic modality, which changed the biotech market rapidly. Hundreds of clinical trials are currently underway using mRNA for prophylaxis and therapy of infectious diseases and cancers, in regenerative medicine, and genome editing. The potential of IVT mRNA to induce an innate immune response favors its use for vaccination and immunotherapy. Nonetheless, in non-immunotherapy applications, the intrinsic immunostimulatory activity of mRNA directly hinders the desired therapeutic effect since it can seriously impair the target protein expression. Targeting the same innate immune factors can increase the effectiveness of mRNA therapeutics for some indications and decrease it for others, and vice versa. The review aims to present the innate immunity-related 'barriers' or 'springboards' that may affect the development of immunotherapies and non-immunotherapy applications of mRNA medicines.
Collapse
Affiliation(s)
- Albert Muslimov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
- Laboratory of Nano- and Microencapsulation of Biologically Active Substances, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia;
- RM Gorbacheva Research Institute, Pavlov University, L’va Tolstogo 6-8, 197022 St. Petersburg, Russia;
| | - Valeriy Tereshchenko
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| | - Daniil Shevyrev
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| | - Anna Rogova
- Laboratory of Nano- and Microencapsulation of Biologically Active Substances, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia;
- Saint-Petersburg Chemical-Pharmaceutical University, Professora Popova 14, 197376 St. Petersburg, Russia
- School of Physics and Engineering, ITMO University, Lomonosova 9, 191002 St. Petersburg, Russia
| | - Kirill Lepik
- RM Gorbacheva Research Institute, Pavlov University, L’va Tolstogo 6-8, 197022 St. Petersburg, Russia;
| | - Vasiliy Reshetnikov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Prospekt Akad. Lavrentyeva 10, 630090 Novosibirsk, Russia
| | - Roman Ivanov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| |
Collapse
|
46
|
Lindblad C, Rostami E, Helmy A. Interleukin-1 Receptor Antagonist as Therapy for Traumatic Brain Injury. Neurotherapeutics 2023; 20:1508-1528. [PMID: 37610701 PMCID: PMC10684479 DOI: 10.1007/s13311-023-01421-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2023] [Indexed: 08/24/2023] Open
Abstract
Traumatic brain injury is a common type of acquired brain injury of varying severity carrying potentially deleterious consequences for the afflicted individuals, families, and society. Following the initial, traumatically induced insult, cellular injury processes ensue. These are believed to be amenable to treatment. Among such injuries, neuroinflammation has gained interest and has become a specific focus for both experimental and clinical researchers. Neuroinflammation is elicited almost immediately following trauma, and extend for a long time, possibly for years, after the primary injury. In the acute phase, the inflammatory response is characterized by innate mechanisms such as the activation of microglia which among else mediates cytokine production. Among the earliest cytokines to emerge are the interleukin- (IL-) 1 family members, comprising, for example, the agonist IL-1β and its competitive antagonist, IL-1 receptor antagonist (IL-1ra). Because of its early emergence following trauma and its increased concentrations also after human TBI, IL-1 has been hypothesized to be a tractable treatment target following TBI. Ample experimental data supports this, and demonstrates restored neurological behavior, diminished lesion zones, and an attenuated inflammatory response following IL-1 modulation either through IL-1 knock-out experiments, IL-1β inhibition, or IL-1ra treatment. Of these, IL-1ra treatment is likely the most physiological. In addition, recombinant human IL-1ra (anakinra) is already approved for utilization across a few rheumatologic disorders. As of today, one randomized clinical controlled trial has utilized IL-1ra inhibition as an intervention and demonstrated its safety. Further clinical trials powered for patient outcome are needed in order to demonstrate efficacy. In this review, we summarize IL-1 biology in relation to acute neuroinflammatory processes following TBI with a particular focus on current evidence for IL-1ra treatment both in the experimental and clinical context.
Collapse
Affiliation(s)
- Caroline Lindblad
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
- Department of Neurosurgery, Uppsala University Hospital, entrance 85 floor 2, Akademiska Sjukhuset, 751 85, Uppsala, Sweden.
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Elham Rostami
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Department of Neurosurgery, Uppsala University Hospital, entrance 85 floor 2, Akademiska Sjukhuset, 751 85, Uppsala, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Adel Helmy
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Mészáros Á, Molnár K, Fazakas C, Nógrádi B, Lüvi A, Dudás T, Tiszlavicz L, Farkas AE, Krizbai IA, Wilhelm I. Inflammasome activation in peritumoral astrocytes is a key player in breast cancer brain metastasis development. Acta Neuropathol Commun 2023; 11:155. [PMID: 37749707 PMCID: PMC10521486 DOI: 10.1186/s40478-023-01646-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/27/2023] [Indexed: 09/27/2023] Open
Abstract
Inflammasomes, primarily responsible for the activation of IL-1β, have emerged as critical regulators of the tumor microenvironment. By using in vivo and in vitro brain metastasis models, as well as human samples to study the role of the NLRP3 inflammasome in triple-negative breast cancer (TNBC) brain metastases, we found NLRP3 inflammasome components and IL-1β to be highly and specifically expressed in peritumoral astrocytes. Soluble factors from TNBC cells induced upregulation and activation of NLRP3 and IL-1β in astrocytes, while astrocyte-derived mediators augmented the proliferation of metastatic cells. In addition, inhibition of NLRP3 inflammasome activity using MCC950 or dampening the downstream effect of IL-1β prevented the proliferation increase in cancer cells. In vivo, MCC950 reduced IL-1β expression in peritumoral astrocytes, as well as the levels of inflammasome components and active IL-1β. Most importantly, significantly retarded growth of brain metastatic tumors was observed in mice treated with MCC950. Overall, astrocytes contribute to TNBC progression in the brain through activation of the NLRP3 inflammasome and consequent IL-1β release. We conclude that pharmacological targeting of inflammasomes may become a novel strategy in controlling brain metastatic diseases.
