1
|
Fan Z, Su D, Li ZC, Sun S, Ge Z. Metformin attenuates central sensitization by regulating neuroinflammation through the TREM2-SYK signaling pathway in a mouse model of chronic migraine. J Neuroinflammation 2024; 21:318. [PMID: 39627853 PMCID: PMC11613737 DOI: 10.1186/s12974-024-03313-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 11/27/2024] [Indexed: 12/08/2024] Open
Abstract
BACKGROUND Chronic migraine (CM) is a serious neurological disorder. Central sensitization is one of the important pathophysiological mechanisms underlying CM, and microglia-induced neuroinflammation conduces to central sensitization. Triggering receptor expressed on myeloid cells 2 (TREM2) is presented solely in microglia residing within the central nervous system and plays a key role in neuroinflammation. Metformin has been shown to regulate inflammatory responses and exert analgesic effects, but its relationship with CM remains unclear. In the study, we investigated whether metformin modulates TREM2 to improve central sensitization of CM and clarified the potential molecular mechanisms. METHODS A CM mouse model was induced by administration of nitroglycerin (NTG). Behavioral evaluations were conducted using von Frey filaments and hot plate experiments. Western blot and immunofluorescence techniques were employed to investigate the molecular mechanisms. Metformin and the SYK inhibitor R406 were administered to mice to assess their regulatory effects on neuroinflammation and central sensitization. To explore the role of TREM2-SYK in regulating neuroinflammation with metformin, a lentivirus encoding TREM2 was injected into the trigeminal nucleus caudalis (TNC). In vitro experiments were conducted to evaluate the regulation of TREM2-SYK by metformin, involving interventions with LPS, metformin, R406, siTREM2, and TREM2 plasmids. RESULTS Metformin and R406 pretreatment can effectively improve hyperalgesia in CM mice. Both metformin and R406 significantly inhibit c-fos and CGRP expression in CM mice, effectively suppressing the activation of microglia and NLRP3 inflammasome induced by NTG. With the administration of NTG, TREM2 expression gradually increased in TNC microglia. Additionally, we observed that metformin significantly inhibits TREM2 and SYK expression in CM mice. Lv-TREM2 attenuated metformin-mediated anti-inflammatory responses. In vitro experiments, knockdown of TREM2 inhibited LPS-induced SYK pathway activation and alleviated inflammatory responses. After the sole overexpression of TREM2, the SYK signaling pathway is activated, resulting in the activation of the NLRP3 inflammasome and an increased expression of pro-inflammatory cytokines; nevertheless, this consequence can be reversed by R406. The overexpression of TREM2 attenuates the inhibition of SYK activity mediated by metformin, and this effect can be reversed by R406. CONCLUSIONS Our findings suggest that metformin attenuates central sensitization in CM by regulating the activation of microglia and NLRP3 inflammasome through the TREM2-SYK pathway.
Collapse
Affiliation(s)
- Zhenzhen Fan
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, 730000, China
| | - Dandan Su
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, 730000, China
| | - Zi Chao Li
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, 730000, China
| | - Songtang Sun
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, China.
| | - Zhaoming Ge
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, 730000, China.
- Gansu Provincial Neurology Clinical Medical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China.
- Expert Workstation of Academician Wang Longde, Lanzhou University Second Hospital, Lanzhou, 730000, China.
| |
Collapse
|
2
|
Leinung N, Mentrup T, Hodzic S, Schröder B. Molecular and functional in vivo characterisation of murine Dectin-1 isoforms. Eur J Immunol 2024; 54:e2451092. [PMID: 39194380 DOI: 10.1002/eji.202451092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 08/29/2024]
Abstract
Dectin-1 is a C-type lectin-receptor involved in sensing fungi by innate immune cells. Encoded by the Clec7a gene, Dectin-1 exists in two major splice isoforms, Dectin-1a and 1b, which differ in the presence of a membrane-proximal stalk domain. As reported previously, this domain determines degradative routes for Dectin-1a and 1b leading to the generation of a stable N-terminal fragment exclusively from Dectin-1a. Here, we narrow down the responsible part of the stalk and demonstrate the stabilisation of the Dectin-1a N-terminal fragment in tetraspanin-enriched microdomains. C57BL/6 and BALB/c mice show divergent Dectin-1 isoform expression patterns, which are caused by a single nucleotide polymorphism in exon 3 of the Clec7a gene, leading to a non-sense Dectin-1a mRNA in C57BL/6 mice. Using backcrossing, we generated mice with the C57BL/6 Clec7a allele on a BALB/c background and compared these to the parental strains. Expression of the C57BL/6 allele leads to the exclusive presence of the Dectin-1b protein. Furthermore, it was associated with higher Dectin-1 mRNA expression, but less Dectin-1 at the cell surface according to flow cytometry. In neutrophils, this altered ROS production induced by Dectin-1 model ligands, while cellular responses in macrophages and dendritic cells were not significantly influenced by the Dectin-1 isoform pattern.
