1
|
Song R, Yin S, Wu J, Yan J. Neuronal regulated cell death in aging-related neurodegenerative diseases: key pathways and therapeutic potentials. Neural Regen Res 2025; 20:2245-2263. [PMID: 39104166 PMCID: PMC11759035 DOI: 10.4103/nrr.nrr-d-24-00025] [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: 01/08/2024] [Revised: 05/13/2024] [Accepted: 06/18/2024] [Indexed: 08/07/2024] Open
Abstract
Regulated cell death (such as apoptosis, necroptosis, pyroptosis, autophagy, cuproptosis, ferroptosis, disulfidptosis) involves complex signaling pathways and molecular effectors, and has been proven to be an important regulatory mechanism for regulating neuronal aging and death. However, excessive activation of regulated cell death may lead to the progression of aging-related diseases. This review summarizes recent advances in the understanding of seven forms of regulated cell death in age-related diseases. Notably, the newly identified ferroptosis and cuproptosis have been implicated in the risk of cognitive impairment and neurodegenerative diseases. These forms of cell death exacerbate disease progression by promoting inflammation, oxidative stress, and pathological protein aggregation. The review also provides an overview of key signaling pathways and crosstalk mechanisms among these regulated cell death forms, with a focus on ferroptosis, cuproptosis, and disulfidptosis. For instance, FDX1 directly induces cuproptosis by regulating copper ion valency and dihydrolipoamide S-acetyltransferase aggregation, while copper mediates glutathione peroxidase 4 degradation, enhancing ferroptosis sensitivity. Additionally, inhibiting the Xc- transport system to prevent ferroptosis can increase disulfide formation and shift the NADP + /NADPH ratio, transitioning ferroptosis to disulfidptosis. These insights help to uncover the potential connections among these novel regulated cell death forms and differentiate them from traditional regulated cell death mechanisms. In conclusion, identifying key targets and their crosstalk points among various regulated cell death pathways may aid in developing specific biomarkers to reverse the aging clock and treat age-related neurodegenerative conditions.
Collapse
Affiliation(s)
- Run Song
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Shiyi Yin
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Jiannan Wu
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Junqiang Yan
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| |
Collapse
|
2
|
Losarwar S, Pancholi B, Babu R, Garabadu D. Mitochondria-dependent innate immunity: A potential therapeutic target in Flavivirus infection. Int Immunopharmacol 2025; 154:114551. [PMID: 40158432 DOI: 10.1016/j.intimp.2025.114551] [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: 01/16/2025] [Revised: 03/10/2025] [Accepted: 03/22/2025] [Indexed: 04/02/2025]
Abstract
Mitochondria, known as the powerhouse of cells, play a crucial role in host innate immunity during flavivirus infections such as Dengue, Zika, West Nile, and Japanese Encephalitis Virus. Mitochondrial antiviral signaling protein (MAVS) resides on the outer mitochondrial membrane which is triggered by viral RNA recognition by RIG-I-like receptors (RLRs). This activation induces IRF3 and NF-κB signaling, resulting in type I interferon (IFN) production and antiviral responses. Upon flavivirus infection, mitochondrial stress and dysfunction may lead to the release of mitochondrial DNA (mtDNA) into the cytoplasm, which serves as a damage-associated molecular pattern (DAMP). Cytosolic mtDNA is sensed by cGAS (cyclic GMP-AMP synthase), leading to the activation of the STING (Stimulator of Interferon Genes) pathway to increase IFN production and expand inflammation. Flaviviral proteins control mitochondrial morphology by controlling mitochondrial fission (MF) and fusion (MFu), disrupting mitochondrial dynamics (MD) to inhibit MAVS signaling and immune evasion. Flaviviral proteins also cause oxidative stress, resulting in the overproduction of reactive oxygen species (ROS), which triggers NLRP3 inflammasome activation and amplifies inflammation. Additionally, flaviviruses drive metabolic reprogramming by shifting host cell metabolism from oxidative phosphorylation (OxPhos) to glycolysis and fatty acid synthesis, creating a pro-replicative environment that supports viral replication and persistence. Thus, the present review explores the complex interaction between MAVS, mtDNA, and the cGAS-STING pathway, which is key to the innate immune response against flavivirus infections. Understanding these mechanisms opens new avenues in therapeutic interventions in targeting mitochondrial pathways to enhance antiviral immunity and mitigate viral infection.
Collapse
Affiliation(s)
- Saurabh Losarwar
- Department of Pharmacology, Central University of Punjab, Bhatinda 151401, India
| | | | - Raja Babu
- Department of Pharmacology, Central University of Punjab, Bhatinda 151401, India
| | - Debapriya Garabadu
- Department of Pharmacology, Central University of Punjab, Bhatinda 151401, India.
| |
Collapse
|
3
|
Lu Y, Wang T, Yu B, Xia K, Guo J, Liu Y, Ma X, Zhang L, Zou J, Chen Z, Zhou J, Qiu T. Mechanism of action of the nucleotide-binding oligomerization domain-like receptor protein 3 inflammasome and its regulation in liver injury. Chin Med J (Engl) 2025; 138:1061-1071. [PMID: 39719693 DOI: 10.1097/cm9.0000000000003309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Indexed: 12/26/2024] Open
Abstract
ABSTRACT Nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) is a cytosolic pattern recognition receptor that recognizes multiple pathogen-associated molecular patterns and damage-associated molecular patterns. It is a cytoplasmic immune factor that responds to cellular stress signals, and it is usually activated after infection or inflammation, forming an NLRP3 inflammasome to protect the body. Aberrant NLRP3 inflammasome activation is reportedly associated with some inflammatory diseases and metabolic diseases. Recently, there have been mounting indications that NLRP3 inflammasomes play an important role in liver injuries caused by a variety of diseases, specifically hepatic ischemia/reperfusion injury, hepatitis, and liver failure. Herein, we summarize new research pertaining to NLRP3 inflammasomes in hepatic injury, hepatitis, and liver failure. The review addresses the potential mechanisms of action of the NLRP3 inflammasome, and its regulation in these liver diseases.
Collapse
Affiliation(s)
- Yifan Lu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Tianyu Wang
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Bo Yu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Kang Xia
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Jiayu Guo
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yiting Liu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Xiaoxiong Ma
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Long Zhang
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Jilin Zou
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Zhongbao Chen
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Jiangqiao Zhou
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Tao Qiu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| |
Collapse
|
4
|
Zhang C, Xiang Z, Yang P, Zhang L, Deng J, Liao X. Advances in Nano-Immunomodulatory Systems for the Treatment of Acute Kidney Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409190. [PMID: 40145715 PMCID: PMC12061249 DOI: 10.1002/advs.202409190] [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] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 01/26/2025] [Indexed: 03/28/2025]
Abstract
Acute kidney injury (AKI) occurs when there is an imbalance in the immune microenvironment, leading to ongoing and excessive inflammation. Numerous immunomodulatory therapies have been suggested for the treatment of AKI, the current immunomodulatory treatment delivery systems are suboptimal and lack efficiency. Given the lack of effective treatment, AKI can result in multi-organ dysfunction and even death, imposing a significant healthcare burden on both the family and society. This underscores the necessity for innovative treatment delivery systems, such as nanomaterials, to better control pathological inflammation, and ultimately enhance AKI treatment outcomes. Despite the modification of numerous immunomodulatory nanomaterials to target the AKI immune microenvironment with promising therapeutic results, the literature concerning their intersection is scarce. In this article, the pathophysiological processes of AKI are outlined, focusing on the immune microenvironment, discuss significant advances in the comprehension of AKI recovery, and describe the multifunctionality and suitability of nanomaterial-based immunomodulatory treatments in managing AKI. The main obstacles and potential opportunities in the swiftly advancing research field are also clarified.
Collapse
Affiliation(s)
- Chenli Zhang
- Department of NephrologyThe Second Affiliated HospitalChongqing Medical UniversityChongqing400016China
- Department of nephrologySecond People's Hospital of YibinYibin644000China
| | - Zeli Xiang
- Department of nephrologySecond People's Hospital of YibinYibin644000China
| | - Pengfei Yang
- Department of NephrologyThe Second Affiliated HospitalChongqing Medical UniversityChongqing400016China
| | - Ling Zhang
- Department of NephrologyThe Second Affiliated HospitalChongqing Medical UniversityChongqing400016China
| | - Jun Deng
- Department of NephrologyThe Second Affiliated HospitalChongqing Medical UniversityChongqing400016China
- Institute of Burn Research, Southwest HospitalState Key Lab of Trauma and Chemical PoisoningArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Xiaohui Liao
- Department of NephrologyThe Second Affiliated HospitalChongqing Medical UniversityChongqing400016China
| |
Collapse
|
5
|
Liu R, Zhao Y, Chen Y, Chen X, Yang G, Li H. NEK7 is an essential regulator in NLRP3 inflammasome assembly of common carp (Cyprinus carpio L.). Int J Biol Macromol 2025; 305:141190. [PMID: 39965690 DOI: 10.1016/j.ijbiomac.2025.141190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 01/28/2025] [Accepted: 02/15/2025] [Indexed: 02/20/2025]
Abstract
The NIMA-related kinase 7 (NEK7), a member of the Never in Mitosis Gene A (NIMA) kinase family, participates in the assembly of the NLRP3 inflammasome in mammalian. However, it is currently unclear that the functions of NEK7 in the activation and assembly of NLRP3 inflammasome in teleost. In this research, the cDNA sequence of NEK7 of common carp (CcNEK7) was cloned and its role in the assembly of CcNLRP3 inflammasome was investigated. CcNEK7 was conserved throughout evolution, with its amino acid sequence, three-dimensional structure, and subcellular localization being similar to those in mammals. qPCR detection showed that CcNEK7 had the highest expression levels in the spleen of healthy common carp and could respond to bacteria and virus infection. It was additionally discovered that CcNEK7 can interact with CcNLRP3 and promote the oligomerization of CcNLRP3 and CcASC. Additionally, CcNEK7 significantly increased the CcNLRP3-induced cytotoxicity and pyroptosis, suggesting that CcNEK7 may exerts a regulatory function in the assembly of the CcNLRP3 inflammasome. These results provide a foundation for further understanding the assembly and regulation mechanisms of the inflammasome in bony fish, and also provides a target and theoretical framework for preventing and controlling of various aquatic animal diseases.
Collapse
Affiliation(s)
- Rongrong Liu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan 250014, PR China
| | - Yue Zhao
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan 250014, PR China
| | - Yixin Chen
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan 250014, PR China
| | - Xinping Chen
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan 250014, PR China
| | - Guiwen Yang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan 250014, PR China
| | - Hua Li
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan 250014, PR China..
| |
Collapse
|
6
|
Zengeler KE, Hollis A, Deutsch TCJ, Samuels JD, Ennerfelt H, Moore KA, Steacy EJ, Sabapathy V, Sharma R, Patel MK, Lukens JR. Inflammasome signaling in astrocytes modulates hippocampal plasticity. Immunity 2025:S1074-7613(25)00170-0. [PMID: 40318630 DOI: 10.1016/j.immuni.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 12/13/2024] [Accepted: 04/08/2025] [Indexed: 05/07/2025]
Abstract
Emerging evidence indicates that a baseline level of controlled innate immune signaling is required to support proper brain function. However, little is known about the function of most innate immune pathways in homeostatic neurobiology. Here, we report a role for astrocyte-dependent inflammasome signaling in regulating hippocampal plasticity. Inflammasomes are multiprotein complexes that promote caspase-1-mediated interleukin (IL)-1 and IL-18 production in response to pathogens and tissue damage. We observed that inflammasome complex formation was regularly detected under homeostasis in hippocampal astrocytes and that its assembly is dynamically regulated in response to learning and regional activity. Conditional ablation of caspase-1 in astrocytes limited hyperexcitability in an acute seizure model and impacted hippocampal plasticity via modulation of synaptic protein density, neuronal activity, and perineuronal net coverage. Caspase-1 and IL-18 regulated hippocampal IL-33 production and related plasticity. These findings reveal a homeostatic function for astrocyte inflammasome activity in regulating hippocampal physiology in health and disease.
Collapse
Affiliation(s)
- Kristine E Zengeler
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA.
| | - Ava Hollis
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Tyler C J Deutsch
- Department of Anesthesiology, University of Virginia, Charlottesville, VA 22908, USA
| | - Joshua D Samuels
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Hannah Ennerfelt
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA 24304, USA
| | - Katelyn A Moore
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Eric J Steacy
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Vikram Sabapathy
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, VA 22908, USA
| | - Rahul Sharma
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, VA 22908, USA
| | - Manoj K Patel
- Department of Anesthesiology, University of Virginia, Charlottesville, VA 22908, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA
| | - John R Lukens
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
7
|
Luo C, Qin X, Xie X, Gao J, Wu Y, Liang W, Wu Z. Cross-platform analysis of atrial fibrillation scientific videos: using composite index and a basic assessment scale. Front Public Health 2025; 13:1507776. [PMID: 40352855 PMCID: PMC12061937 DOI: 10.3389/fpubh.2025.1507776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
Background Currently, video platforms were filled with many low-quality, uncensored scientific videos, and patients who utilize the Internet to gain knowledge about specific diseases are vulnerable to being misled and possibly delaying treatment as a result. Therefore, a large sample survey on the content quality and popularity of online scientific videos was of great significance for future targeted reforms. Objective This study utilized normalization data analysis methods and a basic assessment scale, providing a new aspect for future research across multiple platforms with large sample sizes and for the development of video content quality assessment scales. Methods This cross-sectional study analyzed a sample of 331 videos retrieved from YouTube, BiliBili, TikTok, and Douyin on June 13, 2024. In the analysis of atrial fibrillation scientific videos across four social media platforms, comprehensive metrics and a basic scoring scale revealed associations between platforms, creators, and the popularity and content quality of the videos. Data analysis employed principal component analysis, normalization data processing, non-parametric tests, paired t-tests, and negative binomial regression. Results Analysis of the user engagement data using a composite index revealed a significant difference in the popularity of videos from publishers with a medical background (z = -4.285, p < 0.001), no aforementioned findings were found among video platforms, however, except for the Bilibili platform. As for content quality, while the difference in the total number of videos between the two groups was almost 2-fold (229:102), the difference in qualified videos was only 1.47-fold (47:32), a ratio that was even more unbalanced among the top 30% of videos with the most popularity. Notably, the overall content quality of videos from publishers without a medical background was also significantly higher (z = -2.299, p = 0.02). Conclusion This analysis of atrial fibrillation information on multiple social media platforms found that people prefer videos from publishers with a medical background. However, it appeared that these publishers did not sufficiently create high-quality, suitable videos for the public, and the platforms seemed to lack a rigorous censorship system and policy support for high-quality content. Moreover, the normalized data processing method and the basic assessment scale that we attempted to use in this study provided new ideas for future large-sample surveys and content quality review.