Collapse
Affiliation(s)
- Ádám Mészáros
- Institute of Biophysics, Biological Research Centre, ELKH (Eötvös Loránd Research Network), Temesvári Krt. 62, 6726, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Kinga Molnár
- Institute of Biophysics, Biological Research Centre, ELKH (Eötvös Loránd Research Network), Temesvári Krt. 62, 6726, Szeged, Hungary
| | - Csilla Fazakas
- Institute of Biophysics, Biological Research Centre, ELKH (Eötvös Loránd Research Network), Temesvári Krt. 62, 6726, Szeged, Hungary
| | - Bernát Nógrádi
- Institute of Biophysics, Biological Research Centre, ELKH (Eötvös Loránd Research Network), Temesvári Krt. 62, 6726, Szeged, Hungary
- Department of Neurology, University of Szeged, Szeged, Hungary
| | - Adél Lüvi
- Institute of Biophysics, Biological Research Centre, ELKH (Eötvös Loránd Research Network), Temesvári Krt. 62, 6726, Szeged, Hungary
| | - Tamás Dudás
- Institute of Biophysics, Biological Research Centre, ELKH (Eötvös Loránd Research Network), Temesvári Krt. 62, 6726, Szeged, Hungary
- Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | | | - Attila Elek Farkas
- Institute of Biophysics, Biological Research Centre, ELKH (Eötvös Loránd Research Network), Temesvári Krt. 62, 6726, Szeged, Hungary
| | - István Adorján Krizbai
- Institute of Biophysics, Biological Research Centre, ELKH (Eötvös Loránd Research Network), Temesvári Krt. 62, 6726, Szeged, Hungary.
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, Arad, Romania.
| | - Imola Wilhelm
- Institute of Biophysics, Biological Research Centre, ELKH (Eötvös Loránd Research Network), Temesvári Krt. 62, 6726, Szeged, Hungary.
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, Arad, Romania.
| |
Collapse
|
48
|
Janković T, Pilipović K. Single Versus Repetitive Traumatic Brain Injury: Current Knowledge on the Chronic Outcomes, Neuropathology and the Role of TDP-43 Proteinopathy. Exp Neurobiol 2023; 32:195-215. [PMID: 37749924 PMCID: PMC10569144 DOI: 10.5607/en23008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 07/18/2023] [Accepted: 08/23/2023] [Indexed: 09/27/2023] Open
Abstract
Traumatic brain injury (TBI) is one of the most important causes of death and disability in adults and thus an important public health problem. Following TBI, secondary pathophysiological processes develop over time and condition the development of different neurodegenerative entities. Previous studies suggest that neurobehavioral changes occurring after a single TBI are the basis for the development of Alzheimer's disease, while repetitive TBI is considered to be a contributing factor for chronic traumatic encephalopathy development. However, pathophysiological processes that determine the evolvement of a particular chronic entity are still unclear. Human post-mortem studies have found combinations of amyloid, tau, Lewi bodies, and TAR DNA-binding protein 43 (TDP-43) pathologies after both single and repetitive TBI. This review focuses on the pathological changes of TDP-43 after single and repetitive brain traumas. Numerous studies have shown that TDP-43 proteinopathy noticeably occurs after repetitive head trauma. A relatively small number of available preclinical research on single brain injury are not in complete agreement with the results from the human samples, which makes it difficult to draw specific conclusions. Also, as TBI is considered a heterogeneous type of injury, different experimental trauma models and injury intensities may cause differences in the cascade of secondary injury, which should be considered in future studies. Experimental and post-mortem studies of TDP-43 pathobiology should be carried out, preferably in the same laboratories, to determine its involvement in the development of neurodegenerative conditions after one and repetitive TBI, especially in the context of the development of new therapeutic options.