Collapse
Affiliation(s)
- Nadja Leinung
- Institute of Physiological Chemistry, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Torben Mentrup
- Institute of Physiological Chemistry, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Sajma Hodzic
- Institute of Physiological Chemistry, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Bernd Schröder
- Institute of Physiological Chemistry, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
3
|
Zhao Y, Wang L, Zhang X, Zhang L, Wei F, Li S, Li Y. Identification of neutrophil extracellular traps genes as potential biomarkers in psoriasis based on bioinformatics analysis. Sci Rep 2024; 14:23848. [PMID: 39394253 PMCID: PMC11470069 DOI: 10.1038/s41598-024-75069-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: 12/31/2023] [Accepted: 10/01/2024] [Indexed: 10/13/2024] Open
Abstract
The epidermal infiltration of neutrophils is a hallmark of psoriasis (PSO) and its activation leads to the release of neutrophil extracellular traps (NETs). However, the molecular mechanism of NETs-related genes (NETRGs) has not been extensively studied in PSO. To define NETs-related-biomarkers for PSO. The GSE13355 and GSE78097 datasets, and NETRGs gene set were included in this study. The datasets used in this study were all microarray data. The weighted gene co-expression network analysis (WGCNA) and machine learning algorithms were used to mine key genes. Later on, single-gene gene set enrichment analysis (GSEA) and immune infiltration analysis were implemented. Finally, the expression of key genes was verified using quantitative real-time fluorescence PCR (qRT-PCR). A total of 3 key genes (S100A9, CLEC7A, and CXCR4) were derived, and they all had excellent diagnostic performance. The single-gene GSEA enrichment results indicated that the key genes were mainly enriched in the chemokine signaling pathway and humoral immune response in the high-expression group, while focal adhesion was enriched in the low-expression group. The correlation analysis indicated that all key genes were strongly negatively correlated with resting mast cells and TGF-β family member receptor, while they were strongly positively correlated with activated CD4 memory T cells and antigen processing and presentation. Lastly, the experimental results showed that the expression trends of key genes were consistent with public database. In this study, we successfully screened three potential PSO diagnostic genes (S100A9, CLEC7A and CXCR4) that were closely related to NETs, and these findings not only provided new molecular marker candidates for the precise diagnosis of PSO patients, but also revealed possible future therapeutic targets. However, further in-depth research and validation were necessary.
Collapse
Affiliation(s)
- Yike Zhao
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereology in Hebei Province, Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Ling Wang
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereology in Hebei Province, Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Xiaoguang Zhang
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereology in Hebei Province, Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Lihua Zhang
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereology in Hebei Province, Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Feng Wei
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereology in Hebei Province, Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Suyue Li
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereology in Hebei Province, Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Yanling Li
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereology in Hebei Province, Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.
| |
Collapse
|
4
|
Wang H, Wang H, Zheng W, Wang D, Sun C, Dong J, Yu W, Du Q. OTULIN's influence on neuroinflammation and pain modulation in trigeminal neuralgia. CNS Neurosci Ther 2024; 30:e70006. [PMID: 39169794 PMCID: PMC11339468 DOI: 10.1111/cns.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/28/2024] [Accepted: 07/29/2024] [Indexed: 08/23/2024] Open
Abstract
INTRODUCTION Trigeminal neuralgia (TN), marked by chronic pain from neural damage, is closely associated with inflammation. The role of OTULIN, a key regulator in inflammation and autophagy, is not fully understood in TN. The regulatory mechanism of OTULIN, a key protein involved in modulating inflammatory responses and autophagy processes, remains incompletely elucidated, particularly in the context of TN and neuroinflammation. METHODS An infraorbital nerve ligation-induced rat model of TN was used. OTULIN's expression was modulated using adenovirus vectors and short hairpin RNA. The impact on pain and inflammatory responses was assessed via quantitative real-time polymerase chain reaction, western blot, immunofluorescence, and transcriptomic analysis. RESULTS Enhanced OTULIN expression significantly increased head withdrawal thresholds and reduced pain sensitivity and neuroinflammatory markers in the model. Conversely, silencing OTULIN exacerbated pain and inflammation. Transcriptomic data revealed OTULINs influence on both inflammatory and autophagy pathways, specifically in suppressing NLR family pyrin domain containing 3 (NLRP3) inflammasome and promoting autophagy. In vitro experiments demonstrated OTULIN's inhibition of inflammatory markers in microglia and neurons. CONCLUSION OTULIN is crucial in modulating TN, reducing neuropathic pain and neuroinflammation by activating the autophagy pathway and inhibiting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Haiyang Wang
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, School of MedicineWestlake UniversityHangzhouChina
- Department of NeurosurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Heng Wang
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, School of MedicineWestlake UniversityHangzhouChina