Collapse
Affiliation(s)
- Chong Luo
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoli Qin
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoyu Xie
- Department of Anesthesia, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jie Gao
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Yuwei Wu
- West China School of Public Health, Sichuan University, Chengdu, Sichuan, China
| | - Weitao Liang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhong Wu
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
8
|
Lage SL, Bricker-Holt K, Rocco JM, Rupert A, Donovan FX, Abramzon YA, Chandrasekharappa SC, McNinch C, Cook L, Amaral EP, Rosenfeld G, Dalhuisen T, Eun A, Hoh R, Fehrman E, Martin JN, Deeks SG, Henrich TJ, Peluso MJ, Sereti I. Persistent immune dysregulation and metabolic alterations following SARS-CoV-2 infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.16.25325949. [PMID: 40321289 PMCID: PMC12047922 DOI: 10.1101/2025.04.16.25325949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
SARS-CoV-2 can cause a variety of post-acute sequelae including Long COVID19 (LC), a complex, multisystem disease characterized by a broad range of symptoms including fatigue, cognitive impairment, and post-exertional malaise. The pathogenesis of LC is incompletely understood. In this study, we performed comprehensive cellular and transcriptional immunometabolic profiling within a cohort that included SARS-CoV-2-naïve controls (NC, N=30) and individuals with prior COVID-19 (~4-months) who fully recovered (RC, N=38) or went on to experience Long COVID symptoms (N=58). Compared to the naïve controls, those with prior COVID-19 demonstrated profound metabolic and immune alterations at the proteomic, cellular, and epigenetic level. Specifically, there was an enrichment in immature monocytes with sustained inflammasome activation and oxidative stress, elevated arachidonic acid levels, decreased tryptophan, and variation in the frequency and phenotype of peripheral T-cells. Those with LC had increased CD8 T-cell senescence and a distinct transcriptional profile within CD4 and CD8 T-cells and monocytes by single cell RNA sequencing. Our findings support a profound and persistent immunometabolic dysfunction that follows SARS-CoV-2 which may form the pathophysiologic substrate for LC. Our findings suggest that trials of therapeutics that help restore immune and metabolic homeostasis may be warranted to prevent, reduce, or resolve LC symptoms.
Collapse
Affiliation(s)
- Silvia Lucena Lage
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, USA
| | - Katherine Bricker-Holt
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, USA
| | - Joseph M. Rocco
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, USA
| | - Adam Rupert
- AIDS Monitoring Laboratory, Frederick National Laboratory for Cancer Research; Frederick, USA
| | - Frank X. Donovan
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute; Bethesda, USA
| | - Yevgeniya A. Abramzon
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute; Bethesda, USA
| | | | - Colton McNinch
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
| | - Logan Cook
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, USA
| | - Eduardo Pinheiro Amaral
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, USA
| | - Gabriel Rosenfeld
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
| | - Thomas Dalhuisen
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Avery Eun
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Rebecca Hoh
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Emily Fehrman
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Jeffrey N. Martin
- Department of Epidemiology and Biostatistics, University of California, San Francisco; San Francisco, USA
| | - Steven G. Deeks
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Timothy J. Henrich
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Michael J. Peluso
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Irini Sereti
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, USA
| |
Collapse
|
9
|
Liu L, Wang Y, Wang X, Zhang G, Sha S, Zhou R, Du Y, Wu C, Chen L. Transient receptor potential vanilloid 4 blockage attenuates pyroptosis in hippocampus of mice following pilocarpine‑induced status epilepticus. Acta Neuropathol Commun 2025; 13:73. [PMID: 40205503 PMCID: PMC11983898 DOI: 10.1186/s40478-025-01990-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
Pyroptosis contributes to the neuronal damage that occurs during epilepsy. Calcium-activated neutral protease (calpain) dissociates cysteinyl aspartate specific proteinase-1 (caspase-1, cas-1) from the cytoskeleton, and the activated cas-1 is responsible for the production of N-terminus of gasdermin D (N-GSDMD), the final executor of pyroptosis. Blocking transient receptor potential vanilloid 4 (TRPV4) can reduce neuronal injury in temporal lobe epilepsy (TLE) model mice. This study investigated the role of TRPV4 in pyroptosis during TLE. In the hippocampus of pilocarpine-induced status epilepticus (PISE) mice, the ratio of inactive calpain 1 protein level to its total protein level (inactive/total calpain 1) significantly decreased, while the ratio of inactive calpain 2 protein level to its total protein level remained unchanged. The protein levels of NLRP3, cleaved cas-1 (c-cas-1), interleukin (IL)-1β, and N-GSDMD increased, with more GSDMD-immunofluorescence-positive (GSDMD+) cells and fewer surviving pyramidal neurons observed in the hippocampus of PISE mice. Calpain inhibition with MDL-28170 reversed these changes, except for the elevated NLRP3 levels. Inhibitors targeting NLRP3 (MCC950) and cas-1 (Ac-YVAD-cmk) blocked the increase in c-cas-1, IL-1β, and N-GSDMD levels in the hippocampus of PISE mice. TRPV4 inhibition via HC-067047 increased the inactive/total calpain 1 ratio, decreased NLRP3, c-cas-1, IL-1β, and N-GSDMD protein levels, reduced GSDMD+ cells number, and improved pyramidal neuron survival in the hippocampus of PISE mice. Conversely, TRPV4 activation with GSK1016790A decreased the inactive/total calpain 1 ratio, elevated NLRP3, c-cas-1, IL-1β, and N-GSDMD levels, and increased GSDMD+ cells number in the hippocampus. In the hippocampus of GSK1016790A-injected mice, the inactive/total calpain 1 ratio was increased by MDL-28170, and c-cas-1, IL-1β, and N-GSDMD protein levels were markedly attenuated by MDL-28170, MCC950, and Ac-YVAD-cmk, respectively. In conclusion, TRPV4 inhibition mitigates pyroptosis in PISE mice by downregulating the calpain 1-NLRP3/cas-1-GSDMD pathway, ultimately reducing neuronal damage.
Collapse
Affiliation(s)
- Lihan Liu
- Department of Physiology, Nanjing Medical University, No. 101, Longmian Ave, Nanjing, Jiangsu Province, 211166, P.R. China
| | - Yue Wang
- Department of Physiology, Nanjing Medical University, No. 101, Longmian Ave, Nanjing, Jiangsu Province, 211166, P.R. China
| | - Xiaolin Wang
- Department of Physiology, Nanjing Medical University, No. 101, Longmian Ave, Nanjing, Jiangsu Province, 211166, P.R. China
| | - Guowen Zhang
- Department of Physiology, Nanjing Medical University, No. 101, Longmian Ave, Nanjing, Jiangsu Province, 211166, P.R. China
| | - Sha Sha
- Department of Physiology, Nanjing Medical University, No. 101, Longmian Ave, Nanjing, Jiangsu Province, 211166, P.R. China
| | - Rong Zhou
- Department of Physiology, Nanjing Medical University, No. 101, Longmian Ave, Nanjing, Jiangsu Province, 211166, P.R. China
| | - Yimei Du
- Research Center of Ion Channelopathy, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430022, P.R. China
| | - Chunfeng Wu
- Department of Neurology, Children's Hospital of Nanjing Medical University, No.8, Jiangdong South Road, Nanjing, Jiangsu Province, 211166, P. R. China.
| | - Lei Chen
- Department of Physiology, Nanjing Medical University, No. 101, Longmian Ave, Nanjing, Jiangsu Province, 211166, P.R. China.
| |
Collapse
|
10
|
Cui Y, Cen Q, Feng J, Wei J, Wang L, Chang C, Pang R, Wang J, Zhang A. Sodium butyrate alleviates spinal cord injury via inhibition of NLRP3/Caspase-1/GSDMD-mediated pyroptosis. Metab Brain Dis 2025; 40:157. [PMID: 40126723 PMCID: PMC11933225 DOI: 10.1007/s11011-025-01589-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 03/18/2025] [Indexed: 03/26/2025]
Abstract
NOD-like receptor protein 3 (NLRP3)/cysteinyl aspartate-specific proteinase 1 (Caspase-1)/gasdermin D (GSDMD)-mediated pyroptosis is linked to spinal cord injury (SCI) pathogenesis. The levels of short-chain fatty acids (SCFAs), especially butyric acid, are significantly altered after SCI. Sodium butyrate (NaB) has anti-inflammatory effects on SCI; however, its effect on pyroptosis is unknown. The aim of this study was to determine the role of NaB in SCI functional recovery and its effect on NLRP3/Caspase-1/GSDMD-mediated pyroptosis. SCI model rats were established using aneurysm clips. After SCI, rats were administered NaB (300 mg/kg) via gavage. SCFAs in faeces were measured using gas chromatography-mass spectrometry. Motor function recovery was assessed using cylinder rearing and grooming tests. Histopathological analysis was performed using haematoxylin-eosin staining, transmission electron microscopy, and terminal deoxynucleotidyl transferase dUTP nick-end labelling. The expression of proteins associated with pyroptosis signalling pathways was analysed using enzyme-linked immunosorbent assay, western blotting, and immunohistochemistry. SCFAs levels, particularly butyric acid, significantly decreased after SCI. NaB treatment promoted forelimb motor function recovery and attenuated pathological SCI. NaB also decreased spinal pro-inflammatory factors (interleukin-18 and interleukin-1β) and downregulated pyroptosis-related proteins, including NLRP3, apoptosis-associated speck-like protein, Caspase-1, and GSDMD. NaB inhibits NLRP3/Caspase-1/GSDMD-mediated neuronal pyroptosis and inflammation, exerting protective and therapeutic effects in SCI, suggesting NaB as an effective SCI treatment.
Collapse
Affiliation(s)
- Yanru Cui
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Qiuyu Cen
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Jing Feng
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Juanfang Wei
- College of Physical Education and Health, Geely University of China, Chengdu, China
| | - Linjie Wang
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Cong Chang
- Chengdu Eighth People's Hospital (Geriatric Hospital of Chengdu Medical College), Chengdu, China
| | - Rizhao Pang
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China.
| | - Junyu Wang
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China.
| | - Anren Zhang
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China.
| |
Collapse
|
11
|
Walczyńska-Dragon K, Kurek-Górecka A, Fiegler-Rudol J, Nitecka-Buchta A, Baron S. The Therapeutic Potential of Cannabidiol in the Management of Temporomandibular Disorders and Orofacial Pain. Pharmaceutics 2025; 17:328. [PMID: 40142992 PMCID: PMC11945290 DOI: 10.3390/pharmaceutics17030328] [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: 02/08/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Temporomandibular disorders (TMDs) are a group of conditions affecting the temporomandibular joint (TMJ) and associated muscles, leading to pain, restricted jaw movement, and impaired quality of life. Conventional treatments, including physical therapy, medications, and surgical interventions, have varying degrees of success and potential side effects. Cannabidiol (CBD), a non-psychoactive component of cannabis, has gained attention for its anti-inflammatory, analgesic, and anxiolytic properties. This study explores the potential role of CBD in TMD management. Methods: A review of existing literature was conducted (2007-2024), focusing on preclinical and clinical studies assessing the efficacy of CBD in pain modulation, inflammation reduction, and muscle relaxation. Relevant studies were sourced from PubMed, Scopus, and Web of Science databases. Additionally, potential mechanisms of action, including interactions with the endocannabinoid system, were analyzed. Results: Studies suggest that CBD exerts analgesic and anti-inflammatory effects by modulating CB1 and CB2 receptors, reducing cytokine release, and influencing neurotransmitter pathways. Preliminary clinical evidence indicates that CBD may alleviate TMD-related pain and muscle tension with minimal adverse effects. However, high-quality randomized controlled trials are limited. Conclusions: CBD demonstrates promise as a potential adjunctive treatment for TMD. Further research, including well-designed clinical trials, is necessary to establish its efficacy, optimal dosage, and long-term safety.
Collapse
Affiliation(s)
- Karolina Walczyńska-Dragon
- Department of Temporomandibular Disorders, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland; (A.N.-B.); (S.B.)
| | - Anna Kurek-Górecka
- Department of Microbiology and Immunology, Faculty of Medical Sciences, Medical University of Silesia in Katowice, 41-808 Zabrze, Poland
| | - Jakub Fiegler-Rudol
- Student Scientific Society at the Department of Temporomandibular Disorders, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland;
| | - Aleksandra Nitecka-Buchta
- Department of Temporomandibular Disorders, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland; (A.N.-B.); (S.B.)
| | - Stefan Baron
- Department of Temporomandibular Disorders, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland; (A.N.-B.); (S.B.)
| |
Collapse
|
12
|
Wang Z, Sun X, Lin Y, Fu Y, Yi Z. Stealth in non-tuberculous mycobacteria: clever challengers to the immune system. Microbiol Res 2025; 292:128039. [PMID: 39752805 DOI: 10.1016/j.micres.2024.128039] [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: 08/07/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/19/2025]
Abstract
Non-tuberculous Mycobacteria (NTM) are found extensively in various environments, yet most are non-pathogenic. Only a limited number of these organisms can cause various infections, including those affecting the lungs, skin, and central nervous system, particularly when the host's autoimmune function is compromised. Among these, Non-tuberculous Mycobacteria Pulmonary Diseases (NTM-PD) are the most prevalent. Currently, there is a lack of effective treatments and preventive measures for NTM infections. This article aims to deepen the comprehension of the pathogenic mechanisms linked to NTM and to formulate new intervention strategies by synthesizing current research and detailing the different tactics used by NTM to avoid elimination by the host's immune response. These intricate mechanisms not only affect the innate immune response but also successfully oppose the adaptive immune response, establishing persistent infections within the host. This includes effects on the functions of macrophages, neutrophils, dendritic cells, and T lymphocytes, as well as modulation of cytokine production. The article particularly emphasizes the survival strategies of NTM within macrophages, such as inhibiting phagosome maturation and acidification, resisting intracellular killing mechanisms, and interfering with autophagy and cell death pathways. This review aims to deepen the understanding of NTM's immune evasion mechanisms, thereby facilitating efforts to inhibit its proliferation and spread within the host, ultimately providing new methods and strategies for NTM-related treatments.