Collapse
Affiliation(s)
- Tamara Janković
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia
| | - Kristina Pilipović
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia
| |
Collapse
|
49
|
Nguyen T, Nguyen N, Cochran AG, Smith JA, Al-Juboori M, Brumett A, Saxena S, Talley S, Campbell EM, Obukhov AG, White FA. Repeated closed-head mild traumatic brain injury-induced inflammation is associated with nociceptive sensitization. J Neuroinflammation 2023; 20:196. [PMID: 37635235 PMCID: PMC10464478 DOI: 10.1186/s12974-023-02871-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 08/03/2023] [Indexed: 08/29/2023] Open
Abstract
BACKGROUND Individuals who have experienced mild traumatic brain injuries (mTBIs) suffer from several comorbidities, including chronic pain. Despite extensive studies investigating the underlying mechanisms of mTBI-associated chronic pain, the role of inflammation in long-term pain after mTBIs is not fully elucidated. Given the shifting dynamics of inflammation, it is important to understand the spatial-longitudinal changes in inflammatory processes following mTBIs and their effects on TBI-related pain. METHODS We utilized a recently developed transgenic caspase-1 luciferase reporter mouse model to monitor caspase-1 activation through a thinned skull window in the in vivo setting following three closed-head mTBI events. Organotypic coronal brain slice cultures and acutely dissociated dorsal root ganglion (DRG) cells provided tissue-relevant context of inflammation signal. Mechanical allodynia was assessed by mechanical withdrawal threshold to von Frey and thermal hyperalgesia withdrawal latency to radiant heat. Mouse grimace scale (MGS) was used to detect spontaneous or non-evoked pain. In some experiments, mice were prophylactically treated with MCC950, a potent small molecule inhibitor of NLRP3 inflammasome assembly to inhibit injury-induced inflammatory signaling. Bioluminescence spatiotemporal dynamics were quantified in the head and hind paws, and caspase-1 activation was confirmed by immunoblot. Immunofluorescence staining was used to monitor the progression of astrogliosis and microglial activation in ex vivo brain tissue following repetitive closed-head mTBIs. RESULTS Mice with repetitive closed-head mTBIs exhibited significant increases of the bioluminescence signals within the brain and paws in vivo for at least one week after each injury. Consistently, immunoblotting and immunofluorescence experiments confirmed that mTBIs led to caspase-1 activation, astrogliosis, and microgliosis. Persistent changes in MGS and hind paw withdrawal thresholds, indicative of pain states, were observed post-injury in the same mTBI animals in vivo. We also observed enhanced inflammatory responses in ex vivo brain slice preparations and DRG for at least 3 days following mTBIs. In vivo treatment with MCC950 significantly reduced caspase-1 activation-associated bioluminescent signals in vivo and decreased stimulus-evoked and non-stimulus evoked nociception. CONCLUSIONS Our findings suggest that the inflammatory states in the brain and peripheral nervous system following repeated mTBIs are coincidental with the development of nociceptive sensitization, and that these events can be significantly reduced by inhibition of NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Tyler Nguyen
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Natalie Nguyen
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ashlyn G Cochran
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jared A Smith
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mohammed Al-Juboori
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew Brumett
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Saahil Saxena
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sarah Talley
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL, USA
| | - Edward M Campbell
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL, USA
- Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Alexander G Obukhov
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy, Cellular Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fletcher A White
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA.
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
50
|
Freire MAM, Rocha GS, Bittencourt LO, Falcao D, Lima RR, Cavalcanti JRLP. Cellular and Molecular Pathophysiology of Traumatic Brain Injury: What Have We Learned So Far? BIOLOGY 2023; 12:1139. [PMID: 37627023 PMCID: PMC10452099 DOI: 10.3390/biology12081139] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of long-lasting morbidity and mortality worldwide, being a devastating condition related to the impairment of the nervous system after an external traumatic event resulting in transitory or permanent functional disability, with a significant burden to the healthcare system. Harmful events underlying TBI can be classified into two sequential stages, primary and secondary, which are both associated with breakdown of the tissue homeostasis due to impairment of the blood-brain barrier, osmotic imbalance, inflammatory processes, oxidative stress, excitotoxicity, and apoptotic cell death, ultimately resulting in a loss of tissue functionality. The present study provides an updated review concerning the roles of brain edema, inflammation, excitotoxicity, and oxidative stress on brain changes resulting from a TBI. The proper characterization of the phenomena resulting from TBI can contribute to the improvement of care, rehabilitation and quality of life of the affected people.
Collapse
Affiliation(s)
- Marco Aurelio M. Freire
- Graduate Program in Physiological Sciences, University of the State of Rio Grande do Norte, Mossoró 59607-360, RN, Brazil
| | - Gabriel Sousa Rocha
- Graduate Program in Biochemistry and Molecular Biology, University of the State of Rio Grande do Norte, Mossoró 59607-360, RN, Brazil
| | - Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-900, PA, Brazil
| | - Daniel Falcao
- VCU Health Systems, Virginia Commonwealth University, 23219 Richmond, VA, USA
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-900, PA, Brazil
| | - Jose Rodolfo Lopes P. Cavalcanti
- Graduate Program in Physiological Sciences, University of the State of Rio Grande do Norte, Mossoró 59607-360, RN, Brazil
- Graduate Program in Biochemistry and Molecular Biology, University of the State of Rio Grande do Norte, Mossoró 59607-360, RN, Brazil
| |
Collapse
|