| | - Wenhao Zheng
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, School of MedicineWestlake UniversityHangzhouChina
| | - Ding Wang
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, School of MedicineWestlake UniversityHangzhouChina
| | - Chenglong Sun
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, School of MedicineWestlake UniversityHangzhouChina
| | - Jun Dong
- Department of NeurosurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Wenhua Yu
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, School of MedicineWestlake UniversityHangzhouChina
| | - Quan Du
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, School of MedicineWestlake UniversityHangzhouChina
| |
Collapse
|
5
|
Reynders A, Anissa Jhumka Z, Gaillard S, Mantilleri A, Malapert P, Magalon K, Etzerodt A, Salio C, Ugolini S, Castets F, Saurin AJ, Serino M, Hoeffel G, Moqrich A. Gut microbiota promotes pain chronicity in Myosin1A deficient male mice. Brain Behav Immun 2024; 119:750-766. [PMID: 38710336 DOI: 10.1016/j.bbi.2024.05.010] [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: 08/11/2023] [Revised: 04/23/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024] Open
Abstract
Chronic pain is a heavily debilitating condition and a huge socio-economic burden, with no efficient treatment. Over the past decade, the gut microbiota has emerged as an important regulator of nervous system's health and disease states. Yet, its contribution to the pathogenesis of chronic somatic pain remains poorly documented. Here, we report that male but not female mice lacking Myosin1a (KO) raised under single genotype housing conditions (KO-SGH) are predisposed to develop chronic pain in response to a peripheral tissue injury. We further underscore the potential of MYO1A loss-of-function to alter the composition of the gut microbiota and uncover a functional connection between the vulnerability to chronic pain and the dysbiotic gut microbiota of KO-SGH males. As such, parental antibiotic treatment modifies gut microbiota composition and completely rescues the injury-induced pain chronicity in male KO-SGH offspring. Furthermore, in KO-SGH males, this dysbiosis is accompanied by a transcriptomic activation signature in the dorsal root ganglia (DRG) macrophage compartment, in response to tissue injury. We identify CD206+CD163- and CD206+CD163+ as the main subsets of DRG resident macrophages and show that both are long-lived and self-maintained and exhibit the capacity to monitor the vasculature. Consistently, in vivo depletion of DRG macrophages rescues KO-SGH males from injury-induced chronic pain underscoring a deleterious role for DRG macrophages in a Myo1a-loss-of function context. Together, our findings reveal gene-sex-microbiota interactions in determining the predisposition to injury-induced chronic pain and point-out DRG macrophages as potential effector cells.
Collapse
Affiliation(s)
- Ana Reynders
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, Marseille, France.
| | - Z Anissa Jhumka
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, Marseille, France
| | | | - Annabelle Mantilleri
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, Marseille, France
| | - Pascale Malapert
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, Marseille, France
| | - Karine Magalon
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, Marseille, France
| | - Anders Etzerodt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Chiara Salio
- Department of Veterinary Sciences, University of Turin, Grugliasco, TO, Italy
| | - Sophie Ugolini
- Aix-Marseille-Université, CNRS, INSER, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Francis Castets
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, Marseille, France
| | - Andrew J Saurin
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, Marseille, France
| | - Matteo Serino
- Institut de Recherche en Santé Digestive, Université de Toulouse-Paul Sabatier, INSERM, INRAe, ENVT, UPS, Toulouse, France
| | - Guillaume Hoeffel
- Aix-Marseille-Université, CNRS, INSER, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Aziz Moqrich
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, Marseille, France.
| |
Collapse
|
6
|
Zhao Y, Li Q, Niu J, Guo E, Zhao C, Zhang J, Liu X, Wang L, Rao L, Chen X, Yang K. Neutrophil Membrane-Camouflaged Polyprodrug Nanomedicine for Inflammation Suppression in Ischemic Stroke Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311803. [PMID: 38519052 DOI: 10.1002/adma.202311803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/17/2024] [Indexed: 03/24/2024]
Abstract
Neuroinflammation has emerged as a major concern in ischemic stroke therapy because it exacebates neurological dysfunction and suppresses neurological recovery after ischemia/reperfusion. Fingolimod hydrochloride (FTY720) is an FDA-approved anti-inflammatory drug which exhibits potential neuroprotective effects in ischemic brain parenchyma. However, delivering a sufficient amount of FTY720 through the blood-brain barrier into brain lesions without inducing severe cardiovascular side effects remains challenging. Here, a neutrophil membrane-camouflaged polyprodrug nanomedicine that can migrate into ischemic brain tissues and in situ release FTY720 in response to elevated levels of reactive oxygen species. This nanomedicine delivers 15.2-fold more FTY720 into the ischemic brain and significantly reduces the risk of cardiotoxicity and infection compared with intravenously administered free drug. In addition, single-cell RNA-sequencing analysis identifies that the nanomedicine attenuates poststroke inflammation by reprogramming microglia toward anti-inflammatory phenotypes, which is realized via modulating Cebpb-regulated activation of NLRP3 inflammasomes and secretion of CXCL2 chemokine. This study offers new insights into the design and fabrication of polyprodrug nanomedicines for effective suppression of inflammation in ischemic stroke therapy.