Collapse
Affiliation(s)
- Zhenghao Wang
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Xiurong Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Yuli Lin
- School of Medical Laboratory, Shandong Second Medical University, Weifang 261053, China
| | - Yurong Fu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China.
| | - Zhengjun Yi
- School of Medical Laboratory, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
13
|
Chen K, Ying J, Zhu J, Chen L, Liu R, Jing M, Wang Y, Zhou K, Wu L, Wu C, Xiao J, Ni W. Urolithin A alleviates NLRP3 inflammasome activation and pyroptosis by promoting microglial mitophagy following spinal cord injury. Int Immunopharmacol 2025; 148:114057. [PMID: 39827665 DOI: 10.1016/j.intimp.2025.114057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/02/2025] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Spinal cord injury (SCI) is a potentially fatal condition that often results in loss of motor and sensory functions, thereby significantly burdening global health initiatives. Urolithin A (UA), an intestinal microbial metabolite of ellagic acid, is known for its potent anti-inflammatory properties in chronic inflammation contexts. UA treatment in humans induces a molecular signature of improved mitochondrial and cellular health. Yet, its effects on acute inflammation following SCI remain unclear. In this study, we developed an impact-induced mouse model for SCI and treated the injured mice with UA (50 mg/kg/d, till 8 weeks) via intragastric administration. Furthermore, we subjected BV2 cells to lipopolysaccharide and adenosine 5'-triphosphate to simulate the post-injury inflammatory response. Our results demonstrated that pre-treatment with UA (10 μM) effectively inhibited NLRP3 inflammasome activation in LPS-primed BV2 cells. This inhibition was evidenced by reduced cleaved Caspase-1 and mature IL-1β release, diminished ASC speck formation, and decreased gasdermin D (GSDMD)-mediated pyroptosis. Additionally, UA treatment restored mitochondrial activity and ROS production attenuated by NLRP3 activation, increased LC3-II expression, and enhanced LC3 co-localization with mitochondria. 3-Methyladenine (3-MA), an autophagy inhibitor, can partially reverse the stimulatory effect of UA on mitophagy, as well as the inhibitory effect of UA on pyroptosis. This study highlighted the protective role of UA against SCI through its promotion of mitophagy, which in turn inhibits NLRP3 inflammasome activation and pyroptosis.
Collapse
Affiliation(s)
- Kongbin Chen
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China
| | - Jiahao Ying
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China
| | - Jiangwei Zhu
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000 China
| | - Liang Chen
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China
| | - Rongjie Liu
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China
| | - Mengqi Jing
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000 China
| | - Yuchao Wang
- Department of Orthopedic, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116600, China
| | - Kailiang Zhou
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China
| | - Long Wu
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China.
| | - Chenyu Wu
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China; Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000 China.
| | - Jian Xiao
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China; Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000 China.
| | - Wenfei Ni
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China; Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000 China.
| |
Collapse
|
14
|
McCutcheon CR, Gaddy JA, Aronoff DM, Manning SD, Petroff MG. Group B Streptococcal Membrane Vesicles Induce Proinflammatory Cytokine Production and Are Sensed in an NLRP3 Inflammasome-Dependent Mechanism in a Human Macrophage-like Cell Line. ACS Infect Dis 2025; 11:453-462. [PMID: 39761308 PMCID: PMC11833861 DOI: 10.1021/acsinfecdis.4c00641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 02/20/2025]
Abstract
Group B Streptococcus (GBS) is a major cause of fetal and neonatal mortality worldwide. Many of the adverse effects of invasive GBS are associated with inflammation; therefore, understanding bacterial factors that promote inflammation is of critical importance. Membrane vesicles (MVs), which are produced by many bacteria, may modulate host inflammatory responses. While it is known that mice injected intra-amniotically with GBS MVs exhibit large-scale leukocyte infiltration, preterm birth, and subsequent fetal death, the immune effectors driving this response remain unclear. Here, we hypothesized that THP-1 macrophage-like cells respond to GBS-derived MVs by producing proinflammatory cytokines and are recognized through one or more pattern recognition receptors. We show that THP-1s produce high levels of neutrophil- and monocyte-specific chemokines in response to MVs derived from different clinical isolates of GBS. Using antibody microarrays and multiplex Luminex assays, we found that GBS MVs elicit significantly (p < 0.05) higher levels of CCL1, CCL2, CCL20, CXCL1, CXCL10, and IL-1β relative to untreated THP-1s. Using chemical inhibitors in combination with caspase-1 activity assays and Luminex assays, we further demonstrate that GBS MVs upregulated IL-1β production in a caspase-1 and NLRP3-dependent manner, ultimately identifying NLRP3 as a sensor of GBS MVs. These data indicate that MVs contain one or more pathogen-associated molecular patterns that can be sensed by the immune system and show that the NLRP3 inflammasome is a novel sensor of GBS MVs. Our data additionally indicate that MVs may serve as immune effectors that can be targeted for immunotherapeutics.
Collapse
Affiliation(s)
- Cole R. McCutcheon
- Department
of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Jennifer A. Gaddy
- Department
of Medicine, Division of Infectious Disease, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department
of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Tennessee
Valley Healthcare System, Department of
Veterans Affairs, Nashville, Tennessee 37212, United States
| | - David M. Aronoff
- Department
of Medicine, Indiana University School of
Medicine, Indianapolis, Indiana 46202, United States
| | - Shannon D. Manning
- Department
of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Margaret G. Petroff
- Department
of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Pathobiology and Diagnostic Investigation, Michigan State University, East
Lansing, Michigan 48824, United States
| |
Collapse
|
15
|
Rahman MA, Datta S, Lakkakula H, Koka S, Boini KM. Acid Sphingomyelinase and Ceramide Signaling Pathway Mediates Nicotine-Induced NLRP3 Inflammasome Activation and Podocyte Injury. Biomedicines 2025; 13:416. [PMID: 40002829 PMCID: PMC11852453 DOI: 10.3390/biomedicines13020416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/23/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Recent studies have shown that Nlrp3 inflammasome activation is importantly involved in podocyte dysfunction induced by nicotine. The present study was designed to test whether acid sphingomyelinase (Asm) and ceramide signaling play a role in mediating nicotine-induced Nlrp3 inflammasome activation and subsequent podocyte damage. Methods and Results: Nicotine treatment significantly increased the Asm expression and ceramide production compared to control cells. However, prior treatment with amitriptyline, an Asm inhibitor significantly attenuated the nicotine-induced Asm expression and ceramide production. Confocal microscopic and biochemical analyses showed that nicotine treatment increased the colocalization of NLRP3 with Asc, Nlrp3 vs. caspase-1, IL-1β production, caspase-1 activity, and desmin expression in podocytes compared to control cells. Pretreatment with amitriptyline abolished the nicotine-induced colocalization of NLRP3 with Asc, Nlrp3 with caspase-1, IL-1β production, caspase-1 activity and desmin expression. Immunofluorescence analyses showed that nicotine treatment significantly decreased the podocin expression compared to control cells. However, prior treatment with amitriptyline attenuated the nicotine-induced podocin reduction. In addition, nicotine treatment significantly increased the cell permeability, O2 production, and apoptosis compared to control cells. However, prior treatment with amitriptyline significantly attenuated the nicotine-induced cell permeability, O2 production and apoptosis in podocytes. Conclusions: Asm is one of the important mediators of nicotine-induced inflammasome activation and podocyte injury. Asm may be a therapeutic target for the treatment or prevention of glomerulosclerosis associated with smoking.
Collapse
Affiliation(s)
- Mohammad Atiqur Rahman
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd, Houston, TX 77204, USA (H.L.)
| | - Sayantap Datta
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd, Houston, TX 77204, USA (H.L.)
| | - Harini Lakkakula
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd, Houston, TX 77204, USA (H.L.)
- Novi High School, Novi, MI 48375, USA
| | - Saisudha Koka
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A & M University, Kingsville, TX 78363, USA
| | - Krishna M. Boini
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd, Houston, TX 77204, USA (H.L.)
| |
Collapse
|
16
|
Cai L, Fan Q, Pang R, Chen C, Zhang Y, Xie H, Huang J, Wang Y, Li P, Huang D, Jin X, Zhou Y, Li Y. Microglia programmed cell death in neurodegenerative diseases and CNS injury. Apoptosis 2025; 30:446-465. [PMID: 39656359 PMCID: PMC11799081 DOI: 10.1007/s10495-024-02041-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2024] [Indexed: 02/06/2025]
Abstract
Programmed cell death (PCD) has emerged as a critical regulatory mechanism in the initiation and progression of various pathological conditions. PCD in microglia, including necroptosis, pyroptosis, apoptosis, ferroptosis, and autophagy, occurs in a variety of central nervous system (CNS) diseases. Dysregulation of microglia can lead to excessive tissue damage or neuronal death in CNS injury. Various injury stimuli trigger aberrant activation of the PCD pathway of microglia, which then further leads to inflammatory cascades that exacerbates CNS pathology in a vicious cycle. Therefore, targeting PCD in microglia is considered an important avenue for the treatment of various neurodegenerative diseases and CNS injury. In this review, we summarize the major and recent findings focusing on the mechanisms of PCD in microglia modulating functions in neurodegenerative diseases and CNS injury and provide a systematic overview of the current inhibitors targeting various PCD pathways, which may provide important therapeutic targets that merit further investigation.
Collapse
Affiliation(s)
- Ling Cai
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiuyue Fan
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Pang
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Chen
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yueman Zhang
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyi Xie
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingyi Huang
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Wang
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Huang
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia Jin
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yuxi Zhou
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yan Li
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
17
|
Karmakar V, Chain M, Majie A, Ghosh A, Sengupta P, Dutta S, Mazumder PM, Gorain B. Targeting the NLRP3 inflammasome as a novel therapeutic target for osteoarthritis. Inflammopharmacology 2025; 33:461-484. [PMID: 39806051 DOI: 10.1007/s10787-024-01629-2] [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: 08/20/2024] [Accepted: 12/07/2024] [Indexed: 01/16/2025]
Abstract
Osteoarthritis, the most common arthritic condition, is an age-related progressive disease characterized by the loss of cartilage and synovial inflammation in the knees and hips. Development of pain, stiffness, and considerably restricted mobility of the joints are responsible for the production of matrix metalloproteinases and cytokines. Although several treatments are available for the management of this disease condition, they possess limitations at different levels. Recently, efforts have focused on regulating the production of the NLRP3 inflammasome, which plays a critical role in the disease's progression due to its dysregulation. Inhibition of NLRP3 inflammasome has shown the potential to modulate the production of MMP-13, caspase-1, IL-1β, etc., which has been reflected by positive responses in different preclinical and clinical studies. Aiming inhibition of this NLRP3 inflammasome, several compounds are in different stages of research owing to bring a novel agent for the treatment of osteoarthritis. This review summarizes the mechanistic pathways linking NLRP3 activation to osteoarthritis development and discusses the progress in new therapeutics aimed at effective treatment.
Collapse
Affiliation(s)
- Varnita Karmakar
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Mayukh Chain
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Ankit Majie
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Arya Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Pallav Sengupta
- Department of Biomedical Sciences, College of Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Sulagna Dutta
- Basic Medical Sciences Department, College of Medicine, Ajman University, Ajman, United Arab Emirates
| | - Papiya Mitra Mazumder
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Bapi Gorain
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India.
| |
Collapse
|
18
|
Lv G, Wang D, Huang Y, Shi R, Qin C, Chen X, Zeng X, Luo H, Yang P, Chen S, Wang J. High serum uric acid levels are associated with increased prevalence of gallstones in adult women: a cross-sectional study based on NHANES. Front Med (Lausanne) 2025; 12:1487974. [PMID: 39897593 PMCID: PMC11782260 DOI: 10.3389/fmed.2025.1487974] [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: 08/29/2024] [Accepted: 01/02/2025] [Indexed: 02/04/2025] Open
Abstract
Objective We investigated the association between serum uric acid (SUA) levels and gallstone (GS) prevalence in adult women. Methods Participants' information were taken from the United States National Health and Nutrition Examination Survey (2017-2020). Logistic regression analysis and dose-response curve were used to assess the association between SUA levels and the prevalence of GS in adult women. Subgroup analyses were performed to investigate associations between SUA levels and age, ethnicity, body mass index, hypertension, and diabetes. Results A total of 600 participants self-reported a history of GS. After adjusting for confounding, the prevalence of GS in adult women increased by 14% for every 1 mg/dL increase in SUA (odds ratio [OR]: 1.14, 95% confidence interval [CI]: 1.06, 1.22). Testing SUA as a categorical variable for sensitivity analyses indicated a 1.6-fold increase in the prevalence of GS in tertile 3 (OR=1.60, 95% CI: 1.25, 2.04) compared to tertile 1. Dose-response curves showed a nonlinear correlation between SUA levels and the prevalence of GS. Subgroup analyses indicated that SUA level was associated with an increased prevalence of GS in most subgroups, although subtle differences existed. Conclusion SUA was positively and non-linearly associated with the prevalence of GS in adult females. Despite the inability to clarify the causal relationship between them, our results remain interesting.
Collapse
Affiliation(s)
- Guozheng Lv
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Decai Wang
- Department of Urology, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
- NHC Key Laboratory of Nuclear Technology Medical Transformation, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Yu Huang
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Ruizi Shi
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Chuan Qin
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Xi Chen
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Xintao Zeng
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Hua Luo
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Pei Yang
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Sirui Chen
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Jianjun Wang
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
- NHC Key Laboratory of Nuclear Technology Medical Transformation, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| |
Collapse
|
19
|
Jia C, Zhang M, Wu X, Zhang X, Lv Z, Zhao K, Zhang J, Su Y, Zhu F. HERV-W Env Induces Neuron Pyroptosis via the NLRP3-CASP1-GSDMD Pathway in Recent-Onset Schizophrenia. Int J Mol Sci 2025; 26:520. [PMID: 39859234 PMCID: PMC11765033 DOI: 10.3390/ijms26020520] [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: 11/29/2024] [Revised: 01/01/2025] [Accepted: 01/05/2025] [Indexed: 01/30/2025] Open
Abstract
HERVs (Human endogenous retroviruses) are remnants of ancient exogenous retroviruses that have integrated into the human genome, particularly in germ-line cells. Among these, the envelope protein gene HERV-W env (Human endogenous retroviruses W family envelope protein), located on chromosome 7 and primarily expressed in the human placenta, has been closely linked to various neuropsychiatric disorders, including schizophrenia, as well as autoimmune diseases and cancer. Recent studies have highlighted the abnormal expression of cytokines as a key factor in the pathophysiology of schizophrenia. Notably, elevated serum levels of IL-1β (interleukin 1 beta) in schizophrenia, a cytokine associated with inflammation, are a characteristic feature of pyroptosis-a form of pro-inflammatory programmed cell death. Although previous research has observed significant upregulation of pyroptosis-related genes such as CASP1 (Caspase-1), NLRP3 (NLR family pyrin domain containing 3), and IL1B (interleukin 1 beta) in the serum of schizophrenia patients, and extensive neuron pyroptosis has been documented in various neuropsychiatric disorders, including Alzheimer's disease, epilepsy, and multiple sclerosis, the occurrence of neuron pyroptosis in schizophrenia remains uncertain. Furthermore, the mechanisms underlying pyroptosis in schizophrenia and its potential connection with HERV-W env have yet to be fully elucidated. In this study, we found that the expression levels of pyroptosis-related genes, specifically CASP1, GSDMD (Gasdermin D), and IL1B, were significantly elevated in patients with schizophrenia compared to healthy controls. Furthermore, our analysis revealed a strong positive correlation between HERV-W env expression and the levels of CASP1/GSDMD/IL1B in these patients. Experimental evidence further demonstrated that HERV-W env promoted the activation of Caspase-1 and the cleavage of Gasdermin D, leading to increased release of LDH (lactate dehydrogenase) and IL-1β. Importantly, inhibitors targeting NLRP3, CASP1, and GSDMD significantly reduced the releases of LDH and IL-1β induced by HERV-W env, whereas BID (BH3 interacting domain death agonist) inhibitors did not have a notable effect. This suggests that HERV-W env induces CASP1-GSDMD-dependent pyroptosis through the NLRP3-CASP1-GSDMD signaling pathway. As pyroptosis is increasingly recognized for its connection to neurodegenerative diseases, this study provides insights into the molecular mechanisms of neuronal pyroptosis mediated by the NLRP3 inflammasome in the context of HERV-W env. Additionally, it explores the potential facilitation of HERV-W env in the development of schizophrenia via pyroptosis, proposing that certain pyroptosis indicators could serve as potential biomarkers for schizophrenia. Based on our existing research results and the findings of previous researchers, we infer that HERV-W env acts as a bridge in the onset and progression of schizophrenia. Furthermore, HERV-W env may serve as a potential target for the clinical treatment of schizophrenia, suggesting that monoclonal antibody therapy targeting HERV-W env could represent a novel approach to managing this disease.