Collapse
Affiliation(s)
- Ya Zhao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, 150080, P. R. China
| | - Qian Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Jingyan Niu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Erliang Guo
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, P. R. China
| | - Chenchen Zhao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, P. R. China
| | - Jian Zhang
- Biofunctional Experiment Teaching Center, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Xue Liu
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Lihua Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, P. R. China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Kuikun Yang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, 150080, P. R. China
| |
Collapse
|
7
|
Yao Q, Long C, Yi P, Zhang G, Wan W, Rao X, Ying J, Liang W, Hua F. C/EBPβ: A transcription factor associated with the irreversible progression of Alzheimer's disease. CNS Neurosci Ther 2024; 30:e14721. [PMID: 38644578 PMCID: PMC11033503 DOI: 10.1111/cns.14721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 03/20/2024] [Accepted: 03/27/2024] [Indexed: 04/23/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder distinguished by a swift cognitive deterioration accompanied by distinctive pathological hallmarks such as extracellular Aβ (β-amyloid) peptides, neuronal neurofibrillary tangles (NFTs), sustained neuroinflammation, and synaptic degeneration. The elevated frequency of AD cases and its proclivity to manifest at a younger age present a pressing challenge in the quest for novel therapeutic interventions. Numerous investigations have substantiated the involvement of C/EBPβ in the progression of AD pathology, thus indicating its potential as a therapeutic target for AD treatment. AIMS Several studies have demonstrated an elevation in the expression level of C/EBPβ among individuals afflicted with AD. Consequently, this review predominantly delves into the association between C/EBPβ expression and the pathological progression of Alzheimer's disease, elucidating its underlying molecular mechanism, and pointing out the possibility that C/EBPβ can be a new therapeutic target for AD. METHODS A systematic literature search was performed across multiple databases, including PubMed, Google Scholar, and so on, utilizing predetermined keywords and MeSH terms, without temporal constraints. The inclusion criteria encompassed diverse study designs, such as experimental, case-control, and cohort studies, restricted to publications in the English language, while conference abstracts and unpublished sources were excluded. RESULTS Overexpression of C/EBPβ exacerbates the pathological features of AD, primarily by promoting neuroinflammation and mediating the transcriptional regulation of key molecular pathways, including δ-secretase, apolipoprotein E4 (APOE4), acidic leucine-rich nuclear phosphoprotein-32A (ANP32A), transient receptor potential channel 1 (TRPC1), and Forkhead BoxO (FOXO). DISCUSSION The correlation between overexpression of C/EBPβ and the pathological development of AD, along with its molecular mechanisms, is evident. Investigating the pathways through which C/EBPβ regulates the development of AD reveals numerous multiple vicious cycle pathways exacerbating the pathological progression of the disease. Furthermore, the exacerbation of pathological progression due to C/EBPβ overexpression and its molecular mechanism is not limited to AD but also extends to other neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), and multiple sclerosis (MS). CONCLUSION The overexpression of C/EBPβ accelerates the irreversible progression of AD pathophysiology. Additionally, C/EBPβ plays a crucial role in mediating multiple pathways linked to AD pathology, some of which engender vicious cycles, leading to the establishment of feedback mechanisms. To sum up, targeting C/EBPβ could hold promise as a therapeutic strategy not only for AD but also for other degenerative diseases.