Collapse
Affiliation(s)
- Chen Jia
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Mengqi Zhang
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xiulin Wu
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xu Zhang
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Zhao Lv
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Kexin Zhao
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Jiahang Zhang
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yaru Su
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Fan Zhu
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan 430071, China
| |
Collapse
|
20
|
Platt N, Shepherd D, Smith DA, Smith C, Wallom KL, Luqmani R, Churchill GC, Galione A, Platt FM. Dysregulation of the NLRP3 Inflammasome and Promotion of Disease by IL-1β in a Murine Model of Sandhoff Disease. Cells 2025; 14:35. [PMID: 39791736 PMCID: PMC11720672 DOI: 10.3390/cells14010035] [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: 11/14/2024] [Revised: 12/12/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
Sandhoff disease (SD) is a progressive neurodegenerative lysosomal storage disorder characterized by GM2 ganglioside accumulation as a result of mutations in the HEXB gene, which encodes the β-subunit of the enzyme β-hexosaminidase. Lysosomal storage of GM2 triggers inflammation in the CNS and periphery. The NLRP3 inflammasome is an important coordinator of pro-inflammatory responses, and we have investigated its regulation in murine SD. The NLRP3 inflammasome requires two signals, lipopolysaccharide (LPS) and ATP, to prime and activate the complex, respectively, leading to IL-1β secretion. Peritoneal, but not bone-marrow-derived, macrophages from symptomatic SD mice, but not those from pre-symptomatic animals, secrete the cytokine following priming with LPS with no requirement for activation with ATP, suggesting that such NLRP3 deregulation is related to the extent of glycosphingolipid storage. Dysregulated production of IL-1β was dependent upon caspase activity but not cathepsin B. We investigated the role of IL-1β in SD pathology using two approaches: the creation of hexb-/-Il1r1-/- double knockout mice or by treating hexb-/- animals with anakinra, a recombinant form of the IL-1 receptor antagonist, IL-1Ra. Both resulted in modest but significant extensions in lifespan and improvement of neurological function. These data demonstrate that IL-1β actively participates in the disease process and provides proof-of-principle that blockade of the pro-inflammatory cytokine IL-1β may provide benefits to patients.
Collapse
Affiliation(s)
- Nick Platt
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK; (N.P.); (D.S.); (D.A.S.); (C.S.); (K.-L.W.); (G.C.C.); (A.G.)
| | - Dawn Shepherd
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK; (N.P.); (D.S.); (D.A.S.); (C.S.); (K.-L.W.); (G.C.C.); (A.G.)
| | - David A. Smith
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK; (N.P.); (D.S.); (D.A.S.); (C.S.); (K.-L.W.); (G.C.C.); (A.G.)
| | - Claire Smith
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK; (N.P.); (D.S.); (D.A.S.); (C.S.); (K.-L.W.); (G.C.C.); (A.G.)
| | - Kerri-Lee Wallom
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK; (N.P.); (D.S.); (D.A.S.); (C.S.); (K.-L.W.); (G.C.C.); (A.G.)
| | - Raashid Luqmani
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Windmill Road, Oxford OX3 7LD, UK;
| | - Grant C. Churchill
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK; (N.P.); (D.S.); (D.A.S.); (C.S.); (K.-L.W.); (G.C.C.); (A.G.)
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK; (N.P.); (D.S.); (D.A.S.); (C.S.); (K.-L.W.); (G.C.C.); (A.G.)
| | - Frances M. Platt
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK; (N.P.); (D.S.); (D.A.S.); (C.S.); (K.-L.W.); (G.C.C.); (A.G.)
| |
Collapse
|
21
|
Hollis A, Lukens JR. Role of inflammasomes and neuroinflammation in epilepsy. Immunol Rev 2025; 329:e13421. [PMID: 39523682 PMCID: PMC11744240 DOI: 10.1111/imr.13421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Epilepsy is a brain disorder characterized by recurrent seizures, which are brief episodes of abnormal electrical activity in the brain and involuntary movement that can lead to physical injury and loss of consciousness. Seizures are canonically accompanied by increased inflammatory cytokine production that promotes neuroinflammation, brain pathology, and seizure propagation. Understanding the source of pro-inflammatory cytokines which promote seizure pathogenesis could be a gateway to precision epilepsy drug design. This review discusses the inflammasome in epilepsy including its role in seizure propagation and negative impacts on brain health. The inflammasome is a multiprotein complex that coordinates IL-1β and IL-18 production in response to tissue damage, cellular stress, and infection. Clinical evidence for inflammasome signaling in epileptogenesis is reviewed followed by a discussion of emerging strategies to modulate inflammasome activity in epilepsy.
Collapse
Affiliation(s)
- Ava Hollis
- Center for Brain Immunology and Glia (BIG), Department of NeuroscienceUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - John R. Lukens
- Center for Brain Immunology and Glia (BIG), Department of NeuroscienceUniversity of VirginiaCharlottesvilleVirginiaUSA
| |
Collapse
|
22
|
Quan J, Chang X, Liu S, He T, Zhong G, Liu Z, Yu W. Long-term copper exposure induced pyroptosis and inflammation of rat spleen through intestinal-splenic axis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117423. [PMID: 39622129 DOI: 10.1016/j.ecoenv.2024.117423] [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: 07/09/2024] [Revised: 11/12/2024] [Accepted: 11/25/2024] [Indexed: 01/26/2025]
Abstract
Copper is an essential micro-element in animal growth. Tribasic copper chloride (TBCC), as an important source of animal feed copper, is widely used in agricultural production. Multiple studies have shown that excessive intake of copper can affect the immune function of animals and lead to disease or death. The intestine-spleen axis, which is the interaction between the spleen, intestines and gut microbiota, is closely linked to immune function in the body. However, the mechanism of intestine-spleen axis affecting TBCC induced immune dysfunction in rats has not been revealed. In this study, we analyzed the impacts of different doses of TBCC on the intestine, gut microbiota and spleen of rats, and investigated their relationship in the process. Our results demonstrated that under copper exposure, the structural integrity of the intestinal epithelium was compromised, resulting in a significant downregulation of genes and proteins expression levels related to the intestinal barrier (Zonula occludens-1, Claudin-1 and Occludin). Additionally, copper perturbed the composition of the rat intestinal microbiota, altered the abundance and diversity of the microbial community. Copper entered the spleen via the intestine-spleen axis, leading to structural damage in the spleen and activation of the NFκB signaling pathway, and increased pro-inflammatory cytokines (IFN-γ, TNF-α, IL-18, and IL-1β), ultimately causing pyroptosis and inflammation in the spleen. These findings offer a novel perspective on how copper may induce spleen injury through the intestine-spleen axis.
Collapse
Affiliation(s)
- Jinwen Quan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, PR China
| | - Xiaoyue Chang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, PR China
| | - Siying Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, PR China
| | - Ting He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, PR China
| | - Gaolong Zhong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, PR China
| | - Zhonghua Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, PR China; Laboratory Animal Center, South China Agriculture University, Guangzhou, Guangdong 510642, PR China
| | - Wenlan Yu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, PR China; Laboratory Animal Center, South China Agriculture University, Guangzhou, Guangdong 510642, PR China.
| |
Collapse
|
23
|
Yin G, Guo W, Wang R, Li N, Chen X, Zhang Y, Huang Z. Analysis of the role of IL-1 family and related genes in head and neck squamous cell carcinoma. Braz J Otorhinolaryngol 2025; 91:101484. [PMID: 39461030 PMCID: PMC11543642 DOI: 10.1016/j.bjorl.2024.101484] [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: 06/09/2024] [Accepted: 08/03/2024] [Indexed: 10/29/2024] Open
Abstract
OBJECTIVE The Interleukin-1 (IL-1) family plays an important regulatory role in the development of tumors, but its function is still unclear in head and neck squamous cell carcinoma (HNSCC). Analyzing the IL-1 family can help to understand the tumor mechanism. METHODS Using GEPIA2, UALCAN, cBioprotocol and HPA databases, the IL-1 family and related genes (IL-1α, IL-1β, IL1RN, IL1R1, IL1R2, IL1RL1, IL1RL2, IL1RAP, IL1RAPL1, IL1RAPL2, IL1F10, IL18, IL18BP, IL18R1, IL18RAP, IL36A, IL36B, IL36G, IL36RN, IL33, IL37, SIGIRR, CASP1, AIM2) were analyzed for their expression and prognostic relevance in HNSCC. The Kaplan-Meier, log-rank test and Spearman correlation were used to analysis. RESULTS In tumors, IL-1α, IL-1β, IL1R1, IL1RL1, IL1F10, IL33, CASP1, and AIM2 are highly expressed, while IL1RN, IL1RAPL1, IL1RAPL2, IL18BP, IL18R1 and IL18RAP are poorly expressed. IL-1α, IL1RAP, and IL1RAPL2 were prognostic risk factors in at least two databases, while IL18RAP, IL36A, and SIGIRR were prognostic protective factors. SIGIRR was confirmed in all three databases. Compared to HPV- tumors, IL18RAP and SIGIRR are highly expressed in HPV+ tumors. In addition, IL-1α, IL-1β, IL1RL2, IL1RAP were negatively correlated with CD8A/B expression, while IL1R2, IL18R1, IL18RAP, IL33, SIGIRR, CASP1, AIM2 were positively correlated with CD8A/B expression. CONCLUSION The differential expression of the IL-1 family and related genes affects the microenvironment changes and survival prognosis of HNSCC patients. Among them, IL-1α, IL1RAP, IL18RAP, and SIGIRR may affect the prognosis of patients by affecting local CD8+ T cell infiltration in the tumor. LEVEL OF EVIDENCE: 3
Collapse
Affiliation(s)
- Gaofei Yin
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Wei Guo
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Rong Wang
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Nuan Li
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xiaohong Chen
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yang Zhang
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Zhigang Huang
- Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
24
|
Anand PK. From fat to fire: The lipid-inflammasome connection. Immunol Rev 2025; 329:e13403. [PMID: 39327931 PMCID: PMC11744241 DOI: 10.1111/imr.13403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Inflammasomes are multiprotein complexes that play a crucial role in regulating immune responses by governing the activation of Caspase-1, the secretion of pro-inflammatory cytokines, and the induction of inflammatory cell death, pyroptosis. The inflammasomes are pivotal in effective host defense against a range of pathogens. Yet, overt activation of inflammasome signaling can be detrimental. The most well-studied NLRP3 inflammasome has the ability to detect a variety of stimuli including pathogen-associated molecular patterns, environmental irritants, and endogenous stimuli released from dying cells. Additionally, NLRP3 acts as a key sensor of cellular homeostasis and can be activated by disturbances in diverse metabolic pathways. Consequently, NLRP3 is considered a key player linking metabolic dysregulation to numerous inflammatory disorders such as gout, diabetes, and atherosclerosis. Recently, compelling studies have highlighted a connection between lipids and the regulation of NLRP3 inflammasome. Lipids are integral to cellular processes that serve not only in maintaining the structural integrity and subcellular compartmentalization, but also in contributing to physiological equilibrium. Certain lipid species are known to define NLRP3 subcellular localization, therefore directly influencing the site of inflammasome assembly and activation. For instance, phosphatidylinositol 4-phosphate plays a crucial role in NLRP3 localization to the trans Golgi network. Moreover, new evidence has demonstrated the roles of lipid biosynthesis and trafficking in activation of the NLRP3 inflammasome. This review summarizes and discusses these emerging and varied roles of lipid metabolism in inflammasome activation. A deeper understanding of lipid-inflammasome interactions may open new avenues for therapeutic interventions to prevent or treat chronic inflammatory and autoimmune conditions.
Collapse
Affiliation(s)
- Paras K. Anand
- Department of Infectious Disease, Faculty of MedicineImperial College LondonLondonUK
| |
Collapse
|
25
|
Sun JQ, Sheng B, Gao S, Liu XZ, Cui Y, Peng Z, Chen XX, Ding PF, Zhuang Z, Wu LY, Hang CH, Li W. SIRT2 Promotes NLRP3-Mediated Microglia Pyroptosis and Neuroinflammation via FOXO3a Pathway After Subarachnoid Hemorrhage. J Inflamm Res 2024; 17:11679-11698. [PMID: 39741753 PMCID: PMC11687285 DOI: 10.2147/jir.s487716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 12/14/2024] [Indexed: 01/03/2025] Open
Abstract
Purpose This study primarily elucidating the specific mechanism of SIRT2 on neuroinflammation and microglial pyroptosis in a mouse model of SAH. Patients and Methods CSF were collected from 57 SAH patients and 11 healthy individuals. C57BL/6 mouse SAH model was established using prechiasmatic cistern blood injection and the in vitro hemoglobin (Hb) stimulation microglia model. Lentivirus was used as a vector for RNA interference technology to knock down the SIRT2 gene expression. Small interfering RNA was used to knockdown the expression of FOXO3a. The tools included measurements of brain water content, neurological scores, Western blot, PCR, ELISA, TEM, immunofluorescence, LDH assay, modified Garcia score, and balance beam tests to evaluate changes in pyroptosis and neuroinflammatory responses. Results In CSF samples from SAH patients, elevated levels of SIRT2 and GSDMD were observed, with SIRT2 demonstrating particular diagnostic value for predicting prognosis at the 3-month follow-up. SIRT2 upregulation exacerbated neurological deficits, brain edema, and blood-brain barrier disruption in mice following SAH. SIRT2 increased GSDMD, caspase-1, and IL-1β/IL-18 expression, and amplified GSDMD-positive microglia. FOXO3a was also upregulated post-SAH. siRNA-mediated SIRT2 knockdown ameliorated microglial pyroptosis after SAH. FOXO3a siRNA reduced NLRP3 inflammasome activation and microglial pyroptosis severity, along with neuroinflammation post-SAH. Conclusion In summary, SIRT2 promoted microglial pyroptosis, primarily by increasing the expression and activity of Foxo3a, thereby exacerbating neuroinflammatory damage following subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Jia-Qing Sun
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Drum Tower Hospital Clinical College, Xuzhou Medical University, Nanjing, People’s Republic of China
- Neurosurgery Institute of Nanjing University, Nanjing, People’s Republic of China
| | - Bin Sheng
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
- Neurosurgery Institute of Nanjing University, Nanjing, People’s Republic of China
| | - Sen Gao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
- Neurosurgery Institute of Nanjing University, Nanjing, People’s Republic of China
| | - Xun-Zhi Liu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
- Neurosurgery Institute of Nanjing University, Nanjing, People’s Republic of China
| | - Yue Cui
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- Neurosurgery Institute of Nanjing University, Nanjing, People’s Republic of China
| | - Zheng Peng
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
- Neurosurgery Institute of Nanjing University, Nanjing, People’s Republic of China
| | - Xiang-Xin Chen
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
- Neurosurgery Institute of Nanjing University, Nanjing, People’s Republic of China
| | - Peng-Fei Ding
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
- Neurosurgery Institute of Nanjing University, Nanjing, People’s Republic of China
| | - Zong Zhuang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
- Neurosurgery Institute of Nanjing University, Nanjing, People’s Republic of China
| | - Ling-Yun Wu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
- Neurosurgery Institute of Nanjing University, Nanjing, People’s Republic of China
| | - Chun-Hua Hang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Drum Tower Hospital Clinical College, Xuzhou Medical University, Nanjing, People’s Republic of China
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- Neurosurgery Institute of Nanjing University, Nanjing, People’s Republic of China
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- Neurosurgery Institute of Nanjing University, Nanjing, People’s Republic of China
| |
Collapse
|
26
|
Zhang X, Wu J, Wang M, Chen L, Wang P, Jiang Q, Yang C. The role of gene mutations and immune responses in sensorineural hearing loss. Int Immunopharmacol 2024; 143:113515. [PMID: 39486181 DOI: 10.1016/j.intimp.2024.113515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/12/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024]
Abstract
Sensorineural hearing loss (SNHL) is a prevalent clinical condition primarily attributed to dysfunction within various components of the auditory pathway, spanning from the inner ear to the auditory cortex. Recent research has illuminated immune and inflammation-mediated disorders of the inner ear as critical contributors to SNHL. Disruptions in the equilibrium of inflammatory mediators, chemokines, the complement system, and inflammatory vesicles within the cochlea provoke aberrations in immune cell activity, fostering a chronic pro-inflammatory milieu that detrimentally affects the structural and functional integrity of the inner ear, culminating in hearing impairment. Specific genetic mutations, especially those affecting auditory structures, play an important role in SNHL. These mutations regulate inflammatory mediators and cellular responses, thereby altering the inflammatory dynamics within the cochlea. This review delves into the pathogenesis of sensorineural hearing loss, emphasizing the impact of genetic alterations, immune responses within the inner ear, and inflammatory mediators on auditory function. It highlights the significance of Transmembrane Serine Protease 3 (TMPRSS3) and connexin gene mutations as pivotal genetic elements in SNHL, underscoring the central role of inflammatory responses in cochlear damage. Furthermore, the paper discusses the promise of gene therapy and targeted molecular interventions, underscoring the necessity for continued exploration into the specific actions of various inflammatory agents to refine personalized therapeutic strategies.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Junyi Wu
- Department of Otolaryngology-Head and Neck Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu Province, China
| | - Maohua Wang
- Department of Otolaryngology, Head and Neck Surgery, The First People's Hospital of Foshan, Hearing and Balance Medical Engineering Technology Center of Guangdong, Foshan, 528000, China
| | - Li Chen
- Department of Otolaryngology-Head and Neck Surgery, The Second People's Hospital of Yibin City, Sichuan Province, 644000, China
| | - Peng Wang
- Department of Otolaryngology-Head and Neck Surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Jiangsu Province, 225200, China
| | - Qiao Jiang
- Department of Neurology, Deyang Fifth Hospital, Sichuan Province, 618000, China.
| | - Chunping Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China.
| |
Collapse
|
27
|
Chen D, Plott T, Wiest M, Van Trump W, Komalo B, Nguyen D, Marsh C, Heinrich J, Fuller CJ, Nicolaisen L, Cambronero E, Nguyen A, Elabd C, Rubbo F, DeVay Jacobson R. A combined AI and cell biology approach surfaces targets and mechanistically distinct Inflammasome inhibitors. iScience 2024; 27:111404. [PMID: 39687021 PMCID: PMC11648265 DOI: 10.1016/j.isci.2024.111404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/20/2024] [Accepted: 11/13/2024] [Indexed: 12/18/2024] Open
Abstract
Inflammasomes are protein complexes that mediate innate immune responses whose dysregulation has been linked to a spectrum of acute and chronic human conditions, which dictates therapeutic development that is aligned with disease variability. We designed a scalable, physiologic high-content imaging assay in human PBMCs that we analyzed using a combination of machine-learning and cell biology methods. This resulted in a set of biologically interpretable readouts that can resolve a spectrum of cellular states associated with inflammasome activation and inhibition. These methods were applied to a phenotypic screen that surfaced mechanistically distinct inflammasome inhibitors from an annotated 12,000 compound library. A set of over 100 inhibitors, including an array of Raf-pathway inhibitors, were validated in downstream functional assays. This approach demonstrates how complementary machine learning-based methods can be used to generate profiles of cellular states associated with different stages of complex biological pathways and yield compound and target discovery.
Collapse
Affiliation(s)
- Daniel Chen
- Spring Discovery, Inc., 1125 Industrial Road, San Carlos, CA 94070, USA
| | - Tempest Plott
- Spring Discovery, Inc., 1125 Industrial Road, San Carlos, CA 94070, USA
| | - Michael Wiest
- Spring Discovery, Inc., 1125 Industrial Road, San Carlos, CA 94070, USA
| | - Will Van Trump
- Spring Discovery, Inc., 1125 Industrial Road, San Carlos, CA 94070, USA
| | - Ben Komalo
- Spring Discovery, Inc., 1125 Industrial Road, San Carlos, CA 94070, USA
| | - Dat Nguyen
- Spring Discovery, Inc., 1125 Industrial Road, San Carlos, CA 94070, USA
| | - Charlie Marsh
- Spring Discovery, Inc., 1125 Industrial Road, San Carlos, CA 94070, USA
| | - Jarred Heinrich
- Spring Discovery, Inc., 1125 Industrial Road, San Carlos, CA 94070, USA
| | - Colin J. Fuller
- Spring Discovery, Inc., 1125 Industrial Road, San Carlos, CA 94070, USA
| | - Lauren Nicolaisen
- Spring Discovery, Inc., 1125 Industrial Road, San Carlos, CA 94070, USA
| | - Elisa Cambronero
- Spring Discovery, Inc., 1125 Industrial Road, San Carlos, CA 94070, USA
| | - An Nguyen
- Spring Discovery, Inc., 1125 Industrial Road, San Carlos, CA 94070, USA
| | - Christian Elabd
- Spring Discovery, Inc., 1125 Industrial Road, San Carlos, CA 94070, USA
| | - Francesco Rubbo
- Spring Discovery, Inc., 1125 Industrial Road, San Carlos, CA 94070, USA
| | | |
Collapse
|
28
|
Pan W, Jia Z, Du J, Chang K, Liu Y, Liu W, Zhao X, Tan W. NLRP3 Inflammasome Upregulates PD-L1 in Ovarian Cancer and Contributes to an Immunosuppressive Microenvironment. Immunotargets Ther 2024; 13:775-788. [PMID: 39703562 PMCID: PMC11656484 DOI: 10.2147/itt.s495564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/04/2024] [Indexed: 12/21/2024] Open
Abstract
Introduction The NLRP3 inflammasome has been implicated in the initiation of inflammation and tumorigenesis; however, its role in epithelial ovarian cancer (EOC) remains unclear. Methods This study employed high-throughput sequencing data, ELISA, clone formation assay, Western blot, and flow cytometric analysis to investigate the specific role of the NLRP3 inflammasome in EOC. Results NLRP3 was highly expressed in human EOC tissues and correlated with an unfavorable prognosis. Activation of the NLRP3 inflammasome by LPS and ATP promoted EOC cell proliferation and increased IL-1 and PD-L1 levels. MCC950, a NLRP3 inflammasome blocker, reduced IL-1 and PD-L1 levels and diminished tumor-immune suppressive cells, such as myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), and PD-1+ CD4+ T cells, in a murine model of ovarian cancer. This intervention also suppressed tumor growth. Conclusion Our investigation revealed the pro-tumorigenic role of the NLRP3 inflammasome and its regulation of PD-L1 expression in EOC. Blockade of the NLRP3 inflammasome led to reduced PD-L1 expression, fewer immunosuppressive cells, and suppressed tumor growth. These findings suggest that targeting the NLRP3 inflammasome-PD-L1 axis could be a novel treatment approach for ovarian cancer.
Collapse
Affiliation(s)
- Wenjing Pan
- Department of Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Zhaoyang Jia
- Department of Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Jingtong Du
- Department of Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Kexin Chang
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Yiming Liu
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Wei Liu
- Department of Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Xibo Zhao
- Department of Gynecological Oncology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, 510120, People’s Republic of China
| | - Wenhua Tan
- Department of Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| |
Collapse
|
29
|
Lin Y, Cheng L, Chen Y, Li W, Guo Q, Miao Y. TFEB signaling promotes autophagic degradation of NLRP3 to attenuate neuroinflammation in diabetic encephalopathy. Am J Physiol Cell Physiol 2024; 327:C1481-C1496. [PMID: 39437446 DOI: 10.1152/ajpcell.00322.2024] [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: 05/14/2024] [Revised: 09/18/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024]
Abstract
Diabetic encephalopathy (DE), a neurological complication of diabetes mellitus, has an unclear etiology. Shreds of evidence show that the nucleotide-binding oligomerization domain-like receptor family protein 3 (NLRP3) inflammasome-induced neuroinflammation and transcription factor EB (TFEB)-mediated autophagy impairment may take part in DE development. The cross talk between these two pathways and their contribution to DE remains to be explored. A mouse model of type 2 diabetes mellitus (T2DM) exhibiting cognitive dysfunction was created, along with high-glucose (HG) cultured BV2 cells. Following, 3-methyladenine (3-MA) and rapamycin were used to modulate autophagy. To evaluate the potential therapeutic benefits of TFEB in DE, we overexpressed and knocked down TFEB in both mice and cells. Autophagy impairment and NLRP3 inflammasome activation were noticed in T2DM mice and HG-cultured BV2 cells. The inflammatory response caused by NLRP3 inflammasome activation was decreased by rapamycin-induced autophagy enhancement, while 3-MA treatment further deteriorated it. Nuclear translocation and expression of TFEB were hampered in HG-cultured BV2 cells and T2DM mice. Exogenous TFEB overexpression boosted NLRP3 degradation via autophagy, which in turn alleviated microglial activation as well as ameliorated cognitive deficits and neuronal damage. In addition, TFEB knockdown exacerbated neuroinflammation by decreasing autophagy-mediated NLRP3 degradation. Our findings have unraveled the pathogenesis of a previously underappreciated disease, implying that the activation of NLRP3 inflammasome and impairment of autophagy in microglia are significant etiological factors in the DE. The TFEB-mediated autophagy pathway can reduce neuroinflammation by enhancing NLRP3 degradation. This could potentially serve as a viable and innovative treatment approach for DE.NEW & NOTEWORTHY This article delves into the intricate connections between inflammation, autophagy, diabetes, and neurodegeneration, with a particular focus on a disease that is not yet fully understood-diabetic encephalopathy (DE). TFEB emerges as a pivotal regulator in balancing autophagy and inflammation in DE. Our findings highlight the crucial function of the TFEB-mediated autophagy pathway in mitigating inflammatory damage in DE, suggesting a new treatment strategy.
Collapse
Affiliation(s)
- Yijia Lin
- Department of Geriatrics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lizhen Cheng
- Department of Geriatrics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yixin Chen
- Department of Geriatrics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Li
- Department of Geriatrics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qihao Guo
- Department of Geriatrics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ya Miao
- Department of Geriatrics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
30
|
Song D, Cen Y, Qian Z, Wu XS, Rivera K, Wee TL, Demerdash OE, Chang K, Pappin D, Vakoc CR, Tonks NK. PTPN23-dependent ESCRT machinery functions as a cell death checkpoint. Nat Commun 2024; 15:10364. [PMID: 39609437 PMCID: PMC11604704 DOI: 10.1038/s41467-024-54749-2] [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: 06/28/2023] [Accepted: 11/20/2024] [Indexed: 11/30/2024] Open
Abstract
Cell death plasticity is crucial for modulating tissue homeostasis and immune responses, but our understanding of the molecular components that regulate cell death pathways to determine cell fate remains limited. Here, a CRISPR screen of acute myeloid leukemia cells identifies protein tyrosine phosphatase non-receptor type 23 (PTPN23) as essential for survival. Loss of PTPN23 activates nuclear factor-kappa B, apoptotic, necroptotic, and pyroptotic pathways by causing the accumulation of death receptors and toll-like receptors (TLRs) in endosomes. These effects are recapitulated by depletion of PTPN23 co-dependent genes in the endosomal sorting complex required for transport (ESCRT) pathway. Through proximity-dependent biotin labeling, we show that NAK-associated protein 1 interacts with PTPN23 to facilitate endosomal sorting of tumor necrosis factor receptor 1 (TNFR1), sensitizing cells to TNF-α-induced cytotoxicity. Our findings reveal PTPN23-dependent ESCRT machinery as a cell death checkpoint that regulates the spatiotemporal distribution of death receptors and TLRs to restrain multiple cell death pathways.