Collapse
Affiliation(s)
- Qing Yao
- Department of AnesthesiologyThe Second Affiliated Hospital of Nanchang UniversityNanchang CityJiangxi ProvinceChina
- Key Laboratory of Anesthesiology of Jiangxi ProvinceNanchang CityJiangxi ProvinceChina
| | - Chubing Long
- Department of AnesthesiologyThe Second Affiliated Hospital of Nanchang UniversityNanchang CityJiangxi ProvinceChina
- Key Laboratory of Anesthesiology of Jiangxi ProvinceNanchang CityJiangxi ProvinceChina
| | - Pengcheng Yi
- Department of AnesthesiologyThe Second Affiliated Hospital of Nanchang UniversityNanchang CityJiangxi ProvinceChina
- Key Laboratory of Anesthesiology of Jiangxi ProvinceNanchang CityJiangxi ProvinceChina
| | - Guangyong Zhang
- Department of AnesthesiologyThe Second Affiliated Hospital of Nanchang UniversityNanchang CityJiangxi ProvinceChina
- Key Laboratory of Anesthesiology of Jiangxi ProvinceNanchang CityJiangxi ProvinceChina
| | - Wei Wan
- Department of AnesthesiologyThe Second Affiliated Hospital of Nanchang UniversityNanchang CityJiangxi ProvinceChina
- Key Laboratory of Anesthesiology of Jiangxi ProvinceNanchang CityJiangxi ProvinceChina
| | - Xiuqin Rao
- Department of AnesthesiologyThe Second Affiliated Hospital of Nanchang UniversityNanchang CityJiangxi ProvinceChina
- Key Laboratory of Anesthesiology of Jiangxi ProvinceNanchang CityJiangxi ProvinceChina
| | - Jun Ying
- Department of AnesthesiologyThe Second Affiliated Hospital of Nanchang UniversityNanchang CityJiangxi ProvinceChina
- Key Laboratory of Anesthesiology of Jiangxi ProvinceNanchang CityJiangxi ProvinceChina
| | - Weidong Liang
- Department of AnesthesiologyThe First Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxi ProvinceChina
| | - Fuzhou Hua
- Department of AnesthesiologyThe Second Affiliated Hospital of Nanchang UniversityNanchang CityJiangxi ProvinceChina
- Key Laboratory of Anesthesiology of Jiangxi ProvinceNanchang CityJiangxi ProvinceChina
| |
Collapse
|
8
|
Cui X, Liu J, Uniyal A, Xu Q, Zhang C, Zhu G, Yang F, Sivanesan E, Linderoth B, Raja SN, Guan Y. Enhancing spinal cord stimulation-induced pain inhibition by augmenting endogenous adenosine signalling after nerve injury in rats. Br J Anaesth 2024; 132:746-757. [PMID: 38310069 PMCID: PMC10925891 DOI: 10.1016/j.bja.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND The mechanisms for spinal cord stimulation (SCS) to alleviate chronic pain are only partially known. We aimed to elucidate the roles of adenosine A1 and A3 receptors (A1R, A3R) in the inhibition of spinal nociceptive transmission by SCS, and further explored whether 2'-deoxycoformycin (dCF), an inhibitor of adenosine deaminase, can potentiate SCS-induced analgesia. METHODS We used RNAscope and immunoblotting to examine the distributions of adora1 and adora3 expression, and levels of A1R and A3R proteins in the spinal cord of rats after tibial-spared nerve injury (SNI-t). Electrophysiology recording was conducted to examine how adenosine receptor antagonists, virus-mediated adora3 knockdown, and dCF affect SCS-induced inhibition of C-fibre-evoked spinal local field potential (C-LFP). RESULTS Adora1 was predominantly expressed in neurones, whereas adora3 is highly expressed in microglial cells in the rat spinal cord. Spinal application of antagonists (100 μl) of A1R (8-cyclopentyl-1,3-dipropylxanthine [DPCPX], 50 μM) and A3R (MRS1523, 200 nM) augmented C-LFP in SNI-t rats (DPCPX: 1.39 [0.18] vs vehicle: 0.98 [0.05], P=0.046; MRS1523: 1.21 [0.07] vs vehicle: 0.91 [0.03], P=0.002). Both drugs also blocked inhibition of C-LFP by SCS. Conversely, dCF (0.1 mM) enhanced SCS-induced C-LFP inhibition (dCF: 0.60 [0.04] vs vehicle: 0.85 [0.02], P<0.001). In the behaviour study, dCF (100 nmol 15 μl-1, intrathecal) also enhanced inhibition of mechanical hypersensitivity by SCS in SNI-t rats. CONCLUSIONS Spinal A1R and A3R signalling can exert tonic suppression and also contribute to SCS-induced inhibition of spinal nociceptive transmission after nerve injury. Inhibition of adenosine deaminase may represent a novel adjuvant pharmacotherapy to enhance SCS-induced analgesia.
Collapse
Affiliation(s)
- Xiang Cui
- Department of Anaesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Jing Liu
- Department of Anaesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Ankit Uniyal
- Department of Anaesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Qian Xu
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, MD, USA; Howard Hughes Medical Institute, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Chi Zhang
- Department of Anaesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Guangwu Zhu
- Department of Anaesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Fei Yang
- Department of Anaesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Eellan Sivanesan
- Department of Anaesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Bengt Linderoth
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Srinivasa N Raja
- Department of Anaesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Yun Guan
- Department of Anaesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA; Department of Neurological Surgery, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
9
|
Agarwal G, Roy A, Singh AA, Kumar H, Mandoli A, Srivastava A. BM-MSC-Loaded Graphene-Collagen Cryogels Ameliorate Neuroinflammation in a Rat Spinal Cord Injury Model. ACS APPLIED BIO MATERIALS 2024; 7:1478-1489. [PMID: 38354406 DOI: 10.1021/acsabm.3c00876] [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: 02/16/2024]
Abstract
A major obstacle to axonal regeneration following spinal cord injury (SCI) is neuroinflammation mediated by astrocytes and microglial cells. We previously demonstrated that graphene-based collagen hydrogels alone can decrease neuroinflammation in SCI. Their regenerative potential, however, is poorly understood and incomplete. Furthermore, stem cells have demonstrated both neuroprotective and regenerative properties in spinal cord regeneration, although there are constraints connected with the application of stem cell-based therapy. In this study, we have analyzed the regeneration capability of human bone marrow mesenchymal stem cell (BM-MSC)-loaded graphene-cross-linked collagen cryogels (Gr-Col) in a thoracic (T10-T11) hemisection model of SCI. Our study found that BM-MSC-loaded Gr-Col improves axonal regeneration, reduces neuroinflammation by decreasing astrocyte reactivity, and promotes M2 macrophage polarization. BM-MSC-loaded-Gr-Col demonstrated enhanced regenerative potential compared to Gr-Col and the injury group control. Next-generation sequencing (NGS) analysis revealed that BM-MSC-loaded-Gr-Col modulates the JAK2-STAT3 pathway, thus decreasing the reactive and scar-forming astrocyte phenotype. The decrease in neuroinflammation in the BM-MSC-loaded-Gr-Col group is attributed to the modulation of Notch/Rock and STAT5a/b and STAT6 signaling. Overall, Gene Set Enrichment Analysis suggests the promising role of BM-MSC-loaded-Gr-Col in promoting axonal regeneration after SCI by modulating molecular pathways such as the PI3/Akt pathway, focal adhesion kinase, and various inflammatory pathways.
Collapse
Affiliation(s)
- Gopal Agarwal
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Abhishek Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Abhishek A Singh
- Department of Molecular Biology, Radboud University, Postbus 9101, Nijmegen 6500 HB, The Netherlands
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Amit Mandoli
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Akshay Srivastava
- Department of Medical Device, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat 382355, India
| |
Collapse
|
10
|
Wu J, Jin M, Tran Q, Kim M, Kim SI, Shin J, Park H, Shin N, Kang H, Shin HJ, Lee SY, Cui SB, Lee CJ, Lee WH, Kim DW. Employing the sustained-release properties of poly(lactic-co-glycolic acid) nanoparticles to reveal a novel mechanism of sodium-hydrogen exchanger 1 in neuropathic pain. Transl Res 2024; 263:53-72. [PMID: 37678757 DOI: 10.1016/j.trsl.2023.09.003] [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: 05/15/2023] [Revised: 08/16/2023] [Accepted: 09/01/2023] [Indexed: 09/09/2023]
Abstract
Neuropathic pain is caused by injury or disease of the somatosensory system, and its course is usually chronic. Several studies have been dedicated to investigating neuropathic pain-related targets; however, little attention has been paid to the persistent alterations that these targets, some of which may be crucial to the pathophysiology of neuropathic pain. The present study aimed to identify potential targets that may play a crucial role in neuropathic pain and validate their long-term impact. Through bioinformatics analysis of RNA sequencing results, we identified Slc9a1 and validated the reduced expression of sodium-hydrogen exchanger 1 (NHE1), the protein that Slc9a1 encodes, in the spinal nerve ligation (SNL) model. Colocalization analysis revealed that NHE1 is primarily co-localized with vesicular glutamate transporter 2-positive neurons. In vitro experiments confirmed that poly(lactic-co-glycolic acid) nanoparticles loaded with siRNA successfully inhibited NHE1 in SH-SY5Y cells, lowered intracellular pH, and increased intracellular calcium concentrations. In vivo experiments showed that sustained suppression of spinal NHE1 expression by siRNA-loaded nanoparticles resulted in delayed hyperalgesia in naïve and SNL model rats, whereas amiloride-induced transient suppression of NHE1 expression yielded no significant changes in pain sensitivity. We identified Slc9a1, which encodes NHE1, as a key gene in neuropathic pain. Utilizing the sustained release properties of nanoparticles enabled us to elucidate the chronic role of decreased NHE1 expression, establishing its significance in the mechanisms of neuropathic pain.