Collapse
MESH Headings
- Humans
- Endosomal Sorting Complexes Required for Transport/metabolism
- Endosomal Sorting Complexes Required for Transport/genetics
- Endosomes/metabolism
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Apoptosis
- NF-kappa B/metabolism
- Cell Death
- Protein Tyrosine Phosphatases, Non-Receptor/metabolism
- Protein Tyrosine Phosphatases, Non-Receptor/genetics
- Toll-Like Receptors/metabolism
- Tumor Necrosis Factor-alpha/metabolism
- Signal Transduction
- Cell Line, Tumor
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- HEK293 Cells
- Receptors, Death Domain/metabolism
Collapse
Affiliation(s)
- Dongyan Song
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Molecular and Cellular Biology Graduate Program, Stony Brook University, Stony Brook, NY, USA
| | - Yuxin Cen
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Molecular and Cellular Biology Graduate Program, Stony Brook University, Stony Brook, NY, USA
| | - Zhe Qian
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Molecular and Cellular Biology Graduate Program, Stony Brook University, Stony Brook, NY, USA
| | - Xiaoli S Wu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY, USA
| | - Keith Rivera
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Tse-Luen Wee
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Osama E Demerdash
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Kenneth Chang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Darryl Pappin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | | | - Nicholas K Tonks
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA.
| |
Collapse
|
31
|
Kim G, Lee SY, Oh S, Jang JW, Lee J, Kim HS, Son KH, Byun K. Anti-Inflammatory Effects of Extracellular Vesicles from Ecklonia cava on 12-O-Tetradecanoylphorbol-13-Acetate-Induced Skin Inflammation in Mice. Int J Mol Sci 2024; 25:12522. [PMID: 39684233 DOI: 10.3390/ijms252312522] [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: 09/05/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Steroids, which are often used to treat the inflammation associated with various skin diseases, have several negative side effects. As Ecklonia cava extract has anti-inflammatory effects in various diseases, we evaluated the efficacy of Ecklonia cava-derived extracellular vesicles (EVEs) in decreasing 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced inflammation. We determined the effect of the EVEs on the TLR4/NF-κB/NLRP3 inflammasome in human keratinocytes and mouse ear skin. TPA-treated human keratinocytes showed an increased expression of TLR4 and its ligands HMGB1 and S100A8. TPA also increased the expression of (1) NF-κB; (2) the NLRP3 inflammasome components NLRP3, ASC, and caspase 1; and (3) the pyroptosis-related factors GSDMD-NT, IL-18, and IL-1β. However, the expression of these molecules decreased in the TPA-treated human keratinocytes after EVE treatment. Similar to the in vitro results, TPA increased the expression of these molecules in mouse ear skin, and EVE treatment decreased their expression. The TPA treatment of skin increased edema, redness, neutrophil infiltration, and epidermal thickness, and EVE reduced these symptoms of inflammation. In conclusion, the EVEs decreased TPA-induced skin inflammation, which was associated with a decrease in the TLR4/NF-κB/NLRP3 inflammasome.
Collapse
Affiliation(s)
- Geebum Kim
- Misogain Dermatology Clinic, Gimpo 10108, Republic of Korea
| | - So Young Lee
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon University, Incheon 21565, Republic of Korea
| | - Seyeon Oh
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Jong-Won Jang
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
| | - Jehyuk Lee
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Doctorbom Clinic, Seoul 06614, Republic of Korea
| | - Hyun-Seok Kim
- Kim Hyun Seok Plastic Surgery Clinic, Seoul 06030, Republic of Korea
| | - Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon University, Incheon 21565, Republic of Korea
| | - Kyunghee Byun
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
| |
Collapse
|
32
|
Vallese A, Cordone V, Ferrara F, Guiotto A, Gemmo L, Cervellati F, Hayek J, Pecorelli A, Valacchi G. NLRP3 inflammasome-mitochondrion loop in autism spectrum disorder. Free Radic Biol Med 2024; 225:581-594. [PMID: 39433111 DOI: 10.1016/j.freeradbiomed.2024.10.297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social communication and the presence of restricted interests and repetitive behavior. To date, no single cause has been demonstrated but both genetic and environmental factors are believed to be involved in abnormal brain development. In recent years, immunological and mitochondrial dysfunctions acquired particular interest in the study of the molecular mechanisms underlying the pathophysiology of ASD. For this reason, our study focused on evaluating the mitochondrial component and activation of the NLRP3 inflammasome, a critical player of the innate immune system. The assembly of NLRP3 with ASC mediates activation of Caspase-1, which in turn, by proteolytic cleavage, activates Gasdermin D and the proinflammatory cytokines IL-1β/IL-18 with their subsequent secretion. Using primary fibroblasts of autistic and control patients we studied basal and stimulated conditions. Specifically, LPS and ATP were used to activate the NLRP3 inflammasome and MCC950 for its inhibition. In addition, FCCP was used as a mitochondrial stressor and MitoTEMPO as a scavenger of mitochondrial ROS. Our results showed a hyperactivation of NLRP3 inflammasome in ASDs, as evidenced by the co-localization of the two main components, NLRP3 and ASC, by the higher levels of ASC specks, oligomers and dimers and by the increased amounts of active Caspase-1 and IL-1β. In addition, increased mitochondrial superoxide anion and reduced mitochondrial membrane potential were detected in ASD cells. These data are in accordance with the abnormal mitochondrial morphology evidenced by transmission electron microscopy analysis. Interestingly, NLRP3 inflammasome inhibition with MCC950 improved mitochondrial parameters, while the use of MitoTEMPO, in addition to decrease mitochondrial ROS production, was able to prevent NLRP3 inflammasome activation suggesting for the first time an abnormal bidirectional crosstalk between mitochondria and NLRP3 inflammasome in ASD.
Collapse
Affiliation(s)
- Andrea Vallese
- Dept. of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy; Animal Science Dept., Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, USA
| | - Valeria Cordone
- Dept. of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy
| | - Francesca Ferrara
- Dept. of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Anna Guiotto
- Dept. of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy; Animal Science Dept., Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, USA
| | - Laura Gemmo
- Dept. of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy
| | - Franco Cervellati
- Dept. of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | | | - Alessandra Pecorelli
- Dept. of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy; Dept. of Food, Bioprocessing and Nutrition Sciences, Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, USA.
| | - Giuseppe Valacchi
- Dept. of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy; Animal Science Dept., Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, USA; Dept. of Food and Nutrition, Kyung Hee University, Seoul, South Korea.
| |
Collapse
|
33
|
Papantoniou K, Aggeletopoulou I, Michailides C, Pastras P, Triantos C. Understanding the Role of NLRP3 Inflammasome in Acute Pancreatitis. BIOLOGY 2024; 13:945. [PMID: 39596901 PMCID: PMC11592098 DOI: 10.3390/biology13110945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/31/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024]
Abstract
Acute pancreatitis (AP) remains a serious clinical condition, with current treatment options being largely supportive. The discovery of inflammasomes, particularly the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, has significantly advanced our knowledge regarding many inflammatory diseases' pathogenesis, including AP. The NLRP3 inflammasome is central in mediating the inflammatory process in AP through its diverse activation mechanisms and its involvement in multiple signal transduction pathways. This has made NLRP3 an appealing target for novel therapeutic strategies aimed at modulating inflammation in AP. Despite the growing interest in NLRP3 as a therapeutic target, there remains a notable gap in clinical research, with few clinical trials exploring the efficacy of NLRP3 inhibitors in AP. Results of several preclinical studies and animal models are promising and suggest that the use of NLRP3 inhibitors could result in reduced inflammation and improved patient outcomes in AP. Further research is urgently needed to assess their potential benefits, safety, and applicability in human patients and address the underlying inflammatory processes driving AP.
Collapse
Affiliation(s)
- Konstantinos Papantoniou
- Department of Internal Medicine, University Hospital of Patras, 26504 Patras, Greece; (K.P.); (C.M.)
| | - Ioanna Aggeletopoulou
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, 26504 Patras, Greece; (I.A.); (P.P.)
| | - Christos Michailides
- Department of Internal Medicine, University Hospital of Patras, 26504 Patras, Greece; (K.P.); (C.M.)
| | - Ploutarchos Pastras
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, 26504 Patras, Greece; (I.A.); (P.P.)
| | - Christos Triantos
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, 26504 Patras, Greece; (I.A.); (P.P.)
| |
Collapse
|
34
|
Zheng Y, Zhang X, Wang Z, Zhang R, Wei H, Yan X, Jiang X, Yang L. MCC950 as a promising candidate for blocking NLRP3 inflammasome activation: A review of preclinical research and future directions. Arch Pharm (Weinheim) 2024; 357:e2400459. [PMID: 39180246 DOI: 10.1002/ardp.202400459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/19/2024] [Accepted: 07/30/2024] [Indexed: 08/26/2024]
Abstract
The NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome is a key component of the innate immune system that triggers inflammation and pyroptosis and contributes to the development of several diseases. Therefore, blocking the activation of the NLRP3 inflammasome has therapeutic potential for the treatment of these diseases. MCC950, a selective small molecule inhibitor, has emerged as a promising candidate for blocking NLRP3 inflammasome activation. Ongoing research is focused on elucidating the specific targets of MCC950 as well as assessfing its metabolism and safety profile. This review discusses the diseases that have been studied in relation to MCC950, with a focus on stroke, Alzheimer's disease, liver injury, atherosclerosis, diabetes mellitus, and sepsis, using bibliometric analysis. It then summarizes the potential pharmacological targets of MCC950 and discusses its toxicity. Furthermore, it traces the progression from preclinical to clinical research for the treatment of these diseases. Overall, this review provides a solid foundation for the clinical therapeutic potential of MCC950 and offers insights for future research and therapeutic approaches.
Collapse
Affiliation(s)
- Yujia Zheng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Xiaolu Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Ziyu Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Ruifeng Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Huayuan Wei
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Xu Yan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Lin Yang
- School of Medicial Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, Jinghai, China
| |
Collapse
|
35
|
Yoladi FB, Palabiyik-Yucelik SS, Bahador Zirh E, Halici Z, Baydar T. Effects of idebenone and coenzyme Q10 on NLRP3/caspase-1/IL-1β pathway regulation on ethanol-induced hepatotoxicity in rats. Drug Chem Toxicol 2024; 47:1205-1217. [PMID: 38804209 DOI: 10.1080/01480545.2024.2351191] [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: 12/16/2023] [Accepted: 04/29/2024] [Indexed: 05/29/2024]
Abstract
Chronic and excessive alcohol consumption leads to liver toxicity. There is a need to investigate effective therapeutic strategies to alleviate alcohol-induced liver injury, which remains the leading cause of liver-related morbidity and mortality worldwide. Therefore here, we looked into and evaluated how ethanol-induced hepatotoxicity was affected by coenzyme Q10 (CoQ10) and its analog, idebenone (IDE), on the NLRP3/caspase-1/IL-1 pathway. Hepatotoxicity induced in rats through the oral administration of gradually increasing dosages of ethanol (from 2 to 6 g/kg/day) over 30 days and the effect of CoQ10 (10 or 20 mg/kg) and IDE (50 or 100 mg/kg) were evaluated. Serum hepatotoxicity markers (ALT, AST, GGT, ALP, and TBIL), tissue oxidative stress markers and the mRNA expressions of IL-1β, IL-18, TGF-β, NF-κB, NLRP3, and caspase-1 were evaluated. Masson's trichrome staining was also used to visualize fibrosis in the liver tissue. The results indicated that ethanol exposure led to hepatotoxicity as well as considerable NLRP3/caspase-1/IL-1β pathway activation. Moreover, CoQ10 or IDE treatment reduced measured parameters in a dosage-dependent manner. Thus, by inhibiting the NLRP3/caspase-1/IL-1 pathway, CoQ10 and IDE can prevent the hepatotoxicity caused by ethanol, although CoQ10 is more effective than IDE. This study will provide insight into new therapeutic avenues that take advantage of the anti-inflammatory and antioxidant properties of CoQ10 and IDE in ethanol-induced liver diseases.
Collapse
Affiliation(s)
- Fatma Betül Yoladi
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Atatürk University, Erzurum, Turkey
- Department of Toxicology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Saziye Sezin Palabiyik-Yucelik
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Atatürk University, Erzurum, Turkey
- Clinical Research, Development and Design Application and Research Center, Atatürk University, Erzurum, Turkey
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Ondokuz Mayıs University, Samsun, Turkey
| | - Elham Bahador Zirh
- Department of Histology and Embryology, Faculty of Medicine, TOBB University of Economics and Technology, Ankara, Turkey
| | - Zekai Halici
- Clinical Research, Development and Design Application and Research Center, Atatürk University, Erzurum, Turkey
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Terken Baydar
- Department of Toxicology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
36
|
El-Kashef DH, Abdel-Rahman N, Sharawy MH. Apocynin alleviates thioacetamide-induced acute liver injury: Role of NOX1/NOX4/NF-κB/NLRP3 pathways. Cytokine 2024; 183:156747. [PMID: 39236429 DOI: 10.1016/j.cyto.2024.156747] [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: 03/05/2024] [Revised: 07/01/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
The liver has a distinctive capacity to regenerate, yet severe acute injury can be life-threatening if not treated appropriately. Inflammation and oxidative stress are central processes implicated in the pathophysiology of acute livery injury. NOX isoforms are important enzymes for ROS generation, NF-κB and NLRP3 activation, its inhibition could be vital in alleviating acute liver injury (ALI). Here in our study, we used apocynin, a natural occurring potent NOX inhibitor, to exploreits potential protective effect against thioacetamide (TAA)-induced ALI through modulating crucial oxidative and inflammatory pathways. Rats were injected once with TAA (500 mg/kg/i.p) and treated with apocynin (10 mg/kg/i.p) twice before TAA challenge. Sera and hepatic tissues were collected for biochemical, mRNA expression, western blot analysis and histopathological assessments. Pretreatment with apocynin improved liver dysfunction evidenced by decreased levels of aminotransferases, ALP, GGT and bilirubin. Apocynin reduced mRNA expression of NOX1 and NOX4 which in turn alleviated oxidative stress, as shown by reduction in MDA and NOx levels, and elevation in GSH levels andcatalase and SOD activities. Moreover, apocynin significantly reduced MPO gene expression. We also demonstrate that apocynin ameliorated inflammation through activating IκBα and suppressing IKKα, IKKβ, NF-κBp65 and p-NF-κBp65, IL-6 andTNF-α. Additionally, apocynin potentiated the gene expression of anti-inflammatory IL-10 and reduced levels of hepatic NLRP3, Caspase-1 and IL-1β. These results suggest that apocynin protects against ALI in association with the inhibition of NOX1 and NOX4 and regulating oxidative and inflammatory pathways.