Collapse
Affiliation(s)
- Junhua Wu
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Neurology, Yanji Hospital, Yanji, China
| | - Meiling Jin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Quangdon Tran
- Molecular Biology Laboratory, Department of Medical Laboratories, Hai Phong International Hospital, Hai Phong City, Vietnam
| | - Minwoo Kim
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Song I Kim
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Juhee Shin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Hyewon Park
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Nara Shin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Hyunji Kang
- Center for Cognition and Sociality, Life Science Cluster, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Hyo Jung Shin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Sun Yeul Lee
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anesthesia and Pain Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Song-Biao Cui
- Department of Neurology, Affiliated Hospital of Yanbian University, Yanji, China
| | - C Justin Lee
- Center for Cognition and Sociality, Life Science Cluster, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Won Hyung Lee
- Department of Anesthesia and Pain Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea.
| |
Collapse
|
11
|
Li L, Li T, Qu X, Sun G, Fu Q, Han G. Stress/cell death pathways, neuroinflammation, and neuropathic pain. Immunol Rev 2024; 321:33-51. [PMID: 37688390 DOI: 10.1111/imr.13275] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/14/2023] [Accepted: 08/27/2023] [Indexed: 09/10/2023]
Abstract
Neuropathic pain is a common and debilitating modality of chronic pain induced by a lesion or disease of the somatosensory nervous system. Albeit the elucidation of numerous pathophysiological mechanisms and the development of potential treatment compounds, safe and reliable therapies of neuropathic pain remain poor. Multiple stress/cell death pathways have been shown to be implicated in neuroinflammation during neuropathic pain. Here, we summarize the current knowledge of stress/cell death pathways and present an overview of the roles and molecular mechanisms of stress/cell death pathways in neuroinflammation during neuropathic pain, covering intrinsic and extrinsic apoptosis, autophagy, mitophagy, ferroptosis, pyroptosis, necroptosis, and phagoptosis. Small molecule compounds that modulate stress/cell death pathways in alleviating neuropathic pain are discussed mainly based on preclinical neuropathic pain models. These findings will contribute to in-depth understanding of the pathological processes during neuropathic pain as well as bridge the gap between basic and translational research to uncover new neuroprotective interventions.
Collapse
Affiliation(s)
- Lu Li
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xinyu Qu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Guangwei Sun
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qi Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Guang Han
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
12
|
Chen XY, Feng SN, Bao Y, Zhou YX, Ba F. Identification of Clec7a as the therapeutic target of rTMS in alleviating Parkinson's disease: targeting neuroinflammation. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166814. [PMID: 37495085 DOI: 10.1016/j.bbadis.2023.166814] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/25/2023] [Accepted: 07/20/2023] [Indexed: 07/28/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease. Repetitive transcranial magnetic stimulation (rTMS) is a therapeutic tool in PD. High-throughput sequencing was performed to screen potential therapeutic targets in unilaterally 6-hydroxydopamine (6-OHDA)-lesioned rats. The candidate gene, Clec7a, was screened out and validated. Clec7a is a pattern recognition receptor involved in neuroinflammation. The higher expression of Clec7a was observed in the substantia nigra (SN) and striatum of PD rats with dopaminergic neurons damage and was mainly localized in the microglial. Adeno-associated virus (AAV)-mediated specific knockdown of Clec7a in microglial alleviated 6-OHDA induced motor deficits and nigrostriatal dopaminergic neuron damage of rats, as evidenced by the increase of tyrosine hydroxylase (TH) -positive neurons in SN, as well as dopaminergic nerve fibers in the striatum. Clec7a knockdown restrained the neuroinflammation by suppressing inflammatory factors (IFN-γ, TNF-α, IL-1β, IL-18, and IL-6) release in SN, which might result from enhanced Arg-1 expression (M2 polarization) and defective inducible nitric oxide synthase (iNOS) expression (M1 polarization). The same phenomena were also observed in the LPS inflammatory rat model of PD. In vitro, α-synuclein fibrils induced upregulation of Clec7a expression and microglia polarization to a pro-inflammatory state of BV2 cells, leading to increased release of cytokines. However, Clec7a knockdown reversed those changes and induced a shift to an anti-inflammatory phenotype in BV2 cells. In conclusion, our study suggested that Clec7a was involved in PD pathogenesis, and its inhibition might protect rats from PD by depressing neuroinflammation through microglial polarization.
Collapse
Affiliation(s)
- Xue-Yun Chen
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Si-Ning Feng
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Yin Bao
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Yu-Xin Zhou
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Fang Ba
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China.
| |
Collapse
|
13
|
Wu D, Wang P, Zhao C, Su J, Zhang J, Ma W, Zhang Y, Xu H. Levo-tetrahydropalmatine ameliorates neuropathic pain by inhibiting the activation of the Clec7a-MAPK/NF-κB-NLRP3 inflammasome axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155075. [PMID: 37741158 DOI: 10.1016/j.phymed.2023.155075] [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: 11/20/2022] [Revised: 08/27/2023] [Accepted: 09/09/2023] [Indexed: 09/25/2023]
Abstract
BACKGROUND Because of the complex pathogenesis of neuropathic pain (NP), the therapeutic efficacy of existing drugs is not satisfactory. Accumulating studies have indicated that neuroinflammation may play a key role in NP onset and progression. Levo-tetrahydropalmatine (l-THP) has been extensively used for relieving chronic pain for decades. However, its potential mechanisms against NP have not yet been fully elucidated. PURPOSE Exploring and elucidating the therapeutic effect and pharmacological mechanism of l-THP in treating NP. METHODS RNA-seq and bioinformatics analyses were carried out to identify effective target profiling of I-THP in chronic constrictive injury (CCI) rats. The I-THP related hub targets and signaling pathways were obtained via bioinformatics analysis, then subjected to in-depth analyses through experiments in vivo. A gain-of-function study further confirmed the role of Clec7a in l-THP-mediated pain relief. Finally, the interaction between l-THP and Clec7a was verified through molecular docking and surface plasmon resonance (SPR). RESULTS l-THP treatment effectively alleviated mechanical and thermal allodynia in NP model rats. Functionally, the I-THP effective targets were mainly enriched in inflammatory response-related pathways. Furthermore, Clec7a-MAPK/NF-κB-NLRP3 inflammasome axis was selected as one of the potential pathways of l-THP against NP. Mechanically, l-THP markedly reduced CCI-induced Clec7a overexpression, significantly inhibited the Clec7a-triggered phosphorylation of MAPK and NF-κB-p65, and decreased the expression of pyroptosis-related protein NLRP3 and Caspase-1-p20. The analgesic effect of l-THP on NP was partly eliminated when transfecting the overexpression vector virus pLVSO5Clec7a. Importantly, molecular docking and SPR data revealed that l-THP directly binds with the Clec7a protein. CONCLUSION This study is the first to indicate that l-THP may exert an analgesic effect through inhibiting neuroinflammation via the Clec7a-MAPK/NF-κB-NLRP3 inflammasome axis, supporting the clinical utility of l-THP in NP therapy.
Collapse
Affiliation(s)
- Dan Wu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, PR China
| | - Ping Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, PR China
| | - Chunhui Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, PR China
| | - Jin Su
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, PR China
| | - Junhong Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, PR China
| | - Wangming Ma
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, PR China
| | - Yanqiong Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, PR China.
| | - Haiyu Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, PR China.
| |
Collapse
|
14
|
Li B, Guo J, Zhou X, Li W, Wang N, Cao R, Cui S. The emerging role of pyroptosis in neuropathic pain. Int Immunopharmacol 2023; 121:110562. [PMID: 37364324 DOI: 10.1016/j.intimp.2023.110562] [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: 05/15/2023] [Revised: 06/10/2023] [Accepted: 06/22/2023] [Indexed: 06/28/2023]
Abstract
Neuropathic pain caused by somatosensory system injuries is notoriously difficult to treat. Previous research has shown that neuroinflammation and cell death have been implicated in the pathophysiology of neuropathic pain. Pyroptosis is a form of programmed cell death associated with inflammatory processes, as it can enhance or sustain the inflammatory response by releasing pro-inflammatory cytokines. This review presents the current knowledge on pyroptosis and its underlying mechanisms, including the canonical and noncanonical pathways. Moreover, we discuss recent findings on the role of pyroptosis in neuropathic pain and its potential as a therapeutic target. In conclusion, this review highlights the potential significance of pyroptosis as a promising target for developing innovative therapies to treat neuropathic pain.
Collapse
Affiliation(s)
- Baolong Li
- Department of Hand and Foot Surgery, The Third Bethune Hospital of Jilin University, Changchun, China; Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China
| | - Jin Guo
- Department of Hand and Foot Surgery, The Third Bethune Hospital of Jilin University, Changchun, China; Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China
| | - Xiongyao Zhou
- Department of Hand and Foot Surgery, The Third Bethune Hospital of Jilin University, Changchun, China; Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China
| | - Weizhen Li
- Department of Hand and Foot Surgery, The Third Bethune Hospital of Jilin University, Changchun, China; Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China
| | - Ningning Wang
- Department of Hand and Foot Surgery, The Third Bethune Hospital of Jilin University, Changchun, China; Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China
| | - Rangjuan Cao
- Department of Hand and Foot Surgery, The Third Bethune Hospital of Jilin University, Changchun, China; Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China.
| | - Shusen Cui
- Department of Hand and Foot Surgery, The Third Bethune Hospital of Jilin University, Changchun, China; Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China.
| |
Collapse
|