Collapse
Affiliation(s)
- Dalia H El-Kashef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Noha Abdel-Rahman
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| | - Maha H Sharawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
37
|
Hinds A, Ward P, Archer N, Leigh J. Priming from within: TLR2 dependent but receptor independent activation of the mammary macrophage inflammasome by Streptococcus uberis. Front Cell Infect Microbiol 2024; 14:1444178. [PMID: 39463761 PMCID: PMC11502467 DOI: 10.3389/fcimb.2024.1444178] [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: 06/05/2024] [Accepted: 09/12/2024] [Indexed: 10/29/2024] Open
Abstract
Introduction Streptococcus uberis is a member of the pyogenic cluster of Streptococcus commonly associated with intramammary infection and mastitis in dairy cattle. It is a poorly controlled globally endemic pathogen responsible for a significant cause of the disease worldwide. The ruminant mammary gland provides an atypical body niche in which immune cell surveillance occurs on both sides of the epithelial tissue. S. uberis does not cause disease in non-ruminant species and is an asymptomatic commensal in other body niches. S. uberis exploits the unusual niche of the mammary gland to initiate an innate response from bovine mammary macrophage (BMMO) present in the secretion (milk) in which it can resist the host immune responses. As a result - and unexpectedly - the host inflammatory response is a key step in the pathogenesis of S.uberis, without which colonisation is impaired. In contrast to other bacteria pathogenic to the bovine mammary gland, S. uberis does not elicit innate responses from epithelial tissues; initial recognition of infection is via macrophages within milk. Methods We dissected the role of the bacterial protein SUB1154 in the inflammasome pathway using ex vivo bovine mammary macrophages isolated from milk, recombinant protein expression, and a panel of inhibitors, agonists, and antagonists. We combine this with reverse-transcription quantitative real-time PCR to investigate the mechanisms underlying SUB1154-mediated priming of the immune response. Results Here, we show that SUB1154 is responsible for priming the NLRP3 inflammasome in macrophages found in the mammary gland. Without SUB1154, IL-1β is not produced, and we were able to restore IL-1β responses to a sub1154 deletion S. uberis mutant using recombinant SUB1154. Surprisingly, only by blocking internalisation, or the cytoplasmic TIR domain of TLR2 were we able to block SUB1154-mediated priming. Discussion Together, our data unifies several contrasting past studies and provides new mechanistic understanding of potential early interactions between pyogenic streptococci and the host.
Collapse
Affiliation(s)
- Abbie Hinds
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
- Department of Infection Biology and Microbiomes, University of Liverpool, Cheshire, United Kingdom
| | - Philip Ward
- The Division of Structural Biology (STRUBI) for Genomic Medicine, Oxford, United Kingdom
| | - Nathan Archer
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | - James Leigh
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
38
|
Lin J, Chen X, Du Y, Li J, Guo T, Luo S. Mitophagy in Cell Death Regulation: Insights into Mechanisms and Disease Implications. Biomolecules 2024; 14:1270. [PMID: 39456203 PMCID: PMC11506020 DOI: 10.3390/biom14101270] [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: 06/18/2024] [Revised: 08/15/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Mitophagy, a selective form of autophagy, plays a crucial role in maintaining optimal mitochondrial populations, normal function, and intracellular homeostasis by monitoring and removing damaged or excess mitochondria. Furthermore, mitophagy promotes mitochondrial degradation via the lysosomal pathway, and not only eliminates damaged mitochondria but also regulates programmed cell death-associated genes, thus preventing cell death. The interaction between mitophagy and various forms of cell death has recently gained increasing attention in relation to the pathogenesis of clinical diseases, such as cancers and osteoarthritis, neurodegenerative, cardiovascular, and renal diseases. However, despite the abundant literature on this subject, there is a lack of understanding regarding the interaction between mitophagy and cell death. In this review, we discuss the main pathways of mitophagy, those related to cell death mechanisms (including apoptosis, ferroptosis, and pyroptosis), and the relationship between mitophagy and cell death uncovered in recent years. Our study offers potential directions for therapeutic intervention and disease diagnosis, and contributes to understanding the molecular mechanism of mitophagy.
Collapse
Affiliation(s)
| | | | | | | | | | - Sai Luo
- The 1st Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin 150000, China; (J.L.); (X.C.); (Y.D.); (J.L.); (T.G.)
| |
Collapse
|
39
|
Schermuly II, Romanet S, Patra AK, Mastrototaro L, Lemme A, Pieper R, Zentek J, Aschenbach JR. Transport of Neutral Amino Acids in the Jejunum of Pigs with Special Consideration of L-Methionine. Nutrients 2024; 16:3418. [PMID: 39408384 PMCID: PMC11478682 DOI: 10.3390/nu16193418] [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: 09/20/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Methionine (Met) is a popular nutritional supplement in humans and animals. It is routinely supplemented to pigs as L-Met, DL-Met, or DL-2-hydroxy-4-(methylthio) butanoic acid (DL-HMTBA). Methods: We investigated the effect of these Met supplements on jejunal amino acid (AA) transport in male castrated Piétrain × Danbred pigs, also including a non-supplemented group. The mucosal-to-serosal flux of ten [14C]-labeled AAs (L-glutamine, glycine, L-leucine, L-lysine, L-Met, L-serine, L-threonine, L-tryptophan, L-tyrosine and L-valine) was investigated at two concentrations (50 µM and 5 mM). Inhibition of apical uptake by mucosal L-Met was also measured for these AAs. The intestinal expression of apical AA transporters, angiotensin-converting enzyme II and inflammation-related genes were compared with those of a previous study. Results: Except for tryptophan and lysine at 5 mM, all AA fluxes were Na+-dependent (p ≤ 0.05), and the uptake of most AAs, except glycine and lysine, was inhibited by L-Met (p < 0.001). A correlation network existed between Na+-dependent fluxes of most AAs (except tryptophan and partly glycine). We observed the upregulation of B0AT1 (SLC6A19) (p < 0.001), the downregulation of ATB0,+ (SLC6A14) (p < 0.001) and a lower expression of CASP1, IL1β, IL8, TGFβ and TNFα in the present vs. the previous study (p < 0.001). Conclusions: The correlating AAs likely share the same Na+-dependent transporter(s). A varying effect of the Met supplement type on AA transport in the two studies might be related to a different level of supplementation or a different inflammatory status of the small intestine.
Collapse
Affiliation(s)
- Isabel I. Schermuly
- Institute of Veterinary Physiology, Freie Universität Berlin, Königsweg 56, 14163 Berlin, Germany; (I.I.S.); (L.M.)
| | - Stella Romanet
- Institute of Veterinary Physiology, Freie Universität Berlin, Königsweg 56, 14163 Berlin, Germany; (I.I.S.); (L.M.)
| | - Amlan K. Patra
- American Institute for Goat Research, Langston University, Langston, OK 73050, USA;
| | - Lucia Mastrototaro
- Institute of Veterinary Physiology, Freie Universität Berlin, Königsweg 56, 14163 Berlin, Germany; (I.I.S.); (L.M.)
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - Andreas Lemme
- Animal Nutrition Services, Evonik Operations GmbH, Rodenbacher Chausee 4, 63457 Hanau-Wolfgang, Germany;
| | - Robert Pieper
- Institute of Animal Nutrition, Freie Universität Berlin, Königin-Luise-Straße 49, 14195 Berlin, Germany
| | - Jürgen Zentek
- Institute of Animal Nutrition, Freie Universität Berlin, Königin-Luise-Straße 49, 14195 Berlin, Germany
| | - Jörg R. Aschenbach
- Institute of Veterinary Physiology, Freie Universität Berlin, Königsweg 56, 14163 Berlin, Germany; (I.I.S.); (L.M.)
| |
Collapse
|
40
|
Zhao L, Li S, Wang X, Zhang L, Zhang J, Liu X, Hu Y, Xian X, Zhang F, Li W, Zhang M. The AGEs/RAGE Signaling Pathway Regulates NLRP3-Mediated Neuronal Pyroptosis After MCAO Injury in Lepr-/- Obese Rats. J Inflamm Res 2024; 17:6935-6954. [PMID: 39372588 PMCID: PMC11453143 DOI: 10.2147/jir.s476458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024] Open
Abstract
Background Obesity is recognized as a primary risk factor for cerebral ischemia, which has shown a significant increase in its incidence among obese patients. The exact mechanism by which obesity exacerbates cerebral ischemic injury is not fully understood though. The present study validated the hypothesis that obesity mediates pyroptosis by the AGEs/RAGE signaling pathway to exacerbate cerebral ischemic injury. Methods Leptin receptor knockout (Lepr-/- ) rats were used in this study to construct an obesity model, and the middle cerebral artery occlusion (MCAO) models of ischemic stroke were established in Lepr-/- obese rats and their wild-type (WT) littermates respectively. Zea-Longa score, TTC and H&E staining were utilized to evaluate the neurological impairment. Western Blot, immunohistochemistry, and immunofluorescence were used to detect protein expressions. Transmission electron microscopy was used to observe the pores in the neuronal cell membrane in the ischemic penumbra cortex. Results Compared with WT littermates, Lepr-/- obese rats exhibited exacerbated neuronal injury after MCAO, with higher expressions of NLRP3 inflammasome and pyroptosis-related proteins in the cortical tissue of the penumbra. Moreover, more GSDMD pores were observed on the neuronal cell membranes of Lepr-/- obese rats according to the electron microscopy. Inhibition of NLRP3 inflammasome expression with MCC950 inhibited neuronal pyroptosis after cerebral ischemia in Lepr-/- obese rats, thus reducing neuronal injury. We also found that compared with WT littermates, the levels of AGEs and RAGE in the cortex of Lepr-/- obese rats are significantly higher, with further increase after cerebral ischemia. Inhibition of AGEs/RAGE signaling pathway with FPS-ZM1 reduced the NLRP3 inflammasome-mediated neuronal pyroptosis in Lepr-/- obese rats, thereby mitigating the neuronal damage after cerebral ischemia. Conclusion The AGEs/RAGE signaling pathway is involved in the exacerbation of cerebral ischemic injury in Lepr-/- obese rats via regulating NLRP3-mediated neuronal pyroptosis.
Collapse
Affiliation(s)
- Ling Zhao
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
- Department of Obstetrics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050000, People’s Republic of China
| | - Shichao Li
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
- Experimental Diagnostic Center for Infectious Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050000, People’s Republic of China
| | - Xiaoyu Wang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
| | - Lingyan Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
| | - Jingge Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
| | - Xiyun Liu
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
| | - Yuyan Hu
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
| | - Xiaohui Xian
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050051, People’s Republic of China
| | - Wenbin Li
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, Hebei Province, 050017, People’s Republic of China
| |
Collapse
|
41
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Overview of pyroptosis mechanism and in-depth analysis of cardiomyocyte pyroptosis mediated by NF-κB pathway in heart failure. Biomed Pharmacother 2024; 179:117367. [PMID: 39214011 DOI: 10.1016/j.biopha.2024.117367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
The pyroptosis of cardiomyocytes has become an essential topic in heart failure research. The abnormal accumulation of these biological factors, including angiotensin II, advanced glycation end products, and various growth factors (such as connective tissue growth factor, vascular endothelial growth factor, transforming growth factor beta, among others), activates the nuclear factor-κB (NF-κB) signaling pathway in cardiovascular diseases, ultimately leading to pyroptosis of cardiomyocytes. Therefore, exploring the underlying molecular biological mechanisms is essential for developing novel drugs and therapeutic strategies. However, our current understanding of the precise regulatory mechanism of this complex signaling pathway in cardiomyocyte pyroptosis is still limited. Given this, this study reviews the milestone discoveries in the field of pyroptosis research since 1986, analyzes in detail the similarities, differences, and interactions between pyroptosis and other cell death modes (such as apoptosis, necroptosis, autophagy, and ferroptosis), and explores the deep connection between pyroptosis and heart failure. At the same time, it depicts in detail the complete pathway of the activation, transmission, and eventual cardiomyocyte pyroptosis of the NF-κB signaling pathway in the process of heart failure. In addition, the study also systematically summarizes various therapeutic approaches that can inhibit NF-κB to reduce cardiomyocyte pyroptosis, including drugs, natural compounds, small molecule inhibitors, gene editing, and other cutting-edge technologies, aiming to provide solid scientific support and new research perspectives for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
42
|
Didilescu AC, Chinthamani S, Scannapieco FA, Sharma A. NLRP3 inflammasome activity and periodontal disease pathogenesis-A bidirectional relationship. Oral Dis 2024; 30:4069-4077. [PMID: 38817019 PMCID: PMC11480888 DOI: 10.1111/odi.15005] [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: 11/20/2023] [Revised: 03/09/2024] [Accepted: 05/07/2024] [Indexed: 06/01/2024]
Abstract
OBJECTIVE Periodontitis is an inflammatory oral disease that occurs as a result of the damaging effects of the immune response against the subgingival microflora. Among the mechanisms involved, the nucleotide-binding oligomerization domain, leucine-rich repeat-containing proteins family member NLRP3 (NLR family pyrin domain-containing 3), proposed as the key regulator of macrophage-induced inflammation, is strongly associated with periodontal disease due to the bacterial activators. This paper aimed to present key general concepts of NLRP3 inflammasome activation and regulation in periodontal disease. METHOD A narrative review was conducted in order to depict the current knowledge on the relationship between NLRP3 inflammasome activity and periodontal disease. In vitro and in situ studies were retrieved and commented based on their relevance in the field. RESULTS The NLRP3 inflammasome activity stimulated by periodontal microbiota drive periodontal disease pathogenesis and progression. This occurs through the release of proinflammatory cytokines IL-1β, IL-18, and DAMPs (damage-associated molecular pattern molecules) following inflammasome activation. Moreover, the tissue expression of NLRP3 is dysregulated by oral microbiota, further exacerbating periodontal inflammation. CONCLUSION The review provides new insights into the relationship between the NLRP3 inflammasome activity and periodontal disease pathogenesis, highlighting the roles and regulatory mechanism of inflammatory molecules involved in the disease process.
Collapse
Affiliation(s)
- Andreea C. Didilescu
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, State University of New York, Buffalo, New York, USA
- Department of Embryology, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Sreedevi Chinthamani
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Frank A. Scannapieco
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Ashu Sharma
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, State University of New York, Buffalo, New York, USA
| |
Collapse
|
43
|
Xu DW, Tate MD. Taking AIM at Influenza: The Role of the AIM2 Inflammasome. Viruses 2024; 16:1535. [PMID: 39459869 PMCID: PMC11512208 DOI: 10.3390/v16101535] [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: 08/30/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
Influenza A viruses (IAV) are dynamic and highly mutable respiratory pathogens that present persistent public health challenges. Inflammasomes, as components of the innate immune system, play a crucial role in the early detection and response to infections. They react to viral pathogens by triggering inflammation to promote immune defences and initiate repair mechanisms. While a strong response is necessary for early viral control, overactivation of inflammasomes can precipitate harmful hyperinflammatory responses, a defining characteristic observed during severe influenza infections. The Absent in Melanoma 2 (AIM2) inflammasome, traditionally recognised for its role as a DNA sensor, has recently been implicated in the response to RNA viruses, like IAV. Paradoxically, AIM2 deficiency has been linked to both enhanced and reduced vulnerability to IAV infection. This review synthesises the current understanding of AIM2 inflammasome activation during IAV and explores its clinical implications. Understanding the nuances of AIM2's involvement could unveil novel therapeutic avenues for mitigating severe influenza outcomes.
Collapse
Affiliation(s)
- Dianne W. Xu
- Center for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Michelle D. Tate
- Center for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia
| |
Collapse
|
44
|
Sachdeva C, Satyamoorthy K, Murali TS. Pseudomonas aeruginosa: metabolic allies and adversaries in the world of polymicrobial infections. Crit Rev Microbiol 2024:1-20. [PMID: 39225080 DOI: 10.1080/1040841x.2024.2397359] [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: 09/25/2023] [Revised: 08/10/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Pseudomonas aeruginosa (PA), an opportunistic human pathogen that is frequently linked with chronic infections in immunocompromised individuals, is also metabolically versatile, and thrives in diverse environments. Additionally, studies report that PA can interact with other microorganisms, such as bacteria, and fungi, producing unique metabolites that can modulate the host immune response, and contribute to disease pathogenesis. This review summarizes the current knowledge related to the metabolic interactions of PA with other microorganisms (Staphylococcus, Acinetobacter, Klebsiella, Enterococcus, and Candida) and human hosts, and the importance of these interactions in a polymicrobial context. Further, we highlight the potential applications of studying these metabolic interactions toward designing better diagnostic tools, and therapeutic strategies to prevent, and treat infections caused by this pathogen.
Collapse
Affiliation(s)
- Chandni Sachdeva
- Department of Public Health Genomics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kapaettu Satyamoorthy
- Department of Cell & Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
- SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara (SDM) University, Sattur, Karnataka, India
| | - Thokur Sreepathy Murali
- Department of Public Health Genomics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
45
|
Li X, Zeng Q, Yao R, Zhang L, Kong Y, Shen B. Rapamycin mitigates organ damage by autophagy-mediated NLRP3 inflammasome inactivation in sepsis. Histol Histopathol 2024; 39:1167-1177. [PMID: 38288570 DOI: 10.14670/hh-18-706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Autophagy activation can alleviate sepsis-induced organ injuries. Rapamycin (Rap) has emerged as an autophagy regulator in multiple forms of organ injuries. This study aimed to assess whether Rap protects rats from cecal ligation and puncture (CLP)-induced sepsis through autophagy-mediated inactivation of the NLRP3 inflammasome. Rats were allocated to the sham, CLP, Rap (10 mg/kg), or 3-Methyladenine (3-MA) (15 mg/kg) groups. A rat CLP model was established. The survival of rats and lung wet-to-dry weight ratio in each group was assessed. Blood biochemical indexes and oxidative stress-related factors were analyzed with an automatic biochemical analyzer. The bacterial counts of blood and organs were monitored. The degrees of myeloperoxidase of the ileum, inflammation-related indexes, and pathological changes in the tissues were detected by ELISA and hematoxylin-eosin staining. The levels of NLRP3 inflammasome and autophagy-related factors were analyzed by Western blot. Rap increased the survival and SOD activity, and repressed ALT, AST, BUN, SCr, MDA, and inflammation-related marker levels in CLP rats, it also restrained the bacterial counts of blood, lung, liver, and kidney in CLP rats; the effects of 3-MA on CLP rats on the above-mentioned indicators were opposite to those of Rap. Additionally, Rap alleviated the pathological injury of the lung, liver, and kidney, which was the opposite to the effect of 3-MA on CLP rats. Furthermore, Rap mitigated the ASC, Pro-caspase 1, and NLRP3 levels and increased the Beclin-1 levels and the LC3II/LC3I ratio in the organ tissues. Collectively, autophagy activation can mitigate organ damage by suppressing the NLRP3 inflammasome in sepsis rats.
Collapse
Affiliation(s)
- Xiaofeng Li
- Department of Infectious Diseases, Huzhou Central Hospital, Huzhou, PR China
| | - Qingqiu Zeng
- Department of Infectious Diseases, Huzhou Central Hospital, Huzhou, PR China
| | - Rui Yao
- Department of Intensive Care Units, Huzhou Central Hospital, Huzhou, PR China
| | - Lingyan Zhang
- Department of Infectious Diseases, Huzhou Central Hospital, Huzhou, PR China
| | - Ying Kong
- Department of Intensive Care Units, Changxing Traditional Chinese Medicine (TCM) Hospital, Changxing, PR China
| | - Bin Shen
- Department of Infectious Diseases, Huzhou Central Hospital, Huzhou, PR China.
| |
Collapse
|
46
|
Xu S, Zhang Y, Zheng Z, Sun J, Wei Y, Ding G. Mesenchymal stem cells and their extracellular vesicles in bone and joint diseases: targeting the NLRP3 inflammasome. Hum Cell 2024; 37:1276-1289. [PMID: 38985391 DOI: 10.1007/s13577-024-01101-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: 04/16/2024] [Accepted: 07/04/2024] [Indexed: 07/11/2024]
Abstract
The nucleotide-binding oligomerization domain-like-receptor family pyrin domain-containing 3 (NLRP3) inflammasome is a cytosolic multi-subunit protein complex, and recent studies have demonstrated the vital role of the NLRP3 inflammasome in the pathological and physiological conditions, which cleaves gasdermin D to induce inflammatory cell death called pyroptosis and mediates the release of interleukin-1 beta and interleukin-18 in response to microbial infection or cellular injury. Over-activation of the NLRP3 inflammasome is associated with the pathogenesis of many disorders affecting bone and joints, including gouty arthritis, osteoarthritis, rheumatoid arthritis, osteoporosis, and periodontitis. Moreover, mesenchymal stem cells (MSCs) have been discovered to facilitate the inhibition of NLRP3 and maybe ideal for treating bone and joint diseases. In this review, we implicate the structure and activation of the NLRP3 inflammasome along with the detail on the involvement of NLRP3 inflammasome in bone and joint diseases pathology. In addition, we focused on MSCs and MSC-extracellular vesicles targeting NLRP3 inflammasomes in bone and joint diseases. Finally, the existing problems and future direction are also discussed.
Collapse
Affiliation(s)
- Shuangshuang Xu
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, Shandong Province, China
| | - Ying Zhang
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, Shandong Province, China
| | - Zejun Zheng
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, Shandong Province, China
| | - Jinmeng Sun
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, Shandong Province, China
| | - Yanan Wei
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, Shandong Province, China
| | - Gang Ding
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, Shandong Province, China.
| |
Collapse
|
47
|
van Eyll J, Prior R, Celanire S, Van Den Bosch L, Rombouts F. Therapeutic indications for HDAC6 inhibitors in the peripheral and central nervous disorders. Expert Opin Ther Targets 2024; 28:719-737. [PMID: 39305025 DOI: 10.1080/14728222.2024.2404571] [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: 04/16/2024] [Accepted: 09/06/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION Inhibition of the enzymatic function of HDAC6 is currently being explored in clinical trials ranging from peripheral neuropathies to cancers. Advances in selective HDAC6 inhibitor discovery allowed studying highly efficacious brain penetrant and peripheral restrictive compounds for treating PNS and CNS indications. AREAS COVERED This review explores the multifactorial role of HDAC6 in cells, the common pathological hallmarks of PNS and CNS disorders, and how HDAC6 modulates these mechanisms. Pharmacological inhibition of HDAC6 and genetic knockout/knockdown studies as a therapeutic strategy in PNS and CNS indications were analyzed. Furthermore, we describe the recent developments in HDAC6 PET tracers and their utility in CNS indications. Finally, we explore the advancements and challenges with HDAC6 inhibitor compounds, such as hydroxamic acid, fluoromethyl oxadiazoles, HDAC6 degraders, and thiol-based inhibitors. EXPERT OPINION Based on extensive preclinical evidence, pharmacological inhibition of HDAC6 is a promising approach for treating both PNS and CNS disorders, given its involvement in neurodegeneration and aging-related cellular processes. Despite the progress in the development of selective HDAC6 inhibitors, safety concerns remain regarding their chronic administration in PNS and CNS indications, and the development of novel compound classes and modalities inhibiting HDAC6 function offer a way to mitigate some of these safety concerns.
Collapse
Affiliation(s)
| | | | - Sylvain Celanire
- Augustine Therapeutics, Research and Development, Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, Leuven, Belgium
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | | |
Collapse
|
48
|
Erdem M, Erdem Ş, Alver A, Kıran TR, Karahan SC. β 2-adrenoceptor agonist formoterol attenuates NLRP3 inflammasome activation and GSDMD-mediated pyroptosis in microglia through enhancing IκBα/NF-κB inhibition, SQSTM1/p62-dependent selective autophagy and ESCRT-III-mediated plasma membrane repair. Mol Cell Neurosci 2024; 130:103956. [PMID: 39097250 DOI: 10.1016/j.mcn.2024.103956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/05/2024] Open
Abstract
Microglia are immune cells that play important roles in the formation of the innate immune response within the central nervous system (CNS). The NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is a multiple protein complex that is crucial for innate immunity, and excessive activation of the inflammasome for various reasons contributes to the pathogenesis of neurodegenerative diseases (NDs). β2-adrenoceptor agonists have become the focus of attention in studies on NDs due to the high synthesis of β2-adrenoceptors in the central nervous system (CNS). Promising results have been obtained from these studies targeting anti-inflammatory and neuroprotective effects. Formoterol is an effective, safe for long-term use, and FDA-approved β2-adrenoceptor agonist with demonstrated anti-inflammatory features in the CNS. In this study, we researched the effects of formoterol on LPS/ATP-stimulated NLRP3 inflammasome activation, pyroptosis, NF-κB, autophagy, and ESCRT-III-mediated plasma membrane repair pathways in the N9 microglia cells. The results showed that formoterol, through the IκBα/NF-κB axis, significantly inhibited NLRP3 inflammasome activation, reduced the level of active caspase-1, secretion of IL-1β and IL-18 proinflammatory cytokine levels, and the levels of pyroptosis. Additionally, we showed that formoterol activates autophagy, autophagosome formation, and ESCRT-III-mediated plasma membrane repair, which are significant pathways in the inhibition of NLRP3 inflammasome activation and pyroptosis. Our study suggests that formoterol efficaciously prevents the NLRP3 inflammasome activation and pyroptosis in microglial cells regulation through IκBα/NF-κB, autophagy, autophagosome formation, and ESCRT-III-mediated plasma membrane repair.
Collapse
Affiliation(s)
- Mehmet Erdem
- Department of Medical Biochemistry, Faculty of Medicine, Malatya Turgut Özal University, Malatya 44900, Turkey; Department of Medical Biochemistry, Graduate School of Medical Science, Karadeniz Technical University, Trabzon 61080, Turkey.
| | - Şeniz Erdem
- Department of Medical Biochemistry, Graduate School of Medical Science, Karadeniz Technical University, Trabzon 61080, Turkey; Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, Trabzon 61080, Turkey
| | - Ahmet Alver
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, Trabzon 61080, Turkey
| | - Tuğba Raika Kıran
- Department of Medical Biochemistry, Faculty of Medicine, Malatya Turgut Özal University, Malatya 44900, Turkey
| | - Süleyman Caner Karahan
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, Trabzon 61080, Turkey
| |
Collapse
|
49
|
Xu X, Jin W, Chang R, Ding X. Research progress of SREBP and its role in the pathogenesis of autoimmune rheumatic diseases. Front Immunol 2024; 15:1398921. [PMID: 39224584 PMCID: PMC11366632 DOI: 10.3389/fimmu.2024.1398921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Autoimmune rheumatic diseases comprise a group of immune-related disorders characterized by non-organ-specific inflammation. These diseases include systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), ankylosing spondylitis (AS), gout, among others. Typically involving the hematologic system, these diseases may also affect multiple organs and systems. The pathogenesis of autoimmune rheumatic immune diseases is complex, with diverse etiologies, all associated with immune dysfunction. The current treatment options for this type of disease are relatively limited and come with certain side effects. Therefore, the urgent challenge remains to identify novel therapeutic targets for these diseases. Sterol regulatory element-binding proteins (SREBPs) are basic helix-loop-helix-leucine zipper transcription factors that regulate the expression of genes involved in lipid and cholesterol biosynthesis. The expression and transcriptional activity of SREBPs can be modulated by extracellular stimuli such as polyunsaturated fatty acids, amino acids, glucose, and energy pathways including AKT-mTORC and AMP-activated protein kinase (AMPK). Studies have shown that SREBPs play roles in regulating lipid metabolism, cytokine production, inflammation, and the proliferation of germinal center B (GCB) cells. These functions are significant in the pathogenesis of rheumatic and immune diseases (Graphical abstract). Therefore, this paper reviews the potential mechanisms of SREBPs in the development of SLE, RA, and gout, based on an exploration of their functions.
Collapse
Affiliation(s)
| | | | | | - Xinghong Ding
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
50
|
Wang RY, Wang MG, Tang HZ, Du H, Luo Y, Li Q, Zhang XH, Fu J, Lv CZ. The Protective Effects of Ruscogenin Against Lipopolysaccharide-Induced Myocardial Injury in Septic Mice. J Cardiovasc Pharmacol 2024; 84:175-187. [PMID: 38547523 DOI: 10.1097/fjc.0000000000001563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/09/2024] [Indexed: 08/09/2024]
Abstract
ABSTRACT Sepsis-induced myocardial dysfunction commonly occurs in individuals with sepsis and is a severe complication with high morbidity and mortality rates. This study aimed to investigate the effects and potential mechanisms of the natural steroidal sapogenin ruscogenin (RUS) against lipopolysaccharide (LPS)-induced myocardial injury in septic mice. We found that RUS effectively alleviated myocardial pathological damage, normalized cardiac function, and increased survival in septic mice. RNA sequencing demonstrated that RUS administration significantly inhibited the activation of the NOD-like receptor signaling pathway in the myocardial tissues of septic mice. Subsequent experiments further confirmed that RUS suppressed myocardial inflammation and pyroptosis during sepsis. In addition, cultured HL-1 cardiomyocytes were challenged with LPS, and we observed that RUS could protect these cells against LPS-induced cytotoxicity by suppressing inflammation and pyroptosis. Notably, both the in vivo and in vitro findings indicated that RUS inhibited NOD-like receptor protein-3 (NLRP3) upregulation in cardiomyocytes stimulated with LPS. As expected, knockdown of NLRP3 blocked the LPS-induced activation of inflammation and pyroptosis in HL-1 cells. Furthermore, the cardioprotective effects of RUS on HL-1 cells under LPS stimulation were abolished by the novel NLRP3 agonist BMS-986299. Taken together, our results suggest that RUS can alleviate myocardial injury during sepsis, at least in part by suppressing NLRP3-mediated inflammation and pyroptosis, highlighting the potential of this molecule as a promising candidate for sepsis-induced myocardial dysfunction therapy.
Collapse
Affiliation(s)
- Rui-Yu Wang
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ming-Gui Wang
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Han-Zhang Tang
- Department of Cardiology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui Du
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yue Luo
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Quan Li
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiao-Hong Zhang
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jing Fu
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Chuan-Zhu Lv
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China; and
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, China
| |
Collapse
|